1
|
Li X, Zhang Y, Liu A, Li L, Yang X, Wang Y, Zhao Y, Zvyagin AV, Wang T, Lin Q. Nanozyme as tumor energy homeostasis disruptor mediated ferroptosis for high-efficiency radiotherapy. J Colloid Interface Sci 2025; 688:44-58. [PMID: 39987840 DOI: 10.1016/j.jcis.2025.02.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Radioresistance in tumors, driven by the insufficiency and rapid depletion of reactive oxygen species (ROS), limits the efficacy of radiotherapy (RT). This study introduces an Ir@Au nanozyme that enhances tumor radiosensitivity by disrupting energy homeostasis and inducing ferroptosis in tumor cells. The Ir@Au nanozyme mimics glucose oxidase to block the tumor's energy supply, continuously produces hydrogen peroxide (H2O2), and lowers the pH to optimize Fenton reactions. Acting as a peroxidase (POD), it generates additional ROS for chemodynamic therapy (CDT), depletes glutathione (GSH), and perturbs the tumor's antioxidant defenses. Upon exposure to ionizing radiation, the nanozyme absorbs photons and emits electrons, interacting with water to amplify ROS production. This ROS accumulation, combined with radiation, enhances DNA damage and lipid peroxidation, reversing radioresistance and promoting ferroptosis. Additionally, Ir@Au serves as a contrast agent for computed tomography, enabling precise RT through the delineation of tumor boundaries. In summary, the Ir@Au nanozyme effectively disrupts tumor energy homeostasis, initiating ROS-based cascades that inhibit tumor growth. It thus offers a promising strategy for overcoming radioresistance during cancer therapy.
Collapse
Affiliation(s)
- Xingchen Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yuxuan Zhang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Lei Li
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Ziqiang Street No. 218, Changchun 130041, China
| | - Yuan Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Yuechen Zhao
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Andrei V Zvyagin
- School of Mathematical and Physical Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW 2109, Australia; Research Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun 130041, China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun 130012, China.
| |
Collapse
|
2
|
Song K, Ming J, Tao B, Zhao F, Huang S, Wu W, Jiang C, Li X. Emerging glucose oxidase-delivering nanomedicines for enhanced tumor therapy. J Control Release 2025; 381:113580. [PMID: 40024341 DOI: 10.1016/j.jconrel.2025.02.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Abnormalities in glucose metabolism have been shown to characterize malignant tumors. Glucose depletion by glucose oxidase (GOD) has shown great potential in tumor therapy by causing tumor starvation. Since 2017, nanomedicines have been designed and utilized to deliver GOD for more precise and effective glucose modulation, which can overcome intrinsic limitations of different cancer therapeutic modalities by remodeling the tumor microenvironment to enhance antitumor therapy. To date, the topic of GOD-delivering nanomedicines for enhancing tumor therapy has not been comprehensively summarized. Herein, this review aims to provide an overview and discuss in detail recent advances in GOD delivery and directly involved starvation therapy strategies, GOD-sensitized various tumor therapy strategies, and GOD-mediated multimodal antitumor strategies. Finally, the challenges and outlooks for the future progress of the emerging tumor therapeutic nanomedicines are discussed. This review provides intuitive and specific insights to a broad audience in the fields of nanomedicines, biomaterials, and cancer therapy.
Collapse
Affiliation(s)
- Kaiyue Song
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Jiang Ming
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai 200433, China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Feng Zhao
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical College, Nanchang 330006, China.
| | - Wencheng Wu
- Central Laboratory and Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Cong Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200092, China.
| | - Xianglong Li
- Jiangxi Provincial Key Laboratory of Organic Functional Molecules, Institute of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang 330013, China.
| |
Collapse
|
3
|
Fu Y, Sun J, Yang C, Li W, Wang Y. Diversified nanocarrier design to optimize glucose oxidase-mediated anti-tumor therapy: Strategy and progress. Int J Biol Macromol 2025; 306:141581. [PMID: 40023419 DOI: 10.1016/j.ijbiomac.2025.141581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Given the inherent complexity and heterogeneity of tumors, current therapeutic approaches often fall short in meeting prognostic requirements. Starvation therapy (ST) utilizing glucose oxidase (GOx) has emerged as a promising strategy, specifically targeting tumor glucose consumption to disrupt nutrient supply. However, the therapeutic potential of GOx is significantly hampered by its inherent limitations as a protein, particularly its poor stability and short in vivo half-life. In recent years, the development of nanocarriors has provided an effective platform for intravenous and local tumor delivery of GOx. This review systematically examines three key strategies in GOx delivery: stimulus-response, biofilm modification, and local delivery. The progress in various carrier systems for GOx-mediated tumor therapy is comprehensively summarized, providing valuable insights for nanocarrier design. Furthermore, the existing challenges and future directions to advance the development of GOx-based tumor therapies are critically analyzed.
Collapse
Affiliation(s)
- Yuhan Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jialin Sun
- Department of medicine, Heilongjiang Minzu College, Harbin, Heilongjiang Province, China
| | - Chunyu Yang
- Department of Pathology, Harbin 242 Hospital, Harbin, Heilongjiang Province, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
4
|
Wu T, Zhou J, He W, Jin L, Li T, Gong T, Liu X. An injectable multimodal thiolated carboxymethyl cellulose hydrogel for advanced gastric cancer treatment. Int J Biol Macromol 2025; 306:141546. [PMID: 40020807 DOI: 10.1016/j.ijbiomac.2025.141546] [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: 09/17/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Chemotherapy is a standard preoperative treatment for locally advanced resectable gastric cancer (GC). However, its efficacy is often limited by the high intracellular glutathione (GSH) levels in tumor cells, which diminish the effectiveness of chemotherapeutic agents. In this study, we developed a GSH-sensitive injectable hydrogel, T-CMC@Fe3+(5-FU + GOx), incorporating ferric ions (Fe3+), glucose oxidase (GOx), and 5-fluorouracil (5-FU) for advanced GC multimodal treatment. The hydrogel synergistically enhances anti-tumor activity through multiple mechanisms, including chemodynamic therapy, starvation therapy, and chemotherapy. Thiolated carboxymethyl cellulose (T-CMC) was synthesized by grafting L-cysteine onto carboxymethyl cellulose, with successful preparation confirmed by XPS, FTIR, and 13C NMR characterizations. The T-CMC@Fe3+ hydrogel demonstrated responsive degradation of GSH and exhibited favorable biocompatibility in co-culture experiments with GES-1 cells. In vitro analyses revealed that the T-CMC@Fe3+(5-FU + GOx) hydrogel significantly enhanced anti-tumor efficacy compared to chemotherapy alone, reducing cell viability to 12.28 ± 2.88 % after 48 h. In vivo studies using cell-derived and patient-derived xenograft models further confirmed its potent anti-tumor effects, with a marked reduction in tumor volume by 14 days (8.12 ± 4.89 mm3). These findings highlight the potential of the T-CMC@Fe3+(5-FU + GOx) hydrogel as a novel and effective strategy for enhancing GC treatment through its multimodal therapeutic approach.
Collapse
Affiliation(s)
- Tao Wu
- College of Medicine and Biological Information Engineering, Northeastern University; Shenyang 110169, China; Department of Biomedical Engineering, Shenyang University of Technology, Shenyang 110870, China; Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, China
| | - Jingqiu Zhou
- College of Medicine and Biological Information Engineering, Northeastern University; Shenyang 110169, China; Department of Biomedical Engineering, Shenyang University of Technology, Shenyang 110870, China; Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, China
| | - Wanli He
- Department of Biomedical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Lei Jin
- Department of Biomedical Engineering, Shenyang University of Technology, Shenyang 110870, China
| | - Tenghui Li
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University; Shenyang 110001, China.
| | - Tianxing Gong
- Department of Biomedical Engineering, Shenyang University of Technology, Shenyang 110870, China.
| | - Xu Liu
- Department of Gastroenterology, General Hospital of Northern Theater Command, Shenyang 110840, China.
| |
Collapse
|
5
|
Chen Y, Cao H, Jiang C, Li Y. Tumor-microenvironment-mediated second near-infrared light activation multifunctional cascade nanoenzyme for self-replenishing O 2/H 2O 2 multimodal tumor therapy. J Colloid Interface Sci 2025; 683:930-943. [PMID: 39755017 DOI: 10.1016/j.jcis.2024.12.228] [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: 09/23/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
Developing a catalytic nanoenzyme activated by the tumor microenvironment (TME) shows excellent potential for in situ cancer treatment. However, the rational design of a cascade procedure to achieve high therapeutic efficiency remains challenging. In this study, the colorectal TME-responsive multifunctional cascade nanoenzyme Cu2-xO@MnO2@glucose oxidase (GOx)@hyaluronic acid (HA) was developed to target in situ cancer starvation/chemodynamic therapy (CDT)/photothermal therapy (PTT). First, the MnO2 nanolayer specifically decomposes within the acidic TME to generate Mn2+ and oxygen (O2), thereby alleviating the hypoxic TME. Subsequently, Cu2-xO can be vulcanized into Cu2-xS by overexpressing sulfuretted hydrogen (H2S) gas in the colorectal tumor for a second near-infrared (NIR-II) light-triggered deep tissue PTT. Cu2-xS nanoparticles can react with hydrogen peroxide (H2O2) to generate hydroxyl radical (OH) for the CDT. In addition, GOx catalyzes the conversion of glucose into H2O2 for starvation therapy and enhances the CDT efficiency by self-supplying H2O2. Interestingly, the generated reactive oxygen species (ROS) induce immunogenic cell death (ICD), which further activates adaptive cancer immunity for anti-tumor immunotherapy. Finally, therapeutic efficiency was greatly improved after coating with tumor-targeted HA. Collectively, these TME-responsive cascade nanoenzymes can realize PTT, CDT starvation therapy, and immunotherapy, paving the way for the design of TME-responsive cascade nanoenzymes for synergistically enhanced tumor-specific therapy.
Collapse
Affiliation(s)
- Yu Chen
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Haiqiong Cao
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Chaoqun Jiang
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, PR China
| | - Youbin Li
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, PR China.
| |
Collapse
|
6
|
Xie Q, Sun T, Zhang L, Gong M, Zhang W, Liu X, Zhao Y, Wang M, Yang X, Zhang Z, Liu G, Zhou C, Zhang D. Responsive plasmonic hybrid nanorods enables metabolism reprogramming via cuproptosis-photothermal combined cancer therapy. Biomaterials 2025; 315:122971. [PMID: 39577035 DOI: 10.1016/j.biomaterials.2024.122971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Abnormal tumor metabolism leads to tumor growth, metastasis, and recurrence, reprogramming tumor metabolism and activating potent anti-tumor immune response have been demonstrated to have good therapeutic effects on tumor elimination. Copper-based nanomaterials involved in cuproptosis show great prospects in these two aspects, but their efficiency is restricted by Cu homeostasis and the toxicity of the chelator. Here, the pH-responsive AuNRs@Cu2O core-shell plasmonic hybrid nanorods (ACNRs) have been successfully fabricated to realize microenvironment-controlled release at the tumor site for the combined therapy of cuproptosis and photothermal treatment. The AuNRs core exhibited excellent NIR-II photothermal property, which boost the intracellular concentration of copper to trigger severe cuproptosis and induce immunogenic cell death of tumor cells. In vivo studies demonstrated the ACNR exhibited efficient tumor therapy for primary, metastatic, and recurrent tumors. ACNRs-induced cuproptosis and PTT were capable of reprogramming energy metabolism, leading to a decreased production of lactic acid. This potential of metabolic reprogramming assisted in reshaping the immunosuppressive tumor microenvironment to facilitate the infiltration of immune cells and boost the immune responses triggered by PTT. The therapeutic mechanism was further verified by metabolomics analysis, which indicated that ACNRs + PTT treatment led to the inhibition of the Pentose Phosphate Pathway and Glycolysis pathways in tumor cells. The suppression of glycolytic reduced ATP synthesis, thereby hindering energy-dependent copper efflux, which in turn promoted cuproptosis. Taken together, this study offers promising insights for cuproptosis-based cancer treatment and sheds new light on nanomedicine-mediated metabolic modulation for future tumor therapy.
