1
|
Huang Y, Jia L, Zhang S, Yan L, Li L. Bimetallic doped carbon dot nanozymes for enhanced sonodynamic and nanocatalytic therapy. J Mater Chem B 2025; 13:588-598. [PMID: 39575676 DOI: 10.1039/d4tb01916d] [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: 12/06/2024]
Abstract
Conventional inorganic semiconductors are not suitable for acting as nanozymes or sonosensitizers for in vivo therapeutic nanomedicine owing to the lack of excellent biocompatibility. Biocompatible carbon dots (CDs) exhibit a variety of biological activities due to their adjustable size and surface chemical modification; however, the simultaneous sonodynamic activity and multiple enzyme-mimicking catalytic activity of a single CD have not been reported. Herein, we report the development of bimetallic doped CDs as a high-efficiency nanozyme and sonosensitizer for enhanced sonodynamic therapy (SDT) and nanocatalytic therapy (NCT). By selecting metal-organic complexes like EDTA-FeNa as the carbon source, we ensure that the coordination environments of metal atoms are preserved throughout the low-temperature calcination process. Compared with the single metal doped CDs including Fe-CDs or Ni-CDs, the obtained Fe and Ni co-doped CDs (Fe-Ni-CDs) not only exhibit enhanced sonodynamic activity owing to the decreased bandgap, but also possess augmented dual enzyme-mimicking catalytic activities due to the synergistic effect of bimetallic ions. The Fe-Ni-CD-mediated cascade amplification of ROS generation could lead to the production of 1O2 and O2˙- through SDT, the generation of ˙OH through POD-mimicking catalytic activity, and the provision of more O2 for SDT through CAT-mimicking catalytic activity. Through the integrated multifunctionality of Fe-Ni-CDs, we successfully enhanced the effectiveness of antitumor treatment with a single drug injection and a single US irradiation for enhanced SDT and NCT. This work provides a distinct paradigm of endowing CDs with sonodynamic and multiple enzyme-mimicking catalytic activities for enhanced SDT and NCT through bimetallic ion doping.
Collapse
Affiliation(s)
- Yandong Huang
- Department of Ultrasound, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Lanting Jia
- Department of Ultrasound, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Shiqi Zhang
- Department of Ultrasound, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Lang Yan
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China.
| | - Lei Li
- Department of Emergency, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
2
|
Li X, Tang WJ, Wang W, Yue S, Yao H, Zhu JJ. Acid-responsive liposomal nanodrug with promoted tumor penetration for photoacoustic imaging-guided sonodynamic therapy. Chem Commun (Camb) 2024; 60:15023-15026. [PMID: 39605026 DOI: 10.1039/d4cc05043f] [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: 11/29/2024]
Abstract
Herein, an acid-responsive liposomal nanodrug was developed for photoacoustic (PA) imaging-guided oxygen (O2)-independent sonodynamic therapy (SDT). This liposomal nanodrug offers several advantages: (i) it facilitates O2-independent alkyl radical generation upon ultrasound irradiation, (ii) it exhibits acid-responsive charge reversion that enhances tumor penetration, and (iii) it enables activated PA imaging for therapeutic feedback.
Collapse
Affiliation(s)
- Xiangli Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Wen-Jing Tang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Wei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Shuzhen Yue
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Huiqin Yao
- Department of Medical Chemistry, College of Basic Medicine, Ningxia Medical University, Yinchuan 750004, China.
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
3
|
Liu X, Zhang W, Wei S, Liang X, Luo B. Targeting cuproptosis with nano material: new way to enhancing the efficacy of immunotherapy in colorectal cancer. Front Pharmacol 2024; 15:1451067. [PMID: 39691393 PMCID: PMC11649426 DOI: 10.3389/fphar.2024.1451067] [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: 06/18/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Colorectal cancer has emerged as one of the predominant malignant tumors globally. Immunotherapy, as a novel therapeutic methodology, has opened up new possibilities for colorectal cancer patients. However, its actual clinical efficacy requires further enhancement. Copper, as an exceptionally crucial trace element, can influence various signaling pathways, gene expression, and biological metabolic processes in cells, thus playing a critical role in the pathogenesis of colorectal cancer. Recent studies have revealed that cuproptosis, a novel mode of cell death, holds promise to become a potential target to overcome resistance to colorectal cancer immunotherapy. This shows substantial potential in the combination treatment of colorectal cancer. Conveying copper into tumor cells via a nano-drug delivery system to induce cuproptosis of colorectal cancer cells could offer a potential strategy for eliminating drug-resistant colorectal cancer cells and vastly improving the efficacy of immunotherapy while ultimately destroy colorectal tumors. Moreover, combining the cuproptosis induction strategy with other anti-tumor approaches such as photothermal therapy, photodynamic therapy, and chemodynamic therapy could further enhance its therapeutic effect. This review aims to illuminate the practical significance of cuproptosis and cuproptosis-inducing nano-drugs in colorectal cancer immunotherapy, and scrutinize the current challenges and limitations of this methodology, thereby providing innovative thoughts and references for the advancement of cuproptosis-based colorectal cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Xiangdong Liu
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Wanqiu Zhang
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Shaozhong Wei
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinjun Liang
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Abdominal Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Luo
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| |
Collapse
|
4
|
Xu Y, Zhu K, Wu J, Zheng S, Zhong R, Zhou W, Cao Y, Liu J, Wang H. HBOC alleviated tumour hypoxia during radiotherapy more intensely in large solid tumours than regular ones. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:1-14. [PMID: 37994792 DOI: 10.1080/21691401.2023.2276768] [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: 05/16/2023] [Accepted: 09/07/2023] [Indexed: 11/24/2023]
Abstract
Radiotherapy (RT) is a highly valuable method in cancer therapy, but its therapeutic efficacy is limited by its side effects and tumour radiation resistance. The resistance is mainly induced by hypoxia in the tumour microenvironment (TME). As a nano-oxygen carrier, Haemoglobin-based oxygen carriers (HBOCs) administration is a promising strategy to alleviate tumour hypoxia which may remodel TME to ameliorate radiation resistance and enable RT more effective. In this study, we administered fractionated RT combined with HBOC to treat Miapaca-2 cell and Hela cell xenografts on nude mice. The study found that HBOC relieved hypoxic environment and down-regulate expression of hypoxia-inducible factor-1α (Hif-1α) both in regular (100 mm3) and large (360/400 mm3) tumours. The proliferation and metastasis of tumour tissue also decreased after HBOC application. Nevertheless, in vivo RT combined with HBOC performed more effectively to suppress tumour growth in large tumours than in regular tumours. This is due to more severe hypoxic regions exist in the large solid tumours compared to the regular counterparts, and HBOC administration may be more effective in alleviating hypoxia in large tumours. Thus, HBOC sensitization therapy is more suitable for large solid tumours.
Collapse
Affiliation(s)
- Yingcan Xu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Kehui Zhu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Jiakang Wu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Shifan Zheng
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Rui Zhong
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Wentao Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Ye Cao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Jiaxin Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| | - Hong Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Peking Union Medical College, Chengdu, China
| |
Collapse
|
5
|
Xu S, Zhang H, Qian Z, Yuan W. pH-Responsive injectable self-healing hydrogels loading Au nanoparticles-decorated bimetallic organic frameworks for synergistic sonodynamic-chemodynamic-starvation-chemo therapy of cancer. J Colloid Interface Sci 2024; 675:746-760. [PMID: 38996704 DOI: 10.1016/j.jcis.2024.07.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
A novel and efficient cancer therapy was developed using a smart hydrogel containing multifunctional bimetallic organic frameworks and anticancer drugs. The injectable self-healing hydrogel with pH-responsiveness was constructed through borate ester and imine bonds among dopamine-grafted sodium alginate (SADA), hydroxypropyl chitosan (HPCS) and 2-formylphenylboronic acid (2-FPBA). The Au nanoparticles-decorated Ti/Fe bimetallic organic framework tetragonal nanosheets (Au/TF-MOF TNS) were synthesized and incorporated into the hydrogel with the anticancer drugs doxorubicin (DOX). Upon intratumoral injection of nanocomposite hydrogel, the acidic tumor microenvironment triggered the cleavage of borate ester and imine bonds, causing the hydrogel to break down and accelerating the release of both Au/TF-MOF TNS and DOX. These Au/TF-MOF TNS functioned as nanozymes, producing hydroxyl radicals (·OH) for chemodynamic therapy (CDT), generating oxygen (O2) to support sonodynamic therapy (SDT), and depleting glucose for starvation therapy (ST). Additionally, the Au/TF-MOF TNS served as sonosensitizers, capable of converting O2 into singlet oxygen (1O2) upon ultrasound irradiation to achieve SDT. Therefore, this nanocomposite hydrogel system enabled synergistic sonodynamic-chemodynamic-starvation-chemo therapy (SDT-CDT-ST-CT) of cancer, presenting a promising platform for advanced cancer therapy strategies.
Collapse
Affiliation(s)
- Sicheng Xu
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Hanyan Zhang
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Zhiyi Qian
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Weizhong Yuan
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China.
| |
Collapse
|
6
|
Qu R, Jiang X, Zhen X. Light/X-ray/ultrasound activated delayed photon emission of organic molecular probes for optical imaging: mechanisms, design strategies, and biomedical applications. Chem Soc Rev 2024; 53:10970-11003. [PMID: 39380344 DOI: 10.1039/d4cs00599f] [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: 10/10/2024]
Abstract
Conventional optical imaging, particularly fluorescence imaging, often encounters significant background noise due to tissue autofluorescence under real-time light excitation. To address this issue, a novel optical imaging strategy that captures optical signals after light excitation has been developed. This approach relies on molecular probes designed to store photoenergy and release it gradually as photons, resulting in delayed photon emission that minimizes background noise during signal acquisition. These molecular probes undergo various photophysical processes to facilitate delayed photon emission, including (1) charge separation and recombination, (2) generation, stabilization, and conversion of the triplet excitons, and (3) generation and decomposition of chemical traps. Another challenge in optical imaging is the limited tissue penetration depth of light, which severely restricts the efficiency of energy delivery, leading to a reduced penetration depth for delayed photon emission. In contrast, X-ray and ultrasound serve as deep-tissue energy sources that facilitate the conversion of high-energy photons or mechanical waves into the potential energy of excitons or the chemical energy of intermediates. This review highlights recent advancements in organic molecular probes designed for delayed photon emission using various energy sources. We discuss distinct mechanisms, and molecular design strategies, and offer insights into the future development of organic molecular probes for enhanced delayed photon emission.
