1
|
Feng Y, Shi Y, Zhao Q, Gao G, Wang Z, Zhi J. Onion-like carbon based single-atom iron nanozyme for photothermal and catalytic synergistic antibacterial application. J Colloid Interface Sci 2025; 681:205-214. [PMID: 39602971 DOI: 10.1016/j.jcis.2024.11.139] [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: 08/26/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Nanozymes with oxidase (OXD)-like activity have emerged as promising antibacterial agents due to their capability of catalyzing atmospheric O2 to generate highly active free radicals. However, the precise engineering of functional nanozyme at the atomic level for antibacterial therapy presents a challenge. Here, atomically dispersed Fe atoms were loaded onto onion-like carbon (OLC) through a ligand-assisted calcination strategy, yielding a single-atom nanozyme (FeSA-OLC) with enhanced oxidase-like activity. The FeSA-OLC could catalyze the decomposition of O2 to produce active hydroxyl radicals (·OH) owing to the fully exposed Fe atoms and a highly curved carbon shell. Density functional theory calculation revealed that the single-atom Fe sites facilitated the generation of free radical species by promoting the adsorption and cleavage of OO bond. Meanwhile, the FeSA-OLC exhibited a notable photothermal conversion efficiency of 66.48% under near-infrared laser irradiation. Furthermore, in vitro experimental results demonstrated a synergistic antibacterial effect towards Escherichia coli due to the photothermal-enhanced oxidase-like activity. Overall, this work introduced a strategy to develop OLC-based single-atom nanozyme, thereby offering new avenues for photothermal-augmented antibacterial therapy.
Collapse
Affiliation(s)
- Yuchen Feng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yuxi Shi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qi Zhao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| | - Guanyue Gao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhiqiang Wang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, PR China; Henan Huifeng Diamond Co., Ltd, Zhengzhou 450016, PR China
| | - Jinfang Zhi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
2
|
Zhao H, Jin S, Liu Y, Wang Q, Tan BSN, Wang S, Han WK, Niu X, Zhao Y. A Second Near-Infrared Window-Responsive Metal-Organic-Framework-Based Photosensitizer for Tumor Immunotherapy via Synergistic Ferroptosis and STING Activation. J Am Chem Soc 2025. [PMID: 39854684 DOI: 10.1021/jacs.4c13241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Photodynamic therapy (PDT) holds promise as a cancer treatment modality due to its potential for enhanced therapy precision and safety. To enhance deep tissue penetration and minimize tissue adsorption and phototoxicity, developing photosensitizers activated by second near-infrared window (NIR-II) light shows significant potential. However, the efficacy of PDT is often impeded by tumor microenvironment hypoxia, primarily caused by irregular tumor vasculature. Fortunately, the stimulator of interferon genes (STING) pathway, known for immune activation, has been linked to vasculature normalization. In this study, we developed a nanoplatform (Fe-THBQ/SR) by loading a STING agonist (SR-717) into an iron-tetrahydroxy-1,4-benzoquinone (Fe-THBQ) metal-organic framework. Fe-THBQ was proven to be an effective NIR-II photosensitizer, generating numerous reactive oxygen species (ROS) under 1064 nm laser irradiation. These ROS downregulated heat shock protein expression, consequently promoting mild-photothermal therapy (mild-PTT), and facilitated ferroptosis by depleting glutathione (GSH)/glutathione peroxidase 4. Moreover, Fe-THBQ/SR released SR-717 upon GSH stimulation, synergizing with the ROS-mediated double-stranded DNA leakage to enhance STING activation. This process contributed to tumor vasculature normalization and hypoxia alleviation, thereby enhancing the PDT efficacy. Overall, we presented a versatile single-laser-triggered nanoplatform (Fe-THBQ/SR) for NIR-II PDT and NIR-II mild-PTT and simultaneously coupled it with the effective activation of STING to form a reinforcing cycle. These synergistic enhancements increased the immunogenicity of tumor cells, remodeled the immunosuppressive tumor microenvironment, increased T lymphocyte infiltration, and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Huan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Shujuan Jin
- Senior Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, Fuxin Road, No. 28, Haidian District, Beijing, 100853, China
| | - Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Qian Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, China
| | - Brynne Shu Ni Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Shihuai Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
- Yunnan Key Laboratory for Micro/Nano Materials & Technology, School of Materials and Energy, Yunnan University, Kunming, Yunnan 650091, China
| | - Wang-Kang Han
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Xuping Niu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, State Key Breeding Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan City Central Hospital, Taiyuan, Shanxi 030009, China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| |
Collapse
|
3
|
Zhao L, Tong Y, Yin J, Li H, Du L, Li J, Jiang Y. Photo-Activated Oxidative Stress Amplifier: A Strategy for Targeting Glutathione Metabolism and Enhancing ROS-Mediated Therapy in Triple-Negative Breast Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2403861. [PMID: 39096062 DOI: 10.1002/smll.202403861] [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: 05/13/2024] [Revised: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Amplifying oxidative stress within tumor cells can effectively inhibit the growth and metastasis of triple-negative breast cancer (TNBC). Therefore, the development of innovative nanomedicines that can effectively disrupt the redox balance represents a promising yet challenging therapeutic strategy for TNBC. In this study, an oxidative stress amplifier, denoted as PBCH, comprising PdAg mesoporous nanozyme and a CaP mineralized layer, loaded with GSH inhibitor L-buthionine sulfoximine (BSO), and further surface-modified with hyaluronic acid that can target CD44, is introduced. In the acidic tumor microenvironment, Ca2+ is initially released, thereby leading to mitochondrial dysfunction and eventually triggering apoptosis. