1
|
Deng C, Ye Z, Zheng CJ, Cheng H, Ge J. Iron-based nanozymes induced ferroptosis for tumor therapy. NANOSCALE 2025. [PMID: 40370315 DOI: 10.1039/d5nr00880h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Iron-based nanozymes are an emerging class of nanomaterials demonstrating significant potential in tumor therapy by inducing ferroptosis-a regulated form of cell death marked by iron-mediated lipid peroxidation (LPO). These nanozymes exhibit unique enzymatic activities, including peroxidase, oxidase, and glutathione oxidase-like functions, enabling them to generate reactive oxygen species (ROS) and disrupt tumor microenvironment homeostasis. Leveraging Fenton chemistry, iron-based nanozymes amplify oxidative stress within tumor cells, thereby overcoming therapeutic challenges such as drug resistance and nonspecific toxicity. Despite significant advancements, the precise mechanisms by which iron-based nanozymes influence ferroptosis and their therapeutic efficacy remain underexplored. This review systematically categorizes these iron-based nanozymes, including iron oxides, single-atom enzymes, and metal-organic frameworks. We further elucidate their mechanisms in enhancing ferroptosis, focusing on their structural attributes, ROS generation pathways, and their enzymatic activities. Additionally, we summarized their biochemical applications alongside challenges in biosafety, nanozyme specificity, and advanced design and analysis approaches essential for maximizing their therapeutic efficacy.
Collapse
Affiliation(s)
- Chi Deng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong 999077, China.
| | - Zichen Ye
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong 999077, China.
| | | | - Hongfei Cheng
- Institute of New Energy for Vehicles, School of Material Science and Engineering, Tongji University, Shanghai 201804, P.R. China
| | - Jingjie Ge
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong 999077, China.
| |
Collapse
|
2
|
Sun R, Liu R, Tian Y, Li Y, Fan B, Li S. Removing Barriers to Tumor 'Oxygenation': Depleting Glutathione Nanozymes in Cancer Therapy. Int J Nanomedicine 2025; 20:5613-5643. [PMID: 40331231 PMCID: PMC12051984 DOI: 10.2147/ijn.s515734] [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: 01/04/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Nanozymes are nanomaterials capable of mimicking natural enzyme catalysis in the complex biological environment of the human body. Due to their good stability and strong catalytic properties, nanozymes are widely used in various fields of biomedicine. Among them, nanozymes that trigger intracellular reactive oxygen species (ROS) levels for cancer therapy have gained significant attention. However, the 'explosion' of ROS in tumor cells was prevented by the high levels of glutathione (GSH) in the tumor microenvironment (TME). GSH, a prominent endogenous antioxidant, increases the resistance of tumor cells to oxidative stress by scavenging ROS. Certain nanozymes can deplete intracellular GSH levels by mimicking GSH oxidase (GSHOx), GSH peroxidase (GPx) or by interfering with the reduction of oxidized glutathione (GSSG). On the one hand, elevated the level of intracellular ROS and induced lipid peroxidation reaction leading to ferroptosis. On the other hand, it creates favorable conditions for the treatment of tumors with photodynamic therapy (PDT), sonodynamic therapy (SDT), chemodynamical therapy (CDT) and targeted therapy. In this paper, we present a comprehensive analysis of GSH-depleting nanozymes reported in recent years, including classification, mechanism, responsiveness to TME and their roles in cancer therapy, and look forward to future applications and developments.
Collapse
Affiliation(s)
- Ruilong Sun
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
- Gansu Provincial Key Laboratory of Stem Cells and Gene Drugs, Lanzhou, People’s Republic of China
| | - Ruitang Liu
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yongzheng Tian
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Yunfei Li
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Bo Fan
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| | - Songkai Li
- Spine Surgery, The 940th Hospital of the Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou, People’s Republic of China
| |
Collapse
|
3
|
Xiong K, Luo G, Zeng W, Wen G, Wang C, Ding A, Qi M, Liu Y, Zhang J. Magnetic Microbubbles Combined with ICG-Loaded Liposomes for Synergistic Mild-Photothermal and Ferroptosis-Enhanced Photodynamic Therapy of Melanoma. Int J Nanomedicine 2025; 20:2901-2921. [PMID: 40093542 PMCID: PMC11908402 DOI: 10.2147/ijn.s503753] [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: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background Melanoma poses a significant threat to human health due to the lack of effective treatment options. Previous studies have demonstrated that the combination of photothermal therapy (PTT) and photodynamic therapy (PDT) can enhance therapeutic efficacy. However, conventional PTT/PDT combination strategies face various challenges, including complex preparation processes, potential damage to healthy tissues, and insufficient generation of reactive oxygen species (ROS). This study aims to design a rational and efficient PTT/PDT therapeutic strategy for melanoma and to explore its underlying mechanisms. Methods We first synthesized two target materials, indocyanine green-targeted liposomes (ICG-Lips) and magnetic microbubbles (MMBs), using the thin-film hydration method, followed by characterization and performance evaluation of both materials. Subsequently, we evaluated the synergistic therapeutic effects and underlying mechanisms of ICG-Lips combined with MMBs in melanoma treatment through in vitro experiments using cellular models and in vivo experiments using animal models. Results Herein, we developed a multifunctional system comprising ICG-Lips and MMBs. ICG-Lips enhance targeted delivery through specific binding to the S100B protein on melanoma cells, while MMBs, via ultrasound (US)-induced cavitation effects, shorten the uptake time of ICG-Lips by melanoma cells and improve uptake efficiency. Furthermore, the combination of ICG-Lips and MMBs induces significant reactive oxygen species (ROS) generation. Under 808 nm laser irradiation, the accumulation of ICG-Lips in melanoma cells achieves mild photothermal therapy (mPTT) and PDT effects. The elevated temperature and excessive ROS generated during these processes result in glutathione (GSH) depletion, ultimately triggering ferroptosis. The occurrence of ferroptosis further amplifies PDT efficacy, creating a synergistic effect that effectively suppresses melanoma growth. Additionally, the combined therapeutic strategy of ICG-Lips and MMBs demonstrates excellent biosafety. Conclusion In summary, this study presents a novel and straightforward strategy that integrates mPTT, PDT, and ferroptosis synergistically to combat melanoma, thereby laying a solid foundation for improving melanoma treatment outcomes.