Collapse
Affiliation(s)
- Qian Xie
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China; Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Wansu Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xu Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yue Zhao
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Miaomiao Wang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Xiaofeng Yang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Zhipeng Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
7
|
Lu F, Jang MS, Jiang W, Liu C, Wang B, Lee JH, Fu Y, Yang HY. A multifunctional hyaluronic acid-engineered mesoporous nanoreactor with H 2O 2/O 2 self-sufficiency for pH-triggered endo-lysosomal escape and synergetic cancer therapy. BIOMATERIALS ADVANCES 2025; 169:214161. [PMID: 39721571 DOI: 10.1016/j.bioadv.2024.214161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Monotherapy has poor accuracy and is easily restricted by tumor microenvironment (TME). Remodeling components of the TME to activate multimodal cancer therapy with high precision and efficiency is worth exploring. A multifunctional nanoreactor was fabricated by decorating chlorin e6-modified and PEGylated hyaluronic acid bearing diethylenetriamine-conjugated dihydrolipoic acid on the surface of glucose oxidase (GOx)-loaded hollow mesoporous CuS nanoparticles (labeled as GOx@HCuS@HA). This nanoreactor efficiently targets tumor sites, enhances cellular internalization, and swiftly escapes from endo-lysosomes after intravenous injection. Subsequently, GOx@HCuS@HA was activated in hyaluronidase and H + -rich TME to produce H2O2 and gluconic acid through the oxidation of glucose, which not only blocks the energy supply of cancer cells, executing starvation treatment (ST), but also bolsters hydroxyl radicals (•OH)-based chemodynamic therapy (CDT) by Fenton-like reaction between HCuS and H2O2. Furthermore, reductive Cu ions could catalyze H2O2 to produce O2 to alleviate the limitation of photodynamic therapy (PDT) for tumor hypoxia. Additionally, the photothermal effect of HCuS under NIR irradiation could increase the temperature of tumor tissues to perform photothermal therapy (PTT). This synergistic antitumor strategy could ultimately achieve precise tumor cell destruction and maintain excellent biosafety. Hence, this nanoreactor offer promising prospects for efficient tumor treatment.
Collapse
Affiliation(s)
- Fei Lu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Moon-Sun Jang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Wei Jiang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Changling Liu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Bo Wang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, PR China
| | - Jung Hee Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea.
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China.
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China.
| |
Collapse
|
8
|
Mei L, Ding Q, Xie Y, Liu H, Li H, Kim E, Shen X, Zhang Y, Zhang S, Kim JS. Self-propelling intelligent nanomotor: A dual-action photothermal and starvation strategy for targeted deep tumor destruction. Biomaterials 2025; 315:122968. [PMID: 39561474 DOI: 10.1016/j.biomaterials.2024.122968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Delivering nanoparticles to deep tumor tissues while maintaining high therapeutic efficacy and minimizing damage to surrounding tissues has long posed a significant challenge. To address this, we have developed innovative self-propelling bowl-shaped nanomotors MSLA@GOx-PDA composed of mesoporous silica loaded with l-arginine and polydopamine, along with glucose oxidase (GOx). These nanomotors facilitate the generation of hydrogen peroxide through GOx-catalyzed glucose oxidation, thereby initiating nitric oxide production from l-arginine. This dual mechanism equips MSLA@GOx-PDA with the robust motility required for deep tumor tissue penetration while depleting essential nutrients necessary for tumor growth, consequently impeding tumor progression. In addition, near-infrared lasers have the significant advantage of being depth-penetrating and non-invasive, allowing real-time fluorescence imaging and guiding dopamine-mediated mild photothermal therapy. Notably, starvation therapy depletes intracellular adenosine triphosphate and inhibits the synthesis of heat shock proteins, thus overcoming the Achilles' heel of mild photothermal therapy and significantly enhancing the efficacy of this therapy with encouraging synergistic anti-tumour effects. Overall, the integration of biochemical and optics strategies in this nanomotor platform represents a significant advancement in deep-tissue tumor therapy. It has substantial clinical translational value and is expected to have a transformative impact on future cancer treatments.
Collapse
Affiliation(s)
- Ling Mei
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Yuxin Xie
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Haowei Liu
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Hongping Li
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Eunji Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Xue Shen
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Yibin Zhang
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610106, China.
| | - Shuai Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road Nangang District, Harbin, Heilongjiang Province, 150040, China.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
9
|
Zhao RN, Ke YY, Sun HY, Quan C, Xu Q, Li J, Guan JQ, Zhang YM. Achievements and challenges in glucose oxidase-instructed multimodal synergistic antibacterial applications. Microbiol Res 2025; 297:128149. [PMID: 40187057 DOI: 10.1016/j.micres.2025.128149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 04/07/2025]
Abstract
Glucose oxidase (GOx) with unique catalytic properties and inherent biocompatibility can effectively oxidize both endogenous and exogenous glucose with oxygen (O2) into gluconic acid and hydrogen peroxide (H2O2). Accordingly, the GOx-based catalytic chemistry offers new possibilities for designing and constructing multimodal synergistic antibacterial systems. The consumption of glucose permanently downregulates bacterial cell metabolism by blocking essential energy supplies, inhibiting their growth and survival. Additionally, the production of gluconic acid could downregulates the pH within the bacterial infection microenvironment, enhancing the production of hydroxyl radicals (∙OH) from H2O2 via enhanced Fenton or Fendon-like reactions and triggering the pH-responsive release of drugs. Furthermore, the generated H2O2 in situ avoids the addition of exogenous hydrogen peroxide. Therefore, it is possible to design GOx-based multimodal antibacterial synergistic therapies by combining GOx-instructed cascade reactions with other therapeutic approaches such as chemodynamic therapies (CDT), hypoxia-activated prodrugs, photosensitizers, and stimuli-responsive drug release. Such multimodal strategies are expected to exhibit better therapeutic effects than single therapeutic modes. This tutorial review highlights recent advancements in GOx-instructed multimodal synergistic antibacterial systems, focusing on design philosophy and construction strategies. Current challenges and future prospects for advancing GOx-based multimodal antibacterial synergistic therapies are discussed.
Collapse
Affiliation(s)
- Rui-Nan Zhao
- College of Life Science, Dalian Minzu University, Economical and Technological Development Zone, Dalian 116600, China; Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, China
| | - Yi-Yin Ke
- College of Life Science, Dalian Minzu University, Economical and Technological Development Zone, Dalian 116600, China; Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, China
| | - Hui-Yan Sun
- College of Life Science, Dalian Minzu University, Economical and Technological Development Zone, Dalian 116600, China; Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, China
| | - Chunshan Quan
- College of Life Science, Dalian Minzu University, Economical and Technological Development Zone, Dalian 116600, China; Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, China
| | - Qingsong Xu
- College of Life Science, Dalian Minzu University, Economical and Technological Development Zone, Dalian 116600, China; Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, China
| | - Jun Li
- State Key Laboratory of Catalysis, Dalian Institute of Chemical Physics, Chinese Academy of Science, P. O. Box 110, Dalian 116023, China.
| | - Jing-Qi Guan
- Institute of Physical Chemistry, College of Chemistry, Jilin University, 2519 Jiefang Road, Changchun 130021, China.
| | - Yan-Mei Zhang
- College of Life Science, Dalian Minzu University, Economical and Technological Development Zone, Dalian 116600, China; Key Laboratory of Biotechnology and Bioresources Utilization (Dalian Minzu University), Ministry of Education, China.
| |
Collapse
|
10
|
Lin H, Gao Y, Zhu L, Guo Y, Zhang L, Xie J, Yang D, Liu J, Dong Q, Zhu Z. Rational Design of Single‐Atom Nanozymes for Combination Cancer Immunotherapy. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202416563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Indexed: 02/03/2025]
Abstract
AbstractRemodeling of the tumor immune microenvironment and enhancement of antitumor immune responses are necessary to overcome immunotherapy resistance in tumors. However, tumor heterogeneity and complexity of immune evasion mechanisms pose significant therapeutic challenges. Nanozymes exhibit enzyme‐like characteristics and unique nanomaterial properties, showing potential for tumor therapy. However, design of effective nanozymes remains complex, inefficient, and functionally limited. Therefore, in this study, a novel strategy combining rationally designed single‐atom nanozymes (SAzymes) with immune checkpoint blockade (ICB) therapy is established. Molybdenum SAzymes supported on graphitic carbon nitride (Mo SAs) are constructed using 25 transition metal candidates from the 4th to 6th periods based on high‐throughput calculations and optimal piezoelectric‐enhanced multienzyme‐like activities. Upon activation by ultrasound, Mo SAs exerted potent therapeutic effects against ICB‐resistant tumors and remodeled the tumor immune microenvironment by inducing tumor immunogenic cell death, alleviating tumor hypoxia, and modulating chemokine expression in tumors. Combination of Mo SAs with anti‐programmed death protein‐1 antibodies further enhanced their antitumor efficacy, highlighting their potential to treat ICB‐resistant tumors.
Collapse
Affiliation(s)
- Hanchao Lin
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Yonghui Gao
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Le Zhu
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
| | - Yu Guo
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Lumin Zhang
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
| | - Jiali Xie
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Dongqin Yang
- Department of Laboratory Medicine Huashan Hospital Fudan University 12 Middle Urumqi Road Shanghai 200040 China
| | - Jing Liu
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| | - Qiongzhu Dong
- Key Laboratory of Whole‐Period Monitoring and Precise Intervention of Digestive Cancer Shanghai Municipal Health Commission Minhang Hospital Fudan University 170 Xingsong Road Shanghai 201199 China
- Department of General Surgery Huashan Hospital & Cancer Metastasis Institute Fudan University 12 Middle Urumuqi Road Shanghai 200040 China
| | - Zhiling Zhu
- College of Materials Science and Engineering Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
- Key Laboratory of Optic‐electric Sensing and Analytical Chemistry for Life Science MOE; Shandong Key Laboratory of Biochemical Analysis Qingdao University of Science and Technology 53 Zhengzhou Road Qingdao Shandong 266042 China
| |
Collapse
|
11
|
Shi J, Han W, Wang J, Kong X. Anti-Tumor Strategies Targeting Nutritional Deprivation: Challenges and Opportunities. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415550. [PMID: 39895165 DOI: 10.1002/adma.202415550] [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: 10/12/2024] [Revised: 01/04/2025] [Indexed: 02/04/2025]
Abstract
Higher and richer nutrient requirements are typical features that distinguish tumor cells from AU: cells, ensuring adequate substrates and energy sources for tumor cell proliferation and migration. Therefore, nutrient deprivation strategies based on targeted technologies can induce impaired cell viability in tumor cells, which are more sensitive than normal cells. In this review, nutrients that are required by tumor cells and related metabolic pathways are introduced, and anti-tumor strategies developed to target nutrient deprivation are described. In addition to tumor cells, the nutritional and metabolic characteristics of other cells in the tumor microenvironment (including macrophages, neutrophils, natural killer cells, T cells, and cancer-associated fibroblasts) and related new anti-tumor strategies are also summarized. In conclusion, recent advances in anti-tumor strategies targeting nutrient blockade are reviewed, and the challenges and prospects of these anti-tumor strategies are discussed, which are of theoretical significance for optimizing the clinical application of tumor nutrition deprivation strategies.
Collapse
Affiliation(s)
- Jinsheng Shi
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Wei Han
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Jie Wang
- Pharmacy Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, Shandong, 266000, China
| | - Xiaoying Kong
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong, 266071, China
| |
Collapse
|
12
|
Li Y, Fu B, Jiang W. Emerging Roles of Nanozyme in Tumor Metabolism Regulation: Mechanisms, Applications, and Future Directions. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11552-11577. [PMID: 39936939 DOI: 10.1021/acsami.4c20417] [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: 02/13/2025]
Abstract
Nanozymes, nanomaterials with intrinsic enzyme activity, have garnered significant attention in recent years due to their catalytic abilities comparable to natural enzymes, cost-effectiveness, high catalytic activities, and stability against environmental fluctuations. As functional analogs of natural enzymes, nanozymes participate in various critical metabolic processes, including glucose metabolism, lactate metabolism, and the maintenance of redox homeostasis, all of which are essential for normal cellular functions. However, disruptions in these metabolic pathways frequently promote tumorigenesis and progression, making them potential therapeutic targets. While several therapies targeting tumor metabolism are currently in clinical or preclinical stages, their efficacy requires further enhancement. Consequently, nanozymes that target tumor metabolism are regarded as a promising therapeutic strategy. Despite extensive studies investigating the application of nanozymes in tumor metabolism, relevant reviews are relatively scarce. This article first introduces the physicochemical properties and biological behaviors of nanozymes. Subsequently, we analyze the role of nanozymes in tumor metabolism and explore their potential applications in tumor therapy. In conclusion, this review aims to foster innovative research in related fields and advance the development of nanozyme-based strategies for cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Yikai Li
- The First Bethune Hospital of Jilin University, Jilin University, Changchun, Jilin 130000, China
| | - Bowen Fu
- The First Bethune Hospital of Jilin University, Jilin University, Changchun, Jilin 130000, China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450002, China
| |
Collapse
|
13
|
Mao L, Ma Y, Wen X, Luo Z, Zhu H, Kong J, Liu S, Fan X, Wang J, He C, Wu YL. Iron-glucose oxidase nanogel assembly for amplified starvation-ferroptosis anti-tumor therapy. Int J Biol Macromol 2025; 289:138804. [PMID: 39689793 DOI: 10.1016/j.ijbiomac.2024.138804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Developing advanced and effective enzyme-drug systems for cancer treatment is of significant interest. Herein, a novel approach is reported to create a highly active and robust enzyme-drug system. Glucose oxidase nanogels (nGOx) are first synthesized by polymerization on the surface of GOx using vinylimidazole as comonomers. Fe3+ are utilized to induce self-assembly of nGOx through the imidazole-metal coordination interaction to form GOx nanogel clusters (Fe@nGOx), enhancing the permeability and retention of nGOx into tumor cells by EPR effect. nGOx can deplete glucose in the presence of oxygen and generate H2O2, which is converted to highly cytotoxic hydroxyl radical (·OH) by Fe3+ and GSH, and the proximity between Fe3+ and GOx act in tandem for enhanced tumor therapy. The FeIII/FeII redox cycle reacts with GSH and H2O2, enabling continuous generation of ·OH within tumor cells, thus facilitating the anticancer effect. Moreover, the generation of H2O2 and ·OH can further promote the repolarization of tumor-associated macrophages from an M2 phenotype towards an M1 phenotype polarization, thus enhancing immune response. The cascade reaction between GOx and Fe3+/Fe2+ endows Fe@nGOx with excellent anti-tumor efficacy in mice models, highlighting its potential as a promising anticancer drug for clinical applications. This work establishes a new platform for utilizing enzyme/protein and metal ion complexes in versatile applications, advancing the field of enzyme-based cancer therapies.