Collapse
Affiliation(s)
- Rui Qu
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.
| | - Xu Zhen
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
7
|
Li J, Zhao Z, Tian Y, Liu W, Zhang P, Chen L. Tumor Microenvironment-Responsive Zn(II)-Porphyrin Nanotheranostics for Targeted Sonodynamic Therapy. ACS Biomater Sci Eng 2024; 10:6984-6994. [PMID: 39388140 DOI: 10.1021/acsbiomaterials.4c00344] [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: 10/15/2024]
Abstract
As a novel noninvasive tumor therapy, sonodynamic therapy (SDT) attracts booming concerns. However, the limited water solubility, inadequate biocompatibility, and low targeting ability of conventional sonosensitizers significantly hinder their potential for clinical application. Herein, novel zinc(II)-porphyrin nanotheranostics (HA@Zn-TCPP) were fabricated in which the zinc(II)-porphyrin (TCPP) metal-organic framework was first constructed by a simple thermal reaction, followed by the addition of hyaluronic acid (HA) for modification. The specific targeting ability of HA facilitated the internalization of HA@Zn-TCPP within tumor cells, resulting in its preferential accumulation in tumor tissues that exhibit CD44 receptor overexpression. The acidic tumor microenvironment induced the rapid decomposition of HA@Zn-TCPP, releasing free TCPP for activating SDT. This controllable generation of reactive oxygen species (ROS) could effectively decrease damage to normal tissues. The HA@Zn-TCPP exhibited remarkable antitumor effects in experiments, achieving a tumor inhibition rate of up to 82.1% when under ultrasound. This finding provides an imperative strategy to develop novel sonosensitizers for enhanced SDT.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Zhitong Zhao
- Department of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Yongchang Tian
- Department of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Wenchang Liu
- Department of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Li Chen
- Department of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| |
Collapse
|
8
|
Jeong YG, Park JH, Khang D. Sonodynamic and Acoustically Responsive Nanodrug Delivery System: Cancer Application. Int J Nanomedicine 2024; 19:11767-11788. [PMID: 39553460 PMCID: PMC11566213 DOI: 10.2147/ijn.s496028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/31/2024] [Indexed: 11/19/2024] Open
Abstract
The advent of acoustically responsive nanodrugs that are specifically optimized for sonodynamic therapy (SDT) is a novel approach for clinical applications. Examining the therapeutic applications of sono-responsive drug delivery systems, understanding their dynamic response to acoustic stimuli, and their crucial role in enhancing targeted drug delivery are intriguing issues for current cancer treatment. Specifically, the suggested review covers SDT, a modality that enhances the cytotoxic activity of specific compounds (sonosensitizers) using ultrasound (US). Notably, SDT offers significant advantages in cancer treatment by utilizing US energy to precisely target and activate sonosensitizers toward deep-seated malignant sites. The potential mechanisms underlying SDT involve the generation of radicals from sonosensitizers, physical disruption of cell membranes, and enhanced drug transport into cells via US-assisted sonoporation. In particular, SDT is emerging as a promising modality for noninvasive, site-directed elimination of solid tumors. Given the complexity and diversity of tumors, many studies have explored the integration of SDT with other treatments to enhance the overall efficacy. This trend has paved the way for SDT-based multimodal synergistic cancer therapies, including sonophototherapy, sonoimmunotherapy, and sonochemotherapy. Representative studies of these multimodal approaches are comprehensively presented, with a detailed discussion of their underlying mechanisms. Additionally, the application of audible sound waves in biological systems is explored, highlighting their potential to influence cellular processes and enhance therapeutic outcomes. Audible sound waves can modulate enzyme activities and affect cell behavior, providing novel avenues for the use of sound-based techniques in medical applications. This review highlights the current challenges and prospects in the development of SDT-based nanomedicines in this rapidly evolving research field. The anticipated growth of this SDT-based therapeutic approach promises to significantly improve the precision of cancer treatment.
Collapse
Affiliation(s)
- Yong-Gyu Jeong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Joo-Hwan Park
- Division of Medical Oncology, Department of Internal Medicine, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon, 21565, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, South Korea
| |
Collapse
|
9
|
Hu J, Yan L, Cao Z, Geng B, Cao X, Liu B, Guo J, Zhu J. Tumor Microenvironment Activated Cu Crosslinked Near-Infrared Sonosensitizers for Visualized Cuproptosis-Enhanced Sonodynamic Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407196. [PMID: 39331855 DOI: 10.1002/advs.202407196] [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: 06/27/2024] [Revised: 09/19/2024] [Indexed: 09/29/2024]
Abstract
Reactive oxygen species (ROS)-mediated sonodynamic therapy (SDT) holds increasing potential in treating deep-seated tumor owing to the high tissue-penetration depth. However, the inevitable accumulation of sonosensitizers in normal tissues not only make it difficult to realize the in situ SDT, but also induces sonodynamic effects in normal tissues. Herein, this work reports the passivation and selective activation strategies for the sonodynamic and near-infrared (NIR) imaging performances of an intelligent antitumor theranostic platform termed Cu-IR783 nanoparticles (NPs). Owing to the ruptured coordination bond between IR783 with Cu ions by responding to tumor microenvironment (TME), the selective activation of IR783 only occurred in tumor tissues to achieve the visualized in-situ SDT. The tumor-specific released Cu ions not only realized the cascade amplification of ROS generation through Cu+-mediated Fenton-like reaction, but also triggered cuproptosis through Cu+-induced DLAT oligomerization and mitochondrial dysfunction. More importantly, the immunosuppressive TME can be reversed by the greatly enhanced ROS levels and high-efficiency cuproptosis, ultimately inducing immunogenic cell death that promotes robust systemic immune responses for the eradication of primary tumors and suppression of distant tumors. This work provides a distinct paradigm of the integration of SDT, CDT, and cuproptosis in a controlled manner to achieve visualized in-situ antitumor therapy.
Collapse
Affiliation(s)
- Jinyan Hu
- 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
| | - Zhi Cao
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Bijiang Geng
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xiqian Cao
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Bing Liu
- Department of Urology, The Third Affiliated Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jiaming Guo
- Department of Radiation Medicine, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jiangbo Zhu
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
10
|
Du J, Chen X, Xu X, Que Z, Zhai M, Xiang Q, Zhang Z, Zhang Z, Shao Y, Yang X, Miao F, Zhang J, Xie J, Ju S. Enhancing the tissue penetration to improve sonodynamic immunotherapy for pancreatic ductal adenocarcinoma using membrane-camouflaged nanoplatform. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06952-y. [PMID: 39422735 DOI: 10.1007/s00259-024-06952-y] [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: 06/19/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Sonodynamic therapy (SDT) is a promising strategy as an "in situ vaccine" to enhance activation of antitumor immune responses in solid tumors. However, the dense extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) lead to hypoxia and limited penetration of most drugs, aggravating the immunosuppressive tumor microenvironment and limiting the efficacy of synergistic sonodynamic immunotherapy. Therefore, it is essential to regulate ECM in order to alleviate tumor hypoxia and enhance the efficacy of sonodynamic immunotherapy for PDAC. METHODS The CPIM nanoplatform, consisting of a macrophage membrane-coated oxygen and drug delivery system (CM@PFOB-ICG-α-Mangostin), was synthesized using ultrasound and extrusion methods. The in vivo homologous targeting and hypoxia alleviation capabilities of CPIM were evaluated through near-infrared (NIR) imaging and photoacoustic (PA) imaging. The tumor growth inhibition potential and ability to reprogram the tumor microenvironment by the CPIM nanoplatform were also investigated. RESULTS Co-delivery of α-Mangostin inhibits CAFs and enhances stromal depletion, thereby facilitating better infiltration of macromolecules. Additionally, the nanoemulsion containing perfluorocarbon (PFC) can target tumor cells and accumulate within them through homologous targeting. The US irradiation results in the rapid release of oxygen, serving as a potential source of sonodynamic therapy for hypoxic tumors. Moreover, CPIM reshapes the immunosuppressive microenvironment increasing the population of cytotoxic T lymphocytes (CTLs), and enhancing their anti-tumor immune response through the use of anti-PDL1 antibodies to block immune checkpoints. CONCLUSION The present study offers a potential strategy for the co-delivery of oxygen and α-Mangostin, aiming to enhance the penetration of tumors to improve SDT. This approach effectively addresses the existing limitations of immune checkpoint blockade (ICB) treatment in solid tumors, while simultaneously boosting the immune response through synergistic sonodynamic immunotherapy.
Collapse
Affiliation(s)
- Jiawei Du
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Xin Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, P.R. China
| | - Xiaoxuan Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Ziting Que
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Mengyan Zhai
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Qinyanqiu Xiang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Zhiwei Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Zhiqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Yong Shao
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Xue Yang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
| | - Fengqin Miao
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Jianqiong Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China
- Department of Microbiology and Immunology, Medical School, Southeast University, 87th DingJiaQiao Road, Nanjing, 210009, P.R. China
| | - Jinbing Xie
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China.
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Southeast University, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, P.R. China.
| |
Collapse
|
11
|
Deng C, Zhang J, Hu F, Han S, Zheng M, An F, Wang F. A GSH-Responsive Prodrug with Simultaneous Triple-Activation Capacity for Photodynamic/Sonodynamic Combination Therapy with Inhibited Skin Phototoxicity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400667. [PMID: 38837658 DOI: 10.1002/smll.202400667] [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: 01/28/2024] [Revised: 04/17/2024] [Indexed: 06/07/2024]
Abstract
Herein, a dual-sensitizer prodrug, named pro-THPC, has been designed to function as both a photosensitizer and a sonosensitizer prodrug for precise antitumor combination therapy with minimized skin phototoxicity. Pro-THPC could be activated by glutathione (GSH) to release the dual-sensitizer, THPC, which simultaneously switches on fluorescence emission and combined capabilities of photodynamic therapy (PDT) and sonodynamic therapy (SDT). Pro-THPC is further formulated into nanoparticles (NPs) for water dispersity to enable in vivo applications. In vivo fluorescence imaging shows that the pro-THPC NPs group exhibits a significantly higher tumor-to-normal tissue ratio (T/N) (T/N = 5.2 ± 0.55) compared to the "always on" THPC NPs group (T/N = 2.9 ± 0.47) and the pro-THPC NPs group co-administrated with GSH synthesis inhibitor (buthionine sulfoximine, BSO) (T/N = 3.2 ± 0.63). In addition, the generation of the designed dual-sensitizer's reactive oxygen species (ROS) is effectively confined within the tumor tissues due to the relatively strong correlation between ROS generation and fluorescence emission. In vivo studies further demonstrate the remarkable efficacy of the designed pro-THPC NPs to eradicate tumors through the combination of PDT and SDT while significantly reducing skin phototoxicity.
Collapse
Affiliation(s)
- Caiting Deng
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Jingjing Zhang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Fanchun Hu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Shupeng Han
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Meichen Zheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Feifei An
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- School of Public Health, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi, 710061, China
| | - Fu Wang
- Institute of Medical Engineering, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No. 76 Yanta West Road, Xi'an, Shaanxi, 710061, China
- Xianyang Key Laboratory of Molecular Imaging and Drug Synthesis, School of Pharmacy, Shaanxi University of International Trade & Commerce, Xianyang, Shaanxi, 712046, China
| |
Collapse
|
12
|
Yang R, Tang S, Xie X, Jin C, Tong Y, Huang W, Zan X. Enhanced Ocular Delivery of Beva via Ultra-Small Polymeric Micelles for Noninvasive Anti-VEGF Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314126. [PMID: 38819852 DOI: 10.1002/adma.202314126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 05/15/2024] [Indexed: 06/01/2024]
Abstract
Pathological ocular neovascularization resulting from retinal ischemia constitutes a major cause of vision loss. Current anti-VEGF therapies rely on burdensome intravitreal injections of Bevacizumab (Beva). Herein ultrasmall polymeric micelles encapsulating Beva (P@Beva) are developed for noninvasive topical delivery to posterior eye tissues. Beva is efficiently loaded into 11 nm micelles fabricated via self-assembly of hyperbranched amphiphilic copolymers. The neutral, brush-like micelles demonstrate excellent drug encapsulation and colloidal stability. In vitro, P@Beva enhances intracellular delivery of Beva in ocular cells versus free drug. Ex vivo corneal and conjunctival-sclera-choroidal tissues transport after eye drops are improved 23-fold and 7.9-fold, respectively. Anti-angiogenic bioactivity is retained with P@Beva eliciting greater inhibition of endothelial tube formation and choroid sprouting over Beva alone. Remarkably, in an oxygen-induced retinopathy (OIR) model, topical P@Beva matching efficacy of intravitreal Beva injection, is the clinical standard. Comprehensive biocompatibility verifies safety. Overall, this pioneering protein delivery platform holds promise to shift paradigms from invasive intravitreal injections toward simplified, noninvasive administration of biotherapeutics targeting posterior eye diseases.