Collapse
Affiliation(s)
- Kaifen Xiong
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Guanghong Luo
- Department of Radiation Oncology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Wei Zeng
- Department of Ultrasonography, Shenzhen People’s Hospital, second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Guanxi Wen
- Department of Ultrasonography, Shenzhen People’s Hospital, second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Chong Wang
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, Guangdong, People’s Republic of China
- Department of Geriatrics, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Aijia Ding
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Min Qi
- Department of Plastic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, People’s Republic of China
| | - Yingying Liu
- Department of Ultrasonography, Shenzhen People’s Hospital, second Clinical Medical College of Jinan University, First Clinical Medical College of Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, Guangdong, People’s Republic of China
- Department of Geriatrics, Shenzhen People’s Hospital, The second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
4
|
Yang L, Zhang WH, Li Y, An YL, Wu YM, Gu N, Teng GJ. Oxygen Nanobubbles Enhance ICG/Fe(III)-Mediated Dual-Modal Therapy To Induce Ferroptosis in Tumor Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11718-11730. [PMID: 39963718 DOI: 10.1021/acsami.4c19604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Noninvasive therapies such as photodynamic therapy (PDT) and chemodynamic therapy (CDT), which rely on reactive oxygen species (ROS), are gaining attention for their low toxicity. However, single-modal treatments have individual limitations that restrict the therapeutic efficacy. Fe(III) can coordinate with the hydrophilic regions of indocyanine green (ICG) molecules to form the ICG/Fe(III) complex, making it a promising dual-modal agent for combined PDT and CDT. However, coordination with Fe(III) leads to the aggregation quenching of ICG, hindering its application in dual-modal therapy. We innovatively utilize oxygen nanobubbles, prepared solely from water and oxygen, to significantly reverse the aggregation-induced quenching of the ICG/Fe(III) complex, thereby enhancing its stability in aqueous environments. In this system, Fe(III) assembles at the nanobubble interface, coordinating with ICG's hydrophilic regions to form the ICG/Fe(III)-NBs. The oxygen nanobubbles boost PDT efficiency by improving the ICG/Fe(III) complex stability and oxygen content, while Fe(III) achieves CDT by generating hydroxyl radicals (•OH) through the Fenton reaction. This dual-modality treatment significantly disrupts the tumor's redox balance, induces ferroptosis, and demonstrates strong antitumor efficacy, reducing tumor volume to 34% of its initial size in mice. The strategy offers a promising and clinically viable approach to cancer treatment.
Collapse
Affiliation(s)
- Li Yang
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Wei-Hua Zhang
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Yan Li
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210009, China
| | - Yan-Li An
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Ye-Ming Wu
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210093, China
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| |
Collapse
|
5
|
Wang Y, Xu Y, Qu Y, Jin Y, Cao J, Zhan J, Li Z, Chai C, Huang C, Li M. Ferroptosis: A novel cell death modality as a synergistic therapeutic strategy with photodynamic therapy. Photodiagnosis Photodyn Ther 2025; 51:104463. [PMID: 39736368 DOI: 10.1016/j.pdpdt.2024.104463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
Although there has been significant progress in current comprehensive anticancer treatments centered on surgery, postoperative recurrence and tumor metastasis still significantly affect both prognosis and quality of life of the patient. Hence, the development of precisely targeted tumor therapies and exploration of immunotherapy represent additional strategies for tumor treatment. Photodynamic therapy (PDT) is a relatively safe treatment modality that not only induces multiple modes of tumor cell death but also mediates the secondary immunological responses against tumor resistance and metastasis. Ferroptosis, an iron-dependent type of programmed cell death characterized by accumulation of reactive oxygen species and lipid peroxidation products to lethal levels, has emerged as an attractive target trigger for tumor therapies. Recent research has revealed a close association between PDT and ferroptosis, suggesting that combining ferroptosis inducers with PDT could strengthen their synergistic anti-tumor efficiency. Here in this review, we discuss the rationale for combining PDT with ferroptosis inducers and highlight the progress of single-molecule photosensitizers to induce ferroptosis, as well as the applications of photosensitizers combined with other therapeutic drugs for collaborative therapy. Furthermore, given the current research dilemma, we propose potential therapeutic strategies to advance the combined usage of PDT and ferroptosis inducers, providing the basis and guidelines for prospective clinical translation and research directionality with regard to PDT.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiting Xu
- Central Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan 430022, China
| | - Yong Qu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yifang Jin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Juanmei Cao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Dermatology, First Affiliated Hospital, Shihezi University, Shihezi 832008, China
| | - Jinshan Zhan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhuoxia Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chuxing Chai
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
6
|
Zhu L, Ren Y, Dong M, Sun B, Huang J, Chen L, Xia X, Dong X, Zheng C. Ultrasmall Metal TPZ Complexes with Deep Tumor Penetration for Enhancing Radiofrequency Ablation Therapy and Inducing Antitumor Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311244. [PMID: 38898764 DOI: 10.1002/smll.202311244] [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: 12/04/2023] [Revised: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Radiofrequency ablation (RFA) is one of the most common minimally invasive techniques for the treatment of solid tumors, but residual malignant tissues or small satellite lesions after insufficient RFA (iRFA) are difficult to remove, often leading to metastasis and recurrence. Here, Fe-TPZ nanoparticles are designed by metal ion and (TPZ) ligand complexation for synergistic enhancement of RFA residual tumor therapy. Fe-TPZ nanoparticles are cleaved in the acidic microenvironment of the tumor to generate Fe2+ and TPZ. TPZ, an anoxia-dependent drug, is activated in residual tumors and generates free radicals to cause tumor cell death. Elevated Fe2+ undergoes a redox reaction with glutathione (GSH), inducing a strong Fenton effect and promoting the production of the highly toxic hydroxyl radical (•OH). In addition, the ROS/GSH imbalance induced by this treatment promotes immunogenic cell death (ICD), which triggers the release of damage-associated molecular patterns, macrophage polarization, and lymphocyte infiltration, thus triggering a systemic antitumor immune response and noteworthy prevention of tumor metastasis. Overall, this integrated treatment program driven by multiple microenvironment-dependent pathways overcomes the limitations of the RFA monotherapy approach and thus improves tumor prognosis. Furthermore, these findings aim to provide new research ideas for regulating the tumor immune microenvironment.
