1
|
Chen Y, Sun B, Yu Z, Cheng Z, Chen S, Chang B. Hollow heterojunction nanocages of zeolitic imidazolate framework-8@zinc sulfide@copper sulfide for synergistic tumor therapy. J Colloid Interface Sci 2025; 695:137811. [PMID: 40344729 DOI: 10.1016/j.jcis.2025.137811] [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: 02/12/2025] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
The tumor microenvironment (TME) poses significant challenges for achieving efficient cancer remission through monotherapy, making multimodal synergistic therapy a key strategy in tumor treatment. Here, we designed and constructed a hollow core-shell nanocage based on zeolitic imidazolate framework-8 (ZIF-8), zeolitic imidazolate framework-8@zinc sulfide@copper sulfide (ZIF-8@ZnS@CuS, denoted as ZZCS), for the combined application of chemodynamic therapy (CDT), photothermal therapy (PTT) and photocatalytic therapy (PCT). The Cu2+ ions in ZZCS imparted peroxidase (POD)-like and catalase (CAT)-like activities, enabling the generation of toxic hydroxyl radicals (OH) through POD-like catalysis to enhance CDT effect, while also reacting with overexpressed glutathione (GSH) to elevate intracellular reactive oxygen species (ROS) levels. To overcome the inhibitory effects of hypoxia on PCT in the TME, ZZCS catalyzed the production of O2 from endogenous hydrogen peroxide (H2O2) via CAT-like activity, alleviating hypoxia and enhancing the PCT effect. Furthermore, hollow core-shell heterojunction structure of ZZCS exhibited excellent near-infrared absorption and multiple light reflection effects. Under 808 nm laser irradiation, ZZCS showed a high photothermal conversion efficiency (η = 78.1 %) and generated significant amounts of ROS (OH, O2-, 1O2), enabling the synergistic elimination of tumor cells via PTT and PCT. Benefiting from the combined CDT/PTT/PCT effects, ZZCS demonstrated excellent therapeutic efficacy in vivo, nearly eradicating tumors. This study may provide a promising foundation and potential direction for advancing multimodal tumor therapy. Future studies will focus on conducting long-term investigations to systematically evaluate the efficacy of ZZCS core-shell nanocages in achieving durable tumor eradication.
Collapse
Affiliation(s)
- Yuqin Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Benjian Sun
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhouyu Yu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Si Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
2
|
Lugoloobi I, Wu F, Kang Y, Yuan X, Bi W, Li P, Shi S, Dong H, Zhu J, Zheng B. Red-Light Triggered CO/ROS Release from Porphyrin-Flavonol Hybrid@PC7A Micelles for Eradicating Escherichia coli. Macromol Biosci 2025:e70014. [PMID: 40351109 DOI: 10.1002/mabi.202500079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Photo-triggered carbon monoxide (CO) release, mediated by reactive oxygen species (ROS), shows a significant potential in therapeutic applications. However, the existing photosensitizers predominantly function as type II ROS generators. When ROS are present in excess, they are always wasted due to their short half-lives, which limit their ability to travel significant distances and effectively achieve therapeutic outcomes. In this study, the biological function of a single-component molecule, PdHF, is investigated. This molecule is formed by covalently conjugating 3-hydroxyflavone (3-HF) to a palladium(II) tetraphenyltetrabenzoporphyrin (PdTPTBP) photosensitizer. Subsequently, PdHF is loaded into the 2-hexamethyleneimino ethyl methacrylate (C7A)-modified PEG-b-PCL block copolymer (PC7A) to form a PdHF@PC7A micellar system capable of co-releasing CO and ROS under red-light irradiation. CO/ROS co-release can be attributed to the generation of singlet oxygen species that not only oxidize 3-HF to release CO but are concurrently reduced by the tertiary amine, C7A, to form cytotoxic superoxide anions and hydrogen peroxide. In vitro studies on these positively charged micelles validate a high biosafety and excellent antibacterial effects with competent elimination of Gram-negative bacteria, Escherichia coli. Furthermore, evidence of micelle uptake by bacterial cells supports synergistic photodynamic and gas therapy through intracellular CO/ROS co-release.