Collapse
Affiliation(s)
- Ruhui Yang
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Sicheng Tang
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Xiaoling Xie
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Chaofan Jin
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| | - Yuhua Tong
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang Province, 324000, China
| | - Wenjuan Huang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, China
| | - Xingjie Zan
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou, Zhejiang Province, 325035, China
- Wenzhou Key Laboratory of Perioperative Medicine, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province, 325001, China
| |
Collapse
|
13
|
Qian K, Gao S, Jiang Z, Ding Q, Cheng Z. Recent advances in mitochondria-targeting theranostic agents. EXPLORATION (BEIJING, CHINA) 2024; 4:20230063. [PMID: 39175881 PMCID: PMC11335472 DOI: 10.1002/exp.20230063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/07/2024] [Indexed: 08/24/2024]
Abstract
For its vital role in maintaining cellular activity and survival, mitochondrion is highly involved in various diseases, and several strategies to target mitochondria have been developed for specific imaging and treatment. Among these approaches, theranostic may realize both diagnosis and therapy with one integrated material, benefiting the simplification of treatment process and candidate drug evaluation. A variety of mitochondria-targeting theranostic agents have been designed based on the differential structure and composition of mitochondria, which enable more precise localization within cellular mitochondria at disease sites, facilitating the unveiling of pathological information while concurrently performing therapeutic interventions. Here, progress of mitochondria-targeting theranostic materials reported in recent years along with background information on mitochondria-targeting and therapy have been briefly summarized, determining to deliver updated status and design ideas in this field to readers.
Collapse
Affiliation(s)
- Kun Qian
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Shu Gao
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingChina
| | - Zhaoning Jiang
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingChina
- Shandong Laboratory of Yantai Drug DiscoveryBohai Rim Advanced Research Institute for Drug DiscoveryYantaiShandongChina
| | - Qihang Ding
- Department of ChemistryKorea UniversitySeoulRepublic of Korea
| | - Zhen Cheng
- State Key Laboratory of Drug ResearchMolecular Imaging CenterShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingChina
- Shandong Laboratory of Yantai Drug DiscoveryBohai Rim Advanced Research Institute for Drug DiscoveryYantaiShandongChina
| |
Collapse
|
14
|
Wang F, Fan Y, Liu Y, Lou X, Sutrisno L, Peng S, Li J. Oxygen-carrying semiconducting polymer nanoprodrugs induce sono-pyroptosis for deep-tissue tumor treatment. EXPLORATION (BEIJING, CHINA) 2024; 4:20230100. [PMID: 39175882 PMCID: PMC11335461 DOI: 10.1002/exp.20230100] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/20/2023] [Indexed: 08/24/2024]
Abstract
Sonodynamic therapy (SDT) has been explored for cancer therapy, especially for deep tumors due to its low tissue penetration restriction. The therapeutic efficacy of SDT is limited due to the complicated tumor microenvironment. This study reports the construction of oxygen-carrying semiconducting polymer nanoprodrugs (OSPNpro) for deep tumor treatment via combining amplified SDT with pyroptosis. An oxygen carrier perfluorohexane, sonodynamic semiconducting polymer as the sonosensitizer, and reactive oxygen species (ROS)-responsive prodrug are co-loaded into a nanoparticle system, leading to the formation of these polymer nanoprodrugs. Such OSPNpro show an effective accumulation in tumor tissues after systemic administration, in which they deliver oxygen to relieve tumor hypoxia microenvironment and thus mediate amplified SDT via producing ROS under ultrasound (US) irradiation, even when the tumors are covered with a 2-cm chicken breast tissue. In addition, the ROS-responsive prodrugs are activated by the generated ROS to trigger pyroptosis of tumor cells. Such a sono-pyroptosis induces a strong antitumor immunity with obviously higher level infiltrations of effector immune cells into tumors. Therefore, OSPNpro-based combinational therapy can greatly inhibit the growth of 2-cm chicken breast tissue-covered deep tumors and suppress tumor metastasis. This study offers a prodrug nanoplatform for treatment of deep tumor via sono-pyroptosis strategy.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| | - Yongliang Fan
- Department of Cardiovascular SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yue Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| | - Xiangxin Lou
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| | - Linawati Sutrisno
- World Premier International (WPI) Research Center for Materials Nanoarchitectonics (MANA)National Institute for Materials Science (NIMS)TsukubaJapan
| | - Shaojun Peng
- Zhuhai Institute of Translational MedicineZhuhai Precision Medical CenterZhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University)ZhuhaiGuangdongChina
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| |
Collapse
|
15
|
Zhou W, Chen L, Li H, Wu M, Liang M, Liu Q, Wu W, Jiang X, Zhen X. Membrane Disruption-Enhanced Photodynamic Therapy against Gram-Negative Bacteria by a Peptide-Photosensitizer Conjugate. ACS NANO 2024. [PMID: 39033413 DOI: 10.1021/acsnano.4c05443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Photodynamic therapy (PDT) emerges as a promising strategy for combating bacteria with minimal drug resistance. However, a significant hurdle lies in the ineffectiveness of most photosensitizers against Gram-negative bacteria, primarily attributable to their characteristic impermeable outer membrane (OM) barrier. To tackle this obstacle, we herein report an amphipathic peptide-photosensitizer conjugate (PPC) with intrinsic outer membrane disruption capability to enhance PDT efficiency against Gram-negative bacteria. PPC is constructed by conjugating a hydrophilic ultrashort cationic peptide to a hydrophobic photosensitizer. PPC could efficiently bind to the OM of Gram-negative bacteria through electrostatic adsorption, and subsequently disrupt the structural integrity of the OM. Mechanistic investigations revealed that PPC triggers membrane disruption by binding to both lipopolysaccharide (LPS) and phospholipid leaflet in the OM, enabling effective penetration of PPC into the Gram-negative bacteria interior. Upon light irradiation, PPC inside bacteria generates singlet oxygen not only to effectively decrease the survival of Gram-negative bacteria P. aeruginosa and E. coli to nearly zero in vitro, but also successfully cure the full-thickness skin infection and bacterial keratitis (BK) induced by P. aeruginosa in animal models. Thus, this study provides a broad-spectrum antibacterial phototherapeutic design strategy by the synergistic action of membrane disruption and PDT to combat Gram-negative bacteria.
Collapse
Affiliation(s)
- Wenya Zhou
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Linrong Chen
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Haoze Li
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Min Wu
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Mengke Liang
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin 300192, P. R. China
| | - Wei Wu
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xu Zhen
- MOE Key Laboratory of High Performance Polymer Materials & Technology and State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
16
|
Zhang S, Zhang S, Luo S, Wang R, Di J, Wang Y, Wu D. Four-component of double-layer infinite coordination polymer nanocomposites for large tumor trimodal therapy via multi high-efficiency synergies. J Colloid Interface Sci 2024; 666:259-275. [PMID: 38598998 DOI: 10.1016/j.jcis.2024.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/30/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Multimodal /components tumors synergistic therapy is a crucial approach for enhancing comprehensive efficacy. Our research has identified lots of high efficiency synergies among four suitable components, revealing combinations with remarkably low combination index (CI) values (10-3-10-8). These combinations hold promise for large tumor powerful electrothermal-thermodynamic-multi-chemo trimodal therapy. To implement this approach, we developed four-component of double-layer infinite coordination polymer (ICP) nanocomposites, in which hypoxia-activated AQ4N and thermodynamic agent AIPH coordinated with Cu(Ⅱ) to form initial layer of positively charged ICPs-l NPs, chemotherapeutic agents gossypol-hyaluronic acid (G-HA) and CA4 coordinated with Fe(Ⅲ) to form out layer of negatively charged ICPs-2 NPs, then double-layer infinite coordination polymer nanocomposites (ICPs-1@ICPs-2 CNPs) were fabricated by electrostatic adsorption using ICPs-l NPs and ICPs-2 NPs. Cell experiments have extensively optimized the coordination combinations of the four components and the composition of the two layers. A programmable three-stage therapeutic procedure, assisted by a micro-electrothermal needle (MEN), was developed. Under this procedure the resulting nanocomposites demonstrate the powerful trimodal comprehensive therapeutic outcomes for large tumors using lower components dosage, achieving a tumor inhibition rate nearly reaching 100 % and no recurrence for 60 days. This study offers remarkable potential for tumor multimodal /components synergistic therapy in future.
Collapse
Affiliation(s)
- Shuai Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Shuo Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Siyuan Luo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Rong Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Jingran Di
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Ya Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China.
| |
Collapse
|
17
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [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: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
18
|
Tian Y, Cheng T, Sun F, Zhou Y, Yuan C, Guo Z, Wang Z. Effect of biophysical properties of tumor extracellular matrix on intratumoral fate of nanoparticles: Implications on the design of nanomedicine. Adv Colloid Interface Sci 2024; 326:103124. [PMID: 38461766 DOI: 10.1016/j.cis.2024.103124] [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: 10/23/2023] [Revised: 02/11/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
Nanomedicine has a profound impact on various research domains including drug delivery, diagnostics, theranostics, and regenerative medicine. Nevertheless, the clinical translation of nanomedicines for solid cancer remains limited due to the abundant physiological and pathological barriers in tumor that hinder the intratumoral penetration and distribution of these nanomedicines. In this article, we review the dynamic remodeling of tumor extracellular matrix during the tumor progression, discuss the impact of biophysical obstacles within tumors on the penetration and distribution of nanomedicines within the solid tumor and collect innovative approaches to surmount these obstacles for improving the penetration and accumulation of nanomedicines in tumor. Furthermore, we dissect the challenges and opportunities of the respective approaches, and propose potential avenues for future investigations. The purpose of this review is to provide a perspective guideline on how to effectively enhance the penetration of nanomedicines within tumors using promising methods.
Collapse
Affiliation(s)
- Yachao Tian
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China; Heilongjiang Guoru Biotechnology Co., Ltd., Xiangfang District, Harbin City 150030, China; School of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong 250353, China
| | - Tianfu Cheng
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Fuwei Sun
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yaxin Zhou
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Chao Yuan
- School of Food Science and Engineering, Qilu University of Technology, Jinan, Shandong 250353, China
| | - Zengwang Guo
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| | - Zhongjiang Wang
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| |
Collapse
|
19
|
Chen Y, Tan Q, Tang Y, Pang E, Peng R, Lan M, Bai D. Sorafenib and tetrakis (4-carboxyphenyl) porphyrin assembled nanoparticles for synergistic targeted chemotherapy and sonodynamic therapy of hepatocellular carcinoma. Biomater Sci 2024; 12:1864-1870. [PMID: 38411494 DOI: 10.1039/d3bm01994b] [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: 02/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by a high degree of malignancy and mortality. Sorafenib (SOR), a multi-kinase inhibitor, is clinically used in the treatment of HCC. However, SOR suffers from serious side effects and drug resistance. The development of novel therapeutic strategies for HCC therapy is urgently needed. Sonodynamic therapy (SDT) has unique advantages in treating deep tumors due to the merits of deep tissue penetration, low side effects, and the absence of drug resistance. Here, we developed multifunctional nanoparticles (NPs) termed SOR-TCPP@PEG-FA by assembling SOR, tetrakis (4-carboxyphenyl) porphyrin (TCPP), and folic acid (FA)-modified DSPE-PEG. The FA group enhances the tumor targeting capability of these NPs, while TCPP generates ROS under ultrasound (US) irradiation, which are toxic to tumor cells, and SOR with chemotherapeutic effects is released, thus realizing the synergistic SDT and chemotherapy of tumors.
Collapse
Affiliation(s)
- Yongzhi Chen
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou 225009, P.R. China.
| | - Qiuxia Tan
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China.
| | - Yuanyu Tang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China.
| | - E Pang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China.
| | - Rui Peng
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou 225009, P.R. China.
| | - Minhuan Lan
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China.