Collapse
Affiliation(s)
- Licheng Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengna Dong
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bo Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jia Huang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
7
|
Deng Y, Huang S, Jiang G, Zhou L, Nezamzadeh-Ejhieh A, Liu J, Zhou Z. Current status and prospects of MOFs loaded with H 2O 2-related substances for ferroptosis therapy. RSC Med Chem 2024; 15:2996-3016. [PMID: 39309362 PMCID: PMC11411616 DOI: 10.1039/d4md00261j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a programmed cell death mechanism characterized by the accumulation of iron (Fe)-dependent lipid peroxides within cells. Ferroptosis holds excellent promise in tumor therapy. Metal-organic frameworks (MOFs) offer unique advantages in tumor ferroptosis treatment due to their high porosity, excellent stability, high biocompatibility, and targeting capabilities. Inducing ferroptosis in tumor cells primarily involves the production of reactive oxygen species (ROS), like hydroxyl radicals (˙OH), through iron-mediated Fenton reactions. However, the intrinsic H2O2 levels in tumor cells are often insufficient to sustain prolonged consumption, limiting therapeutic efficacy if ˙OH production is inadequate. Therefore, catalyzing or supplementing the intracellular H2O2 levels in tumor cells is essential for inducing ferroptosis by nanoscale metal-organic frameworks. This article reviews the biological characteristics and molecular mechanisms of ferroptosis, introduces H2O2-related substances, and reviews MOF-based nanoscale strategies for enhancing intracellular H2O2 levels in tumor cells. Finally, the challenges and prospects of this approach are discussed, aiming to provide insights into improving the effectiveness of ferroptosis induced by MOFs.
Collapse
Affiliation(s)
- Yu Deng
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Sida Huang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Guanming Jiang
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital) 78 Wandao Road South Dongguan 523059 Guangdong China
| | - Luyi Zhou
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | | | - Jianqiang Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Zhikun Zhou
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| |
Collapse
|
8
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self-Catalytic NO/O 2 Gas-Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403921. [PMID: 39101290 DOI: 10.1002/adma.202403921] [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: 03/17/2024] [Revised: 06/29/2024] [Indexed: 08/06/2024]
Abstract
Radiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor-targeted "prosthetic-Arginine" coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO-donating Fmoc-protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu-HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2-transporting ability of heme, HRRu-HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu-HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu-HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment, Henan, 450001, China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
9
|
Wang S, Cheng M, Wang S, Jiang W, Yang F, Shen X, Zhang L, Yan X, Jiang B, Fan K. A Self‐Catalytic NO/O 2 Gas‐Releasing Nanozyme for Radiotherapy Sensitization through Vascular Normalization and Hypoxia Relief. ADVANCED MATERIALS 2024. [DOI: doi:10.1002/adma.202403921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Indexed: 04/16/2025]
Abstract
AbstractRadiotherapy (RT), essential for treating various cancers, faces challenges from tumor hypoxia, which induces radioresistance. A tumor‐targeted “prosthetic‐Arginine” coassembled nanozyme system, engineered to catalytically generate nitric oxide (NO) and oxygen (O2) in the tumor microenvironment (TME), overcoming hypoxia and enhancing radiosensitivity is presented. This system integrates the prosthetic heme of nitric oxide synthase (NOS) and catalase (CAT) with NO‐donating Fmoc‐protected Arginine and Ru3+ ions, creating HRRu nanozymes that merge NOS and CAT functionalities. Surface modification with human heavy chain ferritin (HFn) improves the targeting ability of nanozymes (HRRu‐HFn) to tumor tissues. In the TME, strategic arginine incorporation within the nanozyme allows autonomous O2 and NO release, triggered by endogenous hydrogen peroxide, elevating NO and O2 levels to normalize vasculature and improve blood perfusion, thus mitigating hypoxia. Employing the intrinsic O2‐transporting ability of heme, HRRu‐HFn nanozymes also deliver O2 directly to the tumor site. Utilizing esophageal squamous cell carcinoma as a tumor model, the studies reveal that the synergistic functions of NO and O2 production, alongside targeted delivery, enable the HRRu‐HFn nanozymes to combat tumor hypoxia and potentiate radiotherapy. This HRRu‐HFn nanozyme based approach holds the potential to reduce the radiation dose required and minimize side effects associated with conventional radiotherapy.