Collapse
Affiliation(s)
- Ishaq Lugoloobi
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Feng Wu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Ye Kang
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Xinsong Yuan
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Wenjie Bi
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Pan Li
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Shanshan Shi
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Huaze Dong
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Jinmiao Zhu
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| | - Bin Zheng
- School of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, Anhui, 230061, China
| |
Collapse
|
3
|
Chen YC, Chang LC, Liu YL, Chang MC, Liu YF, Chang PY, Manoharan D, Wang WJ, Chen JS, Wang HC, Chiu WT, Li WP, Sheu HS, Su WP, Yeh CS. Redox disruption using electroactive liposome coated gold nanoparticles for cancer therapy. Nat Commun 2025; 16:3253. [PMID: 40188189 PMCID: PMC11972414 DOI: 10.1038/s41467-025-58636-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
Cancer remains a global health challenge necessitating innovative therapies. We introduce a strategy to disrupt cancer cell redox balance using gold nanoparticles (Au NPs) as electron sinks combined with electroactive membranes. Utilizing Shewanella oneidensis MR-1 membrane proteins, we develop liposomes enriched with c-type cytochromes. These, coupled with Au NPs, facilitate autonomous electron transfer from cancer cells, disrupting redox processes and inducing cell death. Effective across various cancer types, larger Au NPs show enhanced efficacy, especially under hypoxic conditions. Oxidative stress from Au@MIL (MIL: membrane-integrated liposome) treatments, including mitochondrial and endoplasmic reticulum lipid oxidation and mitochondrial membrane potential changes, triggers apoptosis, bypassing iron-mediated pathways. Surface plasmon band and X-ray absorption near-edge structure (XANES) analyses confirm electron transfer. A SiO2 insulator coating on Au NPs blocks this transfer, suppressing cancer cell damage. This approach highlights the potential of modulated electron transfer pathways in targeted cancer therapy, offering refined and effective treatments.
Collapse
Grants
- NSTC 113-2740-B-006-002 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- NSTC 112-2113-M-037-014-MY2 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- NSTC 113-2320-B-037-007- Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- NSTC 113-2314-B-006 -014 - Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- NSTC 113-2321-B-006 -010 - Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- C.-S.Y. acknowledges the financial support from the National Science and Technology Council (NSTC), Taiwan (NSTC 113-2113-M-006-015). This research was also partially supported by the Higher Education Sprout Project, Ministry of Education, to the Headquarters of University Advancement at National Cheng Kung University. Additional financial support was provided by the Center of Applied Nanomedicine, National Cheng Kung University, under the Featured Areas Research Center Program within the framework of the Higher Education Sprout Project of the Ministry of Education (MOE) in Taiwan. W.-T.C. acknowledges the financial support from NSTC, Taiwan (NSTC 113-2740-B-006-002). W.-P. L. acknowledges the financial support provided by NSTC, Taiwan (NSTC 112-2113-M-037-014-MY2 and 113-2320-B-037-007-) and the Yushan Young Scholar Program of the Ministry of Education of Taiwan. W.-P.S. thanks the financial support by the NSTC (NSTC 113-2314-B-006 -038 -MY3; 113-2314-B-006 -014 -; 113-2321-B-006 -010 -).
Collapse
Affiliation(s)
- Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Li-Chan Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan
| | - Yan-Ling Liu
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ming-Che Chang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yin-Fen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan
| | - Po-Ya Chang
- National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wen-Jyun Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Jia-Sin Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hsueh-Chun Wang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Wei-Peng Li
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan.
| | - Hwo-Shuenn Sheu
- National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan.
| | - Wen-Pin Su
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan.
- Departments of Oncology and Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
- Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan.
