1
|
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
|
2
|
Liu S, Wei Y, Chen T, Li P, Wu J, Tan X, Huang KJ. A dual-mode nanoplatform based on Cu 2O@Au-Pt nanoenzyme and CHA-HCR DNA framework circuit for sensitive detection of CD122 and CD17. Int J Biol Macromol 2025; 309:142740. [PMID: 40185434 DOI: 10.1016/j.ijbiomac.2025.142740] [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: 02/01/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
A Cu2O@Au-Pt nanoenzyme is prepared and integrated with a DNA nanocircuit to construct an electrochemical/colorimetric dual-mode sensing platform for the sensitive detection of CD122 and CD17, which are biomarkers for immune-related diseases.When CD122 is present, it triggers a catalyzed hairpin assembly (CHA) reaction, forming a double-stranded structure recognized by the DNA framework's "arms" (C-DNA), with methylene blue (MB) adsorbing onto it as an electron receptor. Methylene blue (MB) is adsorbed onto the double-stranded structure and introduced as a cathodic electron receptor. In the presence of the target CD17, it acts as a bridging molecule to engage in a hybridization chain reaction (HCR), producing HCR products that are specifically recognized by the other set of "arms" within the C-DNA, which captures abundant MB and generates a heightened response signal. MB can be electrochemically reduced to its reduced state to cause a change in solution color and an increase in RGB Blue values. This method offers a detection range of 0.0001-10,000 pM, with detection limits for CD122 and CD17 of 23.9 and 36.0 aM (S/N = 3) using the electrochemical method, and 26.2 and 26.6 aM (S/N = 3) in the colorimetric mode, respectively. This study presents a novel, reliable multi-target detection strategy for disease biomarkers.
Collapse
Affiliation(s)
- Shiyu Liu
- Education Department of Guangxi Zhuang Autonomous Region, Laboratory of Optic-electric Chemo/Biosensing and Molecular Recognition, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China
| | - Yashu Wei
- The Reproductive Hospital of Guangxi Zhuang Autonomous Region, , Nanning 530021, China
| | - Ting Chen
- Education Department of Guangxi Zhuang Autonomous Region, Laboratory of Optic-electric Chemo/Biosensing and Molecular Recognition, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China
| | - Peiyuan Li
- Education Department of Guangxi Zhuang Autonomous Region, Laboratory of Optic-electric Chemo/Biosensing and Molecular Recognition, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China
| | - Jiawen Wu
- Education Department of Guangxi Zhuang Autonomous Region, Laboratory of Optic-electric Chemo/Biosensing and Molecular Recognition, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China
| | - Xuecai Tan
- Education Department of Guangxi Zhuang Autonomous Region, Laboratory of Optic-electric Chemo/Biosensing and Molecular Recognition, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China.
| | - Ke-Jing Huang
- Education Department of Guangxi Zhuang Autonomous Region, Laboratory of Optic-electric Chemo/Biosensing and Molecular Recognition, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Key Laboratory of Chemistry and Engineering of Forest Products, State Ethnic Affairs Commission, School of Chemistry and Chemical Engineering, Guangxi Minzu University, Nanning 530006, China.
| |
Collapse
|
3
|
Zhang Q, Chen S, Zhang H, Bao Z, Chen Y, Zhang G, Liu Z, Yang J, He R, Liu Y, Tian X. Optimizing cancer therapy through metal organic frameworks-based nanozymes. Int J Biol Macromol 2025; 306:141409. [PMID: 39993671 DOI: 10.1016/j.ijbiomac.2025.141409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Cancer remains a leading global health challenge, with conventional treatments facing limitations due to drug resistance and adverse effects arising from tumor heterogeneity. Nanozymes, nanomaterials mimicking natural enzymes, have emerged as promising therapeutic agents owing to their catalytic efficiency, stability, and biocompatibility. Among nanozymes, MOFs-based nanozymes are particularly attractive due to the inherent tunability of MOFs, which allows for precise control over their structure, porosity, and catalytic activity. This review comprehensively explores the recent advancements in optimizing cancer therapy through MOFs-based nanozymes. We delve into the classification of these nanozymes based on their enzyme-mimicking activities, including peroxidase, oxidase, catalase, and superoxide dismutase, and discuss their underlying catalytic mechanisms. Additionally, emerging single-atom nanozymes are discussed as a distinct category. Furthermore, we highlight the diverse therapeutic strategies employing MOFs-based nanozymes, such as starvation therapy, oxygen supply, catalytic therapy, glutathione depletion, and activation of therapeutic agents within tumor microenvironment. By exploiting the unique properties of MOFs, these nanozymes demonstrate enhanced therapeutic efficacy in various cancer treatment modalities, including chemotherapy, radiotherapy, photodynamic therapy, and sonodynamic therapy. This review underscores the significant potential of MOFs-based nanozymes as a versatile platform for developing next-generation cancer therapeutics, offering improved targeting, reduced systemic toxicity, and enhanced treatment outcomes.
Collapse
Affiliation(s)
- Qinxin Zhang
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China; Key Laboratory of Carbon Materials of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Sai Chen
- Hebei Key Laboratory of Green Development of Rock and Mineral Materials and Institute of Basalt Fiber Materials, School of Gemmology and Materials Science, Hebei GEO University, Shijiazhuang 050031, China
| | - Hongwei Zhang
- Key Laboratory of Development and Application of Rural Renewable Energy, Biogas Institute of Ministry of Agriculture and Rural Affairs, Chengdu 610041, China
| | - Zitong Bao
- Key Laboratory of Development and Application of Rural Renewable Energy, Biogas Institute of Ministry of Agriculture and Rural Affairs, Chengdu 610041, China
| | - Yangyang Chen
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China; Hebei Key Laboratory of Green Development of Rock and Mineral Materials and Institute of Basalt Fiber Materials, School of Gemmology and Materials Science, Hebei GEO University, Shijiazhuang 050031, China
| | - Guangling Zhang
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China
| | - Zhiyong Liu
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China
| | - Jichun Yang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Runhe He
- Key Laboratory of Carbon Materials of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Yatao Liu
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xuetao Tian
- Hebei Key Laboratory of Medical-Industrial Integration Precision Medicine, School of Clinical Medicine, North China University of Science and Technology, Tangshan 063000, China.
| |
Collapse
|
4
|
Chen J, Wang Y, Huang J, Yang Z, Niu H, Su X, Huang J, Ma H, Zhu Y, Wu C, Shi J. Cascade specific endogenous Fe 3+ interference and in situ catalysis for tumor therapy with stemness suppression. Natl Sci Rev 2025; 12:nwae434. [PMID: 39967605 PMCID: PMC11833684 DOI: 10.1093/nsr/nwae434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/03/2024] [Accepted: 11/26/2024] [Indexed: 02/20/2025] Open
Abstract
Cancer stem-like cells (CSCs), featuring high tumorigenicity and invasiveness, are one of the critical factors leading to the failure of clinical cancer treatment such as metastasis and recurrence. However, current strategies suffer from the low stemness-inhibiting efficacy on CSCs by conventional molecular agents and the poor lethal effects against bulk tumor cells. Here we engineer a coordination nanomedicine by 2,5-dihydroxyterephthalic acid (DHT) complexing zinc ions (Zn2+) as a double-effect nanodisrupter of tumor iron (Fe) and redox homeostasis for catalysis-boosted tumor therapy with stemness inhibition. Taking advantage of the much higher binding force of DHT toward Fe3+, this nanomedicine can specifically chelate endogenous Fe3+ into its nanostructure and release Zn2+, and the in situ formed hexacoordinated Fe-DHT conformation is of much enhanced reducibility in order to promote reactive oxygen species (ROS) production in tumors. The nanomedicine-mediated Fe depletion and ROS generation collectively induce CSC differentiation via downregulating the Wnt signaling and inducing forkhead box O3 (FoxO3) activation, respectively. Notably, the combined tumor-selective ROS generation and Zn2+-induced antioxidation dysfunction potently trigger intratumoral oxidative damage leading to both cellular apoptosis and ferroptosis. This nanomedicine, capable of synchronously treating CSCs and bulk tumor cells, has been demonstrated to effectively inhibit the growth, postoperative recurrence and metastasis of orthotopic triple-negative breast tumors in vivo, offering an encouraging candidate of cancer therapeutic agents for treating CSCs-enriched malignancy.
Collapse
Affiliation(s)
- Jiajie Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yitong Wang
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jian Huang
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Zhibo Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huicong Niu
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai 200032, China
| | - Xiaolian Su
- Department of Radiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Jimin Huang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongshi Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200331, China
| |
Collapse
|
5
|
Agrawal S, Singh GK, Tiwari S. Focused starvation of tumor cells using glucose oxidase: A comprehensive review. Int J Biol Macromol 2024; 281:136444. [PMID: 39389487 DOI: 10.1016/j.ijbiomac.2024.136444] [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: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Starvation therapy targets the high metabolic demand of tumor cells. It primarily leans over the consumption of intracellular glucose and simultaneous blockade of alternative metabolic pathways. The strategy involves the use of glucose oxidase (GOx) for catalyzing the conversion of glucose into gluconic acid and hydrogen peroxide. Under these conditions, metabolic re-programming of tumor cells enables the utilization of substrates such as amino acids, fatty acids and lipids. This can be overcome by co-administration of chemo-, photo- and immuno-therapeutics together with glucose oxidase. Targeted delivery of glucose oxidase at tumor site can be enabled with the use of nanoformulations. In this review, we highlight that the outcomes of starvation therapy can be improved using rationally developed nano-formulations. It is possible to load synergistically acting bioactives in these formulations and deliver in site-specific manner and hence achieve the elimination of tumors cells with greater efficacy.