Collapse
Affiliation(s)
- Liuzhou Mao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China
| | - Yedong Ma
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| | - Xiaoqing Wen
- Drug clinical trial institution, The first affiliated hospital of Xiamen university, Xiamen, Fujian, PR China
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China
| | - Houjuan Zhu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore
| | - Junhua Kong
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore
| | - Siqi Liu
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore
| | - Xiaotong Fan
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 1 Pesek Road, Jurong Island, Singapore 627833, Republic of Singapore.
| | - Jinling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, No. 12 Minyou Road, Xiashan, Zhanjiang, Guangdong 524003, PR China.
| | - Chaobin He
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Republic of Singapore; Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology, and Research (A∗STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Republic of Singapore.
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
14
|
Wang Y, Tang Y, Guo L, Yang X, Wu S, Yue Y, Xu C. Recent advances in zeolitic imidazolate frameworks as drug delivery systems for cancer therapy. Asian J Pharm Sci 2025; 20:101017. [PMID: 39931355 PMCID: PMC11808527 DOI: 10.1016/j.ajps.2025.101017] [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: 04/28/2023] [Revised: 04/16/2024] [Accepted: 11/24/2024] [Indexed: 02/13/2025] Open
Abstract
Biological nanotechnologies based on functional nanoplatforms have synergistically catalyzed the emergence of cancer therapies. As a subtype of metal-organic frameworks (MOFs), zeolitic imidazolate frameworks (ZIFs) have exploded in popularity in the field of biomaterials as excellent protective materials with the advantages of conformational flexibility, thermal and chemical stability, and functional controllability. With these superior properties, the applications of ZIF-based materials in combination with various therapies for cancer treatment have grown rapidly in recent years, showing remarkable achievements and great potential. This review elucidates the recent advancements in the use of ZIFs as drug delivery agents for cancer therapy. The structures, synthesis methods, properties, and various modifiers of ZIFs used in oncotherapy are presented. Recent advances in the application of ZIF-based nanoparticles as single or combination tumor treatments are reviewed. Furthermore, the future prospects, potential limitations, and challenges of the application of ZIF-based nanomaterials in cancer treatment are discussed. We except to fully explore the potential of ZIF-based materials to present a clear outline for their application as an effective cancer treatment to help them achieve early clinical application.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xi Yang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Shanli Wu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
15
|
Liao Y, Zhang Z, Zhao Y, Zhang S, Zha K, Ouyang L, Hu W, Zhou W, Sun Y, Liu G. Glucose oxidase: An emerging multidimensional treatment option for diabetic wound healing. Bioact Mater 2025; 44:131-151. [PMID: 39484022 PMCID: PMC11525048 DOI: 10.1016/j.bioactmat.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/06/2024] [Accepted: 10/06/2024] [Indexed: 11/03/2024] Open
Abstract
The healing of diabetic skin wounds is a complex process significantly affected by the hyperglycemic environment. In this context, glucose oxidase (GOx), by catalyzing glucose to produce gluconic acid and hydrogen peroxide, not only modulates the hyperglycemic microenvironment but also possesses antibacterial and oxygen-supplying functions, thereby demonstrating immense potential in the treatment of diabetic wounds. Despite the growing interest in GOx-based therapeutic strategies in recent years, a systematic summary and review of these efforts have been lacking. To address this gap, this review article outlines the advancements in the application of GOx and GOx-like nanozymes in the treatment of diabetic wounds, including reaction mechanisms, the selection of carrier materials, and synergistic therapeutic strategies such as multi-enzyme combinations, microneedle structures, and gas therapy. Finally, the article looks forward to the application prospects of GOx in aiding the healing of diabetic wounds and the challenges faced in translating these innovations to clinical practice. We sincerely hope that this review can provide readers with a comprehensive understanding of GOx-based diabetic treatment strategies, facilitate the rigorous construction of more robust multifunctional therapeutic systems, and ultimately benefit patients with diabetic wounds.
Collapse
Affiliation(s)
| | | | | | | | - Kangkang Zha
- Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, China
| | - Lizhi Ouyang
- Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, China
| | - Weixian Hu
- Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, China
| | - Wu Zhou
- Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, China
| | - Yun Sun
- Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, China
| | - Guohui Liu
- Wuhan Union Hospital of Tongji Medical College of Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, China
| |
Collapse
|
16
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
17
|
Meng X, Tian L, Zhang J, Wang J, Cao X, Hu Z, Sun Y, Dai Z, Zheng X. Tumor microenvironment-regulated nanoplatform for enhanced chemotherapy, cuproptosis and nonferrous ferroptosis combined cancer therapy. J Mater Chem B 2025; 13:1089-1099. [PMID: 39652201 DOI: 10.1039/d4tb02000f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Therapeutic approaches combining various treatments have attracted intensive interests for tumor therapy. Nevertheless, these strategies still face many obstacles, such as overexpressed GSH and hypoxia, owing to the intricate tumor microenvironment (TME). Herein, a versatile nanoplatform, CeO2@CuO2@DOX-RSL3@HA (CCDRH), was initially constructed for promoting the antitumor efficiency via regulation of the TME. The CCDRH was prepared taking mixed valence CeO2 as the nanocarrier, followed by the attachment of CuO2 nanodots, DOX and RSL3 and the camouflaging of hyaluronic acid. The CuO2 could disassemble in the acidic TME to release Cu2+ and H2O2. The POD- and CAT-mimicking activities of CeO2 could convert H2O2 to ˙OH and O2, leading to the enhancement of chemo-chemodynamic therapy. Meanwhile, RSL3 could effectively suppress GPX4 expression, and the overloaded Cu2+ and Ce4+ could deplete excess GSH, resulting in an intensive accumulation of LPO and significant nonferrous ferroptosis. Additionally, Cu+ induces the oligomerization of lipoylated DLAT and downregulates iron-sulfur cluster proteins, resulting in potent cellular cuproptosis. The experimental results revealed that CCDRH exhibited high performance in tumor inhibition, which is attributed to the combined effect of enhanced chemotherapy, ferroptosis and cuproptosis. The study provides a new approach for improving anticancer efficiency via regulation of the TME.
Collapse
Affiliation(s)
- Xiangyu Meng
- College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, P. R. China.
- Qilu Normal University, Jinan 250000, P. R. China.
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
| | - Lu Tian
- College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, P. R. China.
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
| | - Jingmei Zhang
- School of Chemical Engineering, Hebei Normal University of Science and Technology, Qinhuangdao 066000, P. R. China
| | - Jiaoyu Wang
- College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, P. R. China.
- Qilu Normal University, Jinan 250000, P. R. China.
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
| | - Xuewei Cao
- Qilu Normal University, Jinan 250000, P. R. China.
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
- College of Medicine, Linyi University, Linyi 276000, P. R. China
| | - Zunfu Hu
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
| | - Yunqiang Sun
- College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, P. R. China.
| | - Zhichao Dai
- College of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, P. R. China.
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
| | - Xiuwen Zheng
- Qilu Normal University, Jinan 250000, P. R. China.
- Key Laboratory of Advanced Biomaterials and Nanomedicine in Universities of Shandong, Linyi University, Linyi 276000, P. R. China
| |
Collapse
|
18
|
Mohammed Ameen SS, Bedair A, Hamed M, R Mansour F, Omer KM. Recent Advances in Metal-Organic Frameworks as Oxidase Mimics: A Comprehensive Review on Rational Design and Modification for Enhanced Sensing Applications. ACS APPLIED MATERIALS & INTERFACES 2025; 17:110-129. [PMID: 39772422 DOI: 10.1021/acsami.4c17397] [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: 01/11/2025]
Abstract
Metal-organic frameworks (MOFs) have emerged as innovative nanozyme mimics, particularly in the area of oxidase catalysis, outperforming traditional MOF-based peroxidase and other nanomaterial-based oxidase systems. This review explores the various advantages that MOFs offer in terms of catalytic activity, low-cost, stability, and structural versatility. With a primary focus on their application in biochemical sensing, MOF-based oxidases have demonstrated remarkable utility, prompting a thorough exploration of their design and modification strategies. Moreover, the review aims to provide a comprehensive analysis of the strategies employed in the rational design and modification of MOF structures to optimize key parameters such as sensitivity, selectivity, and stability in the context of biochemical sensors. Through an exhaustive examination of recent research and developments, this article seeks to offer insights into the nuanced interplay between MOF structures and their catalytic performance, shedding light on the mechanisms that underpin their effectiveness as nanozyme mimics. Finally, this review addresses challenges and opportunities associated with MOF-based oxidase mimics, aiming to drive further advancements in MOF structure design and the development of highly effective biochemical sensors for diverse applications.
Collapse
Affiliation(s)
- Sameera Sh Mohammed Ameen
- Department of Chemistry, College of Science, University of Zakho, 46002 Zakho, Kurdistan Region, Iraq
| | - Alaa Bedair
- Department of Analytical Chemistry, Faculty of Pharmacy, University of Sadat City, Sadat City 32958, Egypt
| | - Mahmoud Hamed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Misr International University, Km 28 Ismailia Road, Cairo 44971, Egypt
| | - Fotouh R Mansour
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Khalid M Omer
- Department of Chemistry, College of Science, University of Sulaimani, Qliasan St., 46002 Sulaymaniyah, Kurdistan Region, Iraq
| |
Collapse
|
19
|
Zhang Y, Liu Y, Zhao M, Wang Y, Yi H, Liu D, Hou S, Zhao Q, Ma S. Iron-based theranostic nanoenzyme for combined tumor magneto-photo thermotherapy and starvation therapy. BIOMATERIALS ADVANCES 2025; 166:214038. [PMID: 39306963 DOI: 10.1016/j.bioadv.2024.214038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/14/2024] [Accepted: 09/08/2024] [Indexed: 11/13/2024]
Abstract
To address the issue of high-dose treatment agents in magnetic hyperthermia-mediated multi-model tumor therapy, a unique iron-based theranostic nanoenzyme with excellent magnetothermal and catalytic properties was constructed. By using a high-temperature arc method, the iron carbon nanoparticles (MF1-3) with a particle size between 13.7 and 27.6 nm and shell thickness between 1 and 5 nm were prepared. After screening, we selected MF3 as the magnetic core due to its high Ms. value and excellent thermal properties. Under the magneto-photo dual thermal conditions, MF3 exhibited a remarkable specific absorption rate (SAR) of 4917 W/g, which was 20 times more than that of iron oxide. Notably, MF3 also exhibited best peroxidase (POD)-like catalytic in pH 5.0 and maintained stable catalytic performance at 45 °C. Considering the "starvation" strategy of cutting off the energy supply to tumor cells and killing them, the glucose oxidase (GOX) and chitosan oligosaccharide (COS) was further grafted onto MF3, forming the MF3/GOX/COS. This multifunctional therapeutic nanoenzyme not only exhibited significant peroxidase-like activity, but also had glucose decomposition and glutathione (GSH) consumption capabilities. The thermal effect significantly promoted the uptake of MF3/GOX/COS by 4T1 cells, and the IC50 value of MF3/GOX/COS reached low to 3.75 μg/mL. In vivo anti-tumor experiment, compared with single treatment methods, the combined therapy of MF3/GOX/COS mediated magneto-photo thermotherapy (M-PTT) and starvation therapy (ST) exhibited higher tumor inhibition rate of 82.1 % by increased cell apoptosis through the mitochondrial pathway. Overall, MF3/GOX/COS therapeutic nanoenzyme combined the advantages of nano-catalysis, M-PTT and ST, providing a solution for achieving sustained, stable, and effective tumor inhibition rates at lower dose levels.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of Target Drug design and Research, Ministry of Education, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China
| | - Yaqi Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Mengtong Zhao
- Key Laboratory of Target Drug design and Research, Ministry of Education, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China
| | - Yifei Wang
- Key Laboratory of Target Drug design and Research, Ministry of Education, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China
| | - Hang Yi
- Key Laboratory of Target Drug design and Research, Ministry of Education, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China
| | - Daheng Liu
- Shenyang National Laboratory for Material Science, the Institute of Metal Research (IMR), Chinese Academy of Sciences (CAS), Wenhua Road 72, Shenyang 110015, PR China
| | - Siyu Hou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Song Ma
- Shenyang National Laboratory for Material Science, the Institute of Metal Research (IMR), Chinese Academy of Sciences (CAS), Wenhua Road 72, Shenyang 110015, PR China.