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
4
|
Yao C, Zhang C, Fan D, Li X, Zhang S, Liu D. Advancements in research on the precise eradication of cancer cells through nanophotocatalytic technology. Front Oncol 2025; 15:1523444. [PMID: 40236645 PMCID: PMC11996665 DOI: 10.3389/fonc.2025.1523444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
The rapid development of nanotechnology has significantly advanced the application of nanophotocatalysis in the medical field, particularly for cancer therapy. Traditional cancer treatments, such as chemotherapy and radiotherapy, often cause severe side effects, including damage to healthy tissues and the development of drug resistance. In contrast, nanophotocatalytic therapy offers a promising approach by utilizing nanomaterials that generate reactive oxygen species (ROS) under light activation, allowing for precise tumor targeting and minimizing collateral damage to surrounding tissues. This review systematically explores the latest advancements in highly efficient nanophotocatalysts for cancer treatment, focusing on their toxicological profiles, underlying mechanisms for cancer cell eradication, and potential for clinical application. Recent research shows that nanophotocatalysts, such as TiO2, In2O3, and g-C3N4 composites, along with photocatalysts with high conduction band or high valence band positions, generate ROS under light irradiation, which induces oxidative stress and leads to cancer cell apoptosis or necrosis. These ROS cause cellular damage by interacting with key biological molecules such as DNA, proteins, and lipids, triggering a cascade of biochemical reactions that ultimately result in cancer cell death. Furthermore, strategies such as S-scheme heterojunctions and oxygen vacancies (OVs) have been incorporated to enhance charge separation efficiency and light absorption, resulting in increased ROS generation, which improves photocatalytic performance for cancer cell targeting. Notably, these photocatalysts exhibit low toxicity to healthy cells, making them a safe and effective treatment modality. The review also discusses the challenges associated with photocatalytic cancer therapy, including limitations in light penetration and the need for improved biocompatibility. The findings suggest that nanophotocatalytic technology holds significant potential for precision cancer therapy, paving the way for safer and more effective treatment strategies.
Collapse
Affiliation(s)
- Changyang Yao
- Department of General Surgery, Fengyang County People’s Hospital, Chuzhou, China
| | - Chensong Zhang
- Department of Surgical Oncology Surgery (General Ward), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Dongwei Fan
- Department of General Surgery, Affiliated Hospital of West Anhui Health Vocational College, Lu’an, Anhui, China
| | - Xuanhe Li
- Department of Surgical Oncology Surgery (General Ward), The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shaofa Zhang
- Department of General Surgery, Fengyang County People’s Hospital, Chuzhou, China
| | - Daoxin Liu
- Department of General Surgery, Fengyang County People’s Hospital, Chuzhou, China
| |
Collapse
|
5
|
Yin Y, Guo W, Chen Q, Tang Z, Liu Z, Lin R, Pan T, Zhan J, Ren L. A Single H 2S-Releasing Nanozyme for Comprehensive Diabetic Wound Healing through Multistep Intervention. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18134-18149. [PMID: 40088144 DOI: 10.1021/acsami.5c00889] [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: 03/17/2025]
Abstract
Diabetic wound healing presents a significant medical challenge and requires multistep interventions due to comprehensive wound environments, such as hyperglycemia, bacterial infection, and impaired angiogenesis. However, current multistep interventions are complicated and need on-demand sequential release and synergy of multicomponents. Herein, a H2S-releasing cascade nanozyme (FeS@Au), which is composed of ultrasmall gold nanocluster (AuNC) loaded on ferrous sulfide nanoparticle (FeSNP), is developed as a single component to regulate glucose level, eliminate infection, and promote angiogenesis, achieving multistep interventions for comprehensive diabetic wound treatment. The glucose oxidase-like activity of AuNC catalyzes glucose into gluconic acid and H2O2, which not only lowers the local glucose level but also decreases the local pH and increases H2O2 level to boost the peroxidase-like activity of FeSNP to generate abundant hydroxyl radical (reactive oxygen species, ROS), inducing ferroptosis-like death in drug-resistant bacteria. Additionally, FeSNP release H2S in the acidified environment to upregulate hypoxia-inducible factor-1 to enhance vascularization through upregulating the expression of vascular endothelial growth factor (VEGF) and other angiogenesis-related genes, reducing the damage to endothelial cells caused by excessive ROS produced by the nanozyme. In a full-thickness MRSA-infected diabetic rat model, FeS@Au significantly eliminates bacteria, enhances angiogenesis, promotes collagen deposition, and accelerates wound healing. This work presents a single nanozyme with H2S-release for multistep interventions, providing a versatile strategy for healing extensive tissue damage caused by diabetes.
Collapse
Affiliation(s)
- Ying Yin
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Wentai Guo
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Qiangyu Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Zhimin Tang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Zheng Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Ruibin Lin
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Ting Pan
- Institute of Nanophotonics, College of Physics & Optoelectronic Engineering, Jinan University, 511443 Guangzhou, China
| | - Jiezhao Zhan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
| | - Li Ren
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
6
|
Xue X, Zheng F, Luo Y, Chen W, Gao Y, Wei K. A multifunctional Pt/DMSN nanozyme-based colorimetric-fluorescence sensing platform for breast cancer detection. Mikrochim Acta 2025; 192:228. [PMID: 40080161 DOI: 10.1007/s00604-025-07082-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/01/2025] [Indexed: 03/15/2025]
Abstract
Nanozyme-linked immunosorbent assay has emerged as a promising strategy for sensitive biosensing. However, the catalytic activity and stability of nanozymes affect the accuracy of immunosorbent assays. In this study, we synthesized a Pt/DMSN nanozyme with peroxidase-mimicking activity, which effectively catalyzed the oxidation of peroxidase substrate 3,3',5,5'-tetramethylbenzidin (TMB) in the presence of hydrogen peroxide. Capitalizing on its peroxidase-like activity, the Pt/DMSN nanozyme was functionalized with dual-fluorescent recognition elements (HER2-mAbs and sk6Ea aptamers) to establish a nanozyme-linked immunosorbent assay platform, which exhibited catalytic stability and substrate affinity comparable to horseradish peroxidase. The resulting Multi-Pt/DMSN platform was used to selectively distinguish HER2-positive breast cancer cells from luminal A, triple-negative breast cancer subtypes, and non-neoplastic cells, achieving a detection limit of 50 HER2-positive cells within 30 min. The combination of robust enzyme-like activity and tumor-targeting properties enables fluorescence imaging, providing dual-mode diagnostic functionality. This work presents a prospective platform for differentiating breast cancer subtypes in early diagnosis.
Collapse
Affiliation(s)
- Xinrui Xue
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Fang Zheng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yujia Luo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Wenyu Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yuanyuan Gao
- Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong, 637000, Sichuan, P. R. China.
| | - Kun Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China.
| |
Collapse
|
7
|
Malla P, Wang YM, Su CH. New horizons for the therapeutic application of nanozymes in cancer treatment. J Nanobiotechnology 2025; 23:130. [PMID: 39979897 PMCID: PMC11844087 DOI: 10.1186/s12951-025-03185-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/01/2025] [Indexed: 02/22/2025] Open
Abstract
The advent of nanozymes has revolutionized approaches to cancer diagnosis and therapy, introducing innovative strategies that address the limitations of conventional treatments. Nanozyme nanostructures with enzyme-mimicking catalytic abilities exhibit exceptional stability, biocompatibility, and customizable functions, positioning them as promising tools for cancer theranostics. By emulating natural enzyme reactions, nanozymes can selectively target and eradicate cancer cells, minimizing harm to adjacent healthy tissues. Nanozymes can also be functionalized with specific targeting ligands, allowing for the precise delivery and regulated release of therapeutic agents, improving treatment effectiveness and reducing adverse effects. However, issues such as biocompatibility, selectivity, and regulatory compliance remain critical challenges for the clinical application of nanozymes. This review provides an overview of nanozymes, highlighting their unique properties, various classifications, catalytic activities, and diverse applications in cancer treatments. The strategic oncological deployment of nanozymes could profoundly impact future advancements in personalized medicine, highlighting recent progress and prospective directions in enzyme-mimetic approaches for cancer treatment. This review summarizes an overview of nanozymes, highlighting their unique properties, various classifications, catalytic activities, and diverse applications in cancer treatments.