Collapse
Affiliation(s)
- Shivanshu Agrawal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Gireesh K Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya 824236, India
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
6
|
Guo Y, Cao H, Di W, Gao X. Imprinted membrane-covalent organic framework platform for efficient label-free visual detection of Listeria monocytogenes and Salmonella typhimurium in food samples. Anal Chim Acta 2024; 1320:343002. [PMID: 39142781 DOI: 10.1016/j.aca.2024.343002] [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: 03/30/2024] [Revised: 06/25/2024] [Accepted: 07/21/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Rapid and sensitive detection of foodborne pathogens in food plays a crucial role in controlling outbreaks of foodborne diseases, of which Listeria monocytogenes and Salmonella typhimurium are representative and notable pathogens. Thus, it's of great importance to achieve the effective detection of these pathogens. However, the most common detection methods (culture-based technique, Polymerase Chain Reaction and immunological methods) have disadvantages that cannot be ignored, such as time-consuming, laborious, complex sample preparation process, and the possibility of cross-reaction. Hence, it is essential to develop a facile detection method for the pathogens with high sensitivity and specificity to avoid the above-mentioned disadvantages. RESULTS We report a label-free visual platform for the simultaneous capture and detection of Listeria monocytogenes and Salmonella typhimurium. For the first time, we have prepared polydimethylsiloxane-Chromotrope 2R membrane which serves as the substrate for bacterial capture and enrichment through the formation of specific recognition sites. The positively charged Pt-covalent organic framework combines with the pathogens through surface charge interaction, thereby the label-free sandwich platform is formed. Remarkable peroxidase activity of Pt-covalent organic framework converts the conversion of bacterial quantity into amplified color signal by catalyzing 3,3',5,5'-Tetramethylbenzidine to oxidized 3,3',5,5'-Tetramethylbenzidine. The platform demonstrates the capability to identify two representative food-borne pathogens within a time frame of 100 min, exhibiting high sensitivity and excellent specificity without the interference from non-target bacteria. The limit of detection of the visual platform toward Listeria monocytogenes and Salmonella typhimurium was 1.61 CFU mL-1 and 1.31 CFU mL-1, respectively. And the limit of quantification toward Listeria monocytogenes and Salmonella typhimurium was 4.94 CFU mL-1 and 2.47 CFU mL-1, respectively. The relative standard derivations of the visual platform for both bacteria were lower than 4.9 %. Furthermore, our proposed platform has obtained reliable and satisfactory results on analyzing diverse food samples. SIGNIFICANCE This research expands the application of a label-free platform combined with unlabeled nanocomponents in the rapid isolation and detection of diverse of food-borne pathogens. The platform possesses the advantages of simple operation and real-time monitoring, without complicated sample pretreatment process. The whole detection process can realize the simultaneous monitoring of Listeria monocytogenes and Salmonella typhimurium within 100 min. Furthermore, it is also of reference significance for the detection of other common pathogens.
Collapse
Affiliation(s)
- Yuanyuan Guo
- School of Public Health, Shandong University, Jinan, 250000, Shandong Province, PR China
| | - Hongqian Cao
- School of Public Health, Shandong University, Jinan, 250000, Shandong Province, PR China.
| | - Wenli Di
- School of Public Health, Shandong University, Jinan, 250000, Shandong Province, PR China
| | - Xibao Gao
- School of Public Health, Shandong University, Jinan, 250000, Shandong Province, PR China.
| |
Collapse
|
7
|
Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta 2024; 191:541. [PMID: 39150483 DOI: 10.1007/s00604-024-06583-y] [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: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.
Collapse
Affiliation(s)
- Karim Khanmohammadi Chenab
- Department of Chemistry, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
- Department of Physics, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, 9220, Aalborg, Denmark
| | | | | | | |
Collapse
|
8
|
Zhang X, Li M, Tang YL, Zheng M, Liang XH. Advances in H 2O 2-supplying materials for tumor therapy: synthesis, classification, mechanisms, and applications. Biomater Sci 2024; 12:4083-4102. [PMID: 39010783 DOI: 10.1039/d4bm00366g] [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: 07/17/2024]
Abstract
Hydrogen peroxide (H2O2) as a reactive oxygen species produced by cellular metabolism can be used in antitumor therapy. However, the concentration of intracellular H2O2 limits its application. Some materials could enhance the concentration of intracellular H2O2 to strengthen antitumor therapy. In this review, the recent advances in H2O2-supplying materials in terms of promoting intracellular H2O2 production and exogenous H2O2 supply are summarized. Then the mechanism of H2O2-supplying materials for tumor therapy is discussed from three aspects: reconstruction of the tumor hypoxia microenvironment, enhancement of oxidative stress, and the intrinsic anti-tumor ability of H2O2-supplying materials. In addition, the application of H2O2-supplying materials for tumor therapy is discussed. Finally, the future of H2O2-supplying materials is presented. This review aims to provide a novel idea for the application of H2O2-supplying materials in tumor therapy.
Collapse
Affiliation(s)
- Xu Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, People's Republic of China.
| | - Mao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, No.14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, No.14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, People's Republic of China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China.
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu, Sichuan 610041, People's Republic of China.
| |
Collapse
|
9
|
Yang HZ, Chen JJ, Zhang L, Tian XL, Wang R, Pu L, Yu XQ, Zhang J. A dual responsive nitric oxide / β-lapachone co-delivery platform for redox imbalance-enhanced tumor therapy. Eur J Pharm Biopharm 2024; 201:114348. [PMID: 38844097 DOI: 10.1016/j.ejpb.2024.114348] [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: 03/18/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024]
Abstract
Nitric oxide (NO) / β-Lapachone (Lap) combined therapy by causing oxidative stress is an effective tumor therapy strategy. Herein, a dual-responsive lipid nanoparticles (LNPs) LSNO for NO / Lap co-delivery were constructed from the zinc-coordinated lipid (DSNO(Zn)) and the hydrophobic drug Lap in the presence of helper lipids (DOPE and DSPE-PEG2000). The zinc-coordinated structure in LSNO might elevate the Zn2+ content in tumor cells, contributing to antioxidant imbalance. The fluorescent assays proved the light-triggered NO release and fluorescent self-reporting abilities of LSNO. In addition, the LNPs had good drug release behavior under high concentration of GSH, indicating the NO / drug co-delivery capacity. In vitro antitumor assays showed that the NO / Lap combination treatment group could induce more significant tumor cell growth inhibition and cell apoptosis than individual NO or Lap treatment. The following mechanism studies revealed that NO / Lap combination treatment led to distinct oxidative stress by producing reactive oxygen species (ROS) and peroxynitrite anion (ONOO-). On the other hand, the intracellular redox balance could be further disrupted by Lap-induced NADPH consumption and Zn2+ / NO-induced reductase activities downregulation, thus promoting the degree of cell damage. Besides, it was also found that NO and Lap could directly damage nuclear DNA and induce mitochondrial dysfunction, thereby leading to caspase-3 activation and tumor cell death. These results proved that LSNO could serve as a promising multifunctional tumor therapy platform.
