1
|
Huang C, Li J, Wu R, Li Y, Zhang C. Targeting pyroptosis for cancer immunotherapy: mechanistic insights and clinical perspectives. Mol Cancer 2025; 24:131. [PMID: 40319304 PMCID: PMC12049004 DOI: 10.1186/s12943-025-02344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025] Open
Abstract
Pyroptosis is a distinct form of programmed cell death characterized by the rupture of the cell membrane and robust inflammatory responses. Increasing evidence suggests that pyroptosis significantly affects the tumor microenvironment and antitumor immunity by releasing damage-associated molecular patterns (DAMPs) and pro-inflammatory mediators, thereby establishing it as a pivotal target in cancer immunotherapy. This review thoroughly explores the molecular mechanisms underlying pyroptosis, with a particular focus on inflammasome activation and the gasdermin family of proteins (GSDMs). It examines the role of pyroptotic cell death in reshaping the tumor immune microenvironment (TIME) involving both tumor and immune cells, and discusses recent advancements in targeting pyroptotic pathways through therapeutic strategies such as small molecule modulators, engineered nanocarriers, and combinatory treatments with immune checkpoint inhibitors. We also review recent advances and future directions in targeting pyroptosis to enhance tumor immunotherapy with immune checkpoint inhibitors, adoptive cell therapy, and tumor vaccines. This study suggested that targeting pyroptosis offers a promising avenue to amplify antitumor immune responses and surmount resistance to existing immunotherapies, potentially leading to more efficacious cancer treatments.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruiyan Wu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
2
|
Luo X, Zhou Y, Rao K, Xiang J, Ning S, Zhu D, Li G, Chen H. Biomimetic Cascade Nanozyme Catalytic System for the Treatment of Lymph Node Metastasis in Gastric Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2411576. [PMID: 40123244 DOI: 10.1002/smll.202411576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/18/2025] [Indexed: 03/25/2025]
Abstract
Lymphatic metastasis of gastric cancer is a challenging issue in clinical practice. Recently, copper single-atom nanozymes (SAZ) have gained tremendous attention due to its superior peroxidase (POD) activity that has good nonocatalytic tumor therapy (NCT) capabilities, and photothermal properties. Therefore, using a high-expressing P-selectin platelet membrane (PM) to encapsulate SAZ and cisplatin is proposed, forming PSC nanoparticles. Due to their exquisite nanoscale size and the unique structure of lymphatic vessels, PSC can highly target cancer cells in invasive primary tumors and metastatic lymph nodes that both highly express CD44. It is noteworthy that cisplatin can simultaneously perform chemotherapy and generate H₂O₂ under the action of NADPH oxidases (NOXs) that further enhance the catalytic activity of SAZ and increase intracellular reactive oxygen species (ROS) production. Both in vitro and vivo experiments have demonstrated the superior targeting and elimination capability of the PCS system in primary and metastatic tumor cells. In addition, transcriptomic analysis reveals that PSC + NIR induced apoptosis in MFC cells. This marks the first proposal of combining single-atom nanozymes and chemotherapy drugs for dual-targeting in gastric cancer and lymphatic metastasis, providing new insights into a challenging clinical issue in the treatment of gastric cancer lymphatic metastasis.
Collapse
Affiliation(s)
- Xi Luo
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yingguang Zhou
- Department of Joint Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510900, P. R. China
| | - Kexiang Rao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jingfeng Xiang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, P. R. China
| | - Daoming Zhu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Guoxin Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
- Cancer Center of Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, No. 168 Litang Road, Changping District, Beijing, 102218, P. R. China
| | - Hao Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|
3
|
Du Q, Jiang H, Wu D, Song C, Hu W, Lu Q, Sun C, Liu J, Wu G, Wang S. Radiation-Activated Cobalt-Based Zeolite Imidazolate Frameworks for Tumor Multitherapy. Biomater Res 2025; 29:0164. [PMID: 40236956 PMCID: PMC11997308 DOI: 10.34133/bmr.0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 04/17/2025] Open
Abstract
Radiation dynamic therapy (RDT) is known to induce cancer apoptosis and death with minimal side effects and high accuracy. However, low efficiency of radiation sensitization and persistent hypoxic environment in tumors pose marked challenges for successful RDT. To address these challenges, a novel biodegradable drug delivery system was developed, using quercetin and sorafenib-loaded ZIF67 nanoparticles (QSZP NPs) coated with polydopamine. This system effectively controlled the tumor microenvironment (TME), overcame hypoxia, and was thus utilized for collaborative RDT and radiotherapy (RT). The QSZP NPs demonstrated great potential in x-ray sensitization and reactive oxygen species (ROS)-mediated effects in vitro. Furthermore, they continuously generated oxygen and increased ROS levels in the TME with x-ray irradiation to achieve RDT. In vivo studies showed that QSZP NPs had no apparent systemic toxicity and showed good therapeutic effect in a HepG2 tumor-bearing model. Due to its unique and outstanding combinational effect of RDT/RT/antiangiogenic cancer therapy, these synthesized NPs offer a promising method for radiation-based cancer treatment.
Collapse
Affiliation(s)
- Qijun Du
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Hongwei Jiang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine,
Henan University of Science and Technology, Luoyang 471003, China
| | - Di Wu
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu 641400, China
| | - Changlong Song
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Wenqi Hu
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Qinrui Lu
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Chenwei Sun
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Jie Liu
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Guohua Wu
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine,
Henan University of Science and Technology, Luoyang 471003, China
| | - Shuqi Wang
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu 641400, China
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
4
|
Meng Q, Ding B, Ma P, Lin J. Inorganic Nanobiomaterials Boost Tumor Immunotherapy: Strategies and Applications. Acc Chem Res 2025; 58:1210-1223. [PMID: 40179239 DOI: 10.1021/acs.accounts.4c00843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
ConspectusTumor immunotherapy, as a new antitumor method to fight cancer by activating or enhancing the body's own immune system, has been extensively studied and applied in clinical practice. However, as an extremely complex system, tumor heterogeneity and complex immunosuppressive tumor microenvironment (TME) lead to poor immune response rate or secondary drug resistance. The advent of nanotechnology has ushered in a new era for immunotherapy. In particular, inorganic nanomaterials, with their unique physicochemical properties and excellent biocompatibility, are becoming an important tool for enhancing immunotherapy. Inorganic nanomaterials can be used as carriers for immune agents, improving drug delivery efficiency and thereby reducing systemic immunotoxicity and enhancing immune responses. Inorganic nanomaterials also trigger tumor immunogenic cell death (ICD), stimulate antitumor immune responses, and alleviate immunosuppressive TME by increasing oxygen levels, modulating metabolic pathways, and altering the secretion of immunosuppressive cytokines. The synergistic integration of inorganic nanomaterials with immunotherapy adeptly navigates around the constraints of conventional treatments, reducing side effects while concurrently augmenting therapeutic efficacy. In this review, we summarize our recent efforts in the design and synthesis of inorganic nanobiomaterials to enhance the efficacy of tumor immunotherapy. These nanomaterials achieve the desired immune efficacy mainly through four strategies, including inducing ICD, developing tumor nanovaccines, activating pyroptosis, and regulating tumor metabolism, providing beneficial implications for tumor immunotherapy. For one thing, due to the deficiency of ICD effect in single therapy, we mainly developed nanocatalysts that integrate multiple therapeutic functions to play a catalytic role in TME, converting tumor substances or metabolites into therapeutic products in situ, and further enhancing ICD. For another, in order to solve the problems of low antigen loading and therapeutic efficiency of existing adjuvants, several novel multifunctional nanoadjuvants were prepared, which combine high antigen loading and multimode therapeutic function in one, and achieve efficient immune activation. Moreover, to attain strong inflammatory responses and immunogenicity, we engineer pyroptosis adjuvants that selectively induce tumor cell pyroptosis by enhancing intracellular oxidative stress or ion overload. Finally, to reverse the immunosuppressive microenvironment, we developed nanoplatforms that target tumor metabolism, altering the levels of nutrients and metabolites in tumor such as glucose, lactic acid, citric acid, and tryptophan to effectively alter the TME, thereby activating and enhancing the body's immune response. The implementation of these strategies not only improves the therapeutic effect but also reduces the side effects and provides valuable insights and references for the development of novel nanomaterials to assist immunotherapy.
Collapse
Affiliation(s)
- Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
5
|
Jin S, Chen Y, Li F, Yan P, Guo G, Xu G, Song W, Zhong W. Photodynamic and Photothermal Co-Induced Efficient Anti-Tumor Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20833-20848. [PMID: 40152855 DOI: 10.1021/acsami.5c00184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Currently, immunotherapy based on photothermal and the application of photodynamic therapy in anti-tumor treatment is showing great potential. Its uniqueness lies in the critical role of small molecule immunomodulators in promoting effective immune responses against tumors, and the use of laser-activated biophysical mechanisms to precisely trigger the swift demise of cancer cells, avoiding damage to surrounding normal tissues. However, the use of photodynamic therapy (PDT) alone is hampered by the tumors' hypoxic environment, resulting in poor antitumor effects, while photothermal therapy (PTT) alone cannot arouse enough antigen presentation. It is of great significance to design photosensitizers (PSs) that possess both PDT and PTT effects. Herein, a series of PSs with both PDT and PTT efficacy are reported, ultimately selecting Cy7-Naph as the star molecule due to its best overall phototherapeutic effect. Upon reactive oxygen species (ROS) production and thermogenesis in tumor cells, Cy7-Naph induced significant apoptosis and eventually boosted the release of damage-associated molecular patterns (DAMPs) under near-infrared (NIR) light irradiation. By combining Cy7-Naph with the Toll-like receptor agonist Resiquimod (R848), a synergistic treatment for bilateral tumor-bearing mice is achieved. This combination promotes dendritic cell (DC) maturation and increases the infiltration of cytotoxic T lymphocytes (CTLs), leading to significant inhibition of both primary and distant tumors.
Collapse
Affiliation(s)
- Shanshan Jin
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Yongkang Chen
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Fahui Li
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Ping Yan
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Guanhong Guo
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Guangzhao Xu
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Weiguo Song
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| | - Wenda Zhong
- School of Pharmacy, Shandong Second Medical University, Weifang 261053, People's Republic of China
| |
Collapse
|
6
|
Li Q, Chen Q, Xiao S, Wang S, Ge X, Wang Q, Zheng L, Wei Q, Du W, Shen W, Wu Y, Song J. A Salidroside-Based Radiosensitizer Regulates the Nrf2/ROS Pathway for X-Ray Activated Synergistic Cancer Precise Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2413226. [PMID: 40195850 DOI: 10.1002/adma.202413226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/26/2025] [Indexed: 04/09/2025]
Abstract
The hypoxic microenvironment and radioresistance of tumor cells, as well as the delay in efficacy evaluation, significantly limit the effect of clinical radiotherapy. Therefore, developing effective radiosensitizers with monitoring of tumor response is of great significance for precise radiotherapy. Herein, a novel radiosensitizer (term as: SCuFs) is developed, consisting of traditional Chinese medicine (TCM) compounds salidroside, Cu2+, and hydroxyl radical (•OH) activated second near-infrared window fluorescence (NIR-II FL) molecules, which make the radiosensitization effect and boosted chemodynamic therapy (CDT) efficacy. The overexpressed glutathione in the tumor induces the SCuFs dissociation, allowing deep penetration of the drug to the whole tumor region. After X-ray irradiation, salidroside inhibits the Nuclear factor erythroid 2-like 2 (Nrf2)protein expression and blocks cells in the G2/M phase with the highest radiosensitivity, which amplifies the reactive oxygen species (ROS) generation to exacerbate DNA damage, thus achieving radiosensitization. Meanwhile, the upregulated ROS provides sufficient chemical fuel for Cu+-mediated CDT to produce more •OH. NIR-II FL imaging can monitor the •OH changes during the therapy process, confirming the radiosensitization effect and CDT process related to •OH. This study not only achieves effective radiosensitization and cascaded ROS-mediated CDT efficacy, but also provides a useful tool for monitoring therapeutic efficacy, showing great prospects for clinical application.