Collapse
Affiliation(s)
- Pravanjan Malla
- Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yu-Ming Wang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.
| | - Chia-Hao Su
- Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
8
|
Meng W, Chen T, Li X, Li Y, Zhang L, Xu Y, Song T, Qi J, Xiong Q, Li W. A Dual-Targeting Biomimetic Nanoplatform Integrates SDT/CDT/Gas Therapy to Boost Synergistic Ferroptosis for Orthotopic Hepatocellular Carcinoma Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413833. [PMID: 39783849 PMCID: PMC11848614 DOI: 10.1002/advs.202413833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/29/2024] [Indexed: 01/12/2025]
Abstract
The development of efficient therapeutic strategies to promote ferroptotic cell death offers significant potential for hepatocellular carcinoma (HCC) treatment. Herein, this study presents an HCC-targeted nanoplatform that integrates bimetallic FeMoO4 nanoparticles with CO-releasing molecules, and further camouflaged with SP94 peptide-modified macrophage membrane for enhanced ferroptosis-driven multi-modal therapy of HCC. Leveraging the multi-enzyme activities of the multivalent metallic elements, the nanoplatform not only decomposes H2O2 to generate oxygen and alleviate tumor hypoxia but also depletes glutathione to inactivate glutathione peroxides 4, which amplify sonodynamic therapy and ferroptotic tumor death under ultrasound (US) irradiation. Meanwhile, the nanoplatform catalyzes the Fenton reaction to produce hydroxyl radicals for chemodynamic therapy. Elevated intracellular reactive oxygen species trigger the cascade release of CO, leading to lethal lipid peroxidation and further enhancing ferroptosis-mediated tumor therapy. This nanoplatform demonstrates robust anti-tumor efficacy under US irradiation with favorable biosafety in both subcutaneous and orthotopic HCC models, representing a promising therapeutic approach for HCC. Additionally, the findings offer new insights into tumor microenvironment modulation to optimize US-triggered multi-modal cancer therapy.
Collapse
Affiliation(s)
- Wen Meng
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Ting Chen
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Xueping Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Yi Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Lu Zhang
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| | - Yigang Xu
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
- Key Laboratory of Bionic EngineeringMinistry of EducationJilin UniversityChangchun130022China
| | - Tianqiang Song
- Department of Hepatobiliary CancerLiver Cancer CenterTianjin Medical University Cancer Institute & HospitalNational Clinical Research Center for CancerTianjin Key Laboratory of Digestive CancerTianjin's Clinical Research Center for CancerTianjin300060China
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive MaterialsMinistry of EducationFrontiers Science Center for Cell Responsesand College of Life SciencesNankai UniversityTianjin300071China
| | - Qingqing Xiong
- Department of Hepatobiliary CancerLiver Cancer CenterTianjin Medical University Cancer Institute & HospitalNational Clinical Research Center for CancerTianjin Key Laboratory of Digestive CancerTianjin's Clinical Research Center for CancerTianjin300060China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer ImmunotherapyInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192China
| |
Collapse
|
9
|
Peng Y, Hu C, Zhang L, Dong F, Li R, Liang H, Dai H, Jang WJ, Cheng HB, Zhou L, Wang Y, Yoon J. Harnessing Dual Phototherapy and Immune Activation for Cancer Treatment: The Development and Application of BODIPY@F127 Nanoparticles. Adv Healthc Mater 2024; 13:e2401981. [PMID: 39073014 DOI: 10.1002/adhm.202401981] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/17/2024] [Indexed: 07/30/2024]
Abstract