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou 225009, P.R. China.
| |
Collapse
|
20
|
Chen S, Ma T, Wang J, Liang S, Liao H, Tan W, Chen M, Zhou X, Xu Y, Wang L, Niu C. Macrophage-derived biomimetic nanoparticles enhanced SDT combined with immunotherapy inhibited tumor growth and metastasis. Biomaterials 2024; 305:122456. [PMID: 38184961 DOI: 10.1016/j.biomaterials.2023.122456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Combination therapy based on sonodynamic therapy (SDT) combined with immune checkpoint blockers anti-PD-L1 provides effective anti-tumor effects. We designed a combination therapy based on M1/PLGA@IR780/CAT NPs of SDT-enhanced immunity combined with immune checkpoint blockers against PD-L1, which was based on M1 macrophage membrane-encapsulated poly (lactic-co-glycolic acid) (PLGA) nanoparticles loaded with the acoustic sensitizer IR780 and catalase (CAT) to successfully realize it. SDT based on M1/PLGA@IR780/CAT NPs could induce tumor cell death by promoting dendritic cell (DC) maturation and modulating the tumor immune microenvironment. In particular, the systemic anti-tumor immune response and potent immune memory induced upon combination with anti-PD-L1 checkpoint blockade not only alleviated the progression of mammary cancer in 4T1 mice and effectively blocked distant metastasis, but also prevented tumor recurrence, providing a promising new therapeutic strategy for clinical tumor therapy.
Collapse
Affiliation(s)
- Sijie Chen
- Department of Ultrasound Diagnosis, 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 Diagnosis and Treatment in Hunan Province, China
| | - Tianliang Ma
- 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
| | - Jiahao 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
| | - Shuailong Liang
- 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
| | - Haiqin Liao
- Department of Ultrasound Diagnosis, 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 Diagnosis and Treatment in Hunan Province, China
| | - Wanlin Tan
- Department of Ultrasound Diagnosis, 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 Diagnosis and Treatment in Hunan Province, China
| | - Mingyu Chen
- Department of Ultrasound Diagnosis, 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 Diagnosis and Treatment in Hunan Province, China
| | - Xiaohui Zhou
- Department of Ultrasound Diagnosis, Changsha Central Hospital, Nanhua University, Changsha, Hunan 410014, China
| | - Yan Xu
- Department of Ultrasound Diagnosis, 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 Diagnosis and Treatment in Hunan Province, 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
| | - Chengcheng Niu
- Department of Ultrasound Diagnosis, 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 Diagnosis and Treatment in Hunan Province, China.
| |
Collapse
|
21
|
Han Y, Ge K, Zhao Y, Bottini M, Fan D, Wu W, Li L, Liu F, Gao S, Liang XJ, Zhang J. Modulating the Coordination Environment of Carbon-Dot-Supported Fe Single-Atom Nanozymes for Enhanced Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306656. [PMID: 37817351 DOI: 10.1002/smll.202306656] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/26/2023] [Indexed: 10/12/2023]
Abstract
Herein, carbon dot (CD)-supported Fe single-atom nanozymes with high content of pyrrolic N and ultrasmall size (ph-CDs-Fe SAzyme) are fabricated by a phenanthroline-mediated ligand-assisted strategy. Compared with phenanthroline-free nanozymes (CDs-Fe SAzyme), ph-CDs-Fe SAzyme exhibit higher peroxidase (POD)-like activity due to their structure similar to that of ferriporphyrin in natural POD. Aberration-corrected high-angle annular dark field scanning transmission electron microscopy (HAADF-STEM) and X-ray absorption fine structure spectroscopy (XAFS) analyses show that metal Fe is dispersed in ph-CDs-Fe SAzyme as single atoms. Steady-state kinetic studies show that the maximum velocity (Vmax ) and turnover number (kcat ) of H2 O2 homolytic cleavage catalyzed by ph-CDs-Fe SAzyme are 3.0 and 6.2 more than those of the reaction catalyzed by CDs-Fe SAzyme. Density functional theory (DFT) calculations show that the energy barrier of the reaction catalyzed by ph-CDs-Fe SAzyme is lower than that catalyzed by CDs-Fe SAzyme. Antitumor efficacy experiments show that ph-CDs-Fe SAzyme can efficiently inhibit the growth of tumor cells both in vitro and in vivo by synergistic chemodynamic and photothermal effects. Here a new paradigm is provided for the development of efficient antitumor therapeutic approaches based on SAzyme with POD-like activity.
Collapse
Affiliation(s)
- Yu Han
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P. R. China
| | - Kun Ge
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P. R. China
| | - Ying Zhao
- College of Science, Hebei Agricultural University, Baoding, 071001, P. R. China
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Sanford Burnham Prebys, La Jolla, CA, 92037, USA
| | - Dehui Fan
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P. R. China
| | - Wenchang Wu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P. R. China
| | - Luwei Li
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P. R. China
| | - Fengsong Liu
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, P. R. China
| | - Shutao Gao
- College of Science, Hebei Agricultural University, Baoding, 071001, P. R. China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P. R. China
| |
Collapse
|
22
|
Yan L, Shang S, Hu J, Zhang X, Chen J, Geng B, Zhao Y, Zhu J. An NIR-II-photoresponsive CoSnO 3 nanozyme for mild photothermally augmented nanocatalytic cancer therapy. J Mater Chem B 2024; 12:710-719. [PMID: 38164065 DOI: 10.1039/d3tb02018e] [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: 01/03/2024]
Abstract
The main challenges of nanozyme-based tumor catalytic therapy (NCT) lie in the unsatisfactory catalytic activity accompanied by a complex tumor microenvironment (TME). A few nanozymes have been designed to possess both enzyme-like catalytic activities and photothermal properties; however, the previously reported nanozymes mainly utilize the inefficient and unsafe NIR-I laser, which has a low maximum permissible exposure limit and a limited penetration depth. Herein, we report for the first time an all-in-one strategy to realize mild NIR-II photothermally amplified NCT by synthesizing amorphous CoSnO3 nanocubes with efficient triple enzyme-like catalytic activities and photothermal conversion properties. The presence of Co2+ and Sn4+ endows CoSnO3 nanocubes with the triple enzyme-like catalytic activities, not only achieving enhanced reactive oxygen species (ROS) generation through the Co2+-mediated peroxidase-like catalytic reaction to generate ˙OH and Sn4+-mediated depletion of overexpressed GSH, but also realizing the catalytic decomposition of endogenous H2O2 for relieving tumor hypoxia. More importantly, the obtained CoSnO3 nanocubes with a high photothermal conversion efficiency of 82.1% at 1064 nm could achieve mild hyperthermia (43 °C), which further improves the triple enzyme-like catalytic activities of the CoSnO3 nanozyme. The synergetic therapeutic efficacy of the NIR-II-responsive CoSnO3 nanozyme through mild NIR-II PTT-enhanced NCT could realize all-in-one multimodal tumor therapy to completely eliminate tumors without recurrence. This study will open a new avenue to explore NIR-II-photoresponsive nanozymes for efficient tumor therapy.
Collapse
Affiliation(s)
- Lang Yan
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Siyu Shang
- Operating Theatre, Department of Anaesthesiology, First Affliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Jinyan Hu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Xiaofang Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Jikuai Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Bijiang Geng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Yin Zhao
- Spine Center, Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jiangbo Zhu
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
23
|
Xu S, Qian Z, Zhao N, Yuan W. Thermoresponsive injectable self-healing hydrogel containing polydopamine-coated Fe/Mo-doped TiO 2 nanoparticles for efficient synergistic sonodynamic-chemodynamic-photothermal-chemo therapy. J Colloid Interface Sci 2024; 654:1431-1446. [PMID: 37922629 DOI: 10.1016/j.jcis.2023.10.145] [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: 08/03/2023] [Revised: 10/20/2023] [Accepted: 10/27/2023] [Indexed: 11/07/2023]
Abstract
A smart hydrogel loading multifunctional nanoparticles and anticancer drugs was designed to achieve synergistic therapy against tumors with high efficiency and specificity. The thermoresponsive injectable self-healing hydrogel was prepared through the Schiff base between aldehyde-functionalized poly(2-(2-methoxyethoxy) ethyl methacrylate)-co-oligo(ethylene glycol) methacrylate-co-2-hydroxyethyl methacrylate) (P(MEO2MA-co-OEGMA-co-HEMA), APMOH) and hydroxypropyl chitosan (HPCS). The polydopamine-coated Fe/Mo-doped titanium dioxide nanoparticles (PDA@dTiO2 NPs) were prepared and dispersed into the hydrogel with anticancer drug doxorubicin (DOX). PDA@dTiO2 NPs as sonosensitizers can convert oxygen into singlet oxygen (1O2) under ultrasound (US) irradiation, achieving sonodynamic therapy (SDT). They were also considered nanoenzymes, generating oxygen to supply an oxygen source for SDT, producing hydroxyl radical (·OH) to achieve chemodynamic therapy (CDT), and eliminating glutathione (GSH) to enhance the level of oxidative stress. After near-infrared (NIR) irradiation, the temperature of the hydrogel increased due to the photothermal ability of the polydopamine (PDA) layer. When the temperature reached the hydrogel's lower critical solution temperature (LCST), the hydrophilic-hydrophobic transformation occurred, and the hydrogel volume contracted. Consequently, the release rate of PDA@dTiO2 NPs and DOX increased, improving the therapeutic effects. The nanocomposite hydrogel system can achieve synergistic sonodynamic-chemodynamic-photothermal-chemo therapy (SDT-CDT-PTT-CT) for tumors, providing a novel platform for synergistic tumor treatment.
Collapse
Affiliation(s)
- Sicheng Xu
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Zhiyi Qian
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Nuoya Zhao
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Weizhong Yuan
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China.
| |
Collapse
|
24
|
Chen X, Xu Z, Li T, Thakur A, Wen Y, Zhang K, Liu Y, Liang Q, Liu W, Qin JJ, Yan Y. Nanomaterial-encapsulated STING agonists for immune modulation in cancer therapy. Biomark Res 2024; 12:2. [PMID: 38185685 PMCID: PMC10773049 DOI: 10.1186/s40364-023-00551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 01/09/2024] Open
Abstract
The cGAS-STING signaling pathway has emerged as a critical mediator of innate immune responses, playing a crucial role in improving antitumor immunity through immune effector responses. Targeting the cGAS-STING pathway holds promise for overcoming immunosuppressive tumor microenvironments (TME) and promoting effective tumor elimination. However, systemic administration of current STING agonists faces challenges related to low bioavailability and potential adverse effects, thus limiting their clinical applicability. Recently, nanotechnology-based strategies have been developed to modulate TMEs for robust immunotherapeutic responses. The encapsulation and delivery of STING agonists within nanoparticles (STING-NPs) present an attractive avenue for antitumor immunotherapy. This review explores a range of nanoparticles designed to encapsulate STING agonists, highlighting their benefits, including favorable biocompatibility, improved tumor penetration, and efficient intracellular delivery of STING agonists. The review also summarizes the immunomodulatory impacts of STING-NPs on the TME, including enhanced secretion of pro-inflammatory cytokines and chemokines, dendritic cell activation, cytotoxic T cell priming, macrophage re-education, and vasculature normalization. Furthermore, the review offers insights into co-delivered nanoplatforms involving STING agonists alongside antitumor agents such as chemotherapeutic compounds, immune checkpoint inhibitors, antigen peptides, and other immune adjuvants. These platforms demonstrate remarkable versatility in inducing immunogenic responses within the TME, ultimately amplifying the potential for antitumor immunotherapy.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Tongfei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, 442000, Shiyan, Hubei, China
| | - Abhimanyu Thakur
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, 60637, Chicago, IL, USA
| | - Yu Wen
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Furong Laboratory, Central South University, 410008, Changsha, Hunan, China
| | - Kui Zhang
- Pritzker School of Molecular Engineering, Ben May Department for Cancer Research, University of Chicago, 60637, Chicago, IL, USA
| | - Yuanhong Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Qiuju Liang
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, 310022, Hangzhou, Zhejiang, China.