Collapse
Affiliation(s)
- Qingqing Li
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qing Chen
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Shenggan Xiao
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Shuhan Wang
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoguang Ge
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qian Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Liting Zheng
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Qiaoqiao Wei
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wei Du
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Wenbin Shen
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Ying Wu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
7
|
Lin TC, Liu IJ, Chih HY, Tzang BS, Liang JA, Kuo CW, Hung CY, Hsu TC, Chiang WH. Photothermal-enhanced ROS storm by hyaluronic acid-conjugated nanocatalysts to amplify tumor-specific photo-chemodynamic therapy and immune response. Int J Biol Macromol 2025; 309:142975. [PMID: 40210075 DOI: 10.1016/j.ijbiomac.2025.142975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Integrating photodynamic therapy (PDT) and chemodynamic therapy (CDT) shows promising potential in tumor treatment. Nevertheless, the lack of specific tumor targeting, serious photobleaching, and poor photothermal effect of photosensitizers, the intracellular low Fenton reaction efficiency, and glutathione (GSH)-elicited reactive oxygen species (ROS) depletion profoundly restrict ROS-mediated cancer therapy. To enhance ROS generation with the assistance of photothermal therapy (PTT), the hyaluronic acid (HA)-decorated Fe-MIL-88B (MIL) nanocatalysts were fabricated for tumor-targeted delivery of photosensitizer IR820. The IR820@HA-coated MIL (IHM) nanocatalysts remarkably enhanced the photothermal conversion efficacy and singlet oxygen (1O2) production of IR820 and lowered IR820 photobleaching. The IHM nanocatalysts promoted the conversion of H2O2 into toxic ·OH upon thermo/acidity-enhanced Fe3+-mediated Fenton reaction and consumed GSH via Fe3+-elicited GSH oxidation. After being internalized by 4 T1 cancer cells via CD44-mediated endocytosis, the IHM nanocatalysts under irradiation of near-infrared (NIR) laser prominently produced hyperthermia and strong ROS storm, thereby causing apoptosis and ferroptosis via mitochondria damage and lipid peroxidation, and inducing immunogenic cell death (ICD). Through HA-mediated tumor targeting, the IHM nanocatalysts effectively accumulated in 4 T1 tumor and inhibited tumor growth and lung metastasis by PTT-enhanced PDT/CDT combined with ferroptosis and ICD-amplified antitumor immune response, showing great promise in future tumor treatment.
Collapse
Affiliation(s)
- Tzu-Chen Lin
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - I-Ju Liu
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Hsiang-Yun Chih
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Bor-Show Tzang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Immunology Research Center, Chung Shan Medical University, Taichung 402, Taiwan; Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ju-An Liang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Chia-Wei Kuo
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Chun-Yu Hung
- Department of Orthopedic Surgery, Jen-Ai Hospital, Taichung 402, Taiwan
| | - Tsai-Ching Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Immunology Research Center, Chung Shan Medical University, Taichung 402, Taiwan; Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan.
| | - Wen-Hsuan Chiang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan; i-Center for Advanced Science and Technology (iCAST), National Chung Hsing University, Taichung 402, Taiwan.
| |
Collapse
|
8
|
Zhu X, Zheng L, Zhao P, Gao L, Wang L, Liu J, Yang X, Wei H, Zhang M, Yan L, Lv H, Gong J, Yang JG, Wang Z. Fe/Cu Bimetallic Nanozyme Co-Assembled with 177Lu and Tanshinone for Quadruple-Synergistic Tumor-Specific Therapy. Adv Healthc Mater 2025; 14:e2402696. [PMID: 39543804 DOI: 10.1002/adhm.202402696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Indexed: 11/17/2024]
Abstract
The co-loading of radionuclides and small-molecule chemotherapeutic drugs as nanotheranostic platforms using nanozymes holds tremendous potential for imaging-guided synergistic therapy. This study presents such nanotheranostic platform (177Lu-MFeCu@Tan) via co-assembling 177Lu radionuclide and tanshinone (Tan) into Fe/Cu dual-metal nanozyme (MFeCu). This platform simultaneously enables single-photon emission computed tomography (SPECT) imaging and a quadruple-synergistic tumor therapy approach, including internal radioisotope therapy (RIT), catalysis therapy, chemotherapy, and MFeCu-mediated ferroptosis and cuproptosis therapy. In this platform, the MFeCu can catalyze excessive intracellular hydrogen peroxide (H2O2) to generate radical oxygen species (ROS) and deplete glutathione (GSH). The excess of H2O2 and GSH are main factors for radioresistance and chemoresistance, reducing them can enhance chemotherapy and RIT. The generated ROS and depleted GSH further induce mitochondrial dysfunction and promote the aggregation of lipoylated dihydrolipoamide S-acetyltransferase and lipid peroxidation, causing the enhance of ferroptosis and cuproptosis. The in vitro and in vivo results demonstrate that this quadruple-synergistic approach shows significant therapeutic efficacy to complete tumor eradication and reduced recurrence in vivo. In conclusion, this work presents a promising strategy for designing SPECT imaging-guided quadruple-synergistic therapy and highlights the feasibility of developing a self-assembled radionuclide and small molecule chemotherapy drugs nanotherapeutic platform for combined treatment of cancer.
Collapse
Affiliation(s)
- Xianyu Zhu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Lingling Zheng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Pengfei Zhao
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Lingli Gao
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing Tiantan Xili No. 1, Beijing, 100050, China
| | - Liang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Jun Liu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Xu Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Hanrui Wei
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing Tiantan Xili No. 1, Beijing, 100050, China
| | - Mingyu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Liang Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100049, China
| | - Han Lv
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Jianhua Gong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing Tiantan Xili No. 1, Beijing, 100050, China
| | - Ji Gang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Rd., Xicheng District, Beijing, 100050, China
| |
Collapse
|
9
|
Jang D, Choi H, Lee J, Chun Y, Heo YH, Lee LP, Ahn DJ, Shin IS, Kim DH, Seo YH, Kim S. Inflamed Tissue-Targeting Polyphenol-Condensed Antioxidant Nanoparticles with Therapeutic Potential. Adv Healthc Mater 2025; 14:e2500495. [PMID: 40033968 DOI: 10.1002/adhm.202500495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Inflammation is essential for pathogen eradication and tissue repair; However, chronic inflammation can bring on multi-organ dysfunction due to an overproduction of reactive oxygen species (ROS). Among various anti-inflammatory agents, polyphenol-based nanotherapeutics offer potential advantages, including enhanced stability, targeted delivery, multiple therapeutic functions, and personalized therapy tailored to the severity. Despite these advantages, the development of biocompatible nanomedicines capable of selective accumulation in inflamed tissues and efficient inhibition of ROS-induced inflammatory signaling pathways remains a considerable challenge. In this study, a novel anti-inflammatory nanotherapeutic is engineered through the temperature-dependent condensation of polyphenolic catechin facilitated by hydrothermal reactions. The resulting catechin-condensed nanotherapeutic (CCN150), synthesized at a relatively low temperature, retains physicochemical and functional properties akin to its precursor, catechin, but with a marked enhancement in water solubility. CCN150 protects cells from oxidative stress by eliminating intracellular ROS and augmenting antioxidant enzymes. In vivo studies reveal that intravenously administered CCN150 predominantly accumulates in inflamed tissues, with minimal distribution to healthy regions. Furthermore, CCN150 effectively reduces systemic inflammation in mouse models by disrupting the cycles of ROS instigated by a pro-inflammatory oxidative milieu. Exhibiting negligible toxicity, CCN150 holds substantial promise for extensive therapeutic applications in the treatment of various ROS-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Dohyub Jang
- Department of Biomicrosystem Technology, Korea University, Seoul, 02792, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Honghwan Choi
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Juhyang Lee
- Biosensor Group, Korea Institute of Science and Technology Europe, Campus E7.1, 66123, Saarbrücken, Germany
| | - Yousun Chun
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoon-Ho Heo
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Luke P Lee
- Harvard Institute of Medicine, Harvard Medical School Brigham and Women's Hospital, Harvard University, Boston, MA, 02115, USA
| | - Dong June Ahn
- Department of Biomicrosystem Technology, Korea University, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ik-Soo Shin
- Department of Chemistry, College of Natural Science, Soongsil University, 3, Seoul, 15674, Republic of Korea
| | - Dong Ha Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
- College of Medicine, Ewha Womans University, 25, Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
- Basic Sciences Research Institute (Priority Research Institute), Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
- Nanobio·Energy Materials Center (National Research Facilities and Equipment Center), Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Young Hun Seo
- Biosensor Group, Korea Institute of Science and Technology Europe, Campus E7.1, 66123, Saarbrücken, Germany
| | - Sehoon Kim
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
10
|
Zhuang J, Hai Y, Lu X, Sun B, Fan R, Zhang B, Wang W, Han B, Luo L, Yang L, Zhang C, Zhao M, Wei G. A Self-Assembled Metabolic Regulator Reprograms Macrophages to Combat Cytokine Storm and Boost Sepsis Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2025; 8:0663. [PMID: 40171016 PMCID: PMC11959697 DOI: 10.34133/research.0663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/03/2025]
Abstract
Sepsis, a life-threatening inflammatory disorder characterized by multiorgan failure, arises from a dysregulated immune response to infection. Modulating macrophage polarization has emerged as a promising strategy to control sepsis-associated inflammation. The endogenous metabolite itaconate has shown anti-inflammatory potential by suppressing the stimulator of interferon genes (STING) pathway, but its efficacy is inhibited by hyperactive glycolysis, which sustains macrophage overactivation. Here, we revealed a critical crosstalk between the itaconate-STING axis and glycolysis in macrophage-mediated inflammation. Building on this interplay, we developed a novel nanoparticle LDO (lonidamine disulfide 4-octyl-itaconate), a self-assembled metabolic regulator integrating an itaconate derivative with the glycolysis inhibitor Lonidamine. By concurrently targeting glycolysis and STING pathways, LDO reprograms macrophages to restore balanced polarization. In sepsis models, LDO effectively attenuates CCL2-driven cytokine storms, alleviates acute lung injury, and significantly enhances survival via metabolic reprogramming. This study offers a cytokine-regulatory strategy rooted in immunometabolism, providing a foundation for the translational development of immune metabolite-based sepsis therapies.
Collapse
Affiliation(s)
- Junyan Zhuang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Yongrui Hai
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Xintong Lu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Borui Sun
- Department of Anesthesiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Renming Fan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Bingjie Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Wenhui Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Bingxue Han
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Li Luo
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi’an 710038, Shaanxi, China
| | - Le Yang
- Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi’an 710038, Shaanxi, China
| | - Chun Zhang
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Minggao Zhao
- Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi’an 710038, Shaanxi, China
| | - Gaofei Wei
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| |
Collapse
|
11
|
Khulood MT, Jijith US, Naseef PP, Kallungal SM, Geetha VS, Pramod K. Advances in metal-organic framework-based drug delivery systems. Int J Pharm 2025; 673:125380. [PMID: 39988215 DOI: 10.1016/j.ijpharm.2025.125380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Metal-organic frameworks (MOFs) are emerging crystalline porous materials with significant potential in biomedical applications, particularly as drug delivery systems (DDS). MOFs, composed of metal ions or clusters linked by organic ligands, feature large surface areas, adjustable pores, and diverse functionalities. This review comprehensively examines MOFs as advanced DDS, detailing their structures, synthesis, and drug loading mechanisms. We highlight high drug loading capacity and controlled release capabilities of MOF. Developments of design strategies for MOF-based DDS, namely, surface functionalization for targeted delivery and stimuli-responsive MOFs for controlled release, have been discussed and explored. The use of MOFs for delivering therapeutic agents such as small molecules, peptides, proteins, nucleic acids, and cancer drugs is discussed. Challenges addressed include stability, degradation in biological environments, potential toxicity, and scalability. Advances in hybrid MOF-based DDS, integrating MOFs with polymers, lipids, or nanoparticles for improved delivery, are also examined.