| |
Collapse
|
20
|
Zhang X, An M, Zhang J, Zhao Y, Liu Y. Nano-medicine therapy reprogramming metabolic network of tumour microenvironment: new opportunity for cancer therapies. J Drug Target 2024; 32:241-257. [PMID: 38251656 DOI: 10.1080/1061186x.2024.2309565] [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: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 01/23/2024]
Abstract
Metabolic heterogeneity is one of the characteristics of tumour cells. In order to adapt to the tumour microenvironment of hypoxia, acidity and nutritional deficiency, tumour cells have undergone extensive metabolic reprogramming. Metabolites involved in tumour cell metabolism are also very different from normal cells, such as a large number of lactate and adenosine. Metabolites play an important role in regulating the whole tumour microenvironment. Taking metabolites as the target, it aims to change the metabolic pattern of tumour cells again, destroy the energy balance it maintains, activate the immune system, and finally kill tumour cells. In this paper, the regulatory effects of metabolites such as lactate, glutamine, arginine, tryptophan, fatty acids and adenosine were reviewed, and the related targeting strategies of nano-medicines were summarised, and the future therapeutic strategies of nano-drugs were discussed. The abnormality of tumour metabolites caused by tumour metabolic remodelling not only changes the energy and material supply of tumour, but also participates in the regulation of tumour-related signal pathways, which plays an important role in the survival, proliferation, invasion and metastasis of tumour cells. Regulating the availability of local metabolites is a new aspect that affects tumour progress. (The graphical abstract is by Figdraw).
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Min An
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Juntao Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yumeng Zhao
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Hui Ethnic Medicine Modernization, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
21
|
Liang S, Liu Y, Zhu H, Liao G, Zhu W, Zhang L. Emerging nitric oxide gas-assisted cancer photothermal treatment. EXPLORATION (BEIJING, CHINA) 2024; 4:20230163. [PMID: 39713202 PMCID: PMC11655315 DOI: 10.1002/exp.20230163] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/08/2024] [Indexed: 12/24/2024]
Abstract
Photothermal therapy (PTT) has garnered significant attention in recent years, but the standalone application of PTT still faces limitations that hinder its ability to achieve optimal therapeutic outcomes. Nitric oxide (NO), being one of the most extensively studied gaseous molecules, presents itself as a promising complementary candidate for PTT. In response, various nanosystems have been developed to enable the simultaneous utilization of PTT and NO-mediated gas therapy (GT), with the integration of photothermal agents (PTAs) and thermally-sensitive NO donors being the prevailing approach. This combination seeks to leverage the synergistic effects of PTT and GT while mitigating the potential risks associated with gas toxicity through the use of a single laser irradiation. Furthermore, additional internal or external stimuli have been employed to trigger NO release when combined with different types of PTAs, thereby further enhancing therapeutic efficacy. This comprehensive review aims to summarize recent advancements in NO gas-assisted cancer photothermal treatment. It commences by providing an overview of various types of NO donors and precursors, including those sensitive to photothermal, light, ultrasound, reactive oxygen species, and glutathione. These NO donors and precursors are discussed in the context of dual-modal PTT/GT. Subsequently, the incorporation of other treatment modalities such as chemotherapy (CHT), photodynamic therapy (PDT), alkyl radical therapy, radiation therapy, and immunotherapy (IT) in the creation of triple-modal therapeutic nanoplatforms is presented. The review further explores tetra-modal therapies, such as PTT/GT/CHT/PDT, PTT/GT/CHT/chemodynamic therapy (CDT), PTT/GT/PDT/IT, PTT/GT/starvation therapy (ST)/IT, PTT/GT/Ca2+ overload/IT, PTT/GT/ferroptosis (FT)/IT, and PTT/GT/CDT/IT. Finally, potential challenges and future perspectives concerning these novel paradigms are discussed. This comprehensive review is anticipated to serve as a valuable resource for future studies focused on the development of innovative photothermal/NO-based cancer nanotheranostics.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yufei Liu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongquan Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guangfu Liao
- College of Material EngineeringFujian Agriculture and Forestry UniversityFuzhouChina
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Zhang
- Department of Critical Care MedicineShenzhen HospitalSouthern Medical UniversityShenzhenGuangdongChina
- Department of ChemistryCity University of Hong KongKowloonHong Kong SARChina
| |
Collapse
|
22
|
Jiang Z, Li J, Liu G, Qiu Q, Zhang J, Hao M, Ren H, Zhang Y. A pH-Sensitive Glucose Oxidase and Hemin Coordination Micelle for Multi-Enzyme Cascade and Amplified Cancer Chemodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2407674. [PMID: 39363789 DOI: 10.1002/smll.202407674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Indexed: 10/05/2024]
Abstract
Chemodynamic therapy (CDT) is an emerging therapeutic paradigm for cancer treatment that utilizes reactive oxygen species (ROS) to induce apoptosis of cancer cells but few biomaterials have been developed to differentiate the cancer cells and normal cells to achieve precise and targeted CDT. Herein, a simple cascade enzyme system is developed, termed hemin-micelles-GOx, based on hemin and glucose oxidase (GOx)-encapsulated Pluronic F127 (F127) micelles with pH-sensitive enzymatic activities. Histidine-tagged GOx can be easily chelated to hemin-F127 micelles via the coordination of histidine and ferrous ions in the center of hemin by simple admixture in an aqueous solution. In tumor microenvironment (TME), hemin-micelles-GOx exhibits enhanced peroxidase (POD)-like activities to generate toxic hydroxyl radicals due to the acidic condition, whereas in normal cells the catalase (CAT)-like, but not POD-like activity is amplified, resulting in the elimination of hydrogen peroxide to generate oxygen. In a murine melanoma model, hemin-micelles-GOx significantly suppresses tumor growth, demonstrating its great potential as a pH-mediated enzymatic switch for tumor management by CDT.
Collapse
Affiliation(s)
- Zhen Jiang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Jiexin Li
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Gengqi Liu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Qian Qiu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Jingyu Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Minchao Hao
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - He Ren
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| |
Collapse
|
23
|
Ren H, Bai Y, Liu Z, Ma C, Tao X, Wang Q, Lian H, Li X. A multifunctional cascade gas-nanoreactor with MnO 2 as a gatekeeper to enhance starvation therapy and provoke antitumor immune response. Acta Biomater 2024:S1742-7061(24)00658-5. [PMID: 39521315 DOI: 10.1016/j.actbio.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Glucose oxidase (GOx)-mediated starvation therapy is an effective tumor treatment that blocks energy and activates the immune response. However, the insufficient tumor immunogenicity and immunosuppressive tumor microenvironment (TME) limited its therapeutic efficacy. To address this, we have designed a multifunctional cascade gas-nanoreactor with a MnO2 coating, which serves as an out gatekeeper to encapsulate both GOx and a carbon monoxide (CO) donor (denoted as GCM). Due to the protective effect of MnO2 coating, GCM maintains better stability in normal physiological environments, enhancing the catalytic activity of GOx and minimizing toxic side effects. Upon accumulation in the tumor, the degradation of MnO2 coating exposes the GOx enzyme, thereby initiating a cascade catalysis reaction to generate hydrogen peroxide (H2O2) and release CO in the hypoxic conditions. Additionally, the released Mn2+ reacts with H2O2 to generate toxic hydroxyl radical (•OH) as chemodynamic therapy (CDT). The synergistic treatments of starvation therapy, CO gas therapy and CDT effectively kill cancer cells and amplify immunogenic cell death (ICD), maturing DC cells and activating anti-tumor immune response. Furthermore, the released CO increases M1 macrophages infiltration and reduces myeloid-derived suppressor cells (MDSCs) infiltration, thus reversing the immunosuppressive TME. This multifunctional gas-nanoreactor provides a strategy for CO gas generation to trigger a robust anti-tumor immune response and has the potential for clinical application in cancer immunotherapy. STATEMENT OF SIGNIFICANCE: A multifunctional cascade gas-nanoreactor with a MnO2 gatekeeper was developed to perform synergistic treatments involving starvation therapy, CO gas therapy and chemodynamic therapy (CDT) for tumor elimination. The MnO2 gatekeeper enhanced the catalytic activity of GOx within the nanoreactor by generating oxygen, thereby minimizing toxic side effects after intravenous injection. The gas-nanoreactor amplified ICD through synergistic treatments to mature DC cells and activate anti-tumor immune response. Furthermore, the released CO could reverse the immunosuppression of the TME to enhance cancer immunotherapy. The combination strategy utilizing the gas-nanoreactor demonstrates clinical potential for facilitating cancer immunotherapy.
Collapse
Affiliation(s)
- Hao Ren
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Yunhao Bai
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Zhangya Liu
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Chenyu Ma
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Xinyue Tao
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China
| | - Qiyue Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China.
| | - Huibo Lian
- Urology & Nephrology Center, Cancer Center, Department of Urology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China.
| | - Xueming Li
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing, Jiangsu 211816, China.
| |
Collapse
|
24
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
25
|
Zheng H, Huang L, An G, Guo L, Wang N, Yang W, Zhu Y. A Nanoreactor Based on Metal-Organic Frameworks With Triple Synergistic Therapy for Hepatocellular Carcinoma. Adv Healthc Mater 2024; 13:e2401743. [PMID: 39015058 DOI: 10.1002/adhm.202401743] [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: 05/11/2024] [Revised: 06/17/2024] [Indexed: 07/18/2024]
Abstract
The transformation of monotherapy into multimodal combined targeted therapy to fully exploit synergistic efficacy is of increasing interest in tumor treatment. In this work, a novel nanodrug-carrying platform based on iron-based MOFs, which is loaded with doxorubicin hydrochloride (DOX), dihydroartemisinin (DHA), and glucose oxidase (GOx), and concurrently covalently linked to the photosensitizer 5,10,15,20-tetrakis(4-carboxyphenyl)porphyrin (TCPP) in polydopamine (PDA)-encapsulated MIL-101(Fe) (denoted as MIL-101(Fe)-DOX-DHA@TCPP/GOx@PDA, MDDTG@P), is successfully developed. Upon entering the tumor microenvironment, MDDTG@P catalyzes the hydrogen peroxide (H2O2) into hydroxyl radicals (·OH) and depletes glutathione (GSH); thus, exerting the role of chemodynamic therapy (CDT). The reduced Fe2+ can also activate DHA, further expanding CDT and promoting tumor cell apoptosis. The introduced GOx will rapidly consume glucose and oxygen (O2) in the tumor; while, replenishing H2O2 for Fenton reaction, starving the cancer cells; and thus, realizing starvation and chemodynamic therapy. In addition, the covalent linkage of TCPP endows MDDTG@P with good photodynamic therapeutic (PDT) properties. Therefore, this study develops a nanocarrier platform for triple synergistic chemodynamic/photodynamic/starvation therapy, which has promising applications in the efficient treatment of tumors.
Collapse
Affiliation(s)
- Heming Zheng
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Lei Huang
- School of Stomatology, Minzhu Clinic of Stomatology Hospital Affiliated to Guangxi Medical University, Guangxi, 530007, China
| | - Guanghui An
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Lianshan Guo
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Nannan Wang
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Wenhui Yang
- Department of Medical Laboratory, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, 530021, China
| | - Yanqiu Zhu
- State Key Laboratory of Featured Metal Materials and Life-cycle Safety for Composite Structures, MOE Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, and School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QF, UK
| |
Collapse
|
26
|
Mercadal P, González A, Beloqui A, Tomé LC, Mecerreyes D, Calderón M, Picchio ML. Eutectogels: The Multifaceted Soft Ionic Materials of Tomorrow. JACS AU 2024; 4:3744-3758. [PMID: 39483226 PMCID: PMC11522931 DOI: 10.1021/jacsau.4c00677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 11/03/2024]
Abstract
Eutectogels, a rising category of soft materials, have recently garnered significant attention owing to their remarkable potential in various domains. This innovative class of materials consists of a eutectic solvent immobilized in a three-dimensional network structure. The use of eco-friendly and cost-effective eutectic solvents further emphasizes the appeal of these materials in multiple applications. Eutectogels exhibit key characteristics of most eutectic solvents, including environmental friendliness, facile preparation, low vapor pressure, and good ionic conductivity. Moreover, they can be tailored to display functionalities such as self-healing capability, adhesiveness, and antibacterial properties, which are facilitated by incorporating specific combinations of the eutectic mixture constituents. This perspective article delves into the current landscape and challenges associated with eutectogels, particularly focusing on their potential applications in CO2 separation, drug delivery systems, battery technologies, biocatalysis, and food packaging. By exploring these diverse realms, we aim to shed light on the transformative capabilities of eutectogels and the opportunities they present to address pressing industrial, academic, and environmental challenges.