Collapse
Affiliation(s)
- Shuyu Wang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Miaomiao Cheng
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Shenghui Wang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Wei Jiang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
| | - Feifei Yang
- College of Chemistry and Chemical Engineering Jiangxi Normal University Nanchang 330022 China
| | - Xiaomei Shen
- College of Chemistry and Chemical Engineering Jiangxi Normal University Nanchang 330022 China
| | - Lirong Zhang
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Henan 450001 China
| | - Xiyun Yan
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
- Nanozyme Laboratory in Zhongyuan Henan Academy of Innovations in Medical Science Zhengzhou Henan 451163 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Biomacromolecules (CAS) CAS Center for Excellence in Biomacromolecules Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| | - Bing Jiang
- Nanozyme Laboratory in Zhongyuan School of Basic Medical Sciences Zhengzhou University Zhengzhou Henan 450001 China
- Nanozyme Laboratory in Zhongyuan Henan Academy of Innovations in Medical Science Zhengzhou Henan 451163 China
| | - Kelong Fan
- Nanozyme Laboratory in Zhongyuan Henan Academy of Innovations in Medical Science Zhengzhou Henan 451163 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Biomacromolecules (CAS) CAS Center for Excellence in Biomacromolecules Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| |
Collapse
|
10
|
Adzavon KP, Zhao W, He X, Sheng W. Ferroptosis resistance in cancer cells: nanoparticles for combination therapy as a solution. Front Pharmacol 2024; 15:1416382. [PMID: 38962305 PMCID: PMC11219589 DOI: 10.3389/fphar.2024.1416382] [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: 04/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Ferroptosis is a form of regulated cell death (RCD) characterized by iron-dependent lipid peroxidation. Ferroptosis is currently proposed as one of the most promising means of combating tumor resistance. Nevertheless, the problem of ferroptosis resistance in certain cancer cells has been identified. This review first, investigates the mechanisms of ferroptosis induction in cancer cells. Next, the problem of cancer cell resistance to ferroptosis, as well as the underlying mechanisms is discussed. Recently discovered ferroptosis-suppressing biomarkers have been described. The various types of nanoparticles that can induce ferroptosis are also discussed. Given the ability of nanoparticles to combine multiple agents, this review proposes nanoparticle-based ferroptosis cell death as a viable method of circumventing this resistance. This review suggests combining ferroptosis with other forms of cell death, such as apoptosis, cuproptosis and autophagy. It also suggests combining ferroptosis with immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Wang Sheng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
11
|
Su Y, Liu B, Wang B, Chan L, Xiong C, Lu L, Zhang X, Zhan M, He W. Progress and Challenges in Tumor Ferroptosis Treatment Strategies: A Comprehensive Review of Metal Complexes and Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310342. [PMID: 38221682 DOI: 10.1002/smll.202310342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/27/2023] [Indexed: 01/16/2024]
Abstract
Ferroptosis is a new form of regulated cell death featuring iron-dependent lipid peroxides accumulation to kill tumor cells. A growing body of evidence has shown the potential of ferroptosis-based cancer therapy in eradicating refractory malignancies that are resistant to apoptosis-based conventional therapies. In recent years, studies have reported a number of ferroptosis inducers that can increase the vulnerability of tumor cells to ferroptosis by regulating ferroptosis-related signaling pathways. Encouraged by the rapid development of ferroptosis-driven cancer therapies, interdisciplinary fields that combine ferroptosis, pharmaceutical chemistry, and nanotechnology are focused. First, the prerequisites and metabolic pathways for ferroptosis are briefly introduced. Then, in detail emerging ferroptosis inducers designed to boost ferroptosis-induced tumor therapy, including metal complexes, metal-based nanoparticles, and metal-free nanoparticles are summarized. Subsequently, the application of synergistic strategies that combine ferroptosis with apoptosis and other regulated cell death for cancer therapy, with emphasis on the use of both cuproptosis and ferroptosis to induce redox dysregulation in tumor and intracellular bimetallic copper/iron metabolism disorders during tumor treatment is discussed. Finally, challenges associated with clinical translation and potential future directions for potentiating cancer ferroptosis therapies are highlighted.
Collapse
Affiliation(s)
- Yanhong Su
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Bing Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Binghan Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Leung Chan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Chan Xiong
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Xuanjun Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
- MOE Frontiers Science Centre for Precision Oncology, University of Macau, Macau SAR, 999078, China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Weiling He
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| |
Collapse
|
12
|
Liu C, Wang G, Han W, Tian Q, Li M. Ferroptosis: a potential therapeutic target for stroke. Neural Regen Res 2024; 19:988-997. [PMID: 37862200 PMCID: PMC10749612 DOI: 10.4103/1673-5374.385284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/05/2023] [Accepted: 08/03/2023] [Indexed: 10/22/2023] Open
Abstract
Ferroptosis is a form of regulated cell death characterized by massive iron accumulation and iron-dependent lipid peroxidation, differing from apoptosis, necroptosis, and autophagy in several aspects. Ferroptosis is regarded as a critical mechanism of a series of pathophysiological reactions after stroke because of iron overload caused by hemoglobin degradation and iron metabolism imbalance. In this review, we discuss ferroptosis-related metabolisms, important molecules directly or indirectly targeting iron metabolism and lipid peroxidation, and transcriptional regulation of ferroptosis, revealing the role of ferroptosis in the progression of stroke. We present updated progress in the intervention of ferroptosis as therapeutic strategies for stroke in vivo and in vitro and summarize the effects of ferroptosis inhibitors on stroke. Our review facilitates further understanding of ferroptosis pathogenesis in stroke, proposes new targets for the treatment of stroke, and suggests that more efforts should be made to investigate the mechanism of ferroptosis in stroke.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Guijun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Wenrui Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
13
|
Chang Q, Wang P, Zeng Q, Wang X. A review on ferroptosis and photodynamic therapy synergism: Enhancing anticancer treatment. Heliyon 2024; 10:e28942. [PMID: 38601678 PMCID: PMC11004815 DOI: 10.1016/j.heliyon.2024.e28942] [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: 12/01/2023] [Revised: 03/11/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Ferroptosis is an iron-dependent programmed cell death modality, which has showed great potential in anticancer treatment. Photodynamic therapy (PDT) is widely used in clinic as an anticancer therapy. PDT combined with ferroptosis-promoting therapy has been found to be a promising strategy to improve anti-cancer therapy efficacy. Fenton reaction in ferroptosis can provide oxygen for PDT, and PDT can produce reactive oxygen species for Fenton reaction to enhance ferroptosis. In this review, we briefly present the importance of ferroptosis in anticancer treatment, mechanism of ferroptosis, researches on PDT induced ferroptosis, and the mechanism of the synergistic effect of PDT and ferroptosis on cancer killing.