Collapse
Affiliation(s)
- M T Khulood
- College of Pharmaceutical Sciences, Government Medical College, Kozhikode 673008 Kerala, India; Kerala University of Health Sciences, Medical College P.O., Thrissur 680596 Kerala, India
| | - U S Jijith
- College of Pharmaceutical Sciences, Government Medical College, Kozhikode 673008 Kerala, India
| | - P P Naseef
- Department of Pharmaceutics, Moulana College of Pharmacy, Perinthalmanna, Malappuram 679321 Kerala, India
| | - Sirajudheen M Kallungal
- Department of Pharmaceutics, Jamia Salafiya Pharmacy College, Pulikkal, Malappuram 673637 Kerala, India
| | - V S Geetha
- College of Pharmaceutical Sciences, Government Medical College, Kozhikode 673008 Kerala, India
| | - K Pramod
- College of Pharmaceutical Sciences, Government Medical College, Kozhikode 673008 Kerala, India.
| |
Collapse
|
12
|
Xiao J, Ge Z, Tan X, Liu Z, Zhang Y, Xiao S, Yi R, Hu Y, Hu W, Chu H, Chen J. Biodegradable Covalent Beta-Cyclodextrin Nanocages for Acidic and Reductive-Responsive Drug Delivery in Enhanced Tumor Therapy. Biotechnol J 2025; 20:e70006. [PMID: 40123422 DOI: 10.1002/biot.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/19/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Traditional beta-cyclodextrin (beta-CD) in biomedical applications faces challenges due to its inherent physical and biochemical limitations. One of the most effective strategies to enhance the properties of beta-CD for drug delivery is the synthesis of supramolecular polycyclodextrins. In this study, we designed a novel beta-CD nanocage-like structure for drug delivery, incorporating imine and disulfide bonds through Schiff base reactions. Aldehyde group-functionalized beta-CD units were used to construct the main backbone of the nanocage, forming dual-dynamic covalent bonds. The chemical structure of the beta-CD nanocage was confirmed using ¹H nuclear magnetic resonance (¹H NMR) and Fourier transform infrared spectroscopy (FTIR). Additionally, atomic force microscopy (AFM) and dynamic light scattering (DLS) revealed that varying amounts of beta-CD crosslinked with cystamine resulted in nanocages approximately 200 nm in size. In vitro drug release experiments demonstrated that doxorubicin (DOX)-loaded beta-CD nanocages exhibited accelerated DOX release in acidic and reductive environments compared to normal physiological conditions, owing to the pH-sensitive imine bond and the glutathione (GSH)-cleavable disulfide bond. The DOX-loaded beta-CD nanocages showed exceptional tumor-killing effects, particularly in acid/reduction-enhanced tumor cells. Both cellular fluorescence imaging and flow cytometry confirmed the potential of the beta-CD nanocages for acid/reduction-specific drug release. Consequently, this precision medicine model using imine/disulfide-linked beta-CD nanocage structures as acidity/reduction-sensitive drug carriers promises to improve oncotherapy through more targeted drug delivery and release, supporting individualized treatment approaches.
Collapse
Affiliation(s)
- Jingyi Xiao
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Zan Ge
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Xiaowei Tan
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Ziyi Liu
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Yafang Zhang
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Shufen Xiao
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Rongyuan Yi
- Fourth Department of Gynecologic Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People's Republic of China
| | - Ye Hu
- Nanjing Institute for Food and Drug Control, Nanjing, Jiangsu, People's Republic of China
| | - Wenyan Hu
- Nanjing Institute for Food and Drug Control, Nanjing, Jiangsu, People's Republic of China
| | - Hui Chu
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| | - Jian Chen
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, Hunan, People's Republic of China
| |
Collapse
|
13
|
Yu Z, Lepoitevin M, Serre C. Iron-MOFs for Biomedical Applications. Adv Healthc Mater 2025; 14:e2402630. [PMID: 39388416 PMCID: PMC11937880 DOI: 10.1002/adhm.202402630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/29/2024] [Indexed: 10/12/2024]
Abstract
Over the past two decades, iron-based metal-organic frameworks (Fe-MOFs) have attracted significant research interest in biomedicine due to their low toxicity, tunable degradability, substantial drug loading capacity, versatile structures, and multimodal functionalities. Despite their great potential, the transition of Fe-MOFs-based composites from laboratory research to clinical products remains challenging. This review evaluates the key properties that distinguish Fe-MOFs from other MOFs and highlights recent advances in synthesis routes, surface engineering, and shaping technologies. In particular, it focuses on their applications in biosensing, antimicrobial, and anticancer therapies. In addition, the review emphasizes the need to develop scalable, environmentally friendly, and cost-effective production methods for additional Fe-MOFs to meet the specific requirements of various biomedical applications. Despite the ability of Fe-MOFs-based composites to combine therapies, significant hurdles still remain, including the need for a deeper understanding of their therapeutic mechanisms and potential risks of resistance and overdose. Systematically addressing these challenges could significantly enhance the prospects of Fe-MOFs in biomedicine and potentially facilitate their integration into mainstream clinical practice.
Collapse
Affiliation(s)
- Zhihao Yu
- Institut des Matériaux Poreux de ParisENSESPCI ParisCNRSPSL UniversityParisFrance
| | - Mathilde Lepoitevin
- Institut des Matériaux Poreux de ParisENSESPCI ParisCNRSPSL UniversityParisFrance
| | - Christian Serre
- Institut des Matériaux Poreux de ParisENSESPCI ParisCNRSPSL UniversityParisFrance
| |
Collapse
|
14
|
Dong J, Ding J, Luo S, Li R, Wang Y, Xiao B, Pei Y, Chen X, Sun W, Pei Z. Remodeling tumor microenvironment using prodrug nMOFs for synergistic cancer therapy. J Nanobiotechnology 2025; 23:123. [PMID: 39972341 PMCID: PMC11837371 DOI: 10.1186/s12951-025-03202-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Metal-organic frameworks (MOFs) hold tremendous potential in cancer therapy due to their remarkable structural and functional adaptability, enabling them to serve as nanocarriers for biopharmaceuticals and nanoreactors for organizing cascade bioreactions. Nevertheless, MOFs are predominantly utilized as biologically inactive carriers in most cases. Developing nanoscale prodrug MOFs suitable for biomedical applications remains a huge challenge. In this study, we have designed a novel prodrug nano-MOFs (nMOFs, named DCCMH) using metformin (Met) and α-cyano-4-hydroxycinnamic acid (CHCA) as ligands for coordination self-assembly with CuCl2, followed by loading of doxorubicin (DOX) and surface modification with hyaluronic acid (HA). Upon internalization by cancer cells, DCCMH releases Cu2+/+, CHCA, Met, and DOX in response to high levels of glutathione (GSH) and hydrogen peroxide (H2O2) within the tumor microenvironment (TME); Cu+ catalyzes the conversion of H2O2 to ·OH via the Fenton reaction while it was oxidized to Cu2+, which was subsequently further de-consumed of GSH; CHCA induces a further decrease in intracellular pH and promotes Fenton reactions by inhibiting lactate efflux; Met up-regulates tyrosine kinase activity and enhances the chemotherapy of DOX. With the ability to synergistically combine chemo/chemodynamic therapy (CT/CDT) and remodel the TME, the DCCMH NPs inhibit murine hepatoma effectively. This study presents a feasible strategy for fabricating prodrug nMOFs which are capable of remodeling TME to improve efficacy through synergistic cancer therapy.
Collapse
Affiliation(s)
- Junliang Dong
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Xiaoshan District, No.733 Jianshe San Road, Hangzhou, 311200, Zhejiang, People's Republic of China
| | - Jindong Ding
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Shifan Luo
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Xiaoshan District, No.733 Jianshe San Road, Hangzhou, 311200, Zhejiang, People's Republic of China
| | - Ruoshui Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Xiaoshan District, No.733 Jianshe San Road, Hangzhou, 311200, Zhejiang, People's Republic of China
| | - Yi Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Xiaoshan District, No.733 Jianshe San Road, Hangzhou, 311200, Zhejiang, People's Republic of China
| | - Bing Xiao
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Xiaoshan District, No.733 Jianshe San Road, Hangzhou, 311200, Zhejiang, People's Republic of China
| | - Yuxin Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore.
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 138667, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore.
| | - Wenjing Sun
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Xiaoshan District, No.733 Jianshe San Road, Hangzhou, 311200, Zhejiang, People's Republic of China.
| | - Zhichao Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
| |
Collapse
|
15
|
Yang N, He Z, Lang T. Drug Delivery Systems Based on Metal-Organic Frameworks for Tumor Immunotherapy. Pharmaceutics 2025; 17:225. [PMID: 40006592 PMCID: PMC11859595 DOI: 10.3390/pharmaceutics17020225] [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: 01/08/2025] [Revised: 02/01/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Metal-organic frameworks (MOFs) are a class of inorganic-organic hybrid nanoparticles formed by the coordination of metal ions/clusters and organic ligands. Due to their high porosities, large surface areas, adjustable structures, and responsiveness to light/sound, etc., MOFs have shown great clinical potential in the field of tumor therapy. Tumor immunotherapy exerts antitumor effects through reshaping tumor immune microenvironment, showing significant preclinical and clinical advantages. Based on the mechanisms of immunity activation, the tumor immunotherapy agents can be divided into chemotherapeutic agents, immunomodulators, enzymes, tumor vaccines and oligonucleotide drugs, etc. Herein, we review the MOFs-based drug delivery systems for tumor immunotherapy. The classification of MOFs, followed by their antitumor immunity activation mechanisms, are first introduced. Drug delivery systems based on MOFs with different immunotherapy agents are also summarized, especially the synergetic immunity activation mechanisms triggered by MOFs and their loadings. Furthermore, the merits and drawbacks of MOFs and the potential strategies for MOFs to promote their clinical applications are discussed.
Collapse
Affiliation(s)
| | | | - Tianqun Lang
- Lin Gang Laboratory, Shanghai 200031, China; (N.Y.); (Z.H.)
| |
Collapse
|
16
|
Xu Y, Reheman A, Feng W. Recent research progress on metal ions and metal-based nanomaterials in tumor therapy. Front Bioeng Biotechnol 2025; 13:1550089. [PMID: 39991139 PMCID: PMC11842396 DOI: 10.3389/fbioe.2025.1550089] [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: 12/22/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Tumors, as a disease that seriously threatens human health, have always been a major challenge in the field of medicine. Currently, the main methods of tumor treatment include surgery, radiotherapy, chemotherapy, etc., but these traditional treatment methods often have certain limitations. In addition, tumor recurrence and metastasis are also difficult problems faced in clinical treatment. In this context, the importance of metal-based nanomaterials in tumor therapy is increasingly highlighted. Metal-based nanomaterials possess unique physical, chemical, and biological properties, providing new ideas and methods for tumor treatment. Metal-based nanomaterials can achieve targeted therapy for tumors through various mechanisms, reducing damage to normal tissues; they can also serve as drug carriers, improving the stability and bioavailability of drugs; at the same time, some metal-based nanomaterials also have photothermal, photodynamic, and other characteristics, which can be used for phototherapy of tumors. This review examines the latest advances in the application of metal-based nanomaterials in tumor therapy within past 5 years, and presents prospective insights into the future applications.