Collapse
Affiliation(s)
- Pablo
A. Mercadal
- Facultad
de Ciencias Químicas, Departamento de Química Orgánica, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Instituto
de Investigación y Desarrollo en Ingeniería de Procesos
y Química Aplicada (IPQA-CONICET), Córdoba 5000, Argentina
- Facultad
de Ciencias Agropecuarias, Departamento de Recursos Naturales, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
| | - Agustín González
- Facultad
de Ciencias Químicas, Departamento de Química Orgánica, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Instituto
de Investigación y Desarrollo en Ingeniería de Procesos
y Química Aplicada (IPQA-CONICET), Córdoba 5000, Argentina
| | - Ana Beloqui
- POLYMAT,
Applied Chemistry Department, University
of the Basque Country UPV/EHU, Paseo Manuel de Lardizábal, 3, 20018 Donostia-San Sebastián, Spain
- IKERBASQUE,
Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Liliana C. Tomé
- CEMMPRE,
ARISE, Department of Chemical Engineering, University of Coimbra, Pólo II, Rua Sílvio Lima, 3030-790 Coimbra, Portugal
| | - David Mecerreyes
- POLYMAT,
Applied Chemistry Department, University
of the Basque Country UPV/EHU, Paseo Manuel de Lardizábal, 3, 20018 Donostia-San Sebastián, Spain
- IKERBASQUE,
Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Marcelo Calderón
- POLYMAT,
Applied Chemistry Department, University
of the Basque Country UPV/EHU, Paseo Manuel de Lardizábal, 3, 20018 Donostia-San Sebastián, Spain
- IKERBASQUE,
Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Matias L. Picchio
- POLYMAT,
Applied Chemistry Department, University
of the Basque Country UPV/EHU, Paseo Manuel de Lardizábal, 3, 20018 Donostia-San Sebastián, Spain
- IKERBASQUE,
Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| |
Collapse
|
27
|
Li Z, Ma X. Multifunctional Nanocomposite Hydrogel with Enhanced Chemodynamic Therapy and Starvation Therapy for Inhibiting Postoperative Tumor Recurrence. Int J Mol Sci 2024; 25:11465. [PMID: 39519017 PMCID: PMC11547091 DOI: 10.3390/ijms252111465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Surgical resection is the primary treatment for melanoma; however, preventing tumor recurrence after resection remains a significant clinical challenge. To address this, we developed a multifunctional nanocomposite hydrogel (H-CPG) composed of glucose oxidase (GOx)-coated CuS@PDA@GOx (CPG) nanoparticles, aminated hyaluronic acid (HA-ADH), and oxidized rhizomatous polysaccharides (OBSP), which are interconnected through hydrogen bonds and dynamic Schiff base linkages. In the acidic tumor micro-environment, the hydrogel releases GOx, catalyzing the production of hydrogen peroxide (H2O2), which enhances chemokinetic activity through a Cu2+-mediated Fenton-like reaction. This process generates hydroxyl radicals that intensify oxidative stress and promote macrophage polarization from the M2 to M1 phenotype. This polarization triggers the release of pro-inflammatory cytokines, thereby inhibiting tumor recurrence. Additionally, the hydrogel induces photothermal effects that help eradicate residual bacteria at the wound site. Overall, the H-CPG hydrogel offers a dual mechanism to prevent melanoma recurrence and reduce resistance to monotherapy, presenting a promising strategy for postoperative tumor management.
Collapse
Affiliation(s)
- Zeliang Li
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi’an 710069, China;
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi’an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi’an 710069, China
- Biotechnology & Biomed, Research Institute, Northwest University, Xi’an 710069, China
| | - Xiaoxuan Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi’an 710069, China;
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi’an 710069, China
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Xi’an 710069, China
- Biotechnology & Biomed, Research Institute, Northwest University, Xi’an 710069, China
| |
Collapse
|
28
|
Henrique RBL, Santos ALF, Pereira MIA, Santos NRM, Pereira G, Cabral Filho PE, Fontes A. Emerging trends on the uptake of fluorescent probes based on glucose analogs by cancer cells: From basic studies to therapeutics. Life Sci 2024; 355:122978. [PMID: 39147317 DOI: 10.1016/j.lfs.2024.122978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/31/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
The cancer cell metabolism, notably characterized by the Warburg effect, has been the focus of intense investigation regarding the mechanisms of the uptake of glucose analogs, opening up perspectives for diagnosis and treatment of cancer disease. In this review, we delve into the ever-evolving landscape of cancer research, centering on fluorescent probes based on glucose analogs. These analogs, resulting from modifications in the carbohydrate structure with functional groups, have stood out as versatile molecules in applications ranging from disease comprehension to therapeutic innovation, especially when combined with fluorescent compounds. Fluorescence-based assays have provided valuable contributions to the revelation of complex biological mechanisms in life sciences. This review presents selected studies from about the past six years up to 2024 related to the use of glucose-based fluorescent probes, for the investigation of their uptake profile as well as for therapeutic purposes. We believe that these investigations offer insights into the intricate interaction between glucose analogs and cancer cell metabolism, guiding future research and clinical applications in this field.
Collapse
Affiliation(s)
- Rafaella B L Henrique
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil.
| | - Ana L F Santos
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Maria I A Pereira
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Natália R M Santos
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Goreti Pereira
- Departamento de Química Fundamental, Universidade Federal de Pernambuco, Recife, Pernambuco, 50740-560, Brazil; Departamento de Química & CESAM, Universidade de Aveiro, Aveiro, 3810-193, Portugal
| | - Paulo E Cabral Filho
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Adriana Fontes
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Recife, Pernambuco, 50670-901, Brazil.
| |
Collapse
|
29
|
Bai T, Xue P, Shao S, Yan S, Zeng X. Cholesterol Depletion-Enhanced Ferroptosis and Immunotherapy via Engineered Nanozyme. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405826. [PMID: 39120559 PMCID: PMC11481222 DOI: 10.1002/advs.202405826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/12/2024] [Indexed: 08/10/2024]
Abstract
Ferroptosis, an iron- and reactive oxygen species (ROS)-dependent cell death, holds significant promise for tumor therapy due to its ability to induce lipid peroxidation (LPO) and trigger antitumor immune responses. However, elevated cholesterol levels in cancer cells impede ferroptosis and compromise immune function. Here, a novel nanozyme, Fe-MOF/CP, composed of iron metal-organic framework (Fe-MOF) nanoparticles loaded with cholesterol oxidase and PEGylation for integrated ferroptosis and immunotherapy is introduced. Fe-MOF/CP depletes cholesterol and generates hydrogen peroxide, enhancing ROS levels and inducing LPO, thereby promoting ferroptosis. This process disrupts lipid raft integrity and downregulates glutathione peroxidase 4 and ferroptosis suppressor protein 1, further facilitating ferroptosis. Concurrently, Fe-MOF/CP augments immunogenic cell death, reduces programmed death-ligand 1 expression, and revitalizes exhausted CD8+ T cells. In vivo studies demonstrate significant therapeutic efficacy in abscopal, metastasis, and recurrent tumor models, highlighting the robust antitumor immune responses elicited by Fe-MOF/CP. This study underscores the potential of Fe-MOF/CP as a multifunctional therapeutic agent that combines ferroptosis and immunotherapy, offering a promising strategy for effective and durable cancer treatment.
Collapse
Affiliation(s)
- Tingjie Bai
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province UniversityBiomedical Research Center of South ChinaCollege of Life SciencesFujian Normal UniversityFuzhou350117China
| | - Panpan Xue
- The Straits Institute of Flexible Electronics (SIFEFuture Technologies)Straits Laboratory of Flexible Electronics (SLoFE)Fujian Normal UniversityFuzhou350117China
| | - Sijie Shao
- The Straits Institute of Flexible Electronics (SIFEFuture Technologies)Straits Laboratory of Flexible Electronics (SLoFE)Fujian Normal UniversityFuzhou350117China
| | - Shuangqian Yan
- The Straits Institute of Flexible Electronics (SIFEFuture Technologies)Straits Laboratory of Flexible Electronics (SLoFE)Fujian Normal UniversityFuzhou350117China
| | - Xuemei Zeng
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province UniversityBiomedical Research Center of South ChinaCollege of Life SciencesFujian Normal UniversityFuzhou350117China
| |
Collapse
|
30
|
Xu X, Zhang Y, Meng C, Zheng W, Wang L, Zhao C, Luo F. Nanozymes in cancer immunotherapy: metabolic disruption and therapeutic synergy. J Mater Chem B 2024; 12:9111-9143. [PMID: 39177061 DOI: 10.1039/d4tb00769g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Over the past decade, there has been a growing emphasis on investigating the role of immunotherapy in cancer treatment. However, it faces challenges such as limited efficacy, a diminished response rate, and serious adverse effects. Nanozymes, a subset of nanomaterials, demonstrate boundless potential in cancer catalytic therapy for their tunable activity, enhanced stability, and cost-effectiveness. By selectively targeting the metabolic vulnerabilities of tumors, they can effectively intensify the destruction of tumor cells and promote the release of antigenic substances, thereby eliciting immune clearance responses and impeding tumor progression. Combined with other therapies, they synergistically enhance the efficacy of immunotherapy. Hence, a large number of metabolism-regulating nanozymes with synergistic immunotherapeutic effects have been developed. This review summarizes recent advancements in cancer immunotherapy facilitated by nanozymes, focusing on engineering nanozymes to potentiate antitumor immune responses by disturbing tumor metabolism and performing synergistic treatment. The challenges and prospects in this field are outlined. We aim to provide guidance for nanozyme-mediated immunotherapy and pave the way for achieving durable tumor eradication.
Collapse
Affiliation(s)
- Xiangrui Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yaowen Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chijun Meng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenzhuo Zheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lingfeng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chenyi Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Feng Luo
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu 610041, China.
| |
Collapse
|
31
|
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
|
32
|
Xie W, Lu Y, Yuan Y, Xiao L, Liu J, Song H, Niu R, Liu Y, Lin J. Hyaluronic Acid-Modified Spherical MgO 2/Pd Nanocomposites Exhibit Superior Antitumor Effect through Tumor Microenvironment-Responsive Ferroptosis Induction and Photothermal Therapy. ACS Biomater Sci Eng 2024; 10:5226-5236. [PMID: 38943566 DOI: 10.1021/acsbiomaterials.4c00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Metal peroxide nanomaterials as efficient hydrogen peroxide (H2O2) self-supplying agents have attracted the attention of researchers for antitumor treatment. However, relying solely on metal peroxides to provide H2O2 is undoubtedly insufficient to achieve optimal antitumor effects. Herein, we construct novel hyaluronic acid (HA)-modified nanocomposites (MgO2/Pd@HA NCs) formed by decorating palladium nanoparticles (Pd NPs) onto the surfaces of a magnesium peroxide (MgO2) nanoflower as a highly effective nanoplatform for the tumor microenvironment (TME)-responsive induction of ferroptosis in tumor cells and tumor photothermal therapy (PTT). MgO2/Pd@HA NC could be well endocytosed into tumor cells with CD44 expression depending on the specific recognition of HA with CD44, and then, the nanocomposites can be rapidly decomposed in mild acid and hyaluronidase overexpressed TME, and plenty of H2O2 was released. Simultaneously, Pd NPs catalyze self-supplied H2O2 to generate abundant hydroxyl radicals (•OH) and catalyze glutathione (GSH) into glutathione disulfide owing to its peroxidase and glutathione oxidase mimic enzyme activities, while the abundant •OH could also consume GSH in tumor cells and disturb the defense pathways of ferroptosis leading to the accumulation of lipid peroxidation and resulting in the occurrence of ferroptosis. Additionally, the superior photothermal conversion performance of Pd NPs in near-infrared II could also be used for PTT, synergistically cooperating with nanocomposite-induced ferroptosis for tumor inhibition. Consequently, the successfully prepared TME-responsive MgO2/Pd@HA NCs exhibited marked antitumor effect without obvious biotoxicity, contributing to thoroughly explore the nanocomposites as a novel and promising treatment for tumor therapy.