Collapse
Affiliation(s)
- Hui-Zhen Yang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China
| | - Jia-Jia Chen
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China
| | - Lan Zhang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China
| | - Xiao-Li Tian
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China
| | - Rong Wang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China
| | - Lin Pu
- Department of Chemistry, University of Virginia, McCormick Rd, Charlottesville, VA 22904, USA
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China; Asymmetric Synthesis and Chiral Technology Key Laboratory of Sichuan Province, Department of Chemistry, Xihua University, Chengdu 610039, PR China
| | - Ji Zhang
- Key Laboratory of Green Chemistry and Technology (Ministry of Education), College of Chemistry, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
10
|
Zhang J, Li M, Liu M, Yu Q, Ge D, Zhang J. Metal-Organic Framework Nanomaterials as a Medicine for Catalytic Tumor Therapy: Recent Advances. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:797. [PMID: 38727391 PMCID: PMC11085591 DOI: 10.3390/nano14090797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/28/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
Nanomaterials, with unique physical, chemical, and biocompatible properties, have attracted significant attention as an emerging active platform in cancer diagnosis and treatment. Amongst them, metal-organic framework (MOF) nanostructures are particularly promising as a nanomedicine due to their exceptional surface functionalities, adsorption properties, and organo-inorganic hybrid characteristics. Furthermore, when bioactive substances are integrated into the structure of MOFs, these materials can be used as anti-tumor agents with superior performance compared to traditional nanomaterials. In this review, we highlight the most recent advances in MOFs-based materials for tumor therapy, including their application in cancer treatment and the underlying mechanisms.
Collapse
Affiliation(s)
- Jiaojiao Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Meiyu Li
- School of Life Science, Jiangsu University, Zhenjiang 212013, China;
| | - Maosong Liu
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Qian Yu
- School of Life Science, Jiangsu University, Zhenjiang 212013, China;
| | - Dengfeng Ge
- Shengli Oilfield Central Hospital, 31 Ji’nan Rd, Dongying 257034, China;
| | - Jianming Zhang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
11
|
Li J, Chen Y, Zha D, Wu C, Li X, Yang L, Cao H, Cai S, Cai Y. Mg-ZIF nanozyme regulates the switch between osteogenic and lipogenic differentiation in BMSCs via lipid metabolism. Lipids Health Dis 2024; 23:88. [PMID: 38528544 DOI: 10.1186/s12944-024-02083-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
The accumulation of reactive oxygen species (ROS) within the bone marrow microenvironment leads to diminished osteogenic differentiation and heightened lipogenic differentiation of mesenchymal stem cells residing in the bone marrow, ultimately playing a role in the development of osteoporosis (OP). Mitigating ROS levels is a promising approach to counteracting OP. In this study, a nanozyme composed of magnesium-based zeolitic imidazolate frameworks (Mg-ZIF) was engineered to effectively scavenge ROS and alleviate OP. The results of this study indicate that Mg-ZIF exhibits significant potential in scavenging ROS and effectively promoting osteogenic differentiation of bone mesenchymal stem cells (BMSCs). Additionally, Mg-ZIF was found to inhibit the differentiation of BMSCs into adipose cells. In vivo experiments further confirmed the ability of Mg-ZIF to mitigate OP by reducing ROS levels. Mechanistically, Mg-ZIF enhances the differentiation of BMSCs into osteoblasts by upregulating lipid metabolic pathways through ROS scavenging. The results indicate that Mg-ZIF has potential as an effective therapeutic approach for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Jinying Li
- Department of Endocrinology, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Yongshao Chen
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Dingsheng Zha
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Chunhui Wu
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Xiaofen Li
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Li Yang
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Hui Cao
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Shexing Cai
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China
| | - Yuebo Cai
- Department of Orthopedics Surgery, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, 528300, P. R. China.