Collapse
Affiliation(s)
- Qihang Chang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Peiru Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qingyu Zeng
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
14
|
Wang W, Gao Y, Xu J, Zou T, Yang B, Hu S, Cheng X, Xia Y, Zheng Q. A NRF2 Regulated and the Immunosuppressive Microenvironment Reversed Nanoplatform for Cholangiocarcinoma Photodynamic-Gas Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307143. [PMID: 38308097 PMCID: PMC11005733 DOI: 10.1002/advs.202307143] [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] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/03/2024] [Indexed: 02/04/2024]
Abstract
Photodynamic therapy (PDT) is a minimally invasive and controllable local cancer treatment for cholangiocarcinoma (CCA). However, the efficacy of PDT is hindered by intratumoral hypoxia and the presence of an antioxidant microenvironment. To address these limitations, combining PDT with gas therapy may be a promising strategy to enhance tumor oxygenation. Moreover, the augmentation of oxidative damage induced by PDT and gas therapy can be achieved by inhibiting NRF2, a core regulatory molecule involved in the antioxidant response. In this study, an integrated nanotherapeutic platform called CMArg@Lip, incorporating PDT and gas therapies using ROS-responsive liposomes encapsulating the photosensitizer Ce6, the NO gas-generating agent L-arginine, and the NRF2 inhibitor ML385, is successfully developed. The utilization of CMArg@Lip effectively deals with challenges posed by tumor hypoxia and antioxidant microenvironment, resulting in elevated levels of oxidative damage and subsequent induction of ferroptosis in CCA. Additionally, these findings suggest that CMArg@Lip exhibits notable immunomodulatory effects, including the promotion of immunogenic cell death and facilitation of dendritic cell maturation. Furthermore, it contributes to the anti-tumor function of cytotoxic T lymphocytes through the downregulation of PD-L1 expression in tumor cells and the activation of the STING signaling pathway in myeloid-derived suppressor cells, thereby reprogramming the immunosuppressive microenvironment via various mechanisms.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Liver Transplant CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yang Gao
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jianjun Xu
- Liver Transplant CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Tianhao Zou
- Liver Transplant CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Bin Yang
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Liver Transplant CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Shaobo Hu
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Liver Transplant CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xiang Cheng
- Department of Digestive Oncology SurgeryCancer CentreUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yun Xia
- Department of General SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Qichang Zheng
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Liver Transplant CenterUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
15
|
Zhu J, Wen T, Qu S, Li Q, Liu B, Zhou W. G-Quadruplex/Hemin DNAzyme-Functionalized Silver Nanoclusters with Synergistic Antibacterial and Wound Healing Capabilities for Infected Wound Management. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307220. [PMID: 37828643 DOI: 10.1002/smll.202307220] [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: 09/03/2023] [Revised: 09/28/2023] [Indexed: 10/14/2023]
Abstract
Systematic management of infected wounds requires simultaneous antiinfection and wound healing, which has become the current treatment dilemma. Recently, a multifunctional silver nanoclusters (AgNCs)-based hydrogel dressing to meet these demands is developed. Here a diblock DNA with a cytosine-rich fragment (as AgNCs template) and a guanine-rich fragment (to form G-quadruplex/hemin DNAzyme, termed G4/hemin) is designed, for G4/hemin functionalization of AgNCs. Inside bacteria, G4/hemin can not only accelerate the oxidative release of Ag+ from AgNCs but also generate reactive oxygen species (ROS) via catalase- and peroxidase-mimic activities, which enhance the antibacterial effect. On the other hand, the AgNCs exhibit robust anti-inflammatory and antioxidative activities to switch M1 macrophages into M2 phenotype, which promotes wound healing. Moreover, the hemin is released to upregulate the heme oxygenase-1, an intracellular enzyme that can relieve oxidative stress, which significantly alleviates the cytotoxicity of silver. As a result, such silver-based dressing achieves potent therapeutic efficacy on infected wounds with excellent biosafety.
Collapse
Affiliation(s)
- Jiaojiao Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Tiao Wen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Shuangquan Qu
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Department of Anesthesiology, Hunan Children's Hospital, Changsha, Hunan, 410007, China
| | - Qingnian Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Biwu Liu
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
16
|
Li Z, Wei R, Yao S, Meng F, Kong L. HIF-1A as a prognostic biomarker related to invasion, migration and immunosuppression of cervical cancer. Heliyon 2024; 10:e24664. [PMID: 38298716 PMCID: PMC10828096 DOI: 10.1016/j.heliyon.2024.e24664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Background The incidence of cervical cancer ranks second among malignant tumors in women, exerting a significant impact on their quality of life and overall well-being. The hypoxic microenvironment plays a pivotal role in the initiation and progression of tumorigenesis. The present study aims to investigate the fundamental genes and pathways associated with the hypoxia-inducible factor (HIF-1A) in cervical cancer, aiming to identify potential downstream targets for diagnostic and therapeutic purposes. Methods We obtained dataset GSE63514 from the Comprehensive Gene Expression Database (GEO). The dataset comprised of 24 patients in the normal group and 28 patients in the tumor group. Gene set difference analysis (GSVA) and gene set enrichment analysis (GSEA) were used to identify the genes related to HIF-1A expression and the specific signaling pathways involved.The association between HIF-1A and tumor immune infiltration was examined in the TCGA dataset. The WGCAN network was constructed to identify key genes within the blue module, and subsequent gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted to determine the pathways and functional annotations associated with HIF-1A. The protein interaction network of the HIF-1A gene was obtained from the STRING database and visualized using Cytoscape in the meantime.The function of HIF-1A and its related gene expression were verified in vivo. Results HIF-1A was a risk factor in both univariate and multivariate Cox regression analysis of cervical cancer patients. A total of 344 genes significantly correlated with the expression of HIF-1A were identified through correlation analysis, and the genes exhibiting the strongest correlation were obtained. The major signaling pathways involved in HIF-1A encompass TNF-α/NF-κB, PI3K/AKT/MTOR, TGF-β, JAK-STAT, and various other signaling cascades. Reinforced by qRT-PCR, we identified Integrin beta-1 (ITGB1), C-C motif chemokine ligand 2 (CCL2), striatin 3 (STRN3), and endothelin-1 (EDN1) as pivotal downstream genes influenced by HIF-1A. HIF-1A is associated with immune infiltration of natural killer (NK) cells, mast cells, CD4+T cells, M0 macrophages, neutrophils, follicular helper T cells, CD8+T cells, and regulatory T cells (Treg). HIF-1A is associated with sensitivity to chemotherapy drugs. The identification of the HIF-1A pathway and its function primarily focuses on cytoplasmic translation, aerobic respiration, cellular respiration, oxidative phosphorylation, thermogenesis, among others. The results of in vivo experiments have confirmed that HIF-1A plays a crucial role in promoting the migration and invasion of cervical cancer cells. Moreover, the overexpression of HIF-1A led to an upregulation in the expressions of ITGB1, CCL2, STRN3, and EDN1. Conclusions The role of HIF-1A in cervical cancer was determined through a combination of bioinformatics analysis and experimental validation. The genes potentially implicated in the tumorigenesis mechanism of HIF-1A were identified. These findings has the potential to enhance our comprehension of the progression of cervical cancer and offer promising therapeutic targets for its clinical management.