Collapse
Affiliation(s)
- Yongcheng Xu
- The Second School of Clinical Medicine, Shenyang Medical College, Shenyang, China
| | - Aikebaier Reheman
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, Fujian, China
| | - Wenhua Feng
- Department of Human Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Liaoning Province Key Laboratory for Phenomics of Human Ethnic Specificity and Critical Illness, Shenyang, China
- Shenyang Key Laboratory for Phenomics, Shenyang Medical College, Shenyang, China
| |
Collapse
|
17
|
Li Z, Xi Z, Fan C, Xi X, Zhou Y, Zhao M, Xu L. Nanomaterials evoke pyroptosis boosting cancer immunotherapy. Acta Pharm Sin B 2025; 15:852-875. [PMID: 40177577 PMCID: PMC11959974 DOI: 10.1016/j.apsb.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 04/05/2025] Open
Abstract
Cancer immunotherapy is currently a very promising therapeutic strategy for treating tumors. However, its effectiveness is restricted by insufficient antigenicity and an immunosuppressive tumor microenvironment (ITME). Pyroptosis, a unique form of programmed cell death (PCD), causes cells to swell and rupture, releasing pro-inflammatory factors that can enhance immunogenicity and remodel the ITME. Nanomaterials, with their distinct advantages and different techniques, are increasingly popular, and nanomaterial-based delivery systems demonstrate significant potential to potentiate, enable, and augment pyroptosis. This review summarizes and discusses the emerging field of nanomaterials-induced pyroptosis, focusing on the mechanisms of nanomaterials-induced pyroptosis pathways and strategies to activate or enhance specific pyroptosis. Additionally, we provide perspectives on the development of this field, aiming to accelerate its further clinical transition.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ziyue Xi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chuanyong Fan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinran Xi
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yao Zhou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ming Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lu Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
18
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [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: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
19
|
Zhao C, Song W, Wang J, Tang X, Jiang Z. Immunoadjuvant-functionalized metal-organic frameworks: synthesis and applications in tumor immune modulation. Chem Commun (Camb) 2025; 61:1962-1977. [PMID: 39774558 DOI: 10.1039/d4cc06510g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy, which leverages the body's immune system to recognize and attack cancer cells, has made significant progress, particularly in the treatment of metastatic tumors. However, challenges such as drug stability and off-target effects still limit its clinical success. To address these issues, metal-organic frameworks (MOFs) have emerged as promising nanocarriers in cancer immunotherapy. MOFs have unique porous structure, excellent drug loading capacity, and tunable surface modification properties. MOFs not only enhance drug delivery efficiency but also allow for precise control of drug release. They reduce off-target effects and significantly improve targeting and therapy efficacy. As research deepens, MOFs' effectiveness as drug carriers has been refined. When combined with immunoadjuvants or anticancer drugs, MOFs further stimulate the immune response. This improves the specificity of immune attacks on tumors. This review provides a comprehensive overview of the applications of MOFs in cancer immunotherapy. It focuses on synthesis, drug loading strategies, and surface modifications. It also analyzes their role in enhancing immunotherapy effectiveness. By integrating current research, we aim to provide insights for the future development of immunoadjuvant-functionalized MOFs, accelerating their clinical application for safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Chen Zhao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Weihua Song
- Xuanwu Hospital Capital Medical University, Beijing, 100037, China
| | - Jianing Wang
- School of Medical Technology, the Qiushi College, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoying Tang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Zhenqi Jiang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
20
|
Wang C, Li J, Jiang X, Ma X, Zhen W, Tillman L, Weichselbaum RR, Lin W. Bifunctional Metal-Organic Framework Synergistically Enhances Radiotherapy and Activates STING for Potent Cancer Radio-Immunotherapy. Angew Chem Int Ed Engl 2025; 64:e202417027. [PMID: 39375150 DOI: 10.1002/anie.202417027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/09/2024]
Abstract
The activation of the stimulator of interferon genes (STING) protein by cyclic dinucleotide metabolites plays a critical role in antitumor immunity. However, synthetic STING agonists like 4-(5,6-dimethoxybenzo[b]thiophen-2-yl)-4-oxobutanoic acid (MSA-2) exhibit suboptimal pharmacokinetics and fail to sustain STING activation in tumors for effective antitumor responses. Here, we report the design of MOF/MSA-2, a bifunctional MSA-2 conjugated nanoscale metal-organic framework (MOF) based on Hf6 secondary building units (SBUs) and hexakis(4'-carboxy[1,1'-biphenyl]-4-yl)benzene bridging ligands, for potent cancer radio-immunotherapy. By leveraging the high-Z properties of the Hf6 SBUs, the MOF enhances the therapeutic effect of X-ray radiation and elicits potent immune stimulation in the tumor microenvironment. MOF/MSA-2 further enhances radiotherapeutic effects of X-rays by enabling sustained STING activation and promoting the infiltration and activation of immune cells in the tumors. MOF/MSA-2 plus low-dose X-ray irradiation elicits strong STING activation and potent tumor regression, and when combined with an immune checkpoint inhibitor, effectively suppresses both primary and distant tumors through systemic immune activation.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Jinhong Li
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
| | - Xiaomin Jiang
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Xin Ma
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
| | - Wenyao Zhen
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Langston Tillman
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, 929 E 57th St, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, 5758 S Maryland Ave, Chicago, IL 60637, USA
| |
Collapse
|
21
|
Wang L, Zhang X, He L, Wei Y, Zhang Y, Wu A, Li J. Iron-Based Nanomaterials for Modulating Tumor Microenvironment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70001. [PMID: 39788569 DOI: 10.1002/wnan.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/29/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Iron-based nanomaterials (IBNMs) have been widely applied in biomedicine applications including magnetic resonance imaging, targeted drug delivery, tumor therapy, and so forth, due to their unique magnetism, excellent biocompatibility, and diverse modalities. Further research on its enormous biomedical potential is still ongoing, and its new features are constantly being tapped and demonstrated. Among them, various types of IBNMs have demonstrated significant cancer therapy capabilities by regulating the tumor microenvironment (TME). In this review, a variety of IBNMs including iron oxide-based nanomaterials (IONMs), iron-based complex conjugates (ICCs), and iron-based single iron atom nanomaterials (ISANMs) will be introduced, and their advantages in regulating TME would also be emphasized. Besides, the recent progress of IBNMs for cancer diagnosis and treatment through the strategy of modulating TME will be summarized, including overcoming hypoxia, modulating acidity, decreasing redox species, and immunoregulation. Finally, the challenges and opportunities in this field are briefly discussed. This review is expected to contribute to the future design and development of next-generation TME-modulate IBNMs for cancer treatment.
Collapse
Affiliation(s)
- Le Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Xiaoting Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Lulu He
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yuanyuan Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Yujie Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Aiguo Wu
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| | - Juan Li
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, China
| |
Collapse
|
22
|
Zeng Q, Zhang S, Leng N, Xing Y. Advancing tumor vaccines: Overcoming TME challenges, delivery strategies, and biomaterial-based vaccine for enhanced immunotherapy. Crit Rev Oncol Hematol 2025; 205:104576. [PMID: 39581246 DOI: 10.1016/j.critrevonc.2024.104576] [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: 10/08/2024] [Revised: 11/03/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024] Open
Abstract
Tumor vaccines, as an immunotherapeutic approach, harness the body's immune cells to provoke antitumor responses, which have shown promising efficacy in clinical settings. However, the immunosuppressive tumor microenvironment (TME) and the ineffective vaccine delivery systems hinder the progression of many vaccines beyond phase II trials. This article begins with a comprehensive review of the complex interactions between tumor vaccines and TME, summarizing the current state of vaccine clinical research. Subsequently, we review recent advancements in targeted vaccine delivery systems and explore biomaterial-based tumor vaccines as a strategy to improve the efficacy of both delivery systems and treatment. Finally, we have presented our perspectives on tumor vaccine development, aiming to advance the field towards the creation of more effective tumor vaccines.
Collapse
Affiliation(s)
- Qingsong Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shibo Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ning Leng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
23
|
Hu Y, Yu Q, Li X, Wang J, Guo L, Huang L, Gao W. Nanoformula Design for Inducing Non-Apoptotic Cell Death Regulation: A Powerful Booster for Cancer Immunotherapy. Adv Healthc Mater 2025; 14:e2403493. [PMID: 39632361 DOI: 10.1002/adhm.202403493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Indexed: 12/07/2024]
Abstract
Cancer treatment has witnessed revolutionary advancements marked by the emergence of immunotherapy, specifically immune checkpoint blockade (ICB). However, the inherent low immunogenicity of tumor cells and the intricate immunosuppressive network within the tumor microenvironment (TME) pose significant challenges to the further development of immunotherapy. Nanotechnology has ushered in unprecedented opportunities and vast prospects for tumor immunotherapy. Nevertheless, traditional nano-formulations often rely on inducing apoptosis to kill cancer cells, which encounters the issue of immune silencing, hindering effective tumor immune activation. The non-apoptotic modes of regulated cell death (RCD), including pyroptosis, ferroptosis, autophagy, necroptosis, and cuproptosis, have gradually garnered attention. These non-apoptotic cell death pathways can induce effective immunogenic cell death (ICD), enhancing cancer immunotherapy. This review comprehensively explores advanced nano-formulation design strategies and their applications in enhancing cancer immunotherapy by promoting non-apoptotic RCD in recent years. It also discusses the potential advantages of these strategies in inducing tumor-specific non-apoptotic RCD. By deeply understanding the significance of non-apoptotic RCD in synergistic cancer immunotherapy, this article provides valuable insights for developing more advanced nano-delivery systems that can robustly induce highly immunogenic non-apoptotic modes, offering novel research and development avenues to address the clinical challenges encountered by immunotherapy represented by ICB.
Collapse
Affiliation(s)
- Yi Hu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Qing Yu
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Xia Li
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Juan Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, P.R. China
| |
Collapse
|
24
|
Liu S, Wu X, Li L, Wang J, Liu W, Yuan SJ, Dai XH. Modulation of the Atomic Spacing of Electrocatalytic for Boosting Reactive Oxygen Species Production to Precise Hepatocellular Carcinoma Cell Apoptosis. ACS NANO 2024; 18:34815-34828. [PMID: 39666311 DOI: 10.1021/acsnano.4c11860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Promoting tumor cell apoptosis through the catalytic regulation of reactive oxygen species (ROS) is an ideal therapeutic option for cancer. However, a stable and controllable exogenous source of ROS is still lacking. Efficient and controllable electrocatalysis has shown tremendous potential for cancer treatment, but its key challenge lies in achieving precise, efficient, and controllable electrocatalytic ROS production at the tumor site. This study describes an electrocatalytic treatment technique for hepatocellular carcinoma (HCC) based on traditional Chinese acupuncture. By attaching a biocompatible electrocatalyst NiO-P700 with optimal atomic spacing to the surface of silver acupuncture needles, a high-concentration ROS microenvironment was generated around tumor cells via ORRs when the needles were electrified. This induction led to the accumulation of inflammatory factors (IL-1β, IL-6, and TNF-α) and macrophage infiltration, accelerating tumor cell apoptosis and necrosis. Both in vitro and in vivo experiments demonstrated that the rate of ROS production can be rapidly controlled by adjusting voltage and current. Importantly, the high concentration of ROS can be safely and effectively confined to the lesion site without affecting the entire body. Our study attempted to integrate electrocatalysis and acupuncture in HCC treatment, successfully regulating NiO-P atomic spacing and enhancing ORR performance, thereby presenting a safe and reliable perspective for HCC therapy.
Collapse
Affiliation(s)
- Shiyu Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Xuan Wu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Central Laboratory and Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200070, China
| | - Lei Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Jingjing Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Weiwei Liu
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Shi-Jie Yuan
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China
| | - Xiao-Hu Dai
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
- Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China
| |
Collapse
|
25
|
Xi ZY, Fan CY, Zhu S, Nie GY, Xi XR, Jiang YY, Zhou Y, Mei YH, Xu L. PAFerroptosis Combined with Metabolic Disturbance of Mito by p52-ZER6 for Enhanced Cancer Immunotherapy induced by Nano-Bacilliform-Enzyme. Adv Healthc Mater 2024; 13:e2402314. [PMID: 39171764 DOI: 10.1002/adhm.202402314] [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: 06/25/2024] [Indexed: 08/23/2024]
Abstract
The confused gene expressions and molecular mechanisms for mitochondrial dysfunction of traditional nanoenzymes is a challenge for tumor therapy. Herein, a nano-bacilliform-enzyme obtains the ability to inhibit p52-ZER6 signal pathway, regulate the genes related to mitochondrial metabolism, and possess the GOx/CAT/POD-like property. NBE acquires catalytic activity from the electronic energy transition. The tannin of NBE as a mitochondrial (Mito)-targeting guide overloads MitoROS, and then metabolic disorder and lipid peroxidation of Mito membrane occurs, thus leading to a novel death pathway called PAFerroptosis (pyroptosis, apoptosis, and Ferroptosis). Simultaneously, in order to refrain from mitophagy, hydroxychloroquine is mixed with NBE to form a combo with strength pyroptosis. As a result, NBE/combo improves the PAFerroptosis obviously by activation of CD8+T cells and inactivation of MDSC cells, up-regulating expression of caspase-3 signal pathway, intercepting DHODH pathway to arrive excellent antitumor effect (93%). Therefore, this study establishes a rational nanoenzyme for mitochondrial dysfunction without mitophagy for effective antitumor therapy.