| |
Collapse
|
12
|
Xu L, Peng M, Gao T, Wang D, Lian X, Sun H, Shi J, Wang Y, Wang P. Nanoenabled Intracellular Metal Ion Homeostasis Regulation for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306203. [PMID: 38063781 PMCID: PMC10870045 DOI: 10.1002/advs.202306203] [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: 08/30/2023] [Revised: 10/27/2023] [Indexed: 02/17/2024]
Abstract
Endogenous essential metal ions play an important role in many life processes, especially in tumor development and immune response. The approval of various metallodrugs for tumor therapy brings more attention to the antitumor effect of metal ions. With the deepening understanding of the regulation mechanisms of metal ion homeostasis in vivo, breaking intracellular metal ion homeostasis becomes a new means to inhibit the proliferation of tumor cells and activate antitumor immune response. Diverse nanomedicines with the loading of small molecular ion regulators or metal ions have been developed to disrupt metal ion homeostasis in tumor cells, with higher safety and efficiency than free small molecular ion regulators or metal compounds. This comprehensive review focuses on the latest progress of various intracellular metal ion homeostasis regulation-based nanomedicines in tumor therapy including calcium ion (Ca2+ ), ferrous ion (Fe2+ ), cuprous ion (Cu+ ), managanese ion (Mn2+ ), and zinc ion (Zn2+ ). The physiological functions and homeostasis regulation processes of ions are summarized to guide the design of metal ion regulation-based nanomedicines. Then the antitumor mechanisms of various ions-based nanomedicines and some efficient synergistic therapies are highlighted. Finally, the challenges and future developments of ion regulation-based antitumor therapy are also discussed, hoping to provide a reference for finding more effective metal ions and synergistic therapies.
Collapse
Affiliation(s)
- Lihua Xu
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Mingzheng Peng
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Tingting Gao
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
| | - Dandan Wang
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Xiaowu Lian
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhou450052China
| | - Huihui Sun
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Jinjin Shi
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
| | - Yafeng Wang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001China
| | - Pengju Wang
- Sino‐British Research Centre for Molecular OncologyNational Centre for International Research in Cell and Gene TherapyState Key Laboratory of Esophageal Cancer Prevention & TreatmentSchool of Basic Medical SciencesAcademy of Medical SciencesZhengzhou UniversityZhengzhou450052China
| |
Collapse
|
13
|
Zhang W, Liu H, Yan L, Mei X, Hou Z. Combining emulsion electrospinning with surface functionalization to fabricate multistructural PLA/CS@ZIF-8 nanofiber membranes toward pH-responsive dual drug delivery. Int J Biol Macromol 2023; 253:126506. [PMID: 37659502 DOI: 10.1016/j.ijbiomac.2023.126506] [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: 06/03/2023] [Revised: 07/19/2023] [Accepted: 08/22/2023] [Indexed: 09/04/2023]
Abstract
Developing of the multifunctional polymeric carrier for controlled drug release is still one of most challenging task. In this work, a pH-responsive dual drug delivery system was designed and prepared based on the zeolitic imidazolate framework-8 (ZIF-8). The poly(lactic acid)/chitosan (PLA/CS) core-shell nanofiber membranes by emulsion electrospinning, which the hydrophilic drug (Astragalus Polysacharin, APS) was encapsulated in the CS core and the hydrophobic drug (Camptothecin, CPT) was loaded into the PLA shell, respectively. Subsequently, ZIF-8 nanoparticles served as the protective layer were immobilized on the surface of PLA/CS to form multi-structural PLA/CS@ZIF-8 nanofiber membranes. In vitro drug release of nanofiber membranes were studied in the acidic and neutral medium, respectively. The results were that the hydrophilicity and surface roughness of nanofiber membranes rose with increasing of 2-MIM concentrations. The nanofiber membranes also had excellent pH-responsive and controlled release property. Furthermore, the drug release of PLA/CS@ZIF-8 for either APS or CPT were all carried out in a coexisting manner of diffusion and skeleton corrosion. In addition, in vitro cytotoxicity assay indicated nanofiber membranes with good cytocompatibility. Therefore, the multi-structured PLA/CS@ZIF-8 nanofiber membranes has been used as a potential pH-responsive dual drug release system.
Collapse
Affiliation(s)
- Wen Zhang
- State Key Laboratory of Separation Membranes and Membrane Processes, College of Materials Science and Engineering, Tiangong University, Tianjin 300387, China.
| | - Hongming Liu
- BeiJing Shidabocheng Technology Co., Ltd., Beijing 102200, China
| | - Li Yan
- College of Humanities, Tiangong University, Tianjin 300387, China
| | - Xi Mei
- State Key Laboratory of Separation Membranes and Membrane Processes, College of Materials Science and Engineering, Tiangong University, Tianjin 300387, China
| | - Zikang Hou
- State Key Laboratory of Separation Membranes and Membrane Processes, College of Materials Science and Engineering, Tiangong University, Tianjin 300387, China
| |
Collapse
|
14
|
Zhang Q, Yan S, Yan X, Lv Y. Recent advances in metal-organic frameworks: Synthesis, application and toxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 902:165944. [PMID: 37543345 DOI: 10.1016/j.scitotenv.2023.165944] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/07/2023]
Abstract
Metal-organic frameworks (MOFs) are a new class of crystalline porous hybrid materials with high porosity, large specific surface area and adjustable channel structure and biocompatibility, which are being investigated with increasing interest for energy storage and conversion, gas adsorption/separation, catalysis, sensing and biomedicine. However, the practical applications of MOFs make them release into the environment inevitable, posing a threat to humans and organisms. In this article, we cover advances in the currently available MOFs synthesis methods and the emerging applications of MOFs, especially in the biomedical field (therapeutic agents and bioimaging). Additionally, after evaluating the current status of main exposure routes and affecting factors in the field of MOFs-toxicity, the molecular mechanism is also clarified and identified. Knowledge gaps are identified from such a summarization and frontier development are explored for MOFs. Afterwards, we also present the limitations, challenges, and future perspectives in the study of the entire life cycle of MOFs. This review emphasizes the need for a more targeted discussion of the latest, widely used and effective versatile material class in order to exploit the full potential of high-performance and non-toxicity MOFs in the future.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Shuguang Yan
- Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| | - Xueting Yan
- Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China.