Collapse
Affiliation(s)
- Zhenyu Li
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ran Wei
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shunyu Yao
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fang Meng
- Department of Oncology &Hematology, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Lingsuo Kong
- Department of Anesthesiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
17
|
Zhu J, Peng L, Jehan S, Wang H, Chen X, Zhao S, Zhou W. Activable Photodynamic DNA Probe with an "AND" Logic Gate for Precision Skin Cancer Therapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0295. [PMID: 38269029 PMCID: PMC10807844 DOI: 10.34133/research.0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/10/2023] [Indexed: 01/26/2024]
Abstract
Photodynamic therapy (PDT) has emerged as a promising approach for squamous cell carcinoma treatment but hindered by tumor hypoxia, acquired resistance, phototoxicity, and so on. To address these issues, we developed a smart strategy utilizing activable photosensitizers delivered by an aptamer-functionalized DNA probe (ADP). The ADP incorporated an AS1411 aptamer for tumor targeting and a linear antisense oligonucleotide (ASO) for recognition of Survivin mRNA. In the absence of the target, PDT remained quenched, thereby avoiding phototoxicity during circulation and nonselective distribution. With the aid of the aptamer, ADP achieved selective targeting of tumors. Upon internalization, ADP targeted recognized Survivin mRNA, triggering PDT activation, and releasing ASO to down-regulate Survivin expression and reverse tumor resistance. Consequently, the activable photosensitizers exhibited an "AND" logic gate, combining tumor-targeting delivery and tumor-related gene activation, thus enhancing its specificity. Additionally, the incorporation of hemin into the ADP provided catalase activity, converting tumor-abundant H2O2 into O2, thereby ameliorating tumor hypoxia. The resulting functionalized G-quadruplex/hemin-DNA probe complex demonstrated targeted delivery and activation, minimized side effects, and enhanced PDT efficacy in both xenograft tumor-bearing mice and patient-derived xenograft models. This study offers a unique and promising platform for efficient and safe PDT, thus holding great potential for future clinical translation and improved cancer therapy.
Collapse
Affiliation(s)
- Jiaojiao Zhu
- Xiangya School of Pharmaceutical Sciences,
Central South University, Changsha, Hunan 410013, China
| | - Lanyuan Peng
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital,
Central South University, Changsha, Hunan 410008, China
| | - Shah Jehan
- Xiangya School of Pharmaceutical Sciences,
Central South University, Changsha, Hunan 410013, China
- Department of Vascular Surgery,
The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Haiyang Wang
- Department of Vascular Surgery,
The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital,
Central South University, Changsha, Hunan 410008, China
| | - Shuang Zhao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital,
Central South University, Changsha, Hunan 410008, China
- Furong Laboratory, Changsha, Hunan, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences,
Central South University, Changsha, Hunan 410013, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Changsha, Hunan 410008, China
| |
Collapse
|
18
|
Wei L, Wang Z, Lu X, Chen J, Zhai Y, Huang Q, Pei S, Liu Y, Zhang W. Interfacial strong interaction-enabling cascade nanozymes for apoptosis-ferroptosis synergistic therapy. J Colloid Interface Sci 2024; 653:20-29. [PMID: 37708728 DOI: 10.1016/j.jcis.2023.09.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
Noble metal nanozymes are promising therapeutic agents due to their good ability of reactive oxygen species generation in response to the tumor microenvironment (TME). Achieving optimal performance of noble metal nanozymes at a minimum dosage is crucial due to potential systemic biotoxicity. In this study, we report the successful anchoring of Ir nanoclusters on Co(OH)2 nanosheets with an Ir content of 6.2 wt% (denoted as Ir6.2-Co(OH)2), which exhibits remarkable peroxidase (POD)- and catalase (CAT)-like activities. The strong electronic interaction at the Ir-O-Co interface endows glutathione peroxidase (GSH-Px)-like activity to the composite, ensuring efficient generation of reactive oxygen species (ROS) and deactivation of glutathione peroxidase 4 (GPX4) by supplementing hydrogen peroxide (H2O2) and depleting glutathione (GSH). Both in vitro and in vivo evaluations demonstrate that Ir6.2-Co(OH)2 nanozymes significantly enhance antitumor efficacy through apoptosis-ferroptosis synergistic therapy. This study highlights the tremendous potential of leveraging strong electronic interactions between noble metals and oxides for modulating enzyme-like activities towards high-efficiency synergistic therapies.