Collapse
Affiliation(s)
- Zi-Yue Xi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chuan-Yong Fan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Shuang Zhu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Gan-Yu Nie
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xin-Ran Xi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ying-Ying Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yao Zhou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yi-Hua Mei
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
26
|
Li J, Yang J, Jiang S, Tian Y, Zhang Y, Xu L, Hu B, Shi H, Li Z, Ran G, Huang Y, Ruan S. Targeted reprogramming of tumor-associated macrophages for overcoming glioblastoma resistance to chemotherapy and immunotherapy. Biomaterials 2024; 311:122708. [PMID: 39047538 DOI: 10.1016/j.biomaterials.2024.122708] [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: 01/04/2024] [Revised: 06/07/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
The resistance of glioblastoma multiforme (GBM) to standard chemotherapy is primarily attributed to the existence of tumor-associated macrophages (TAMs) in the GBM microenvironment, particularly the anti-inflammatory M2 phenotype. Targeted modulation of M2-TAMs is emerging as a promising strategy to enhance chemotherapeutic efficacy. However, combination TAM-targeted therapy with chemotherapy faces substantial challenges, notably in terms of delivery efficiency and targeting specificity. In this study, we designed a pH-responsive hierarchical brain-targeting micelleplex loaded with temozolomide (TMZ) and resiquimod (R848) for combination chemo-immunotherapy against GBM. This delivery system, termed PCPA&PPM@TR, features a primary Angiopep-2 decoration on the outer layer via a pH-cleavable linker and a secondary mannose analogue (MAN) on the middle layer. This pH-responsive hierarchical targeting strategy enables effective BBB permeability while simultaneous GBM- and TAMs-targeting delivery. GBM-targeted delivery of TMZ induces alkylation and triggers an anti-GBM immune response. Concurrently, TAM-targeted delivery of R848 reprograms their phenotype from M2 to pro-inflammatory M1, thereby diminishing GBM resistance to TMZ and amplifying the immune response. In vivo studies demonstrated that targeted modulation of TAMs using PCPA&PPM@TR significantly enhanced anti-GBM efficacy. In summary, this study proposes a promising brain-targeting delivery system for the targeted modulation of TAMs to combat GBM.
Collapse
Affiliation(s)
- Jianan Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Jun Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Shaoping Jiang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Yunxin Tian
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuquan Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Lin Xu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Huiping Shi
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhaohan Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China
| | - Guangyao Ran
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China.
| | - Shaobo Ruan
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
27
|
Wang D, Yao H, Ye J, Gao Y, Cong H, Yu B. Metal-Organic Frameworks (MOFs): Classification, Synthesis, Modification, and Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404350. [PMID: 39149999 DOI: 10.1002/smll.202404350] [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: 05/29/2024] [Revised: 08/02/2024] [Indexed: 08/17/2024]
Abstract
Metal-organic frameworks (MOFs) are a new variety of solid crystalline porous functional materials. As an extension of inorganic porous materials, it has made important progress in preparation and application. MOFs are widely used in various fields such as gas adsorption storage, drug delivery, sensing, and biological imaging due to their high specific surface area, porosity, adjustable pore size, abundant active sites, and functional modification by introducing groups. In this paper, the types of MOFs are classified, and the synthesis methods and functional modification mechanisms of MOFs materials are summarized. Finally, the application prospects and challenges of metal-organic framework materials in the biomedical field are discussed, hoping to promote their application in multidisciplinary fields.
Collapse
Affiliation(s)
- Dayang Wang
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Huanchen Yao
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Jiashuo Ye
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Yan Gao
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
- School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Life Sciences, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
28
|
Zhang Y, Zhang C, Qian W, Lei F, Chen Z, Wu X, Lin Y, Wang F. Recent advances in MOF-based nanozymes: Synthesis, activities, and bioapplications. Biosens Bioelectron 2024; 263:116593. [PMID: 39059178 DOI: 10.1016/j.bios.2024.116593] [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: 02/23/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Nanozymes have garnered considerable research interest for their unique capacity to bridge nanotechnology and biology. Current studies predominantly concentrate on exploring nanozymes with diverse catalytic activities and their potential applications across various disciplines. Among them, nanoscale metal-organic frameworks (MOFs) are promising nanomaterials for constructing nanozymes. In this review, we firstly introduce the general construction strategies for MOF-based nanozymes. In addition, we also classify the MOF-based nanozymes in detail based on their catalytic performance. Thirdly, the recent research progress of MOF-based nanozymes in the field of biosensing, cancer therapy, antibacterial infection, and antioxidation are also comprehensively reviewed. Finally, we discuss the current challenges and future perspectives of MOF-based nanozymes, with the aim of assisting in their construction and maximizing their potential in bioapplications. It is hoped that we could provide scientists in materials science and biomedical research with valuable and comprehensive information, fostering advancements in interdisciplinary fields.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China.
| | - Chengfeng Zhang
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China
| | - Wanlong Qian
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China
| | - Fang Lei
- School of Public Health, Nantong University, Nantong, 226019, China
| | - Zhongping Chen
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China
| | - Xiaomei Wu
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China
| | - Youhui Lin
- Department of Physics, Xiamen University, Xiamen, 361005, China.
| | - Faming Wang
- School of Public Health, Nantong University, Nantong, 226019, China.
| |
Collapse
|
29
|
Yang X, Li C, Ge M, Li X, Zhao W, Guo H, Nie H, Liu J. Mn(II)-Aloe-Emodin Nanoscale Coordination Polymer Enhances Ferroptosis by Synergistically Enhancing Reactive Oxygen Species Generation via the Nrf2-GPX4 Axis. Adv Healthc Mater 2024; 13:e2400474. [PMID: 38875525 DOI: 10.1002/adhm.202400474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Ferroptosis induction is particularly promising for cancer therapy when the apoptosis pathway is compromised. Current strategies in nanomedicine for inducing ferroptosis primarily focus on promoting the accumulation of reactive oxygen species (ROS). However, the presence of intracellular antioxidants, such as nuclear factor erythroid 2-related factor 2 (Nrf2), can limit the effectiveness of such therapy by activating detoxification systems and eliminating ROS. To overcome this challenge, we developed a synergistic ferroptosis-inducing agent by modifying manganese (Mn2+)-1,8-dihydroxy-3-hydroxymethyl-anthraquinone (aloe-emodin, AE) with polyvinyl pyrrolidone (PVP) to create nanoparticles (MAP NPs). In the tumor microenvironment, these NPs degraded and released AE and Mn(II), facilitating the generation of ROS and Mn(IV) through a Fenton-like reaction between hydrogen peroxide (H2O2) and Mn(II). Mn(IV) subsequently interacts with glutathione (GSH) to induce a cyclic catalytic effect, and the depletion of GSH diminished the activation of glutathione-dependent peroxidase 4 (GPX4). Furthermore, AE inhibits the activity of Nrf2 and depleted GSH, thereby synergistically enhancing antitumor efficacy. Here it is demonstrated that MAP NPs effectively generate a robust ROS storm within tumor cells, suggesting that high-performance ferroptosis therapy is effective. Additionally, the inclusion of Mn(II) in the MAP NPs enables real-time monitoring of therapeutic efficacy via magnetic resonance T1-weighted contrast imaging.
Collapse
Affiliation(s)
- Xiaoxin Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Chang Li
- Department of Radiology, Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Mengjun Ge
- Department of Biomedical Sciences College of Biology, Hunan University, Changsha, 410011, China
| | - Xiaoying Li
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Wei Zhao
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Hu Guo
- Siemens Healthineers MR Application China, Changsha, 410000, China
| | - Hemin Nie
- Department of Biomedical Sciences College of Biology, Hunan University, Changsha, 410011, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, 410011, China
| |
Collapse
|
30
|
Xiong Y, Mi BB, Shahbazi MA, Xia T, Xiao J. Microenvironment-responsive nanomedicines: a promising direction for tissue regeneration. Mil Med Res 2024; 11:69. [PMID: 39434177 PMCID: PMC11492517 DOI: 10.1186/s40779-024-00573-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/29/2024] [Indexed: 10/23/2024] Open
Abstract
Severe tissue defects present formidable challenges to human health, persisting as major contributors to mortality rates. The complex pathological microenvironment, particularly the disrupted immune landscape within these defects, poses substantial hurdles to existing tissue regeneration strategies. However, the emergence of nanobiotechnology has opened a new direction in immunomodulatory nanomedicine, providing encouraging prospects for tissue regeneration and restoration. This review aims to gather recent advances in immunomodulatory nanomedicine to foster tissue regeneration. We begin by elucidating the distinctive features of the local immune microenvironment within defective tissues and its crucial role in tissue regeneration. Subsequently, we explore the design and functional properties of immunomodulatory nanosystems. Finally, we address the challenges and prospects of clinical translation in nanomedicine development, aiming to propose a potent approach to enhance tissue regeneration through synergistic immune modulation and nanomedicine integration.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands.
| | - Tian Xia
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
31
|
Huang H, Fu J, Peng H, He Y, Chang A, Zhang H, Hao Y, Xu X, Li S, Zhao J, Ni J, Dong X. Co-delivery of polyphyllin II and IR780 PLGA nanoparticles induced pyroptosis combined with photothermal to enhance hepatocellular carcinoma immunotherapy. J Nanobiotechnology 2024; 22:647. [PMID: 39434141 PMCID: PMC11495104 DOI: 10.1186/s12951-024-02887-6] [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: 06/13/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
The clinical efficacy of immunotherapy for hepatocellular carcinoma (HCC) is significantly limited by the low immunogenicity of the tumor. Recent studies have revealed that both pyroptosis and photothermal therapy can effectively induce tumor immunogenic cell death (ICD) in liver cancer cells. Polyphyllin II (PPII), the major active component of Rhizoma Paridis, has been demonstrated for the first time to induce pyroptosis in tumor cells, while IR780 is activated by 808 nm laser to transform light energy into heat energy, effectively eliminating tumor cells. However, both PPII and IR780 are afflicted with challenges such as low solubility and poor targeting, significantly limiting their utilization. To address these problems, the pyroptosis inducer PPII and photosensitizer IR780 were co-loaded in PLGA nanoparticles by precipitation method, and the aptamer AS1411 was modified on the surface of nanoparticles to construct the targeting nanoparticles (Apt/PPII/IR780-NPs). The nanoparticles exhibit a pH/NIR dual-response intelligent release feature, which realizes the targeted and controlled release of drugs in tumor site. Furthermore, it can rapidly release PPII to induce cell pyroptosis under laser irradiation, combining with IR780-based photothermal therapy exert a significant synergistic anti-tumor effect in vitro and in vivo. This process not only promotes maturation of DCs and activates effector T cells, thereby initiating adaptive immunity, but also generates enduring and effective immune memory. In addition, Apt/PPII/IR780-NPs significantly improved the Anti-PD-1 efficacy. In summary, chemo-photothermal therapy based on Apt/PPII/IR780-NPs can significantly enhance tumor ICD, which provides a promising new strategy for HCC immunotherapy.
Collapse
Affiliation(s)
- Huating Huang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jing Fu
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Hulinyue Peng
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yuanyuan He
- Department of Radiology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Aqian Chang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Huizhong Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yang Hao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130015, China
| | - Xiaohan Xu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Shiman Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jingxia Zhao
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China.
| | - Jian Ni
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Xiaoxv Dong
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
32
|
Wu J, Wang H, Gao P, Ouyang S. Pyroptosis: Induction and inhibition strategies for immunotherapy of diseases. Acta Pharm Sin B 2024; 14:4195-4227. [PMID: 39525577 PMCID: PMC11544194 DOI: 10.1016/j.apsb.2024.06.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 11/16/2024] Open
Abstract
Cell death is a central process for organismal health. Pyroptosis, namely pyroptotic cell death, is recognized as a critical type that disrupts membrane and triggers pro-inflammatory cytokine secretion via gasdermins, providing a robust form of cytolysis. Meanwhile, along with the thorough research, a great deal of evidence has demonstrated the dual effects of pyroptosis in host defense and inflammatory diseases. More importantly, the recent identification of abundant gasdermin-like proteins in bacteria and fungi suggests an ancient origin of pyroptosis-based regulated cell death in the life evolution. In this review, we bring a general overview of pyroptosis pathways focusing on gasdermin structural biology, regulatory mechanisms, and recent progress in induction and inhibition strategies for disease treatment. We look forward to providing an insightful perspective for readers to comprehend the frame and challenges of the pyroptosis field, and to accelerating its clinical application.