Collapse
Affiliation(s)
- Wenhui Xie
- Henan Joint International Research Laboratory of Stem Cell Medicine, College of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Yilin Lu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yilin Yuan
- Henan Joint International Research Laboratory of Stem Cell Medicine, College of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Lulu Xiao
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Jiaqi Liu
- Henan Joint International Research Laboratory of Stem Cell Medicine, College of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
| | - Haofeng Song
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Rongcheng Niu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, College of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, China
- Stem Cell and Biotherapy Technology Research Center, College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
33
|
Zhang J, Zhou J, Tang L, Ma J, Wang Y, Yang H, Wang X, Fan W. Custom-Design of Multi-Stimuli-Responsive Degradable Silica Nanoparticles for Advanced Cancer-Specific Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400353. [PMID: 38651235 DOI: 10.1002/smll.202400353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/24/2024] [Indexed: 04/25/2024]
Abstract
Chemotherapy is crucial in oncology for combating malignant tumors but often encounters obatacles such as severe adverse effects, drug resistance, and biocompatibility issues. The advantages of degradable silica nanoparticles in tumor diagnosis and treatment lie in their ability to target drug delivery, minimizing toxicity to normal tissues while enhancing therapeutic efficacy. Moreover, their responsiveness to both endogenous and exogenous stimuli opens up new possibilities for integrating multiple treatment modalities. This review scrutinizes the burgeoning utility of degradable silica nanoparticles in combination with chemotherapy and other treatment modalities. Commencing the elucidation of degradable silica synthesis and degradation mechanisms, emphasis is placed on the responsiveness of these materials to endogenous (e.g., pH, redox reactions, hypoxia, and enzymes) and exogenous stimuli (e.g., light and high-intensity focused ultrasound). Moreover, this exploration delves into strategies harnessing degradable silica nanoparticles in chemotherapy alone, coupled with radiotherapy, photothermal therapy, photodynamic therapy, gas therapy, immunotherapy, starvation therapy, and chemodynamic therapy, elucidating multimodal synergies. Concluding with an assessment of advances, challenges, and constraints in oncology, despite hurdles, future investigations are anticipated to augment the role of degradable silica in cancer therapy. These insights can serve as a compass for devising more efficacious combined tumor treatment strategies.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, P. R. China
| | - Jiani Zhou
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | | | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Ying Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243032, P. R. China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
34
|
Alsamarat R, Sunoqrot S. A Glucose Oxidase-Curcumin Composite Nanoreactor for Multimodal Synergistic Cancer Therapy. ACS APPLIED BIO MATERIALS 2024; 7:4611-4621. [PMID: 38920441 DOI: 10.1021/acsabm.4c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Glucose oxidase (GOx) selectively oxidizes β-d-glucose into gluconic acid and hydrogen peroxide; thus, it has emerged as a promising anticancer agent by tumor starvation and oxidative therapy. Here, we developed a nanoscale platform or "nanoreactor" that incorporates GOx and the bioactive natural product curcumin (CUR) to achieve a multimodal anticancer nanocomposite. The composite nanoreactor was formed by loading CUR in biodegradable polymeric nanoparticles (NPs) of poly(ethylene glycol)-b-poly(ε-caprolactone) (PEG-PCL). Prime-coating of the NPs with an iron(III)-tannic acid complex enabled facile immobilization of GOx on the NP surface. The NPs were monodisperse with a hydrodynamic diameter of 122 nm and a partially negative surface charge. The NPs were also associated with an excellent CUR loading efficiency and sustained release up to 96 h, which was accelerated by surface-immobilized GOx and followed supercase II transport. Viability assays were conducted on two model cancer cell lines, MCF-7 and MDA-MB-231 cells, as well as human dermal fibroblasts as a representative normal cell line. The assays revealed significantly improved potency of CUR in the composite nanoreactor, with up to 6000- and 1280-fold increase in MCF-7 and MDA-MB-231 cells, respectively, and lower toxicity toward normal cells. The NPs were also able to promote intracellular reactive oxygen species (ROS) generation and dissipation of the mitochondrial membrane potential, providing important clues on the mechanism of action of the nanoreactor. Further investigation of caspase-3 activity revealed that the nanoreactor had no effect or inhibited caspase-3 levels, signifying a caspase-independent mechanism of inducing apoptosis. Our findings present a promising nanocarrier platform that combines therapeutic agents with distinct mechanisms of action acting in synergy for more effective cancer therapy.
Collapse
Affiliation(s)
- Rama Alsamarat
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
35
|
Liang X, Chen W, Wang C, Jiang K, Zhu J, Lu R, Lin Z, Cao Z, Zheng J. A mesoporous theranostic platform for ultrasound and photoacoustic dual imaging-guided photothermal and enhanced starvation therapy for cancer. Acta Biomater 2024; 183:264-277. [PMID: 38815685 DOI: 10.1016/j.actbio.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024]
Abstract
Tumor starvation therapy utilizing glucose oxidase (GOx), has gained traction due to its non-invasive and bio-safe attributes. However, its effectiveness is often hampered by severe hypoxia in the tumor microenvironment (TME), limiting GOx's catalytic activity. To address this issue, a multifunctional nanosystem based on mesoporous polydopamine nanoparticles (MPDA NPs) was developled to alleviate TME hypoxia. This nanosystem integrated GOx modification and oxygenated perfluoropentane (PFP) encapsulation to address hypoxia-related challenges in the TME. Under NIR laser irradiation, the MPDA NPs exhibit significant photothermal conversion efficacy, activating targeted tumor photothermal therapy (PTT), while also serving as proficient photoacoustic (PA) imaging agents. The ensuing temperature rise facilitates oxygen (O2) release and induces liquid-gas conversion of PFP, generating microbubbles for enhanced ultrasound (US) imaging signals. The supplied oxygen alleviates local hypoxia, thereby enhancing GOx-mediated endogenous glucose consumption for tumor starvation. Overall, the integration of ultrasound/photoacoustic dual imaging-guided PTT and starvation therapy within MPDA-GOx@PFP@O2 nanoparticles (MGPO NPs) presents a promising platform for enhancing the efficacay of tumor treatment by overcoming the complexities of the TME. STATEMENT OF SIGNIFICANCE: A multifunctional MPDA-based theranostic nanoagent was developed for US/PAI imaging-guided PTT and starvation therapy against tumor hypoxia by direct O2 delivery. The incorporation of oxygenated perfluoropentane (PFP) within the mesoporous structure of MGPO not only enables efficient US imaging but also helps in alleviating tumor hypoxia. Moreover, the strong near-infrared (NIR) absorption of MGPO NPs promote the generation of PFP microbubbles and release of oxygen, thereby enhancing US imaging and GOx-mediated starvation therapy. Such a multifunctional nanosystem leverages synergistic effects to enhance therapeutic efficacy while incorporating US/PA imaging for precise visualization of the tumor.
Collapse
Affiliation(s)
- Xiaotong Liang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Wenbo Chen
- Ultrasound Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China
| | - Chunan Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Kai Jiang
- Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, 518116, Guangdong, China
| | - Jinjin Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Ruitao Lu
- Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, 518116, Guangdong, China
| | - Zhousheng Lin
- Department of Orthopaedics, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Zhong Cao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, No. 66. Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China; Shenzhen International Institute for Biomedical Research, Longhua District, Shenzhen, 518116, Guangdong, China.
| | - Jian Zheng
- Ultrasound Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Shenzhen, 518172, China.
| |
Collapse
|
36
|
Xing Z, Li L, Liao T, Wang J, Guo Y, Xu Z, Yu W, Kuang Y, Li C. A multifunctional cascade enzyme system for enhanced starvation/chemodynamic combination therapy against hypoxic tumors. J Colloid Interface Sci 2024; 666:244-258. [PMID: 38598997 DOI: 10.1016/j.jcis.2024.04.036] [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: 02/02/2024] [Revised: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024]
Abstract
Starvation therapy has shown promise as a cancer treatment, but its efficacy is often limited when used alone. In this work, a multifunctional nanoscale cascade enzyme system, named CaCO3@MnO2-NH2@GOx@PVP (CMGP), was fabricated for enhanced starvation/chemodynamic combination cancer therapy. CMGP is composed of CaCO3 nanoparticles wrapped in a MnO2 shell, with glucose oxidase (GOx) adsorbed and modified with polyvinylpyrrolidone (PVP). MnO2 decomposes H2O2 in cancer cells into O2, which enhances the efficiency of GOx-mediated starvation therapy. CaCO3 can be decomposed in the acidic cancer cell environment, causing Ca2+ overload in cancer cells and inhibiting mitochondrial metabolism. This synergizes with GOx to achieve more efficient starvation therapy. Additionally, the H2O2 and gluconic acid produced during glucose consumption by GOx are utilized by MnO2 with catalase-like activity to enhance O2 production and Mn2+ release. This process accelerates glucose consumption, reactive oxygen species (ROS) generation, and CaCO3 decomposition, promoting the Ca2+ release. CMGP can alleviate tumor hypoxia by cycling the enzymatic cascade reaction, which increases enzyme activity and combines with Ca2+ overload to achieve enhanced combined starvation/chemodynamic therapy. In vitro and in vivo studies demonstrate that CMGP has effective anticancer abilities and good biosafety. It represents a new strategy with great potential for combined cancer therapy.
Collapse
Affiliation(s)
- Zihan Xing
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Linwei Li
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Tao Liao
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Jinyu Wang
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Yuhao Guo
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Ziqiang Xu
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China
| | - Wenqian Yu
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Ying Kuang
- Hubei Key Laboratory of Industry Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, China.
| | - Cao Li
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, College of Health Science and Engineering, School of Materials Science and Engineering, Hubei University, Wuhan 430062, China; Hubei Key Laboratory of Industry Microbiology, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Glyn O. Phillips Hydrocolloid Research Centre at HBUT, School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, China.
| |
Collapse
|
37
|
Korpidou M, Becker J, Tarvirdipour S, Dinu IA, Becer CR, Palivan CG. Glycooligomer-Functionalized Catalytic Nanocompartments Co-Loaded with Enzymes Support Parallel Reactions and Promote Cell Internalization. Biomacromolecules 2024; 25:4492-4509. [PMID: 38910355 PMCID: PMC11238334 DOI: 10.1021/acs.biomac.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024]
Abstract
A major shortcoming associated with the application of enzymes in drug synergism originates from the lack of site-specific, multifunctional nanomedicine. This study introduces catalytic nanocompartments (CNCs) made of a mixture of PDMS-b-PMOXA diblock copolymers, decorated with glycooligomer tethers comprising eight mannose-containing repeating units and coencapsulating two enzymes, providing multifunctionality by their in situ parallel reactions. Beta-glucuronidase (GUS) serves for local reactivation of the drug hymecromone, while glucose oxidase (GOx) induces cell starvation through glucose depletion and generation of the cytotoxic H2O2. The insertion of the pore-forming peptide, melittin, facilitates diffusion of substrates and products through the membranes. Increased cell-specific internalization of the CNCs results in a substantial decrease in HepG2 cell viability after 24 h, attributed to simultaneous production of hymecromone and H2O2. Such parallel enzymatic reactions taking place in nanocompartments pave the way to achieve efficient combinatorial cancer therapy by enabling localized drug production along with reactive oxygen species (ROS) elevation.
Collapse
Affiliation(s)
- Maria Korpidou
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - Jonas Becker
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Shabnam Tarvirdipour
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - Ionel Adrian Dinu
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
| | - C. Remzi Becer
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Cornelia G. Palivan
- Department
of Chemistry, University of Basel, Mattenstrasse 22, Basel 4002, Switzerland
- NCCR
Molecular Systems Engineering, Mattenstrasse 22, Basel 4002, Switzerland
| |
Collapse
|
38
|
Zhang SY, Zhou XY, Chen HY, Deng LY, Li DW, Lv J, Qian RC. Real time imaging of cell-permeable nanoreactor with SERS for insight into cellular processes. Talanta 2024; 274:126010. [PMID: 38569372 DOI: 10.1016/j.talanta.2024.126010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
Intracellular glucose detection is crucial due to its pivotal role in metabolism and various physiological processes. Precise glucose monitoring holds significance in diabetes management, metabolic studies, and biotechnological applications. In this study, we developed an innovative and expedient cell-permeable nanoreactor for intracellular glucose based on surface-enhanced Raman scattering (SERS). The nanoreactor was designed with gold nanoparticles (AuNPs), which were engineered with glucose oxide (GOx) and a H2O2-responsive Raman reporter 2-mercaptohydroquinone (2-MHQ). The interaction between 2-MHQ and H2O2 generated by glucose and GOx could simultaneously induce the appearance in the peak at 985 cm-1. Our results showed excellent performance in detecting glucose within the concentration range from 0.1 μM to 10 mM, with a low detection limitation of 14.72 nM. In addition, the glucose distribution in single HeLa cells was evaluated by real time SERS mapping. By combining noble metal particles and natural oxidases, the nanoreactor possesses both Raman activity and enzymatic functionality, thus enables sensitive glucose detection and facilitates imaging at a single cell level, which offers an insightful monitoring of cellular processes.