Conventional phototherapeutic agents are typically used in either photodynamic therapy (PDT) or photothermal therapy (PTT). However, efficacy is often hindered by hypoxia and elevated levels of heat shock proteins in the tumor microenvironment (TME). To address these limitations, a formylated, near-infrared (NIR)-absorbing and heavy-atom-free Aza-BODIPY dye is presented that exhibits both type-I and type-II PDT actions with a high yield of reactive oxygen species (ROS) and manifests efficient photothermal conversion by precise adjustments to the conjugate structure and electron distribution, leading to a large amount of ROS production even under severe hypoxia. To improve biosafety and water solubility, the dye with an amphiphilic triblock copolymer (Pluronic F-127), yielding BDP-6@F127 nanoparticles (NPs) is coated. Furthermore, inspired by the fact that phototherapy triggers the release of tumor-associated antigens, a strategy that leverages potential immune activation by combining PDT/PTT with immune checkpoint blockade (ICB) therapy to amplify the systemic immune response and achieve the much-desired abscopal effect is developed. In conclusion, this study presents a promising molecular design strategy that integrates multimodal therapeutics for a precise and effective approach to cancer therapy.
Collapse
Affiliation(s)
- Yang Peng
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue Haidian District, Beijing, 100081, P. R. China
| | - Chenyan Hu
- State Key Laboratory of Organic-Inorganic Composites, Key Lab of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, 15 North Third Ring Road, Beijing, 100029, P. R. China
| | - Ludan Zhang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue Haidian District, Beijing, 100081, P. R. China
| | - Fan Dong
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue Haidian District, Beijing, 100081, P. R. China
| | - Ruwan Li
- State Key Laboratory of Organic-Inorganic Composites, Key Lab of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, 15 North Third Ring Road, Beijing, 100029, P. R. China
| | - Huihui Liang
- Henan Provincial Key Laboratory of Surface & Interface Science, School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, China
| | - Hao Dai
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue Haidian District, Beijing, 100081, P. R. China
| | - Won Jun Jang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea
| | - Hong-Bo Cheng
- State Key Laboratory of Organic-Inorganic Composites, Key Lab of Biomedical Materials of Natural Macromolecules, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, 15 North Third Ring Road, Beijing, 100029, P. R. China
| | - Liming Zhou
- Henan Provincial Key Laboratory of Surface & Interface Science, School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450002, China
| | - Yuguang Wang
- Department of General Dentistry II, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue Haidian District, Beijing, 100081, P. R. China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, South Korea
| |
Collapse
|
10
|
Li W, Chen D, Min C, Ma X, Yang X, Wang J. Atomically Engineered Chlorine Coordination of Iron in Active Centers for Selectively Catalytic H 2O 2 Decomposition Toward Efficient Antitumor-Specific Therapy. Adv Healthc Mater 2024:e2401267. [PMID: 39221675 DOI: 10.1002/adhm.202401267] [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: 04/06/2024] [Revised: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The intervention of endogenous H2O2 via nanozymes provides a potential antitumor-specific therapy; however, the role of the nanozyme structure in relation to the selective decomposition of H2O2 to hydroxyl radicals (•OH) is yet to be fully understood, which limits the development of this therapeutic approaches. Herein, an iron single-atom nanozyme (Fe─N2Cl2─C SAzyme) is reported, which is prepared through precise Fe─Cl coordination based on the construction of a characteristic Fe-containing molecule. Fe─N2Cl2─C exhibits efficient catalytic H2O2 decomposition (2.19 × 106 mm-1 s-1), which is the highest among reported SAzymes. More importantly, it is found that H2O2 selectively decomposed into •OH on the Fe─N2Cl2─C surface, which is attributable to the d orbitals of the Fe active center matching the O-2p electrons of the adsorbed hydroxide (*OH) intermediate. Fe─N2Cl2─C is strongly cytotoxic toward a variety of cancer-cell lines in vitro but not to normal cells. Furthermore, Fe─N2Cl2─C shows an outstanding specific therapeutic effect in vivo; it efficiently destroys solid malignant tumors without injuring normal tissue. Altogether, these findings highlight the selective catalytic decomposition of H2O2 to •OH, which is achieved by engineering the active center on the atomic level, thereby providing an avenue for the development of specific nanomedicines with efficient antitumor activities.