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
25
|
Li J, Yue Z, Tang M, Wang W, Sun Y, Sun T, Chen C. Strategies to Reverse Hypoxic Tumor Microenvironment for Enhanced Sonodynamic Therapy. Adv Healthc Mater 2024; 13:e2302028. [PMID: 37672732 DOI: 10.1002/adhm.202302028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Sonodynamic therapy (SDT) has emerged as a highly effective modality for the treatment of malignant tumors owing to its powerful penetration ability, noninvasiveness, site-confined irradiation, and excellent therapeutic efficacy. However, the traditional SDT, which relies on oxygen availability, often fails to generate a satisfactory level of reactive oxygen species because of the widespread issue of hypoxia in the tumor microenvironment of solid tumors. To address this challenge, various approaches are developed to alleviate hypoxia and improve the efficiency of SDT. These strategies aim to either increase oxygen supply or prevent hypoxia exacerbation, thereby enhancing the effectiveness of SDT. In view of this, the current review provides an overview of these strategies and their underlying principles, focusing on the circulation of oxygen from consumption to external supply. The detailed research examples conducted using these strategies in combination with SDT are also discussed. Additionally, this review highlights the future prospects and challenges of the hypoxia-alleviated SDT, along with the key considerations for future clinical applications. These considerations include the development of efficient oxygen delivery systems, the accurate methods for hypoxia detection, and the exploration of combination therapies to optimize SDT outcomes.
Collapse
Affiliation(s)
- Jialun Li
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Zhengya Yue
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Minglu Tang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Wenxin Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin, 150076, P. R. China
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| | - Chunxia Chen
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
26
|
Zhao Y, Yuan B, Yan L, Wang Z, Xu Z, Geng B, Guo X, Chen X. In Situ Synthesis of Ru/TiO 2- x @TiCN Ternary Heterojunctions for Enhanced Sonodynamic and Nanocatalytic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307029. [PMID: 38032117 PMCID: PMC10811504 DOI: 10.1002/advs.202307029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/31/2023] [Indexed: 12/01/2023]
Abstract
Albeit nanozymes-based tumor catalytic therapy (NCT) relies on endogenous chemical reactions that could achieve tumor microenvironment (TME)-specialized reactive oxygen species (ROS) production, the unsatisfactory catalytic activity of nanozymes accompanied by complex TME poses a barrier to the therapeutic effect of NCT. Herein, a one-step in situ synthesis strategy is reported to construct ternary Ru/TiO2- x @TiCN heterojunctions through oxidative conversion of TiCN nanosheets (NSs) to TiO2- x NSs and reductive deposition of Ru3+ to Ru nanoparticles. The narrow bandgap and existence of heterojunctions enhance the ultrasound-activated ROS generation of Ru/TiO2- x @TiCN because of the accelerated electron transfer and inhibits electron-hole pair recombination. The augmented ROS production efficiency is achieved by Ru/TiO2- x @TiCN with triple enzyme-like activities, which amplifies the ROS levels in a cascade manner through the catalytic decomposition of endogenous H2 O2 to relieve hypoxia and heterojunction-mediated NCT, as well as depletion of overexpressed glutathione. The satisfactory therapeutic effects of Ru/TiO2- x @TiCN heterojunctions are achieved through synergetic sonodynamic therapy and NCT, which achieve the complete elimination of tumors without recurrence. This strategy highlights the potential of in situ synthesis of semiconductor heterojunctions as enhanced sonosensitizers and nanozymes for efficient tumor therapy.
Collapse
Affiliation(s)
- Yin Zhao
- Spine CenterDepartment of OrthopedicsShanghai Changzheng HospitalNaval Medical UniversityShanghai200003China
| | - Bo Yuan
- Spine CenterDepartment of OrthopedicsShanghai Changzheng HospitalNaval Medical UniversityShanghai200003China
| | - Lang Yan
- Department of Health ToxicologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433China
| | - Zhiwei Wang
- Spine CenterDepartment of OrthopedicsShanghai Changzheng HospitalNaval Medical UniversityShanghai200003China
| | - Zheng Xu
- Spine CenterDepartment of OrthopedicsShanghai Changzheng HospitalNaval Medical UniversityShanghai200003China
| | - Bijiang Geng
- School of Environmental and Chemical EngineeringShanghai UniversityShanghai200444China
| | - Xiang Guo
- Spine CenterDepartment of OrthopedicsShanghai Changzheng HospitalNaval Medical UniversityShanghai200003China
| | - Xiongsheng Chen
- Spine CenterDepartment of OrthopedicsShanghai Changzheng HospitalNaval Medical UniversityShanghai200003China
| |
Collapse
|
27
|
Yan L, Cao Z, Ren L, Zhang T, Hu J, Chen J, Zhang X, Liu B, Feng C, Zhu J, Geng B. A Sonoresponsive and NIR-II-Photoresponsive Nanozyme for Heterojunction-Enhanced "Three-in-One" Multimodal Oncotherapy. Adv Healthc Mater 2024; 13:e2302190. [PMID: 37792422 DOI: 10.1002/adhm.202302190] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/30/2023] [Indexed: 10/05/2023]
Abstract
Although low-cost nanozymes with excellent stability have demonstrated the potential to be highly beneficial for nanocatalytic therapy (NCT), their unsatisfactory catalytic activity accompanied by intricate tumor microenvironment (TME) significantly hinders the therapeutic effect of NCT. Herein, for the first time, a heterojunction (HJ)-fabricated sonoresponsive and NIR-II-photoresponsive nanozyme is reported by assembling carbon dots (CDs) onto TiCN nanosheets. The narrow bandgap and mixed valences of Ti3+ and Ti4+ endow TiCN with the capability to generate reactive oxygen species (ROS) when exposed to ultrasound (US), as well as the dual enzyme-like activities of peroxidase and glutathione peroxidase. Moreover, the catalytic activities and sonodynamic properties of the TiCN nanosheets are boosted by the formation of HJs owing to the increased speed of carrier transfer and the enhanced electron-hole separation. More importantly, the introduction of CDs with excellent NIR-II photothermal properties could achieve mild hyperthermia (43 °C) and thereby further improve the NCT and sonodynamic therapy (SDT) performances of CD/TiCN. The synergetic therapeutic efficacy of CD/TiCN through mild hyperthermia-amplified NCT and SDT could realize "three-in-one" multimodal oncotherapy to completely eliminate tumors without recurrence. This study opens a new avenue for exploring sonoresponsive and NIR-II-photoresponsive nanozymes for efficient tumor therapy based on semiconductor HJs.
Collapse
Affiliation(s)
- Lang Yan
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhi Cao
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Lijun Ren
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Tiantian Zhang
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Jinyan Hu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Jikuai Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xiaofang Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Bing Liu
- Depanrtment of Urology, the Third Affiliated Hospital of Naval Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, 201805, China
| | - Chuanqi Feng
- College of Chemistry and Chemical Engineering, Dezhou University, University West Road 566, Dezhou, Shandong, 253023, China
| | - Jiangbo Zhu
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Bijiang Geng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
28
|
Wu J, Pu K. Leveraging Semiconducting Polymer Nanoparticles for Combination Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308924. [PMID: 37864513 DOI: 10.1002/adma.202308924] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/20/2023] [Indexed: 10/23/2023]
Abstract
Cancer immunotherapy has become a promising method for cancer treatment, bringing hope to advanced cancer patients. However, immune-related adverse events caused by immunotherapy also bring heavy burden to patients. Semiconducting polymer nanoparticles (SPNs) as an emerging nanomaterial with high biocompatibility, can eliminate tumors and induce tumor immunogenic cell death through different therapeutic modalities, including photothermal therapy, photodynamic therapy, and sonodynamic therapy. In addition, SPNs can work as a functional nanocarrier to synergize with a variety of immunomodulators to amplify anti-tumor immune responses. In this review, SPNs-based combination cancer immunotherapy is comprehensively summarized according to the SPNs' therapeutic modalities and the type of loaded immunomodulators. The in-depth understanding of existing SPNs-based therapeutic modalities will hopefully inspire the design of more novel nanomaterials with potent anti-tumor immune effects, and ultimately promote their clinical translation.
Collapse
Affiliation(s)
- Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| |
Collapse
|
29
|
Cheng P, Ming S, Cao W, Wu J, Tian Q, Zhu J, Wei W. Recent advances in sonodynamic therapy strategies for pancreatic cancer. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1945. [PMID: 38403882 DOI: 10.1002/wnan.1945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Pancreatic cancer, a prevalent malignancy of the digestive system, has a poor 5-year survival rate of around 10%. Although numerous minimally invasive alternative treatments, including photothermal therapy and photodynamic therapy, have shown effectiveness compared with traditional surgical procedures, radiotherapy, and chemotherapy. However, the application of these alternative treatments is constrained by their depth of penetration, making it challenging to treat pancreatic cancer situated deep within the tissue. Sonodynamic therapy (SDT) has emerged as a promising minimally invasive therapy method that is particularly potent against deep-seated tumors such as pancreatic cancer. However, the unique characteristics of pancreatic cancer, including a dense surrounding matrix, high reductivity, and a hypoxic tumor microenvironment, impede the efficient application of SDT. Thus, to guide the evolution of SDT for pancreatic cancer therapy, this review addresses these challenges, examines current strategies for effective SDT enhancement for pancreatic cancer, and investigates potential future advances to boost clinical applicability. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuai Ming
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wei Cao
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jixiao Wu
- School of Materials and Chemistry, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jing Zhu
- School of Materials and Chemistry, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, Anhui, China
| | - Wei Wei
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
30
|
Yang YC, Zhu Y, Sun SJ, Zhao CJ, Bai Y, Wang J, Ma LT. ROS regulation in gliomas: implications for treatment strategies. Front Immunol 2023; 14:1259797. [PMID: 38130720 PMCID: PMC10733468 DOI: 10.3389/fimmu.2023.1259797] [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: 07/16/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023] Open
Abstract
Gliomas are one of the most common primary malignant tumours of the central nervous system (CNS), of which glioblastomas (GBMs) are the most common and destructive type. The glioma tumour microenvironment (TME) has unique characteristics, such as hypoxia, the blood-brain barrier (BBB), reactive oxygen species (ROS) and tumour neovascularization. Therefore, the traditional treatment effect is limited. As cellular oxidative metabolites, ROS not only promote the occurrence and development of gliomas but also affect immune cells in the immune microenvironment. In contrast, either too high or too low ROS levels are detrimental to the survival of glioma cells, which indicates the threshold of ROS. Therefore, an in-depth understanding of the mechanisms of ROS production and scavenging, the threshold of ROS, and the role of ROS in the glioma TME can provide new methods and strategies for glioma treatment. Current methods to increase ROS include photodynamic therapy (PDT), sonodynamic therapy (SDT), and chemodynamic therapy (CDT), etc., and methods to eliminate ROS include the ingestion of antioxidants. Increasing/scavenging ROS is potentially applicable treatment, and further studies will help to provide more effective strategies for glioma treatment.