Collapse
Affiliation(s)
- Shi-Yi Zhang
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Xin-Yue Zhou
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hua-Ying Chen
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Li-Yuan Deng
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Da-Wei Li
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Jian Lv
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Ruo-Can Qian
- Key Laboratory for Advanced Materials, Feringa Nobel Prize Scientist Joint Research Center, Joint International Laboratory for Precision Chemistry, Frontiers Science Center for Materiobiology & Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| |
Collapse
|
39
|
Yang G, Wang DY, Song J, Ren Y, An Y, Busscher HJ, van der Mei HC, Shi L. Cetyltrimethylammonium-chloride assisted in situ metabolic incorporation of nano-sized ROS-generating cascade-reaction containers in Gram-positive and Gram-negative peptidoglycan layers for the control of bacterially-induced sepsis. Acta Biomater 2024; 181:347-361. [PMID: 38702010 DOI: 10.1016/j.actbio.2024.04.045] [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: 01/26/2024] [Revised: 04/03/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Cascade-reaction containers generating reactive oxygen species (ROS) as an alternative for antibiotic-based strategies for bacterial infection control, require endogenous oxygen-sources and ROS-generation close to or preferably inside target bacteria. Here, this is achieved by cetyltrimethylammonium-chloride (CTAC) assisted in situ metabolic labeling and incorporation of mesoporous SiO2-nanoparticles, dual-loaded with glucose-oxidase and Fe3O4-nanoparticles as cascade-reaction containers, inside bacterial cell walls. First, azide-functionalized d-alanine (D-Ala-N3) was inserted in cell wall peptidoglycan layers of growing Gram-positive pathogens. In Gram-negatives, this could only be achieved after outer lipid-membrane permeabilization, using a low concentration of CTAC. Low concentrations of CTAC had no adverse effect on in vitro blood clotting or hemolysis nor on the health of mice when blood-injected. Next, dibenzocyclooctyne-polyethylene-glycol modified, SiO2-nanoparticles were in situ click-reacted with d-Ala-N3 in bacterial cell wall peptidoglycan layers. Herewith, a two-step cascade-reaction is facilitated inside bacteria, in which glucose-oxidase generates H2O2 at endogenously-available glucose concentrations, while subsequently Fe3O4-nanoparticles catalyze generation of •OH from the H2O2 generated. Generation of •OH inside bacterial cell walls by dual-loaded mesoporous SiO2-nanoparticles yielded more effective in vitro killing of both planktonic Gram-positive and Gram-negative bacteria suspended in 10 % plasma than SiO2-nanoparticles solely loaded with glucose-oxidase. Gram-positive or Gram-negative bacterially induced sepsis in mice could be effectively treated by in situ pre-treatment with tail-vein injected CTAC and d-Ala-N3, followed by injection of dual-loaded cascade-reaction containers without using antibiotics. This makes in situ metabolic incorporation of cascade-reaction containers as described attractive for further investigation with respect to the control of other types of infections comprising planktonic bacteria. STATEMENT OF SIGNIFICANCE: In situ metabolic-incorporation of cascade-reaction-containers loaded with glucose-oxidase and Fe3O4 nanoparticles into bacterial cell-wall peptidoglycan is described, yielding ROS-generation from endogenous glucose, non-antibiotically killing bacteria before ROS inactivates. Hitherto, only Gram-positives could be metabolically-labeled, because Gram-negatives possess two lipid-membranes. The outer membrane impedes direct access to the peptidoglycan. This problem was solved by outer-membrane permeabilization using a quaternary-ammonium compound. Several studies on metabolic-labeling perform crucial labeling steps during bacterial-culturing that in real-life should be part of a treatment. In situ metabolic-incorporation as described, can be applied in well-plates during in vitro experiments or in the body as during in vivo animal experiments. Surprisingly, metabolic-incorporation proceeded unhampered in blood and a murine, bacterially-induced sepsis could be well treated.
Collapse
Affiliation(s)
- Guang Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, PR China; University of Groningen and University Medical Center Groningen, Department of Biomaterials & Biomedical Technology, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Da-Yuan Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, PR China; University of Groningen and University Medical Center Groningen, Department of Biomaterials & Biomedical Technology, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Jianwen Song
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, PR China
| | - Yijin Ren
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Yingli An
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, PR China
| | - Henk J Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomaterials & Biomedical Technology, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| | - Henny C van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomaterials & Biomedical Technology, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
40
|
Wang Z, Wang X, Dai X, Xu T, Qian X, Chang M, Chen Y. 2D Catalytic Nanozyme Enables Cascade Enzyodynamic Effect-Boosted and Ca 2+ Overload-Induced Synergistic Ferroptosis/Apoptosis in Tumor. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312316. [PMID: 38501540 DOI: 10.1002/adma.202312316] [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: 11/17/2023] [Revised: 02/21/2024] [Indexed: 03/20/2024]
Abstract
The introduction of glucose oxidase, exhibiting characteristics of glucose consumption and H2O2 production, represents an emerging antineoplastic therapeutic approach that disrupts nutrient supply and promotes efficient generation of reactive oxygen species (ROS). However, the instability of natural enzymes and their low therapeutic efficacy significantly impede their broader application. In this context, 2D Ca2Mn8O16 nanosheets (CMO NSs) designed and engineered to serve as a high-performance nanozyme, enhancing the enzyodynamic effect for a ferroptosis-apoptosis synergistic tumor therapy, are presented. In addition to mimicking activities of glutathione peroxidase, catalase, oxidase, and peroxidase, the engineered CMO NSs exhibit glucose oxidase-mimicking activities. This feature contributes to their antitumor performance through cascade catalytic reactions, involving the disruption of glucose supply, self-supply of H2O2, and subsequent efficient ROS generation. The exogenous Ca2+ released from CMO NSs, along with the endogenous Ca2+ enrichment induced by ROS from the peroxidase- and oxidase-mimicking activities of CMO NSs, collectively mediate Ca2+ overload, leading to apoptosis. Importantly, the ferroptosis process is triggered synchronously through ROS output and glutathione consumption. The application of exogenous ultrasound stimulation further enhances the efficiency of ferroptosis-apoptosis synergistic tumor treatment. This work underscores the crucial role of enzyodynamic performance in ferroptosis-apoptosis synergistic therapy against tumors.
Collapse
Affiliation(s)
- Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xue Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China
| | - Xinyue Dai
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Tianming Xu
- Department of Orthopedics, 905th Hospital of PLA Navy, Naval Medical University, Shanghai, 200050, P. R. China
| | - Xiaoqin Qian
- Department of Ultrasound Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225009, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, 325088, P. R. China
| |
Collapse
|
41
|
Zou W, Gao F, Meng Z, Cai X, Chen W, Zheng Y, Ying T, Wang L, Wu J. Lactic acid responsive sequential production of hydrogen peroxide and consumption of glutathione for enhanced ferroptosis tumor therapy. J Colloid Interface Sci 2024; 663:787-800. [PMID: 38442520 DOI: 10.1016/j.jcis.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ferroptosis is characterized by the lethal accumulation of lipid reactive oxygen species (ROS), which has great potential for tumor therapy. However, developing new ferroptosis-inducing strategies by combining nanomaterials with small molecule inducers is important. In this study, an enzyme-gated biodegradable natural-product delivery system based on lactate oxidase (LOD)-gated biodegradable iridium (Ir)-doped hollow mesoporous organosilica nanoparticles (HMONs) loaded with honokiol (HNK) (HNK@Ir-HMONs-LOD, HIHL) is designed to enhance ferroptosis in colon tumor therapy. After reaching the tumor microenvironment, the outer LOD dissociates and releases the HNK to induce ferroptosis. Moreover, the released dopant Ir4+ and disulfide-bridged organosilica frameworks deplete intracellular glutathione (GSH), which is followed by GSH-mediated Ir(IV)/Ir(III) conversion. This leads to the repression of glutathione peroxidase 4 (GPX4) activity and decomposition of intratumoral hydrogen peroxide (H2O2) into hydroxyl radicals (•OH) by Ir3+-mediated Fenton-like reactions. Moreover, LOD efficiently depletes lactic acid to facilitate the generation of H2O2 and boost the Fenton reaction, which in turn enhances ROS generation. With the synergistic effects of these cascade reactions and the release of HNK, notable ferroptosis efficacy was observed both in vitro and in vivo. This combination of natural product-induced and lactic acid-responsive sequential production of H2O2 as well as the consumption of glutathione may provide a new paradigm for achieving effective ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Weijuan Zou
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Feng Gao
- Department of Ultrasonic Imaging, the First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zheying Meng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Xiaojun Cai
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Wu Chen
- Department of Ultrasonic Imaging, the First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Tao Ying
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| | - Longchen Wang
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| | - Jianrong Wu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| |
Collapse
|
42
|
Li H, Li Y, Su L, Zheng K, Zhang Y, Li J, Lv F, Huang M, Chen T, Zhang H, Shi Z, Zhu D, Dong X, Zeng W, Mei L. Enzyme-Empowered "Two Birds with One Stone" Strategy for Amplifying Tumor Apoptosis and Metabolic Clearance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308251. [PMID: 38447152 PMCID: PMC11095162 DOI: 10.1002/advs.202308251] [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: 10/31/2023] [Revised: 02/03/2024] [Indexed: 03/08/2024]
Abstract
Nanomedicine has reshaped the landscape of cancer treatment. However, its efficacy is still hampered by innate tumor defense systems that rely on adenosine triphosphate (ATP) for fuel, including damage repair, apoptosis resistance, and immune evasion. Inspired by the naturally enzymatic reaction of glucose oxidase (GOx) with glucose, here a novel "two birds with one stone" technique for amplifying enzyme-mediated tumor apoptosis and enzyme-promoted metabolic clearance is proposed and achieved using GOx-functionalized rhenium nanoclusters-doped polypyrrole (Re@ReP-G). Re@ReP-G reduces ATP production while increasing H2O2 concentrations in the tumor microenvironment through GOx-induced enzymatic oxidation, which in turn results in the downregulation of defense (HSP70 and HSP90) and anti-apoptotic Bcl-2 proteins, the upregulation of pro-apoptotic Bax, and the release of cytochrome c. These processes are further facilitated by laser-induced hyperthermia effect, ultimately leading to severe tumor apoptosis. As an enzymatic byproduct, H2O2 catalyzes the conversion of rhenium nanoclusters in Re@ReP-G nanostructures into rhenate from the outside in, which accelerates their metabolic clearance in vivo. This Re@ReP-G-based "two birds with one stone" therapeutic strategy provides an effective tool for amplifying tumor apoptosis and safe metabolic mechanisms.
Collapse
Affiliation(s)
- Hanyue Li
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Yihui Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120P. R. China
| | - Lina Su
- Department of PharmacyQujing Medical CollegeQujing655000P. R. China
| | - Ke Zheng
- School of Materials Science and EngineeringDongguan University of TechnologyDongguan523808P. R. China
| | - Yue Zhang
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Jing Li
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Feng Lv
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Mengjie Huang
- Department of NephrologyFirst Medical Center of Chinese PLA General HospitalNational Key Laboratory of Kidney DiseasesNational Clinical Research Center for Kidney DiseasesBeijing Key Laboratory of Kidney Diseases ResearchBeijing100853P. R. China
| | - Ting Chen
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Hanjie Zhang
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhaoqing Shi
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Dunwan Zhu
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Xia Dong
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Weiwei Zeng
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Biomedical MaterialsInstitute of Biomedical EngineeringChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin300192P. R. China
- Department of PharmacyQujing Medical CollegeQujing655000P. R. China
| |
Collapse
|
43
|
Wang B, Huang Y, Cai Q, Du Z, Li X. Biomaterials for diabetic bone repair: Influencing mechanisms, multi-aspect progress and future prospects. COMPOSITES PART B: ENGINEERING 2024; 274:111282. [DOI: 10.1016/j.compositesb.2024.111282] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
|
44
|
Xue K, Yang R, An Y, Ding Y, Li S, Miao F, Liu D, Chen D, Tang Q. NIR-promoted ferrous ion regeneration enhances ferroptosis for glioblastoma treatment. J Control Release 2024; 368:595-606. [PMID: 38185333 DOI: 10.1016/j.jconrel.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
Ferroptosis, a unique iron-dependent mode of cell death characterized by lipid peroxide accumulation, holds significant potential for the treatment of glioblastoma (GBM). However, the effectiveness of ferroptosis is hindered by the limited intracellular ferrous ions (Fe2+) and hydrogen peroxide (H2O2). In this study, a novel near-infrared (NIR)-light-responsive nanoplatform (ApoE-UMSNs-GOx/SRF) based on upconversion nanoparticles (UCNPs) was developed. A layer of mesoporous silica and a lipid bilayer were coated on UCNPs sequentially and loaded with glucose oxidase (GOx) and sorafenib, respectively. Further attachment of the ApoE peptide endowed the nanoplatform with BBB penetration and GBM targeting capabilities. Our results revealed that ApoE-UMSNs-GOx/SRF could efficiently accumulated in the orthotopic GBM and induce amplified ferroptosis when combining with NIR irradiation. The UCNPs mediated the photoreduction of Fe3+ to Fe2+ by converting NIR to UV light, and excess H2O2 was produced by the reaction of glucose with the loaded GOx. These processes greatly promoted the production of ROS, which together with inhibition of system Xc- by the loaded sorafenib, leading to enhanced accumulation of lipid peroxides and significantly improved the antiglioma effect both in vitro and in vivo. Our strategy has the potential to enhance the effectiveness of ferroptosis as a therapeutic approach for GBM.