Collapse
Affiliation(s)
- Wei Li
- School of Chemical Sciences and Technology, Yunnan Province Engineering Research Center of Photocatalytic Treatment of Industrial Wastewater, Yunnan University, Kunming, 650091, China
| | - Daomei Chen
- School of Materials and Energy, Yunnan Province Engineering Research Center of Photocatalytic Treatment of Industrial Wastewater, Yunnan University, Kunming, 650091, China
| | - Chungang Min
- Research Center for Analysis and Measurement, Kunming University of Science and Technology, Kunming, 650093, China
| | - Xiaoqian Ma
- School of Chemical Sciences and Technology, Yunnan Province Engineering Research Center of Photocatalytic Treatment of Industrial Wastewater, Yunnan University, Kunming, 650091, China
| | - Xikun Yang
- Research Center for Analysis and Measurement, Kunming University of Science and Technology, Kunming, 650093, China
| | - Jiaqiang Wang
- School of Chemical Sciences and Technology, Yunnan Province Engineering Research Center of Photocatalytic Treatment of Industrial Wastewater, Yunnan University, Kunming, 650091, China
- School of Materials and Energy, Yunnan Province Engineering Research Center of Photocatalytic Treatment of Industrial Wastewater, Yunnan University, Kunming, 650091, China
| |
Collapse
|
11
|
Wang Y, Zhang Q, Kan M, Chang F, He X, Cheng N, Huang K. Multi-omics analysis of Au@Pt nanozyme for the modulation of glucose and lipid metabolism. J Nanobiotechnology 2024; 22:524. [PMID: 39217399 PMCID: PMC11365142 DOI: 10.1186/s12951-024-02807-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Au@Pt nanozyme, a bimetallic core-shell structure Au and Pt nanoparticle, has attracted significant attention due to its excellent catalytic activity and stability. Here, we propose that Au@Pt improves glucose tolerance and reduces TG after four weeks administration. The transcriptomic analysis of mouse liver tissues treated with Au@Pt nanozyme showed changes in genes related to glucose and lipid metabolism signaling pathways, including glycolysis/gluconeogenesis, pyruvate metabolism, PPAR signaling, and insulin signaling. Moreover, analysis of fecal samples from mice treated with Au@Pt nanozyme showed significant changes in the abundance of beneficial gut microbiota such as Dubosiella, Parvibacter, Enterorhabdus, Monoglobus, Lachnospiraceae_UCG-008, Lachnospiraceae_UCG-006, Lachnospiraceae_UCG-001, and Christensenellaceae_R-7_group. Combined multi-omics correlation analyses revealed that the modulation of glucose and lipid metabolism by Au@Pt was strongly correlated with changes in hepatic gene expression profiles as well as changes in gut microbial profiles. Overall, our integrated multi-omics analysis demonstrated that Au@Pt nanozyme could modulate glucose and lipid metabolism by regulating the expression of key genes in the liver and altering the composition of gut microbiota, providing new insights into the potential applications of Au@Pt nanozyme in the treatment of metabolic disorder.
Collapse
Affiliation(s)
- Yanan Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Qi Zhang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Minrui Kan
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Fei Chang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Xiaoyun He
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Nan Cheng
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China
| | - Kunlun Huang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China.
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the PR China, Beijing, China.
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, People's Republic of China.
| |
Collapse
|