Collapse
Affiliation(s)
- Yu-Chen Yang
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Yu Zhu
- College of Health, Dongguan Polytechnic, Dongguan, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Si-Jia Sun
- Department of Postgraduate Work, Xi’an Medical University, Xi’an, China
| | - Can-Jun Zhao
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Jin Wang
- Department of Radiation Protection Medicine, Faculty of Preventive Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China
- Shaanxi Key Laboratory of Free Radical and Medicine, Xi’an, China
| | - Li-Tian Ma
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province, Xi’an, China
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| |
Collapse
|
31
|
Li W, Liang M, Qi J, Ding D. Semiconducting Polymers for Cancer Immunotherapy. Macromol Rapid Commun 2023; 44:e2300496. [PMID: 37712920 DOI: 10.1002/marc.202300496] [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: 08/20/2023] [Revised: 09/09/2023] [Indexed: 09/16/2023]
Abstract
As a monumental breakthrough in cancer treatment, immunotherapy has attracted tremendous attention in recent years. However, one challenge faced by immunotherapy is the low response rate and the immune-related adverse events (irAEs). Therefore, it is important to explore new therapeutic strategies and platforms for boosting therapeutic benefits and decreasing the side effects of immunotherapy. In recent years, semiconducting polymer (SP), a category of organic materials with π-conjugated aromatic backbone, has been attracting considerable attention because of their outstanding characteristics such as excellent photophysical features, good biosafety, adjustable chemical flexibility, easy fabrication, and high stability. With these distinct advantages, SP is extensively explored for bioimaging and photo- or ultrasound-activated tumor therapy. Here, the recent advancements in SP-based nanomedicines are summarized for enhanced tumor immunotherapy. According to the photophysical properties of SPs, the cancer immunotherapies enabled by SPs with the photothermal, photodynamic, or sonodynamic functions are highlighted in detail, with a particular focus on the construction of combination immunotherapy and activatable nanoplatforms to maximize the benefits of cancer immunotherapy. Herein, new guidance and comprehensive insights are provided for the design of SPs with desired photophysical properties to realize maximized effectiveness of required biomedical applications.
Collapse
Affiliation(s)
- 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
| | - Mengyun Liang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- School of Materials Science and Engineering & Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin, 300350, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- School of Materials Science and Engineering & Smart Sensing Interdisciplinary Science Center, Nankai University, Tianjin, 300350, China
| |
Collapse
|
32
|
Di Y, Deng R, Liu Z, Mao Y, Gao Y, Zhao Q, Wang S. Optimized strategies of ROS-based nanodynamic therapies for tumor theranostics. Biomaterials 2023; 303:122391. [PMID: 37995457 DOI: 10.1016/j.biomaterials.2023.122391] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/25/2023]
Abstract
Reactive oxygen species (ROS) play a crucial role in regulating the metabolism of tumor growth, metastasis, death and other biological processes. ROS-based nanodynamic therapies (NDTs) are becoming attractive due to non-invasive, low side effects and tumor-specific advantages. NDTs have rapidly developed into numerous branches, such as photodynamic therapy, chemodynamic therapy, sonodynamic therapy and so on. However, the complexity of the tumor microenvironment and the limitations of existing sensitizers have greatly restricted the therapeutic effects of NDTs, which heavily rely on ROS levels. To address the limitations of NDTs, various strategies have been developed to increase ROS yield, which is an urgent aspect for the positive development of NDTs. In this review, the nanodynamic potentiation strategies in terms of unique properties and universalities of NDTs are comprehensively outlined. We mainly summarize the current dilemmas faced by each NDT and the respective solutions. Meanwhile, the NDTs universalities-based potentiation strategies and NDTs-based combined treatments are elaborated. Finally, we conclude with a discussion of the key issues and challenges faced in the development and clinical transformation of NDTs.
Collapse
Affiliation(s)
- Yifan Di
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Ruizhu Deng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Zhu Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China
| | - Yikun Gao
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China.
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, China.
| |
Collapse
|
33
|
Dai X, Du Y, Li Y, Yan F. Nanomaterials-based precision sonodynamic therapy enhancing immune checkpoint blockade: A promising strategy targeting solid tumor. Mater Today Bio 2023; 23:100796. [PMID: 37766898 PMCID: PMC10520454 DOI: 10.1016/j.mtbio.2023.100796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Burgeoning is an evolution from conventional photodynamic therapy (PDT). Thus, sonodynamic therapy (SDT) regulated by nanoparticles (NPs) possesses multiple advantages, including stronger penetration ability, better biological safety, and not reactive oxygen species (ROS)-dependent tumor-killing effect. However, the limitation to tumor inhibition instead of shrinkage and the incapability of eliminating metastatic tumors hinder the clinical potential for SDT. Fortunately, immune checkpoint blockade (ICB) can revive immunological function and induce a long-term immune memory against tumor rechallenges. Hence, synergizing NPs-based SDT with ICB can provide a promising therapeutic outcome for solid tumors. Herein, we briefly reviewed the progress in NPs-based SDT and ICB therapy. We highlighted the synergistic anti-tumor mechanisms and summarized the representative preclinical trials on SDT-assisted immunotherapy. Compared to other reviews, we provided comprehensive and unique perspectives on the innovative sonosensitizers in each trial. Moreover, we also discussed the current challenges and future corresponding solutions.
Collapse
Affiliation(s)
- Xinlun Dai
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Yangyang Du
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yumei Li
- Department of Pediatric Intensive Care Unit, First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
34
|
Guo L, Yang J, Wang H, Yi Y. Multistage Self-Assembled Nanomaterials for Cancer Immunotherapy. Molecules 2023; 28:7750. [PMID: 38067480 PMCID: PMC10707962 DOI: 10.3390/molecules28237750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Advances in nanotechnology have brought innovations to cancer therapy. Nanoparticle-based anticancer drugs have achieved great success from bench to bedside. However, insufficient therapy efficacy due to various physiological barriers in the body remains a key challenge. To overcome these biological barriers and improve the therapeutic efficacy of cancers, multistage self-assembled nanomaterials with advantages of stimuli-responsiveness, programmable delivery, and immune modulations provide great opportunities. In this review, we describe the typical biological barriers for nanomedicines, discuss the recent achievements of multistage self-assembled nanomaterials for stimuli-responsive drug delivery, highlighting the programmable delivery nanomaterials, in situ transformable self-assembled nanomaterials, and immune-reprogramming nanomaterials. Ultimately, we perspective the future opportunities and challenges of multistage self-assembled nanomaterials for cancer immunotherapy.
Collapse
Affiliation(s)
- Lamei Guo
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| |
Collapse
|
35
|
Sun W, Xiao H, Zhu J, Hao Z, Sun J, Wang D, Wang X, Ramalingam M, Xie S, Wang R. Multifunctional Oxygen-Generating Nanoflowers for Enhanced Tumor Therapy. ACS APPLIED BIO MATERIALS 2023; 6:4998-5008. [PMID: 37880964 DOI: 10.1021/acsabm.3c00678] [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: 10/27/2023]
Abstract
Sonodynamic therapy (SDT) and chemotherapy have received great attention as effective methods for tumor treatment. However, the inherent hypoxia of the tumor greatly hinders its therapeutic efficacy. In this work, a tumor microenvironment-responsive biodegradable nanoplatform SiO2-MnO2-PEG-Ce6&DOX (designated as SMPC&D) is fabricated by encapsulating manganese oxide (MnO2) into silica nanoparticles and anchoring poly(ethylene glycol) (PEG) onto the surface for tumor hypoxia relief and delivery, then loaded with sonosensitizer Chlorin e6 (Ce6) and chemotherapeutic drug doxorubicin (DOX) for hypoxic tumor treatment. We evaluated the physicochemical properties of SMPC&D nanoparticles and the tumor therapeutic effects of chemotherapy and SDT under ultrasound stimulation in vitro and in vivo. After endocytosis by tumor cells, highly expressed glutathione (GSH) triggers biodegradation of the nanoplatform and MnO2 catalyzes hydrogen peroxide (H2O2) to generate oxygen (O2), thereby alleviating tumor hypoxia. Depleting GSH and self-supplying O2 effectively improve the SDT efficiency both in vitro and in vivo. Ultrasonic stimulation promoted the release and cellular uptake of chemotherapy drugs. In addition, the relieved hypoxia reduced the efflux of chemotherapy drugs by downregulating the expression of the P-gp protein, which jointly improved the effect of chemotherapy. This study demonstrates that the degradable SMPC&D as a therapeutic agent can achieve efficient chemotherapy and SDT synergistic therapy for hypoxic tumors.
Collapse
Affiliation(s)
- Wanru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
- Science Fund of Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264000, People's Republic of China
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Huifang Xiao
- Zhongnan Hospital of Wuhan University, Wuhan 430062, People's Republic of China
| | - Jiazhi Zhu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Zhaokun Hao
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Jian Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Deqiang Wang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, People's Republic of China
| | - Xin Wang
- Department of Rehabilitation Medicine, Clinical Medical College, Yangzhou University, Yangzhou 225000, People's Republic of China
| | - Murugan Ramalingam
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country(UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
- Joint Research Laboratory (JRL), Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Drug Formulation Unit 10, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- Bioprinting and Precisión Medicine, Centro de investigación Lascaray Ikergunea, Avenida Miguel de Unamuno, 01006 Vitoria-Gasteiz, Spain
- School of Basic Medical Science, Chengdu University, Chengdu 610106, China
- Department of Metallurgical and Materials Engineering, Atilim University, Ankara 06830, Turkey
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, 78054 Villingen-Schwennigen, Germany
| | - Shuyang Xie
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
- Science Fund of Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yantai 264000, People's Republic of China
| |
Collapse
|
36
|
He S, Yu J, Xu M, Zhang C, Xu C, Cheng P, Pu K. A Semiconducting Iron-Chelating Nano-immunomodulator for Specific and Sensitized Sono-metallo-immunotherapy of Cancer. Angew Chem Int Ed Engl 2023; 62:e202310178. [PMID: 37671691 DOI: 10.1002/anie.202310178] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/07/2023]
Abstract
Sono-immunotherapy holds great potential for deep tumor inhibition; however, smart sono-therapeutic agents to simultaneously eliminate 'domestic' tumor cells and regulate the 'community' tumor immune microenvironment have rarely been developed. Herein, we report a spatiotemporally controllable semiconducting iron-chelated nano-metallomodulator (SINM) for hypersensitive sono-metallo-immunotherapy of cancer. SINM consists of a semiconducting polymer (SP) backbone chelating iron ions (Fe3+ ) with thiophene-based Schiff base structure, and a hydrophilic side chain. Upon accumulation in tumors after systemic administration, SINM specifically arouses ferroptosis and M1 macrophage polarization due to its response toward the tumor redox environment; meanwhile, the chelation of Fe3+ enhances the sono-sensitizing effect of SPs, leading to enhanced generation of reactive oxygen species for immunogenic cell death. Such combined sonodynamic metallo-immunotherapy of SINM efficiently ablates deep tumor and spatiotemporally regulates immunophenotypes.
Collapse
Affiliation(s)
- Shasha He
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jie Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Chi Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore, 636921, Singapore
| |
Collapse
|
37
|
Wang X, Zhu L, Zhou J, Zhao L, Li J, Liu C. Drug-loaded hybrid hydrogels for sonodynamic-chemodyanmic therapy and tumor metastasis suppression. Front Bioeng Biotechnol 2023; 11:1281157. [PMID: 37790250 PMCID: PMC10544978 DOI: 10.3389/fbioe.2023.1281157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction: Although various therapies have been adopted to treat cancer, metastasis of tumor cells still is a big challenge that compromises therapeutic benefits. Methods: We herein report an injectable drug-loaded hybrid hydrogel that can achieve sonodynamic therapy (SDT) and chemodyanmic therapy (CDT) combined action and suppression of tumor metastasis. This alginate (ALG)-based hydrogel (termed as AMPS) contains manganese dioxide (MnO2) nanoparticles as the CDT agents, an organic polymer as the sonosensitizer, and a SIS3 drug as metastasis inhibitor. Results: AMPS is formed via the chelation of ALG by Ca2+ in tumor microenvironment, in which MnO2 nanoparticles mediate CDT via Fenton-like reaction and the organic polymers enable SDT under ultrasound (US) irradiation by generating singlet oxygen (1O2), allowing for combinational action of CDT and SDT. In addition, SIS3 is released from AMPS hydrogels to inhibit the metastasis of tumor cells. As such, the AMPS enables a combinational action of SDT and CDT to greatly inhibit the growths of subcutaneous tumors in living mice and also completely suppress the tumor metastasis in lungs and livers. Conclusion: This study thus offers a hybrid hydrogel platform for combinational therapy and metastasis suppression simultaneously.