| | - Yi Lv
- Key Laboratory of Green Chemistry & Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China; Analytical & Testing Center, Sichuan University, Chengdu, Sichuan 610064, China
| |
Collapse
|
15
|
Qi G, Shi G, Wang S, Hu H, Zhang Z, Yin Q, Li Z, Hao L. A Novel pH-Responsive Iron Oxide Core-Shell Magnetic Mesoporous Silica Nanoparticle (M-MSN) System Encapsulating Doxorubicin (DOX) and Glucose Oxidase (Gox) for Pancreatic Cancer Treatment. Int J Nanomedicine 2023; 18:7133-7147. [PMID: 38054080 PMCID: PMC10695029 DOI: 10.2147/ijn.s436253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction This study developed a pancreatic cancer targeted drug delivery system that responds to changes in acidity. The system was based on iron oxide core-shell magnetic mesoporous silica nanoparticles (M-MSNs) to treat pancreatic cancer through combined chemotherapy and starvation therapy. Methods Glucose oxidase (Gox) was coupled to the cancer cell surface to reduce glucose availability for cancer cells, exacerbating the heterogeneity of the tumor microenvironment. Reduced pH accelerated the depolymerization of pH-sensitive polydopamine (PDA), thereby controlling the spatial distribution of Gox and release of doxorubicin (DOX) within tumor cells. Results Characterization results showed the successful synthesis of DG@M-MSN-PDA-PEG-FA (DG@NPs) with a diameter of 66.02 ± 3.6 nm. In vitro data indicated DG@NPs were highly effective and stable with good cellular uptake shown by confocal laser scanning microscopy (CLSM). DG@NPs exhibited high cytotoxicity and induced apoptosis. Additionally, in vivo experiments confirmed DG@NPs effectively inhibited tumor growth in nude mice with good biosafety. The combination of starvation therapy and chemotherapy facilitated drug release, suggesting DG@NPs as a novel drug delivery system for pancreatic cancer treatment. Conclusion This study successfully constructed a doxorubicin release system responsive to acidity changes for targeted delivery in pancreatic cancer, providing a new strategy for combination therapy.
Collapse
Affiliation(s)
- Guiqiang Qi
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
| | - Guangyue Shi
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
| | - Shengchao Wang
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
| | - Haifeng Hu
- Medical Imaging Center, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, 161000, People’s Republic of China
| | - Zhichen Zhang
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
| | - Qiangqiang Yin
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
| | - Zhongtao Li
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
| | - Liguo Hao
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People’s Republic of China
- Department of Molecular Imaging, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, 161041, People’s Republic of China
| |
Collapse
|
16
|
Li S, Wang Q, Jia Z, Da M, Zhao J, Yang R, Chen D. Recent advances in glucose oxidase-based nanocarriers for tumor targeting therapy. Heliyon 2023; 9:e20407. [PMID: 37780773 PMCID: PMC10539972 DOI: 10.1016/j.heliyon.2023.e20407] [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: 07/16/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
Glucose oxidase (GOx) can specifically catalyze the conversion of β-d-glucose into gluconic acid and hydrogen peroxide (H2O2) in the presence of oxygen, making it promising for tumor starvation therapy and oxidative therapy. However, GOx's immunogenicity, poor in vivo stability, short half-life, and potential systemic toxicity, limit its application in cancer therapy. Nanocarriers are capable of improving the pharmacological properties of therapeutic drugs (e.g. stability, circulating half-life, and tumor accumulation) and lower toxicity, hence resolving GOx issues and enhancing its efficacy. Although the application of targeted nanocarriers based on GOx has recently flourished, this field has not yet been reviewed and evaluated. Herein, we initially examined the mechanism of GOx-based nanocarriers for enhanced tumor therapy. Also, we present a comprehensive and up-to-date review that highlights GOx-based nanocarriers for tumor targeting therapy. This review expands on GOx-based nano-targeted combination therapies from both passive and active targeting perspectives, meanwhile, active targeting is further classified into ligand-mediated targeting and physical-mediated targeting. Furthermore, this review also emphasizes the present challenges and promising advancements.