Collapse
Affiliation(s)
- Lineng Wei
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Ziyu Wang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Xiuxin Lu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Jingqi Chen
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Yujie Zhai
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Qinghua Huang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Department of Breast Surgery, Wuzhou Red Cross Hospital, Wuzhou 543000, China.
| | - Shenglin Pei
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Yan Liu
- Department of Breast, Bone and Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, Nanning, Nanning 530021, China; Laboratory of Breast Cancer Diagnosis and Treatment Research of Guangxi Department of Education, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Weiqing Zhang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China.
| |
Collapse
|
19
|
Pan Y, Liu L, Mou X, Cai Y. Nanomedicine Strategies in Conquering and Utilizing the Cancer Hypoxia Environment. ACS NANO 2023; 17:20875-20924. [PMID: 37871328 DOI: 10.1021/acsnano.3c07763] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Cancer with a complex pathological process is a major disease to human welfare. Due to the imbalance between oxygen (O2) supply and consumption, hypoxia is a natural characteristic of most solid tumors and an important obstacle for cancer therapy, which is closely related to tumor proliferation, metastasis, and invasion. Various strategies to exploit the feature of tumor hypoxia have been developed in the past decade, which can be used to alleviate tumor hypoxia, or utilize the hypoxia for targeted delivery and diagnostic imaging. The strategies to alleviate tumor hypoxia include delivering O2, in situ O2 generation, reprogramming the tumor vascular system, decreasing O2 consumption, and inhibiting HIF-1 related pathways. On the other side, hypoxia can also be utilized for hypoxia-responsive chemical construction and hypoxia-active prodrug-based strategies. Taking advantage of hypoxia in the tumor region, a number of methods have been applied to identify and keep track of changes in tumor hypoxia. Herein, we thoroughly review the recent progress of nanomedicine strategies in both conquering and utilizing hypoxia to combat cancer and put forward the prospect of emerging nanomaterials for future clinical transformation, which hopes to provide perspectives in nanomaterials design.
Collapse
Affiliation(s)
- Yi Pan
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Longcai Liu
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaozhou Mou
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yu Cai
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
20
|
Mo F, Zhong S, You T, Lu J, Sun D. Aptamer and DNAzyme-Functionalized Cu-MOF Hybrid Nanozymes for the Monitoring and Management of Bacteria-Infected Wounds. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37921634 DOI: 10.1021/acsami.3c10682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Metal-organic frameworks (MOFs) with peroxidase (POD)-like activity have great potential for combating drug-resistant bacterial infections. However, the use of POD-like activities is severely limited by low oxygen levels and high levels of glutathione (GSH) within the microenvironment of bacterial infection. Herein, G-quadruplex/hemin DNAzyme-aptamer probes and tannic acid-chelated Au nanoparticle (Au-TA)-decorated Cu-based MOF nanosheets (termed GATC) with triple-enzyme activities were developed for visual detection and efficient antibacterial therapy. First, the monometallic MOFs (Cu-ZIF) showed the best catalytic and loading capacity performance compared with the bimetallic MOFs (CoCu-ZIF and ZnCu-ZIF). Then, Cu-MOFs, Au-TA, and DNAzyme improve the POD-like activity to generate more hydroxyl radicals (•OH) to kill bacteria. GATC can bind to bacteria through aptamer recognition, increasing the bacterial surface contact area for efficient antibacterial activity. GATC can decompose H2O2 into O2 to alleviate hypoxia and improve the microenvironment due to its catalase (CAT)-like activity. In addition, GATC exhibited GSH peroxidase-like activity, which can avoid the loss of •OH and result in bacterial death more easily. Compared with previous studies, GATC exhibited extraordinary bactericidal ability at an extremely low dosage of 3 μg/mL against methicillin-resistant Staphylococcus aureus (MRSA). Notably, the GATC-catalyzed chromogenic reaction could accurately monitor the MRSA infection treatment process. Overall, this work could establish a therapeutic platform for the monitoring and management of bacteria-infected wounds.
Collapse
Affiliation(s)
- Fayin Mo
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Specialty of Clinical Pharmacy, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510699, Guangdong, China
| | - Sheng Zhong
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Tianhui You
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, China
| | - Duanping Sun
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
- Key Specialty of Clinical Pharmacy, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510699, Guangdong, China
| |
Collapse
|
21
|
Zhang X, Xu X, Liu H, Ni N, Liu S, Gong Y, Ma G, Song L, Meng Q, Fan Q, Sun X. CCR2-overexpressing biomimetic carrier-free nanoplatform for enhanced cascade ferroptosis tumor therapy. Acta Biomater 2023; 166:604-614. [PMID: 37156432 DOI: 10.1016/j.actbio.2023.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/18/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023]
Abstract
Ferroptosis-based nanoplatforms have shown great potential in cancer therapy. However, they also face issues such as degradation and metabolism. Carrier-free nanoplatforms consisting of active drugs can effectively avoid the security issues associated with additional carrier ingredients. Herein, a biomimetic carrier-free nanoplatform (HESN@CM) was designed to treat cancer by modulating cascade metabolic pathways of ferroptosis. CCR2-overexpressing macrophage membrane-modified HESN can target cancer cells via the CCR2-CCL2 axis. The acidic tumor microenvironment (TME) can disrupt the supramolecular interaction of HESN, releasing hemin and erastin. Then, erastin could induce cancer cells ferroptosis by inhibiting system XC- pathways, while hemin, a vital component of blood to transport oxygen, could be broken down by heme oxygenase-1 (HO-1), increasing the intracellular Fe2+ concentration to induce cancer cells' ferroptosis further. Meanwhile, erastin could enhance the activity of HO-1, further promoting the release of Fe2+ from hemin. As a result, HESN@CM demonstrated superior therapeutic efficacy in both primary and metastatic tumors in vitro and in vivo. The carrier-free HESN@CM provided cascade ferroptosis tumor therapy strategies for potential clinical application. STATEMENT OF SIGNIFICANCE: CCR2-overexpressing biomimetic carrier-free nanoplatform (HESN@CM) was designed for cancer treatment by modulating metabolic pathways of ferroptosis. HESN modified with CCR2-overexpressing macrophage membrane can target tumor cells via the CCR2-CCL2 axis. HESN was composed of hemin and erastin without additional vectors. Erastin could directly induce ferroptosis, while hemin could be broken down by heme oxygenase-1 (HO-1), increasing the intracellular Fe2+ concentration to enhance ferroptosis further. Meanwhile, erastin could improve the activity of HO-1, promoting the release of Fe2+ from hemin. Therefore, HESN@CM with good bioavailability, stability, and simple preparation can realize cascade ferroptosis tumor therapy and have the potential prospect of clinical translation.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan 250101, China
| | - Huimin Liu
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Shuangqing Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yufang Gong
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Guiqi Ma
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Linlin Song
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| | - Qing Fan
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| | - Xiao Sun
- Department of Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China.