Collapse
Affiliation(s)
- Xiaoying Wang
- Office of Hospital Infection and Disease Control and Prevention, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liyun Zhu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Jianhui Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingchao Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Changcun Liu
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Huang L, Su Y, Zhang D, Zeng Z, Hu X, Hong S, Lin X. Recent theranostic applications of hydrogen peroxide-responsive nanomaterials for multiple diseases. RSC Adv 2023; 13:27333-27358. [PMID: 37705984 PMCID: PMC10496458 DOI: 10.1039/d3ra05020c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/31/2023] [Indexed: 09/15/2023] Open
Abstract
It is well established that hydrogen peroxide (H2O2) is associated with the initiation and progression of many diseases. With the rapid development of nanotechnology, the diagnosis and treatment of those diseases could be realized through a variety of H2O2-responsive nanomaterials. In order to broaden the application prospects of H2O2-responsive nanomaterials and promote their development, understanding and summarizing the design and application fields of such materials has attracted much attention. This review provides a comprehensive summary of the types of H2O2-responsive nanomaterials including organic, inorganic and organic-inorganic hybrids in recent years, and focused on their specific design and applications. Based on the type of disease, such as tumors, bacteria, dental diseases, inflammation, cardiovascular diseases, bone injury and so on, key examples for above disease imaging diagnosis and therapy strategies are introduced. In addition, current challenges and the outlook of H2O2-responsive nanomaterials are also discussed. This review aims to stimulate the potential of H2O2-responsive nanomaterials and provide new application ideas for various functional nanomaterials related to H2O2.
Collapse
Affiliation(s)
- Linjie Huang
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Yina Su
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Dongdong Zhang
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Zheng Zeng
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Xueqi Hu
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Shanni Hong
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| | - Xiahui Lin
- School of Medical Imaging, Fujian Medical University Fuzhou 350122 Fujian P. R. China
| |
Collapse
|
39
|
Wu Q, Zhang J, Pan X, Huang Z, Zhang H, Guo J, Xue Y, Shi R, Liu H. Vacancy Augmented Piezo-Sonosensitizer for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301152. [PMID: 37395638 PMCID: PMC10502820 DOI: 10.1002/advs.202301152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/01/2023] [Indexed: 07/04/2023]
Abstract
Sonodynamic therapy (SDT) has been widely reported as a noninvasive and high-penetration therapy for cancer; however, the design of an efficient sonosensitizer remains an urgent need. To address this issue, molybdenum disulfide nanoflowers (MoS2 NF) as piezo-sonosensitizers and introduced sulfur vacancies on the MoS2 NF (Sv-MoS2 NF) to improve their piezoelectric property for cancer therapy are designed. Under ultrasonic mechanical stress, the Sv-MoS2 NF resulted in piezoelectric polarization and band tilting, which enhanced the charge carrier separation and migration. This resulted in an improved catalytic reaction for reactive oxygen species (ROS) production, ultimately enhancing the SDT performance. Thanks to the high efficiency of ROS generation, the Sv-MoS2 NF have demonstrated a good anticancer effect in vitro and in vivo. Following a systematic evaluation, Sv-MoS2 NF also demonstrated good biocompatibility. This novel piezo-sonosensitizer and vacancy engineering strategy provides a promising new approach for achieving efficient SDT.
Collapse
Affiliation(s)
- Qingyuan Wu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBeijing Laboratory of Biomedical MaterialsBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Jie Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBeijing Laboratory of Biomedical MaterialsBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xueting Pan
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBeijing Laboratory of Biomedical MaterialsBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Zhijun Huang
- Beijing National Laboratory of Molecular Sciences, Beijing National Laboratory of Molecular SciencesInstitute of Chemistry, Chinese Academy of SciencesBeijing100190P. R. China
| | - Haoyuan Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBeijing Laboratory of Biomedical MaterialsBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Juan Guo
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBeijing Laboratory of Biomedical MaterialsBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Yun Xue
- National Center for OrthopaedicsBeijing Research Institute of Traumatology and OrthopaedicsBeijing Jishuitan HospitalBeijing100035P. R. China
| | - Rui Shi
- National Center for OrthopaedicsBeijing Research Institute of Traumatology and OrthopaedicsBeijing Jishuitan HospitalBeijing100035P. R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBeijing Laboratory of Biomedical MaterialsBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing University of Chemical TechnologyBeijing100029P. R. China
| |
Collapse
|
40
|
Jiang Z, Xiao W, Fu Q. Stimuli responsive nanosonosensitizers for sonodynamic therapy. J Control Release 2023; 361:547-567. [PMID: 37567504 DOI: 10.1016/j.jconrel.2023.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/27/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Sonodynamic therapy (SDT) has gained significant attention in the treatment of deep tumors and multidrug-resistant (MDR) bacterial infections due to its high tissue penetration depth, high spatiotemporal selectivity, and noninvasive therapeutic method. SDT combines low-intensity ultrasound (US) and sonosensitizers to produce lethal reactive oxygen species (ROS) and external damage, which is the main mechanism behind this therapy. However, traditional organic small-molecule sonosensitizers display poor water solubility, strong phototoxicity, and insufficient targeting ability. Inorganic sonosensitizers, on the other hand, have low ROS yield and poor biocompatibility. These drawbacks have hindered SDT's clinical transformation and application. Hence, designing stimuli-responsive nano-sonosensitizers that make use of the lesion's local microenvironment characteristics and US stimulation is an excellent alternative for achieving efficient, specific, and safe treatment. In this review, we provide a comprehensive overview of the currently accepted mechanisms in SDT and discuss the application of responsive nano-sonosensitizers in the treatment of tumor and bacterial infections. Additionally, we emphasize the significance of the principle and process of response, based on the classification of response patterns. Finally, this review emphasizes the potential limitations and future perspectives of SDT that need to be addressed to promote its clinical transformation.
Collapse
Affiliation(s)
- Zeyu Jiang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China; Department of Cardiovascular Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266003, China
| | - Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
41
|
Zhao R, Zhu H, Feng L, Zhu Y, Liu B, Yu C, Gai S, Yang P. 2D Piezoelectric BiVO 4 Artificial Nanozyme with Adjustable Vanadium Vacancy for Ultrasound Enhanced Piezoelectric/Sonodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301349. [PMID: 37127877 DOI: 10.1002/smll.202301349] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/06/2023] [Indexed: 05/03/2023]
Abstract
Increasing the yield of reactive oxygen species (ROS) to enhance oxidative stress in cells is an eternal goal in cancer therapy. In this study, BiVO4 artificial nanozyme is developed with adjustable vanadium vacancy for ultrasound (US) enhanced piezoelectric/sonodynamic therapy. Under US excitation, the vanadium vacancy-rich BiVO4 nanosheets (abbreviated Vv -r BiVO4 NSs) facilitate the generation of a large number of electrons to improve the ROS yield. Meanwhile, the mechanical strain imposed by US irradiation makes the Vv -r BiVO4 NSs display a typical piezoelectric response, which tilts the conduction band to be more negative and the valance band more positive than the redox potentials of O2 /O2 •- and H2 O/·OH, boosting the efficiency of ROS generation. Both density functional theory calculations and experiments confirm that the introduction of cationic vacancy can improve the sonodynamic effect. As expected, Vv -r BiVO4 NSs have better peroxidase enzyme catalytic and glutathione depletion activities, resulting in increased intracellular oxidative stress. This triple amplification strategy of oxidative stress induced by US substantially inhibits the growth of cancer cells. The work may open an avenue to achieve a synergetic therapy by introducing cationic vacancy, broadening the biomedical use of piezoelectric materials.
Collapse
Affiliation(s)
- Ruoxi Zhao
- 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
| | - Haixia Zhu
- Cancer Institute, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, P. R. China
| | - Lili Feng
- 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
| | - Yanlin Zhu
- 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
| | - 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
| | - Chenghao Yu
- 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
| | - Shili Gai
- 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
| | - Piaoping Yang
- 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
| |
Collapse
|
42
|
Liu S, Ma J, Xue EY, Wang S, Zheng Y, Ng DKP, Wang A, Zheng N. Polymeric Phthalocyanine-Based Nanosensitizers for Enhanced Photodynamic and Sonodynamic Therapies. Adv Healthc Mater 2023; 12:e2300481. [PMID: 37019442 DOI: 10.1002/adhm.202300481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Indexed: 04/07/2023]
Abstract
Photodynamic therapy and sonodynamic therapy are two highly promising modalities for cancer treatment. The latter holds an additional advantage in deep-tumor therapy owing to the deep penetration of the ultrasonic radiation. The therapeutic efficacy depends highly on the photo/ultrasound-responsive properties of the sensitizers as well as their tumor-localization property and pharmacokinetics. A novel nanosensitizer system based on a polymeric phthalocyanine (pPC-TK) is reported herein in which the phthalocyanine units are connected with cleavable thioketal linkers. Such polymer could self-assemble in water forming nanoparticles with a hydrodynamic diameter of 48 nm. The degradable and flexible thioketal linkers could effectively inhibit the π-π stacking of the phthalocyanine units, rendering the resulting nanoparticles an efficient generator of reactive oxygen species upon light or ultrasonic irradiation. The nanosensitizer could be internalized into cancer cells readily, inducing cell death by efficient photodynamic and sonodynamic effects. The potency is significantly higher than that of the monomeric phthalocyanine (PC-4COOH). The nanosensitizer could also effectively inhibit the growth of tumor in liver tumor-bearing mice by these two therapies without causing noticeable side effects. More importantly, it could also retard the growth of a deep-located orthotopic liver tumor in vivo by sonodynamic therapy.
Collapse
Affiliation(s)
- Shuxin Liu
- School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jinjuan Ma
- Department of Comparative Medicine Laboratory Animal Center, Dalian Medical University, Dalian, 116000, China
| | - Evelyn Y Xue
- Department of Chemistry, The Chinese University of Hong Kong, Shatin N.T., Hong Kong, 999077, China
| | - Shaolei Wang
- Department of Radiology Intervention, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110801, China
| | - Yubin Zheng
- School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Dalian University of Technology Corporation of Changshu Research Institution, Suzhou, 215500, China
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong, Shatin N.T., Hong Kong, 999077, China
| | - Aiguo Wang
- Department of Comparative Medicine Laboratory Animal Center, Dalian Medical University, Dalian, 116000, China
| | - Nan Zheng
- School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Dalian University of Technology Corporation of Changshu Research Institution, Suzhou, 215500, China
| |
Collapse
|
43
|
Dong HQ, Fu XF, Wang MY, Zhu J. Research progress on reactive oxygen species production mechanisms in tumor sonodynamic therapy. World J Clin Cases 2023; 11:5193-5203. [PMID: 37621595 PMCID: PMC10445077 DOI: 10.12998/wjcc.v11.i22.5193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/22/2023] [Indexed: 08/04/2023] Open
Abstract
In recent years, because of the growing desire to improve the noninvasiveness and safety of tumor treatments, sonodynamic therapy has gradually become a popular research topic. However, due to the complexity of the therapeutic process, the relevant mechanisms have not yet been fully elucidated. One of the widely accepted possibilities involves the effect of reactive oxygen species. In this review, the mechanism of reactive oxygen species production by sonodynamic therapy (SDT) and ways to enhance the sonodynamic production of reactive oxygen species are reviewed. Then, the clinical application and limitations of SDT are discussed. In conclusion, current research on sonodynamic therapy should focus on the development of sonosensitizers that efficiently produce active oxygen, exhibit biological safety, and promote the clinical transformation of sonodynamic therapy.