Collapse
Affiliation(s)
- Junjun Wu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hong Wang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Pu Gao
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Songying Ouyang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| |
Collapse
|
33
|
Duan Y, Deng M, Liu B, Meng X, Liao J, Qiu Y, Wu Z, Lin J, Dong Y, Duan Y, Sun Y. Mitochondria targeted drug delivery system overcoming drug resistance in intrahepatic cholangiocarcinoma by reprogramming lipid metabolism. Biomaterials 2024; 309:122609. [PMID: 38754290 DOI: 10.1016/j.biomaterials.2024.122609] [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: 04/08/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
The challenge of drug resistance in intrahepatic cholangiocarcinoma (ICC) is intricately linked with lipid metabolism reprogramming. The hepatic lipase (HL) and the membrane receptor CD36 are overexpressed in BGJ398-resistant ICC cells, while they are essential for lipid uptake, further enhancing lipid utilization in ICC. Herein, a metal-organic framework-based drug delivery system (OB@D-pMOF/CaP-AC, DDS), has been developed. The specifically designed DDS exhibits a successive targeting property, enabling it to precisely target ICC cells and their mitochondria. By specifically targeting the mitochondria, DDS produces reactive oxygen species (ROS) through its sonodynamic therapy effect, achieving a more potent reduction in ATP levels compared to non-targeted approaches, through the impairment of mitochondrial function. Additionally, the DDS strategically minimizes lipid uptake through the incorporation of the anti-HL drug, Orlistat, and anti-CD36 monoclonal antibody, reducing lipid-derived energy production. This dual-action strategy on both mitochondria and lipids can hinder energy utilization to restore drug sensitivity to BGJ398 in ICC. Moreover, an orthotopic mice model of drug-resistant ICC was developed, which serves as an exacting platform for evaluating the multifunction of designed DDS. Upon in vivo experiments with this model, the DDS demonstrated exceptional capabilities in suppressing tumor growth, reprogramming lipid metabolism and improving immune response, thereby overcoming drug resistance. These findings underscore the mitochondria-targeted DDS as a promising and innovative solution in ICC drug resistance.
Collapse
Affiliation(s)
- Yi Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Mengqiong Deng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Bin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xianwei Meng
- Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jinghan Liao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yijie Qiu
- Department of Ultrasound, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092, Shanghai, China
| | - Zhihua Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Jiangtao Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yi Dong
- Department of Ultrasound, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 200092, Shanghai, China.
| | - Yourong Duan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Ying Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
34
|
Ma C, Cheng Z, Tan H, Wang Y, Sun S, Zhang M, Wang J. Nanomaterials: leading immunogenic cell death-based cancer therapies. Front Immunol 2024; 15:1447817. [PMID: 39185425 PMCID: PMC11341423 DOI: 10.3389/fimmu.2024.1447817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
The field of oncology has transformed in recent years, with treatments shifting from traditional surgical resection and radiation therapy to more diverse and customized approaches, one of which is immunotherapy. ICD (immunogenic cell death) belongs to a class of regulatory cell death modalities that reactivate the immune response by facilitating the interaction between apoptotic cells and immune cells and releasing specific signaling molecules, and DAMPs (damage-associated molecular patterns). The inducers of ICD can elevate the expression of specific proteins to optimize the TME (tumor microenvironment). The use of nanotechnology has shown its unique potential. Nanomaterials, due to their tunability, targeting, and biocompatibility, have become powerful tools for drug delivery, immunomodulators, etc., and have shown significant efficacy in clinical trials. In particular, these nanomaterials can effectively activate the ICD, trigger a potent anti-tumor immune response, and maintain long-term tumor suppression. Different types of nanomaterials, such as biological cell membrane-modified nanoparticles, self-assembled nanostructures, metallic nanoparticles, mesoporous materials, and hydrogels, play their respective roles in ICD induction due to their unique structures and mechanisms of action. Therefore, this review will explore the latest advances in the application of these common nanomaterials in tumor ICD induction and discuss how they can provide new strategies and tools for cancer therapy. By gaining a deeper understanding of the mechanism of action of these nanomaterials, researchers can develop more precise and effective therapeutic approaches to improve the prognosis and quality of life of cancer patients. Moreover, these strategies hold the promise to overcome resistance to conventional therapies, minimize side effects, and lead to more personalized treatment regimens, ultimately benefiting cancer treatment.
Collapse
Affiliation(s)
- Changyu Ma
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Beijing, China
| | - Zhe Cheng
- Department of Forensic Medicine, Harbin Medical University, Harbin, China
| | - Haotian Tan
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Beijing, China
| | - Yihan Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Clinical College, Peking University Health Science Center, Beijing, China
| | - Shuzhan Sun
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Clinical College, Peking University Health Science Center, Beijing, China
| | - Mingxiao Zhang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| | - Jianfeng Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
35
|
Huo CM, Zuo YC, Chen Y, Chen L, Zhu JY, Xue W. Natural lignin nanoparticles target tumor by saturating the phagocytic capacity of Kupffer cells in the liver. Int J Biol Macromol 2024; 274:133186. [PMID: 38885858 DOI: 10.1016/j.ijbiomac.2024.133186] [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: 02/25/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
Ligand-receptor recognition serves as the fundamental driving force for active targeting, yet it is still constrained by off-target effects. Herein, we demonstrate that circumventing or blocking the mononuclear phagocyte system (MPS) are both viable strategies to address off-target effects. Naturally derived lignin nanoparticles (LNPs) show great potential to block MPS due to its good stability, low toxicity, and degradability. We further demonstrate the impact of LNPs dosage on in vivo tumor targeting and antitumor efficacy. Our results show that a high dose of LNPs (300 mg/kg) leads to significant accumulation at the tumor site for a duration of 14 days after intravenous administration. In contrast, the low-dose counterparts (e.g., 50, 150 mg/kg) result in almost all LNPs accumulating in the liver. This discovery indicates that the liver is the primary site of LNP capture, leaving only the surplus LNPs the chance to reach the tumor. In addition, although cell membrane-engineered LNPs can rapidly penetrate tumors, they are still prone to capture by the liver during subsequent circulation in the bloodstream. Excitingly, comparable therapeutic efficacy is obtained for the above two strategies. Our findings may offer valuable insights into the targeted delivery of drugs for disease treatment.
Collapse
Affiliation(s)
- Cong-Min Huo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yu-Cheng Zuo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yu Chen
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Liheng Chen
- Guangdong Provincial Key Laboratory of Plant Resources Biorefinery, School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China.
| | - Jing-Yi Zhu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
36
|
Lai JM, Chen PL, Shi QY, Xie YQ, Jiaerheng G, Liu LH. A Self-Delivery Nanodrug Simultaneously Inhibits COX-2/PGE 2 Mediated Inflammation and Downregulates PD-L1 to Boost Photoimmunotherapy. Adv Healthc Mater 2024; 13:e2400367. [PMID: 38704750 DOI: 10.1002/adhm.202400367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/11/2024] [Indexed: 05/07/2024]
Abstract
Phototherapy promotes anti-tumor immunity by inducing immunogenic cell death (ICD), However, the accompanying inflammatory responses also trigger immunosuppression, attenuating the efficacy of photo-immunotherapy. Herein, they co-assembled a cell-membrane targeting chimeric peptide C16-Cypate-RRKK-PEG8-COOH (CCP) and anti-inflammatory diclofenac (DA) to develop a nanodrug (CCP@DA) that both enhances the immune effect of phototherapy and weakens the inflammation-mediated immunosuppression. CCP@DA achieves cell membrane-targeting photodynamic and photothermal synergistic therapies to damage programmed death ligand 1 (PD-L1) and induce a strong ICD to activate anti-tumor response. Simultaneously, the released DA inhibits the cycoperoxidase-2 (COX-2)/prostaglandin E2 (PGE2) pathway in tumor cells to inhibit pro-tumor inflammation and further down-regulate PD-L1 expression to relieve the immunosuppressive microenvironment. CCP@DA significantly inhibited tumor growth and inflammation both in vitro and in vivo, while maintaining a potent anti-tumor immune response. Additionally, it exhibits excellent anti-metastatic capabilities and prolongs mouse survival time with a single dose and low levels of near-infrared (NIR) light exposure. This work provides a valuable strategy to control the therapy-induced inflammation for high-efficiency photoimmunotherapy.
Collapse
Affiliation(s)
- Jin-Mei Lai
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Pei-Ling Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Qun-Ying Shi
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yong-Qi Xie
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - GuliJiayina Jiaerheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Li-Han Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
37
|
Yang Q, Sun X, Ding Q, Qi M, Liu C, Li T, Shi F, Wang L, Li C, Kim JS. An ATP-responsive metal-organic framework against periodontitis via synergistic ion-interference-mediated pyroptosis. Natl Sci Rev 2024; 11:nwae225. [PMID: 39071842 PMCID: PMC11275458 DOI: 10.1093/nsr/nwae225] [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: 03/21/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 07/30/2024] Open
Abstract
Periodontitis involves hyperactivated stromal cells that recruit immune cells, exacerbating inflammation. This study presents an ATP-responsive metal-organic framework (Mg/Zn-MOF) designed for periodontitis treatment, utilizing ion interference to modulate immune responses and prevent tissue destruction. Addressing the challenges of synergistic ion effects and targeted delivery faced by traditional immunomodulatory nanomaterials, the Mg/Zn-MOF system is activated by extracellular ATP-a pivotal molecule in periodontitis pathology-ensuring targeted ion release. Magnesium and zinc ions released from the framework synergistically inhibit membrane pore formation by attenuating Gasdermin D (GSDMD) expression and activation. This action curtails pyroptosis, lactate dehydrogenase and IL-1β release, thwarting the onset of inflammatory cascades. Mechanistically, Mg/Zn-MOF intervenes in both the NLRP3/Caspase-1/GSDMD and Caspase-11/GSDMD pathways to mitigate pyroptosis. In vivo assessments confirm its effectiveness in diminishing inflammatory cell infiltration and preserving collagen integrity, thereby safeguarding against periodontal tissue damage and bone loss. This investigation highlights the promise of ion-interference strategies in periodontitis immunotherapy, representing a significant stride in developing targeted therapeutic approaches.
Collapse
Affiliation(s)
- Qijing Yang
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiaolin Sun
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Qihang Ding
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Manlin Qi
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chengyu Liu
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Tingxuan Li
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Fangyu Shi
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Lin Wang
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chunyan Li
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| |
Collapse
|
38
|
Chen G, Lin L, Mai Z, Tang Y, Zhang Q, Chen G, Li Z, Zhang J, Wang Y, Yang Y, Yu Z. Carrier-Free Photodynamic Bioregulators Inhibiting Lactic Acid Efflux Combined with Immune Checkpoint Blockade for Triple-Negative Breast Cancer Immunotherapy. ACS NANO 2024. [PMID: 39034461 DOI: 10.1021/acsnano.4c07213] [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
Abnormal tumor metabolism creates a complex tumor immune microenvironment that plays a dominant role in the metastasis of triple-negative breast cancer (TNBC). TNBC is insensitive to immune checkpoint blockade (ICB) therapy because of insufficient cytotoxic T lymphocyte (CTL) infiltration and a hyper-lactic acid-suppressive immune microenvironment caused by abnormal glycolysis. Herein, we propose an amplified strategy based on lactic acid regulation to reprogram the immunosuppressive tumor microenvironment (ITM) and combine it with ICB therapy to achieve enhanced antitumor immunotherapy effects. Specifically, we constructed CASN, a carrier-free photodynamic bioregulator, through the self-assembly of the photosensitizer Chlorin e6 and monocarboxylate transporter 1 (MCT1) inhibitor AZD3965. CASN exhibited a uniform structure, good stability, and drug accumulation at the tumor site. CASN-mediated photodynamic therapy following laser irradiation inhibited primary tumor growth and induced immunogenic cell death. Furthermore, CASN reduced lactic acid-mediated regulatory T cell generation and M2 tumor-associated macrophage polarization by blocking MCT1-mediated lactic acid efflux to attenuate immune suppression, inducing the recruitment and activation of CTLs. Ultimately, CASN-mediated immunopotentiation combined with ICB therapy considerably strengthened tumor immunotherapy and effectively inhibited tumor growth and metastasis of TNBC. This synergistic amplification strategy overcomes the limitations of an acidic ITM and presents a potential clinical treatment option for metastatic tumors.