| |
Collapse
|
22
|
Zhang Y, Yu W, Chen M, Zhang B, Zhang L, Li P. The applications of nanozymes in cancer therapy: based on regulating pyroptosis, ferroptosis and autophagy of tumor cells. NANOSCALE 2023; 15:12137-12156. [PMID: 37377098 DOI: 10.1039/d3nr01722b] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Nanozymes are nanomaterials with catalytic properties similar to those of natural enzymes, and they have recently been collectively identified as a class of innovative artificial enzymes. Nanozymes are widely used in various fields, such as biomedicine, due to their high catalytic activity and stability. Nanozymes can trigger changes in reactive oxygen species (ROS) levels in cells and the activation of inflammasomes, leading to the programmed cell death (PCD), including the pyroptosis, ferroptosis, and autophagy, of tumor cells. In addition, some nanozymes consume glucose, starving cancer cells and thus accelerating tumor cell death. In addition, the electric charge of the structure and the catalytic activity of nanozymes are sensitive to external factors such as light and electric and magnetic fields. Therefore, nanozymes can be used with different therapeutic methods, such as chemodynamic therapy (CDT), photodynamic therapy (PDT) and sonodynamic therapy (SDT), to achieve highly efficient antitumor effects. Many cancer therapies induce tumor cell death via the pyroptosis, ferroptosis, and autophagy of tumor cells mediated by nanozymes. We review the mechanisms of pyroptosis, ferroptosis, and autophagy in tumor development, as well as the potential application of nanozymes to regulate pyroptosis, ferroptosis, and autophagy in tumor cells.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China.
| | - Wanpeng Yu
- Medical Collage, Qingdao University, Qingdao, China
| | - Mengmeng Chen
- Qingdao Re-store Life Science Co., Ltd, Qingdao, Shandong, China
| | - Bingqiang Zhang
- Qingdao Re-store Life Science Co., Ltd, Qingdao, Shandong, China
| | - Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China.
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China.
| |
Collapse
|
23
|
Wang H, Qiao C, Guan Q, Wei M, Li Z. Nanoparticle-mediated synergistic anticancer effect of ferroptosis and photodynamic therapy: Novel insights and perspectives. Asian J Pharm Sci 2023; 18:100829. [PMID: 37588992 PMCID: PMC10425855 DOI: 10.1016/j.ajps.2023.100829] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/05/2023] [Accepted: 07/02/2023] [Indexed: 08/18/2023] Open
Abstract
Current antitumor monotherapy has many limitations, highlighting the need for novel synergistic anticancer strategies. Ferroptosis is an iron-dependent form of nonapoptotic cell death that plays a pivotal regulatory role in tumorigenesis and treatment. Photodynamic therapy (PDT) causes irreversible chemical damage to target lesions and is widely used in antitumor therapy. However, PDT's effectiveness is usually hindered by several obstacles, such as hypoxia, excess glutathione (GSH), and tumor resistance. Ferroptosis improves the anticancer efficacy of PDT by increasing oxygen and reactive oxygen species (ROS) or reducing GSH levels, and PDT also enhances ferroptosis induction due to the ROS effect in the tumor microenvironment (TME). Strategies based on nanoparticles (NPs) can subtly exploit the potential synergy of ferroptosis and PDT. This review explores recent advances and current challenges in the landscape of the underlying mechanisms regulating ferroptosis and PDT, as well as nano delivery system-mediated synergistic anticancer activity. These include polymers, biomimetic materials, metal organic frameworks (MOFs), inorganics, and carrier-free NPs. Finally, we highlight future perspectives of this novel emerging paradigm in targeted cancer therapies.
Collapse
Affiliation(s)
- Haiying Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chu Qiao
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qiutong Guan
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zhenhua Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
24
|
Xiao X, Chen M, Zhang Y, Li L, Peng Y, Li J, Zhou W. Correction: Hemin‑incorporating DNA nanozyme enabling catalytic oxygenation and GSH depletion for enhanced photodynamic therapy and synergistic tumor ferroptosis. J Nanobiotechnology 2022; 20:508. [PMID: 36457096 PMCID: PMC9717402 DOI: 10.1186/s12951-022-01678-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Affiliation(s)
- Xiaoxiong Xiao
- grid.452223.00000 0004 1757 7615Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China ,grid.452223.00000 0004 1757 7615Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China ,National Clinical Research Center for Geriatric Disorders, Changsha, China
| | - Min Chen
- Department of Thoracic Surgery, The Second People’s Hospital of Huaihua City, Huaihua, China
| | - Yuchen Zhang
- Department of Pharmacy, Yichun People’s Hospital, Yichun, China
| | - Liang Li
- grid.216417.70000 0001 0379 7164Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Ying Peng
- grid.216417.70000 0001 0379 7164Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Junyu Li
- grid.452533.60000 0004 1763 3891Department of Radiation Oncology, Jiangxi Cancer Hospital, Nanchang, China
| | - Wenhu Zhou
- grid.452223.00000 0004 1757 7615Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China ,grid.216417.70000 0001 0379 7164Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|