Collapse
Affiliation(s)
- Su Li
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, 214002, China
| | - Qinghua Wang
- Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Jiangsu, 214002, China
| | - Zhen Jia
- Department of Obstetrics and Gynecology, Haidong No. 2 People's Hospital, Haidong, 810699, China
| | - Mengting Da
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810001, China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University, Xining, 810001, China
| | - Rui Yang
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, 214002, China
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, 214002, China
- Department of Obstetrics and Gynecology, Haidong No. 2 People's Hospital, Haidong, 810699, China
| |
Collapse
|
17
|
Meng X, Wang L, Zhao N, Zhao D, Shen Y, Yao Y, Jing W, Man S, Dai Y, Zhao Y. Stimuli-responsive cancer nanomedicines inhibit glycolysis and impair redox homeostasis. Acta Biomater 2023:S1742-7061(23)00341-0. [PMID: 37343908 DOI: 10.1016/j.actbio.2023.06.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
The solid tumors are characterized with oxidative stress and metabolic reprogramming, which has been independently used for targeted tumor monotherapy. However, the potential of targeting metabolism-redox circuit in tumor therapy has long been neglected. Herein, we report a hybrid nanocarrier for concurrent targeting of glycolysis and redox balance in the current work. The nanocarriers are made of pH- and ATP-responsive zeolitic imidazolate framework (ZIF-8) as the porous core that was further coated with poloxamer 407 as the steric stabilizer. Two active cargos, glucose oxidase (GOx) and 3-bromopyruvate (3-BrPA) were co-loaded in the core of nanocarrier. GOx is well-known for its ability of producing hydrogen peroxide at the expense of glucose and oxygen. 3-BrPA can reduce oxygen and glucose consumption through glycolysis, which sensitized cancer cells to GOx-induced apoptosis. At the cellular level, the hybrid nanocarrier significantly impaired the redox balance in the liver hepatocellular carcinoma cell line (HepG2), as evidenced by the depletion of glutathione and boost of reactive oxygen species. The potency of hybrid nanocarrier in terms of suppressing HepG2 cell energy metabolism was proven by the exhaustion of ATP. As a consequence, cell viability was greatly reduced. The in vivo efficacy of hybrid nanocarriers was demonstrated in HepG2 tumor-bearing mice. The current work presents an approach of targeting metabolism-redox circuit for tumor treatment, which may enrich the available anti-tumor strategies. STATEMENT OF SIGNIFICANCE: Metabolic alterations and elevated reactive oxygen species (ROS) are two characteristics of cancer. The metabolic patterns of cancer cells are elaborately reprogrammed to enable the rapid propagation of cancer cells. However, the potential of targeting the metabolism-redox circuit in anti-tumor therapy has long been neglected. As a proof-of-concept, we report an engineered stimuli-responsive nanomedicine that can eradicate cancer cells via cooperative glycolysis inhibition and redox impairment. The current work presents an approach of targeting the metabolism-redox circuit for tumor treatment, which may enrich the available anti-tumor strategies.
Collapse
Affiliation(s)
- Xuan Meng
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Lin Wang
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Ning Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Delong Zhao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yongli Shen
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yuan Yao
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Wenjie Jing
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shuli Man
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yujie Dai
- College of Biotechnology, Tianjin Key Laboratory of Industrial Microbiology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
18
|
Li Y, Wang R, Liu X, Li K, Xu Q. Recent advances in MOF-bio-interface: a review. NANOTECHNOLOGY 2023; 34:202002. [PMID: 36796094 DOI: 10.1088/1361-6528/acbc81] [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: 09/20/2022] [Accepted: 02/15/2023] [Indexed: 06/18/2023]
Abstract
Metal-organic frameworks (MOFs), as a class of promising material with adjustable function and controllable structure, have been widely used in the food industry, chemical industry, biological medicine, and sensors. Biomacromolecules and living systems play a critical role in the world. However, the insufficiency in stability, recyclability, and efficiency, significantly impedes their further utilization in slightly harsh conditions. MOF-bio-interface engineering effectively address the above-mentioned shortages of biomacromolecules and living systems, and thereby attracting considerable attentions. Herein, we systematically review the achievements in the area of MOF-bio-interface. In particular, we summarize the interface between MOFs and proteins (enzymes and non-enzymatic proteins), polysaccharides, DNA, cells, microbes, and viruses. Meanwhile, we discuss the limitations of this approach and propose future research directions. We expect that this review could provide new insights and inspire new research efforts towards life science and material science.
Collapse
Affiliation(s)
- Yingfeng Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ru Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Xue Liu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ke Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| |
Collapse
|
19
|
Yu Y, Zhao W, Yuan X, Li R. Progress and prospects of nanozymes for enhanced antitumor therapy. Front Chem 2022; 10:1090795. [PMID: 36531332 PMCID: PMC9755492 DOI: 10.3389/fchem.2022.1090795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 11/21/2022] [Indexed: 09/06/2023] Open
Abstract
Nanozymes are nanomaterials with mimicked enzymatic activity, whose catalytic activity can be designed by changing their physical parameters and chemical composition. With the development of biomedical and material science, artificially created nanozymes have high biocompatibility and can catalyze specific biochemical reactions under biological conditions, thus playing a vital role in regulating physiological activities. Under pathological conditions, natural enzymes are limited in their catalytic capacity by the varying reaction conditions. In contrast, compared to natural enzymes, nanozymes have advantages such as high stability, simplicity of modification, targeting ability, and versatility. As a result, the novel role of nanozymes in medicine, especially in tumor therapy, is gaining increasing attention. In this review, function and application of various nanozymes in the treatment of cancer are summarized. Future exploration paths of nanozymes in cancer therapies based on new insights arising from recent research are outlined.