Collapse
Affiliation(s)
- Guimei Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ling Lin
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ziyi Mai
- Department of Pharmacy, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yan Tang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qiaoling Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Gui Chen
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zibo Li
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiasi Zhang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
| | - Yongxia Wang
- Department of Galactophore, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523058, China
| | - Yuanyuan Yang
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
| | - Zhiqiang Yu
- Department of Laboratory Medicine, Dongguan Institute of Clinical Cancer Research, The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Guangdong 523058, China
| |
Collapse
|
39
|
Wang H, Xia P, Kurboniyon MS, Fang S, Huang K, Ning S, Jin G, Zhang L, Wang C. V-doped MoS 2 nanozymes providing reactive oxygen species and depleting glutathione for photothermally-enhanced nanocatalytic therapy. Front Pharmacol 2024; 15:1448867. [PMID: 39101147 PMCID: PMC11294079 DOI: 10.3389/fphar.2024.1448867] [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: 06/14/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction: The tumor microenvironment and multidrug resistance of tumor cells seriously impair the activity of the nanozymes. Methods: Herein, a polyethylene glycol (PEG)-modified vanadium-doped molybdenum disulfide (V-MoS2@PEG) nanozymes were constructed to enhance anti-tumor activity through multi-enzymatic catalysis and photothermal effect with simultaneous reactive oxygen species replenishment and glutathione depletion. Results and discussion: V-MoS2@PEG nanosheets exerted peroxidase activity by causing molybdenum ion (Mo4+) to react with hydrogen peroxide to form toxic hydroxyl radicals (·OH). Meanwhile, the V-doping can deplete glutathione avoiding ·OH consumption. In addition, the high heat generated by V-MoS2@PEG nanozymes under near-infrared laser irradiation brought about a desirable local temperature gradient, which produced an enhanced catalytic effect by promoting band bending. Furthermore, the photothermally inspired polarized charge increased the permeability of the tumor cell membrane and promoted further aggregation of the nanozymes, which realized the combination of photothermal therapy with multi-enzymatic catalysis, solved the problem of multi-enzyme catalysis, and improved the anti-tumor efficiency.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Pengle Xia
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | | | - Shuhong Fang
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Kunying Huang
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shufang Ning
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guanqiao Jin
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Litu Zhang
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chen Wang
- Department of Research and Guangxi Cancer Molecular Medicine Engineering Research Center and Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
40
|
Zhang C, Deng Z, Sun X, Yuan K, Wang J, Wu X, Zhang Y, Yang K, Zhang J, Yang G. Petaloid Metal-Organic Frameworks for Resiquimod Delivery To Potentiate Antitumor Immunity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33093-33105. [PMID: 38884171 DOI: 10.1021/acsami.4c05290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The morphological features of materials significantly influence their interactions with cells, consequently affecting the cellular uptake of these materials. In this study, we examine the cellular uptake behavior of spherical metal-organic frameworks (MOFs) and petaloid MOFs, both possessing similar sizes and compositions. In comparison to spherical MOFs, dendritic cells (DCs) and macrophages exhibit superior phagocytic uptake of petaloid MOFs. Next, the results demonstrate that R848@petaloid MOFs more effectively promote the repolarization of tumor-associated macrophages (TAMs) from the M2 to M1 phenotype and the maturation of DCs. More importantly, the R848-loaded petaloid MOFs are found to significantly enhance the therapeutic effects of radiotherapy (RT) by eliciting antitumor responses. Furthermore, R848@petaloid MOFs combined with RT and αPD-L1 elicit a potent abscopal effect, effectively suppressing tumor metastasis. Therefore, this work proposes a new strategy to enhance the uptake of immunomodulators by immune cells through modulating the morphology of drug delivery carriers.
Collapse
Affiliation(s)
- Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zheng Deng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xianglong Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kangzhi Yuan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jiadong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xirui Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yifan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, Jiangsu 215004, China
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
41
|
Dou Y, Wang Y, Tian S, Song Q, Deng Y, Zhang Z, Chen P, Sun Y. Metal-organic framework (MOF)-based materials for pyroptosis-mediated cancer therapy. Chem Commun (Camb) 2024; 60:6476-6487. [PMID: 38853690 DOI: 10.1039/d4cc02084g] [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: 06/11/2024]
Abstract
Pyroptosis is regarded as a promising strategy to modulate tumor immune microenvironments for anticancer therapy. Although pyroptosis inducers have been extensively explored in the biomedical field, their drug resistance, off-targeting capacity, and adverse effects do not fulfill the growing demands of therapy. Nowadays, metal-organic frameworks (MOFs) with unique structures and facile synthesis/functionalization characteristics have shown great potential in anticancer therapy. The flexible choices of metal ions and ligands endow MOFs with inherent anti-cancer efficiency, whereas the porous structures in MOFs make them ideal vehicles for delivering various chemodrug-based pyroptosis inducers. In this review, we provide the latest advances in MOF-based materials to evoke pyroptosis and give a brief but comprehensive review of the different types of MOFs for pyroptosis-mediated cancer therapy. Finally, we also discuss the current challenges of MOF-based pyroptosis inducers and their future prospects in this field.
Collapse
Affiliation(s)
- You Dou
- College of Pharmacy, Hubei University of Science & Technology, Xianning 437100, China.
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Yuting Wang
- Key Laboratory of Fermentation Engineering (Ministry of Education), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, China.
| | - Shu Tian
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Qiao Song
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| | - Yun Deng
- Key Laboratory of Optoelectronic Chemical Materials and Devices of Ministry of Education, Jianghan University, Wuhan 430056, China
| | - Zhipeng Zhang
- College of Pharmacy, Hubei University of Science & Technology, Xianning 437100, China.
| | - PeiYao Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning 530021, China
| | - Yao Sun
- State Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
42
|
Lu J, He R, Liu Y, Zhang J, Xu H, Zhang T, Chen L, Yang G, Zhang J, Liu J, Chi H. Exploiting cell death and tumor immunity in cancer therapy: challenges and future directions. Front Cell Dev Biol 2024; 12:1416115. [PMID: 38887519 PMCID: PMC11180757 DOI: 10.3389/fcell.2024.1416115] [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: 04/26/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Cancer remains a significant global challenge, with escalating incidence rates and a substantial burden on healthcare systems worldwide. Herein, we present an in-depth exploration of the intricate interplay between cancer cell death pathways and tumor immunity within the tumor microenvironment (TME). We begin by elucidating the epidemiological landscape of cancer, highlighting its pervasive impact on premature mortality and the pronounced burden in regions such as Asia and Africa. Our analysis centers on the pivotal concept of immunogenic cell death (ICD), whereby cancer cells succumbing to specific stimuli undergo a transformation that elicits robust anti-tumor immune responses. We scrutinize the mechanisms underpinning ICD induction, emphasizing the release of damage-associated molecular patterns (DAMPs) and tumor-associated antigens (TAAs) as key triggers for dendritic cell (DC) activation and subsequent T cell priming. Moreover, we explore the contributions of non-apoptotic RCD pathways, including necroptosis, ferroptosis, and pyroptosis, to tumor immunity within the TME. Emerging evidence suggests that these alternative cell death modalities possess immunogenic properties and can synergize with conventional treatments to bolster anti-tumor immune responses. Furthermore, we discuss the therapeutic implications of targeting the TME for cancer treatment, highlighting strategies to harness immunogenic cell death and manipulate non-apoptotic cell death pathways for therapeutic benefit. By elucidating the intricate crosstalk between cancer cell death and immune modulation within the TME, this review aims to pave the way for the development of novel cancer therapies that exploit the interplay between cell death mechanisms and tumor immunity and overcome Challenges in the Development and implementation of Novel Therapies.
Collapse
Affiliation(s)
- Jiaan Lu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Ru He
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yang Liu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jinghan Zhang
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Heng Xu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Tianchi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Li Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Jun Zhang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| |
Collapse
|
43
|
Wang J, Qiao L, Zhu G, Sun Q, Xie Y, Wang M, Xu Y, Li C. Biodegradable pyroptosis inducer with multienzyme-mimic activity kicks up reactive oxygen species storm for sensitizing immunotherapy. J Control Release 2024; 370:438-452. [PMID: 38701885 DOI: 10.1016/j.jconrel.2024.04.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/06/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
Triggering pyroptosis is a major new weathervane for activating tumor immune response. However, biodegradable pyroptosis inducers for the safe and efficient treatment of tumors are still scarce. Herein, a novel tumor microenvironment (TME)-responsive activation nanoneedle for pyroptosis induction, copper-tannic acid (CuTA), was synthesized and combined with the sonosensitizer Chlorin e6 (Ce6) to form a pyroptosis amplifier (CuTA-Ce6) for dual activation and amplification of pyroptosis by exogenous ultrasound (US) and TME. It was demonstrated that Ce6-triggered sonodynamic therapy (SDT) further enhanced the cellular pyroptosis caused by CuTA, activating the body to develop a powerful anti-tumor immune response. Concretely, CuTA nanoneedles with quadruple mimetic enzyme activity could be activated to an "active" state in the TME, destroying the antioxidant defense system of the tumor cells through self-destructive degradation, breaking the "immunosilent" TME, and thus realizing the pyroptosis-mediated immunotherapy with fewer systemic side effects. Considering the outstanding oxygen-producing capacity of CuTA and the distinctive advantages of US, the sonosensitizer Ce6 was attached to CuTA via an amide reaction, which further amplified the pyroptosis and sensitized pyroptosis-induced immunotherapy with the two-pronged strategy of CuTA enzyme-catalyzed cascade and US-driven SDT pathway to generate a "reactive oxygen species (ROS) storm". Conclusively, this work provided a representative paradigm for achieving safe, reliable and efficient pyroptosis, which was further enhanced by SDT for more robust immunotherapy.
Collapse
Affiliation(s)
- Junrong Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Luying Qiao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Guoqing Zhu
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Qianqian Sun
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China.
| | - Yulin Xie
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Man Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Yaqi Xu
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong 250000, PR China.
| | - Chunxia Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China.
| |
Collapse
|
44
|
Han J, Dong H, Zhu T, Wei Q, Wang Y, Wang Y, Lv Y, Mu H, Huang S, Zeng K, Xu J, Ding J. Biochemical hallmarks-targeting antineoplastic nanotherapeutics. Bioact Mater 2024; 36:427-454. [PMID: 39044728 PMCID: PMC11263727 DOI: 10.1016/j.bioactmat.2024.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 07/25/2024] Open
Abstract
Tumor microenvironments (TMEs) have received increasing attention in recent years as they play pivotal roles in tumorigenesis, progression, metastases, and resistance to the traditional modalities of cancer therapy like chemotherapy. With the rapid development of nanotechnology, effective antineoplastic nanotherapeutics targeting the aberrant hallmarks of TMEs have been proposed. The appropriate design and fabrication endow nanomedicines with the abilities for active targeting, TMEs-responsiveness, and optimization of physicochemical properties of tumors, thereby overcoming transport barriers and significantly improving antineoplastic therapeutic benefits. This review begins with the origins and characteristics of TMEs and discusses the latest strategies for modulating the TMEs by focusing on the regulation of biochemical microenvironments, such as tumor acidosis, hypoxia, and dysregulated metabolism. Finally, this review summarizes the challenges in the development of smart anti-cancer nanotherapeutics for TME modulation and examines the promising strategies for combination therapies with traditional treatments for further clinical translation.