Collapse
Affiliation(s)
- Kangli Xue
- Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Rui Yang
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China
| | - Yanli An
- Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Yinan Ding
- Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Su Li
- Nanjing Medical University, Nanjing 211166, China
| | - Fengqin Miao
- Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, China
| | - Dongfang Liu
- Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, China.
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, China.
| | - Qiusha Tang
- Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, China.
| |
Collapse
|
45
|
Zhang M, Xue H, Yang J, Zhao X, Xue M, Sun W, Qiu J, Zhu Z. Copper(II)-based metal-organic framework delivery of calcium ascorbate for enhanced chemodynamic therapy via H 2O 2 self-supply and glutathione depletion. Biomater Sci 2024; 12:1871-1882. [PMID: 38411574 DOI: 10.1039/d3bm01922e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Chemodynamic therapy (CDT) is a promising cancer treatment strategy. However, mild acidic pH, insufficient H2O2 content, and overexpressed glutathione (GSH) in the tumor microenvironment (TME) severely impair CDT efficiency. In this study, a novel therapeutic nanosystem (Cu/ZIF-8/Vc-Ca/HA) was constructed for H2O2 self-supply and GSH depletion co-enhanced CDT. Typically, calcium ascorbate (Vc-Ca) loaded on the surface of Cu2+-doped ZIF-8 (Cu/ZIF-8) was designed as an original source for H2O2 generation, and a hyaluronic acid (HA) shell was subsequently coated to act as a tumor-targeted "guide" and protective layer. Along with the HA shell disintegrated in the TME, exposed Cu/ZIF-8/Vc-Ca dissociated in the tumor acidic microenvironment, thus triggering the release of Vc-Ca and Cu2+. Vc-Ca selectively produced H2O2 in tumor cells, which provided abundant H2O2 for boosting Fenton-like reactions. Meanwhile, the released Cu2+ could get converted into Cu+ by consuming excess intracellular GSH, which could reduce the tumor antioxidant capability of the nanosystem. Moreover, byproduct Cu+ reacted with abundant H2O2 by a highly efficient Fenton-like reaction to generate toxic ˙OH. Biological assays indicated that the Cu/ZIF-8/HA@Vc-Ca nanosystem showed significant anticancer activity by enhancing the CDT process. This study may provide a new strategy for improving the effectiveness of CDT.
Collapse
Affiliation(s)
- Meng Zhang
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - Hongjin Xue
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - Jiaxin Yang
- School of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Xin Zhao
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - Mei Xue
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - Wei Sun
- Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Jianfeng Qiu
- School of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| | - Zhihong Zhu
- Institute of Nano-science and Nano-technology, College of Physical Science and Technology, Central China Normal University, Wuhan, 430079, China
| |
Collapse
|
46
|
Wang Y, Jia X, An S, Yin W, Huang J, Jiang X. Nanozyme-Based Regulation of Cellular Metabolism and Their Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301810. [PMID: 37017586 DOI: 10.1002/adma.202301810] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Metabolism is the sum of the enzyme-dependent chemical reactions, which produces energy in catabolic process and synthesizes biomass in anabolic process, exhibiting high similarity in mammalian cell, microbial cell, and plant cell. Consequently, the loss or gain of metabolic enzyme activity greatly affects cellular metabolism. Nanozymes, as emerging enzyme mimics with diverse functions and adjustable catalytic activities, have shown attractive potential for metabolic regulation. Although the basic metabolic tasks are highly similar for the cells from different species, the concrete metabolic pathway varies with the intracellular structure of different species. Here, the basic metabolism in living organisms is described and the similarities and differences in the metabolic pathways among mammalian, microbial, and plant cells and the regulation mechanism are discussed. The recent progress on regulation of cellular metabolism mainly including nutrient uptake and utilization, energy production, and the accompanied redox reactions by different kinds of oxidoreductases and their applications in the field of disease therapy, antimicrobial therapy, and sustainable agriculture is systematically reviewed. Furthermore, the prospects and challenges of nanozymes in regulating cell metabolism are also discussed, which broaden their application scenarios.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xiaodan Jia
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Shangjie An
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Wenbo Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Jiahao Huang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Xiue Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
47
|
Peng J, Zhou J, Liu X, Zhang X, Zhou X, Gong Z, Chen Y, Shen X, Chen Y. A biomimetic nanocarrier facilitates glucose consumption and reactive oxide species accumulation in enzyme therapy for colorectal cancer. J Control Release 2024; 367:76-92. [PMID: 38262488 DOI: 10.1016/j.jconrel.2024.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Glucose oxidase (GOx)-based enzyme therapeutics are potential alternatives for colorectal cancer (CRC) treatment via glucose consumption and accumulation of hydrogen peroxide (H2O2). Given that H2O2 can be eliminated by cytoprotective autophagy, autophagy inhibitors that can interrupt autolysosome-induced H2O2 elimination are promising combination drugs of GOx. Here, we developed a multifunctional biomimetic nanocarrier for effective co-delivery of an autophagy inhibitor-chloroquine phosphate (CQP) and GOx to exert their synergistic effect by irreversibly upregulating intracellular reactive oxygen species (ROS) levels. Poly (D, l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were used to encapsulate both GOx and CQP using a W/O/W multi-emulsion method. Calcium phosphate (CaP) was used to "fix" CQP to GOx in the internal water phase, where it served as a pH-sensitive unit to facilitate intracellular drug release. Folic acid-modified red blood cell membranes (FR) were used to camouflage the GOx/CQP/CaP encapsulated PLGA NPs (referred to as PLGA/GCC@FR). In an AOM/DSS-induced CRC mouse model, PLGA/GCC@FR exhibited improved antitumor effects, in which the number of tumor nodes were only a quarter of that in the free drug combination group. The enhanced therapeutic effects of PLGA/GCC@FR were attributed to the prolonged tumor retention which was verified by both dynamic in vivo imaging and drug biodistribution. This multifunctional biomimetic nanocarrier facilitated combined enzyme therapeutics by depleting glucose and augmenting intracellular ROS levels in tumor cells, which exerted a synergistic inhibitory effect on tumor growth. Therefore, this study proposed a novel strategy for the enhancement of combined enzyme therapeutics, which provided a promising method for effective CRC treatment.
Collapse
Affiliation(s)
- Jianqing Peng
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Jia Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xing Liu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xiaobo Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xiang Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Zipeng Gong
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Xiangchun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| |
Collapse
|
48
|
Xu W, Zhao Y, Zhang C, Huo M, Wang L, Wu X, Zhang Y, Li Q, Gai Y. Bimetallic nanoplatform for synergistic sonodynamic-calcium overload therapy utilizing self-supplied hydrogen peroxide and relieved hypoxia. Biomater Sci 2024; 12:1171-1184. [PMID: 38205509 DOI: 10.1039/d3bm01430d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Sonodynamic therapy (SDT) has emerged as a potential alternative to traditional cancer treatments as it offers deep cellular penetration and reduced invasivity. Sonosensitizers generate reactive oxygen species (ROS) under ultrasound activation, focusing the ultrasound energy on malignant sites located deep in tissues and causing cell apoptosis and necrosis. However, due to tumor hypoxia and the limited levels of intracellular endogenous hydrogen peroxide (H2O2 is a fundamental species for supplying oxygen via catalase activity), SDT efficacy is still insufficient. In this study, a bimetallic and multifunctional system (Fe3O4-TAPP@PVP-CaO2) was prepared by using ferrosoferric oxide (Fe3O4) as a carrier loaded with 5,10,15,20-tetrakis(4-aminophenyl), porphyrin (TAPP), that was then coated with polyvinyl pyrrolidone (PVP) and calcium peroxide (CaO2). The CaO2 layer elevated the levels of H2O2 and Ca2+ in the tumor microenvironment when exposed to intracellular acidity, providing essential elements for oxygen generation. Intracellular hypoxia was alleviated via the catalase-like activity of Fe3O4 inducing calcium overload. Under ultrasonic irradiation, SDT generated toxic reactive oxygen species (ROS, singlet oxygen) and activated calcium influx through acoustic cavitation. Meanwhile, calcium overload therapy efficiently induced cell apoptosis at the moment of uncontrollable cellular accumulation of Ca2+. In addition, we modified the PVP on the surface to make it more stable. This study presents a bimetallic nanoplatform that can efficiently induce cancer cell death by synergistic sonodynamic-calcium overload therapy via modulation of O2/ROS/Ca2+ species, indicating its potential for multi-modality cancer therapy.
Collapse
Affiliation(s)
- Wenqian Xu
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Yisheng Zhao
- School of Pharmaceutical Sciences, Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, P.R. China
| | - Chao Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, P.R. China
| | - Mengping Huo
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Lei Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Xuewu Wu
- Department of Urology, The Second Hospital of Lanzhou University, Gansu Nephro-Urological Clinical Center, Lanzhou 730000, P.R. China
| | - Yang Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Qiao Li
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| | - Yonghao Gai
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, P.R. China.
| |
Collapse
|
49
|
Zhang Z, Liang X, Yang X, Liu Y, Zhou X, Li C. Advances in Nanodelivery Systems Based on Metabolism Reprogramming Strategies for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6689-6708. [PMID: 38302434 DOI: 10.1021/acsami.3c15686] [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: 02/03/2024]
Abstract
Tumor development and metastasis are closely related to the complexity of the metabolism network. Recently, metabolism reprogramming strategies have attracted much attention in tumor metabolism therapy. Although there is preliminary success of metabolism therapy agents, their therapeutic effects have been restricted by the effective reaching of the tumor sites of drugs. Nanodelivery systems with unique physical properties and elaborate designs can specifically deliver to the tumors. In this review, we first summarize the research progress of nanodelivery systems based on tumor metabolism reprogramming strategies to enhance therapies by depleting glucose, inhibiting glycolysis, depleting lactic acid, inhibiting lipid metabolism, depleting glutamine and glutathione, and disrupting metal metabolisms combined with other therapies, including chemotherapy, radiotherapy, photodynamic therapy, etc. We further discuss in detail the advantages of nanodelivery systems based on tumor metabolism reprogramming strategies for tumor therapy. As well as the opportunities and challenges for integrating nanodelivery systems into tumor metabolism therapy, we analyze the outlook for these emerging areas. This review is expected to improve our understanding of modulating tumor metabolisms for enhanced therapy.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
50
|
Cheng Y, Xia YD, Sun YQ, Wang Y, Yin XB. "Three-in-One" Nanozyme Composite for Augmented Cascade Catalytic Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308033. [PMID: 37851918 DOI: 10.1002/adma.202308033] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/13/2023] [Indexed: 10/20/2023]
Abstract
Cascade catalytic reaction exhibits simple procedure and high efficiency, such as that from the orderly assembly of different enzymes in biological systems. Mimicking of the natural cascade procedure becomes critical, but the orderly assembly of different enzymes is still challenging. Herein, single Au-Pt nanozyme is reported with "three-in-one" functions to initiate cascade conversions for O2 supply as mimic catalase, H2 O2 production with its glucose oxidase-like property, and • OH generation as mimic peroxidase for chemodynamic therapy (CDT). Thus, the complex assembly and cross-talk among the different enzymes are avoided. To this end, metastable Cu2 O NPs, as scaffolds, are used to anchor ultrasmall Au-Pt nanozyme, while metal-organic framework (MOF) is used to encapsulate the nanozyme for tumor microenvironment response and shielding protein adsorption. Pluronic F127 is then modified on the surface to improve hydrophilicity and biocompatibility of the composite. The endogenous acidity and glutathione in tumor degrade MOF to expose nanozyme for cascade catalytic CDT. The high photothermal conversion ability also enhances the CDT, while Cu2+ ions consume GSH to further improve CDT efficiency as augmented cascade catalytic tumor therapy. Thus, a new paradigm is provided with drug-free single nanozyme for improving tumor therapeutic efficacy and minimizing side effects.
Collapse
Affiliation(s)
- Yue Cheng
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yue-Dang Xia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yi-Qing Sun
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin, 300071, China
- Laboratory Equipment Department, Nankai University, Tianjin, 300071, China
| | - Yong Wang
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
| | - Xue-Bo Yin
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin, 300071, China
- Institute of Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| |
Collapse
|