Collapse
Affiliation(s)
| | | | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Li JJ, Yin L, Wang ZF, Jing YC, Jiang ZL, Ding Y, Wang HS. Enzyme-immobilized metal-organic frameworks: From preparation to application. Chem Asian J 2022; 17:e202200751. [PMID: 36029234 DOI: 10.1002/asia.202200751] [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: 07/18/2022] [Revised: 08/27/2022] [Indexed: 11/09/2022]
Abstract
As a class of widely used biocatalysts, enzymes possess advantages including high catalytic efficiency, strong specificity and mild reaction condition. However, most free enzymes have high requirements on the reaction environment and are easy to deactivate. Immobilization of enzymes on nanomaterial-based substrates is a good way to solve this problem. Metal-organic framework (MOFs), with ultra-high specific surface area and adjustable porosity, can provide a large space to carry enzymes. And the tightly surrounded protective layer of MOFs can stabilize the enzyme structure to a great extent. In addition, the unique porous network structure enables selective mass transfer of substrates and facilitates catalytic processes. Therefore, these enzyme-immobilized MOFs have been widely used in various research fields, such as molecule/biomolecule sensing and imaging, disease treatment, energy and environment protection. In this review, the preparation strategies and applications of enzymes-immobilized MOFs are illustrated and the prospects and current challenges are discussed.
Collapse
Affiliation(s)
- Jia-Jing Li
- China Pharmaceutical University, Pharmaceutical analysis, CHINA
| | - Li Yin
- China Pharmaceutical University, Pharmaceutical analysis, CHINA
| | - Zi-Fan Wang
- China Pharmaceutical University, Pharmaceutical analysis, CHINA
| | - Yi-Chen Jing
- China Pharmaceutical University, Pharmaceutical analysis, CHINA
| | - Zhuo-Lin Jiang
- China Pharmaceutical University, Pharmaceutical analysis, CHINA
| | - Ya Ding
- China Pharmaceutical University, Pharmaceutical analysis, CHINA
| | - Huai-Song Wang
- China Parmaceutical University, Pharmaceutical analysis, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing Jiangsu, CHINA
| |
Collapse
|
21
|
Luo Y, Li Y, Huang Z, Li X, Wang Y, Hou J, Zhou S. A Nanounit Strategy Disrupts Energy Metabolism and Alleviates Immunosuppression for Cancer Therapy. NANO LETTERS 2022; 22:6418-6427. [PMID: 35856800 DOI: 10.1021/acs.nanolett.2c02475] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Aberrant energy metabolism not only endows tumor cells with unlimited proliferative capacity but also contributes to the establishment of the glucose-deficient/lactate-rich immunosuppressive tumor microenvironment (ITM) impairing antitumor immunity. Herein, a novel metabolic nanoregulator (D/B/CQ@ZIF-8@CS) was developed by enveloping 2-deoxy-d-glucose (2-DG), BAY-876, and chloroquine (CQ) into zeolitic imidazolate framework-8 (ZIF-8) to simultaneously deprive the energy/nutrition supply of tumor cells and relieve the ITM for synergetic tumor starvation-immunotherapy. Aerobic glycolysis, glucose uptake, and autophagy flux could be concurrently blocked by D/B/CQ@ZIF-8@CS, cutting off the nutrition/energy supply and the source of lactate. Furthermore, inhibition of glucose uptake and aerobic glycolysis could effectively reverse the glucose-deficient/lactate-rich ITM, thus functionally inactivating regulatory T cells and augmenting anti-CTLA-4 immunotherapy. Such a two-pronged strategy would provide new insights for the design of metabolic intervention-based synergistic cancer therapy.
Collapse
Affiliation(s)
- Yang Luo
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Yingmin Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Zhengjie Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Xinyang Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Jianwen Hou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| |
Collapse
|
22
|
Li K, Xu X, Liu W, Yang S, Huang L, Tang S, Zhang Z, Wang Y, Chen F, Qian K. A Copper-Based Biosensor for Dual-Mode Glucose Detection. Front Chem 2022; 10:861353. [PMID: 35444996 PMCID: PMC9014126 DOI: 10.3389/fchem.2022.861353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/15/2022] [Indexed: 12/02/2022] Open
Abstract
Glucose is a source of energy for daily activities of the human body and is regarded as a clinical biomarker, due to the abnormal glucose level in the blood leading to many endocrine metabolic diseases. Thus, it is indispensable to develop simple, accurate, and sensitive methods for glucose detection. However, the current methods mainly depend on natural enzymes, which are unstable, hard to prepare, and expensive, limiting the extensive applications in clinics. Herein, we propose a dual-mode Cu2O nanoparticles (NPs) based biosensor for glucose analysis based on colorimetric assay and laser desorption/ionization mass spectrometry (LDI MS). Cu2O NPs exhibited excellent peroxidase-like activity and served as a matrix for LDI MS analysis, achieving visual and accurate quantitative analysis of glucose in serum. Our proposed method possesses promising application values in clinical disease diagnostics and monitoring.
Collapse
Affiliation(s)
- Kai Li
- Department of Urology, Tianjin Third Central Hospital Affiliated to Nankai University, Tianjin, China
| | - Xiaoyu Xu
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wanshan Liu
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shouzhi Yang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Huang
- Department of Clinical Laboratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuai Tang
- Department of Urology, Tianjin Third Central Hospital Affiliated to Nankai University, Tianjin, China
| | - Ziyue Zhang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuning Wang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Yuning Wang, ; Fangmin Chen, ; Kun Qian,
| | - Fangmin Chen
- Department of Urology, Tianjin Third Central Hospital Affiliated to Nankai University, Tianjin, China
- *Correspondence: Yuning Wang, ; Fangmin Chen, ; Kun Qian,
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Yuning Wang, ; Fangmin Chen, ; Kun Qian,
| |
Collapse
|