Collapse
Affiliation(s)
- Jing Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - He Dong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Tianyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Yongheng Wang
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Yun Wang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Yu Lv
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Shandeng Huang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Ke Zeng
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, 100 Haining Street, Shanghai, 200080, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
45
|
An G, Zheng H, Guo L, Huang J, Yang C, Bai Z, Wang N, Yang W, Zhu Y. A metal-organic framework (MOF) built on surface-modified Cu nanoparticles eliminates tumors via multiple cascading synergistic therapeutic effects. J Colloid Interface Sci 2024; 662:298-312. [PMID: 38354557 DOI: 10.1016/j.jcis.2024.02.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
Tumors produce a hypoxic environment that greatly influences cancer treatment, and conventional chemotherapeutic drugs cannot selectively accumulate in the tumor region because of the lack of a tumor targeting mechanism, causing increased systemic toxicities and side effects. Hence, designing and developing new nanoplatforms that combine multimodal therapeutic regimens is essential to improve tumor therapeutic efficacy. Herein, we report the synthesis of ultrafine Cu nanoparticles loaded with a drug combination of cisplatin (Pt) and 1-methyl-d-tryptophan (1-MT) and externally coated with 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) photosensitizer, polydopamine (PDA) and CaO2 of MIL-101(Fe) as a new nanoplatform (Cu@MIL-101@PMTPC). The nanoplatform synergistically combined chemodynamic therapy (CDT), photodynamic therapy (PDT), and immunochemotherapy. The Fe3+ in MIL-101(Fe) and the surface Cu nanoparticles exhibited strong ability to consume intracellular glutathione (GSH), thereby generating a Fenton-like response in the tumor microenvironment (TME) with substantial peroxidase (POD)-like and superoxide dismutase (SOD)-like activities. In this design, we used the indoleamine 2,3-dioxygenase (IDO) inhibitor 1-MT to overcome chemotherapy-induced immune escape phenomena including enhanced CD8+ and CD4+ T cell expression, interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) production, and accelerated immunogenic cell death. The targeted release of cisplatin loaded into Cu@MIL-101@PMTPC also reduced toxic side effects of chemotherapy. TCPP generated a large amount of singlet oxygen (1O2) upon specific laser irradiation to effectively kill tumor cells. CaO2 on the outer layer generated oxygen (O2) and hydrogen peroxide (H2O2) to ameliorate hypoxia in the tumor microenvironment, enhance the PDT effect, and provide a continuous supply of H2O2 for the Fenton-like reaction. Thus, this nanocarrier platform exhibited a powerful chemodynamic, photodynamic, and immunochemotherapeutic cascade, providing a new strategy for cancer treatment.
Collapse
Affiliation(s)
- Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Lianshan Guo
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Jingmei Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Congling Yang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Zhihao Bai
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China.
| | - Wenhui Yang
- Department of Medical Laboratory, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang 530021, China.
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning 530004, China; College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter EX4 4QF, UK.
| |
Collapse
|
46
|
Ge J, Zhang Z, Zhao S, Chen Y, Min X, Cai Y, Zhao H, Wu X, Zhao F, Chen B. Nanomedicine-induced cell pyroptosis to enhance antitumor immunotherapy. J Mater Chem B 2024; 12:3857-3880. [PMID: 38563315 DOI: 10.1039/d3tb03017b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Immunotherapy is a therapeutic modality designed to elicit or augment an immune response against malignancies. Despite the immune system's ability to detect and eradicate neoplastic cells, certain neoplastic cells can elude immune surveillance and elimination through diverse mechanisms. Therefore, antitumor immunotherapy has emerged as a propitious strategy. Pyroptosis, a type of programmed cell death (PCD) regulated by Gasdermin (GSDM), is associated with cytomembrane rupture due to continuous cell expansion, which results in the release of cellular contents that can trigger robust inflammatory and immune responses. The field of nanomedicine has made promising progress, enabling the application of nanotechnology to enhance the effectiveness and specificity of cancer therapy by potentiating, enabling, or augmenting pyroptosis. In this review, we comprehensively examine the paradigms underlying antitumor immunity, particularly paradigms related to nanotherapeutics combined with pyroptosis; these treatments include chemotherapy (CT), hyperthermia therapy, photodynamic therapy (PDT), chemodynamic therapy (CDT), ion-interference therapy (IIT), biomimetic therapy, and combination therapy. Furthermore, we thoroughly discuss the coordinated mechanisms that regulate these paradigms. This review is expected to enhance the understanding of the interplay between pyroptosis and antitumor immunotherapy, broaden the utilization of diverse nanomaterials in pyroptosis-based antitumor immunotherapy, and facilitate advancements in clinical tumor therapy.
Collapse
Affiliation(s)
- Jingwen Ge
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Zheng Zhang
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Shuangshuang Zhao
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Yanwei Chen
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Xin Min
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Yun Cai
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Huajiao Zhao
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Xincai Wu
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Feng Zhao
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| | - Baoding Chen
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, P. R. China.
| |
Collapse
|
47
|
Fan Z, Jiang X, Sun T, Zeng F, Huang G, Liang C, Nie L. In vivo visualization of tumor-associated macrophages re-education by photoacoustic/fluorescence dual-modal imaging with a metal-organic frames-based caspase-1 nanoreporter. J Colloid Interface Sci 2024; 659:48-59. [PMID: 38157726 DOI: 10.1016/j.jcis.2023.12.123] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Tumor-associated macrophages (TAMs) are vital in the tumor microenvironment, contributing to immunosuppression and therapy tolerance. Despite their importance, the precise re-education of TAMs in vivo continues to present a formidable challenge. Moreover, the lack of real-time and efficient methods to comprehend the spatiotemporal kinetics of TAMs repolarization remains a significant hurdle, severely hampering the accurate assessment of treatment efficacy and prognosis. Herein, we designed a metal-organic frameworks (MOFs) based Caspase-1 nanoreporter (MCNR) that can deliver a TLR7/8 agonist to the TAMs and track time-sensitive Caspase-1 activity as a direct method to monitor the initiation of immune reprogramming. This nanosystem exhibits excellent TAMs targeting ability, enhanced tumor accumulation, and stimuli-responsive behavior. By inducing the reprogramming of TAMs, they were able to enhance T-cell infiltration in tumor tissue, resulting in inhibited tumor growth and improved survival in mice model. Moreover, MCNR also serves as an activatable photoacoustic and fluorescent dual-mode imaging agent through Caspase-1-mediated specific enzyme digestion. This feature enables non-invasive and real-time antitumor immune activation monitoring. Overall, our findings indicate that MCNR has the potential to be a valuable tool for tumor immune microenvironment remodeling and noninvasive quantitative detection and real-time monitoring of TAMs repolarization to immunotherapy in the early stage.
Collapse
Affiliation(s)
- Zhijin Fan
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xiaoxiao Jiang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Tong Sun
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Fanchu Zeng
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Guojia Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Changhong Liang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; School of Medicine, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Liming Nie
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; School of Medicine, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| |
Collapse
|
48
|
Wang X, Zhang W, Wang Y, Zhu X, Liu Z, Liu M, Wu Z, Li B, Liu S, Liao S, Zhu P, Liu B, Li C, Wang Y, Chen Z. Logic "AND Gate Circuit"-Based Gasdermin Protein Expressing Nanoplatform Induces Tumor-Specific Pyroptosis to Enhance Cancer Immunotherapy. ACS NANO 2024; 18:6946-6962. [PMID: 38377037 DOI: 10.1021/acsnano.3c09405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Pyroptosis mediated by gasdermin protein has shown great potential in cancer immunotherapies. However, the low expression of gasdermin proteins and the systemic toxicity of nonspecific pyroptosis limit its clinical application. Here, we designed a synthetic biology strategy to construct a tumor-specific pyroptosis-inducing nanoplatform M-CNP/Mn@pPHS, in which a pyroptosis-inducing plasmid (pPHS) was loaded onto a manganese (Mn)-doped calcium carbonate nanoparticle and wrapped in a tumor-derived cell membrane. M-CNP/Mn@pPHS showed an efficient tumor targeting ability. After its internalization by tumor cells, the degradation of M-CNP/Mn@pPHS in the acidic endosomal environment allowed the efficient endosomal escape of plasmid pPHS. To trigger tumor-specific pyroptosis, pPHS was designed according to the logic "AND gate circuit" strategy, with Hif-1α and Sox4 as two input signals and gasdermin D induced pyroptosis as output signal. Only in cells with high expression of Hif-1α and Sox4 simultaneously will the output signal gasdermin D be expressed. Since Hif-1α and Sox4 are both specifically expressed in tumor cells, M-CNP/Mn@pPHS induces the tumor-specific expression of gasdermin D and thus pyroptosis, triggering an efficient immune response with little systemic toxicity. The Mn2+ released from the nanoplatform further enhanced the antitumor immune response by stimulating the cGAS-STING pathway. Thus, M-CNP/Mn@pPHS efficiently inhibited tumor growth with 79.8% tumor regression in vivo. We demonstrate that this logic "AND gate circuit"-based gasdermin nanoplatform is a promising strategy for inducing tumor-specific pyroptosis with little systemic toxicity.
Collapse
Affiliation(s)
- Xiaoxi Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenyan Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xueqin Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zimai Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Meiyi Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zixian Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bingyu Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Sijia Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shixin Liao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pingping Zhu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Center for Stem Cell and Regenerative Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Benyu Liu
- Center for Stem Cell and Regenerative Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Chong Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Zhongke Jianlan Medical Research Institute, Beijing 100190, China
| | - Yongchao Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
- Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou 450001, China
- International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
49
|
Liu J, Chen T, Liu X, Li Z, Zhang Y. Engineering materials for pyroptosis induction in cancer treatment. Bioact Mater 2024; 33:30-45. [PMID: 38024228 PMCID: PMC10654002 DOI: 10.1016/j.bioactmat.2023.10.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer remains a significant global health concern, necessitating the development of innovative therapeutic strategies. This research paper aims to investigate the role of pyroptosis induction in cancer treatment. Pyroptosis, a form of programmed cell death characterized by the release of pro-inflammatory cytokines and the formation of plasma membrane pores, has gained significant attention as a potential target for cancer therapy. The objective of this study is to provide a comprehensive overview of the current understanding of pyroptosis and its role in cancer treatment. The paper discusses the concept of pyroptosis and its relationship with other forms of cell death, such as apoptosis and necroptosis. It explores the role of pyroptosis in immune activation and its potential for combination therapy. The study also reviews the use of natural, biological, chemical, and multifunctional composite materials for pyroptosis induction in cancer cells. The molecular mechanisms underlying pyroptosis induction by these materials are discussed, along with their advantages and challenges in cancer treatment. The findings of this study highlight the potential of pyroptosis induction as a novel therapeutic strategy in cancer treatment and provide insights into the different materials and mechanisms involved in pyroptosis induction.
Collapse
Affiliation(s)
- Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - XianLing Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Department of Oncology, Guilin Hospital of the Second Xiangya Hospital, Central South University, Guilin, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Zhang
- Department of Biomedical Engineering, The City University of Hong Kong, Hong Kong Special Administrative Region of China
| |
Collapse
|
50
|
Song J, Tang C, Wang Y, Ba J, Liu K, Gao J, Chang J, Kang J, Yin L. Multifunctional nanoparticles for enhanced sonodynamic-chemodynamic immunotherapy with glutathione depletion. Nanomedicine (Lond) 2024; 19:145-161. [PMID: 38270976 DOI: 10.2217/nnm-2023-0218] [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] [Indexed: 01/26/2024] Open
Abstract
Aim: This study aimed to develop a sonodynamic-chemodynamic nanoparticle functioning on glutathione depletion in tumor immunotherapy. Materials & methods: The liposome-encapsulated 2,2-azobis[2-(2-imidazolin-2-yl) propane] dihydrochloride (AIPH) and copper-cysteine nanoparticles, AIPH/Cu-Cys@Lipo, were synthesized with a one-pot method. 4T1 cells were injected into female BALB/c mice for modeling. Results: AIPH/Cu-Cys@Lipo was well synthesized. It generated alkyl radicals upon ultrasound stimulation. AIPH/Cu-Cys@Lipo promoted the generation of -OH via a Fenton-like reaction. Both in vitro and in vivo experiments verified that AIPH/Cu-Cys@Lipo significantly inhibited tumor development by decreasing mitochondrial membrane potential, activating CD4+ and CD8+ T cells and promoting the expression of IL-2 and TNF-α. Conclusion: AIPH/Cu-Cys@Lipo provides high-quality strategies for safe and effective tumor immunotherapy.
Collapse
Affiliation(s)
- Jianying Song
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Cong Tang
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Yun Wang
- Xuzhou Central Hospital, Xuzhou, Jiangsu Province, 221009, China
| | - Junli Ba
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Kairui Liu
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Jinwei Gao
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Jin Chang
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Jun Kang
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Linling Yin
- Department of stomatology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200080, China
| |
Collapse
|