Collapse
Affiliation(s)
- He-Qin Dong
- School of Medicine, Shaoxing University, Shaoxin 312000, Zhejiang Province, China
| | - Xiao-Feng Fu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Min-Yan Wang
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Jiang Zhu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
44
|
Dong HQ, Fu XF, Wang MY, Zhu J. Research progress on reactive oxygen species production mechanisms in tumor sonodynamic therapy. World J Clin Cases 2023; 11:5187-5197. [DOI: 10.12998/wjcc.v11.i22.5187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/27/2023] [Accepted: 05/22/2023] [Indexed: 08/03/2023] Open
Abstract
In recent years, because of the growing desire to improve the noninvasiveness and safety of tumor treatments, sonodynamic therapy has gradually become a popular research topic. However, due to the complexity of the therapeutic process, the relevant mechanisms have not yet been fully elucidated. One of the widely accepted possibilities involves the effect of reactive oxygen species. In this review, the mechanism of reactive oxygen species production by sonodynamic therapy (SDT) and ways to enhance the sonodynamic production of reactive oxygen species are reviewed. Then, the clinical application and limitations of SDT are discussed. In conclusion, current research on sonodynamic therapy should focus on the development of sonosensitizers that efficiently produce active oxygen, exhibit biological safety, and promote the clinical transformation of sonodynamic therapy.
Collapse
Affiliation(s)
- He-Qin Dong
- School of Medicine, Shaoxing University, Shaoxin 312000, Zhejiang Province, China
| | - Xiao-Feng Fu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Min-Yan Wang
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Jiang Zhu
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
45
|
Chen X, Ruan M, Wu J, Zhu T, Wen Y, Li X, Yang C, Zong X, Yuan P, Li Y, Yan X, Xue W, Dai J. Constructing Spatiotemporally Controllable Biocatalytic Cascade in RBC Nanovesicles for Precise Tumor Therapy Based on Reversibly Induced Glucose Oxidase-Magnetoferritin Dimers. Adv Healthc Mater 2023; 12:e2300205. [PMID: 37052368 DOI: 10.1002/adhm.202300205] [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: 01/19/2023] [Revised: 03/26/2023] [Indexed: 04/14/2023]
Abstract
Chemodynamic therapy is a promising tumor treatment strategy. However, it remains a great challenge to overcome the unavoidable off-target damage to normal tissues. In this work, it is discovered that magnetoferritin (M-HFn, biomimic peroxidase) can form nanocomplexes with glucose oxidase (GOD) in the presence of glucose, thus inhibiting the enzyme activity of GOD. Interestingly, GOD&M-HFn (G-M) nanocomplexes can dissociate under near-infrared (NIR) laser, reactivating the enzyme cascade. Based on this new finding, a spatiotemporally controllable biocatalytic cascade in red blood cell (RBC) nanovesicles (G-M@RBC-A) is fabricated for precise tumor therapy, which in situ inhibits enzyme cascade between GOD and M-HFn during blood circulation and reactivates the cascade activity in tumor site by NIR laser irradiation. In RBC nanovesicles, GOD is grabbed by M-HFn to form G-M nanocomplexes in the presence of glucose, thus inhibiting the Fenton reaction and reducing side effects. However, after NIR laser irradiation, G-M nanocomplexes are spatiotemporally dissociated and the cascade activity is reactivated in the tumor site, initiating reactive oxygen species damage to cancer cells in vivo. Therefore, this work provides new insight into the fabrication of spatiotemporally controllable biocatalytic cascade for precise cancer therapy in the future.
Collapse
Affiliation(s)
- Xinjie Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Miaoliang Ruan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Jinpei Wu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Tianci Zhu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yaoqi Wen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xiaodi Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Caiqi Yang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xiaoqing Zong
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Pengfei Yuan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Yuchao Li
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Xiaodie Yan
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| | - Jian Dai
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Engineering Technology Research Center of Drug Carrier of Guangdong, Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
46
|
Shen J, Chen G, Zhao L, Huang G, Liu H, Liu B, Miao Y, Li Y. Recent Advances in Nanoplatform Construction Strategy for Alleviating Tumor Hypoxia. Adv Healthc Mater 2023; 12:e2300089. [PMID: 37055912 DOI: 10.1002/adhm.202300089] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Indexed: 04/15/2023]
Abstract
Hypoxia is a typical feature of most solid tumors and has important effects on tumor cells' proliferation, invasion, and metastasis. This is the key factor that leads to poor efficacy of different kinds of therapy including chemotherapy, radiotherapy, photodynamic therapy, etc. In recent years, the construction of hypoxia-relieving functional nanoplatforms through nanotechnology has become a new strategy to reverse the current situation of tumor microenvironment hypoxia and improve the effectiveness of tumor treatment. Here, the main strategies and recent progress in constructing nanoplatforms are focused on to directly carry oxygen, generate oxygen in situ, inhibit mitochondrial respiration, and enhance blood perfusion to alleviate tumor hypoxia. The advantages and disadvantages of these nanoplatforms are compared. Meanwhile, nanoplatforms based on organic and inorganic substances are also summarized and classified. Through the comprehensive overview, it is hoped that the summary of these nanoplatforms for alleviating hypoxia could provide new enlightenment and prospects for the construction of nanomaterials in this field.
Collapse
Affiliation(s)
- Jing Shen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Guobo Chen
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Linghao Zhao
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Guoyang Huang
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Hui Liu
- Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Baolin Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Miao
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuhao Li
- School of Materials and Chemistry & Institute of Bismuth, University of Shanghai for Science and Technology, Shanghai, 200093, China
| |
Collapse
|
47
|
Liang S, Yao J, Liu D, Rao L, Chen X, Wang Z. Harnessing Nanomaterials for Cancer Sonodynamic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211130. [PMID: 36881527 DOI: 10.1002/adma.202211130] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/12/2023] [Indexed: 06/18/2023]
Abstract
Immunotherapy has made remarkable strides in cancer therapy over the past decade. However, such emerging therapy still suffers from the low response rates and immune-related adverse events. Various strategies have been developed to overcome these serious challenges. Therein, sonodynamic therapy (SDT), as a non-invasive treatment, has received ever-increasing attention especially in the treatment of deep-seated tumors. Significantly, SDT can effectively induce immunogenic cell death to trigger systemic anti-tumor immune response, termed sonodynamic immunotherapy. The rapid development of nanotechnology has revolutionized SDT effects with robust immune response induction. As a result, more and more innovative nanosonosensitizers and synergistic treatment modalities are established with superior efficacy and safe profile. In this review, the recent advances in cancer sonodynamic immunotherapy are summarized with a particular emphasis on how nanotechnology can be explored to harness SDT for amplifying anti-tumor immune response. Moreover, the current challenges in this field and the prospects for its clinical translation are also presented. It is anticipated that this review can provide rational guidance and facilitate the development of nanomaterials-assisted sonodynamic immunotherapy, helping to pave the way for next-generation cancer therapy and eventually achieve a durable response in patients.
Collapse
Affiliation(s)
- Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jianjun Yao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, 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
- 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
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
48
|
Mohajer F, Mirhosseini-Eshkevari B, Ahmadi S, Ghasemzadeh MA, Mohammadi Ziarani G, Badiei A, Farshidfar N, Varma RS, Rabiee N, Iravani S. Advanced Nanosystems for Cancer Therapeutics: A Review. ACS APPLIED NANO MATERIALS 2023; 6:7123-7149. [DOI: 10.1021/acsanm.3c00859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Affiliation(s)
- Fatemeh Mohajer
- Department of Organic Chemistry, Faculty of Chemistry, Alzahra University, Tehran 19938-93973, Iran
| | | | - Sepideh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | | | - Ghodsi Mohammadi Ziarani
- Department of Organic Chemistry, Faculty of Chemistry, Alzahra University, Tehran 19938-93973, Iran
| | - Alireza Badiei
- School of Chemistry, College of Science, University of Tehran, Tehran 14179-35840, Iran
| | - Nima Farshidfar
- Orthodontic Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Rajender S. Varma
- Institute for Nanomaterials, Advanced Technologies and Innovation (CxI), Technical University of Liberec (TUL), 1402/2, Liberec 1 461 17, Czech Republic
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia 6150, Australia
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| |
Collapse
|
49
|
Jiang J, Zhang M, Lyu T, Chen L, Wu M, Li R, Li H, Wang X, Jiang X, Zhen X. Sono-Driven STING Activation using Semiconducting Polymeric Nanoagonists for Precision Sono-Immunotherapy of Head and Neck Squamous Cell Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2300854. [PMID: 37119091 DOI: 10.1002/adma.202300854] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/09/2023] [Indexed: 06/18/2023]
Abstract
Immunotherapy has offered new opportunities to treat head and neck squamous cell carcinoma (HNSCC); however, its clinical applications are hindered by modest therapeutic outcomes and the "always-on" pharmacological activity of immunomodulatory agents. Strategies for precise spatiotemporal activation of antitumor immunity can tackle these issues but remain challenging. Herein, a semiconducting polymeric nanoagonist (SPNM) with in situ sono-activatable immunotherapeutic effects for precision sono-immunotherapy of HNSCC is reported. SPNM is self-assembled from a sonodynamic semiconducting polymer core conjugated with a stimulator of interferon genes (STING) agonist (MSA-2) via a singlet oxygen cleavable linker. Under sono-irradiation, SPNM produces singlet oxygen not only to eradicate tumor cells to trigger immunogenic cell death but also to unleash caged STING agonists via the cleavage of diphenoxyethene bonds for in situ activation of the STING pathway in the tumor region. Such sono-driven STING activation mediated by SPNM promotes effector T cell infiltration and potentiates systemic antitumor immunity, eventually leading to tumor growth inhibition and long-term immunological memory. This study thus presents a promising strategy for the precise spatiotemporal activation of cancer immunotherapy.
Collapse
Affiliation(s)
- Jianli Jiang
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Miaomiao Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Tao Lyu
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Linrong Chen
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Min Wu
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Ruowei Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Haoze Li
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Xiang Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiqun Jiang
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Xu Zhen
- College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
50
|
Perez-Potti A, Rodríguez-Pérez M, Polo E, Pelaz B, Del Pino P. Nanoparticle-based immunotherapeutics: from the properties of nanocores to the differential effects of administration routes. Adv Drug Deliv Rev 2023; 197:114829. [PMID: 37121275 DOI: 10.1016/j.addr.2023.114829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 05/02/2023]
Abstract
The engagement with the immune system is one of the main cornerstones in the development of nanotechnologies for therapy and diagnostics. Recent advances have made possible the tuning of features like size, shape and biomolecular modifications that influence such interactions, however, the capabilities for immune modulation of nanoparticles are still not well defined and exploited. This review focuses on recent advances made in preclinical research for the application of nanoparticles to modulate immune responses, and the main features making them relevant for such applications. We review and discuss newest evidence in the field, which include in vivo experiments with an extensive physicochemical characterization as well as detailed study of the induced immune response. We emphasize the need of incorporating knowledge about immune response development and regulation in the design and application of nanoparticles, including the effect by parameters such as the administration route and the differential interactions with immune subsets.
Collapse
Affiliation(s)
- André Perez-Potti
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Manuel Rodríguez-Pérez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ester Polo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Pablo Del Pino
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|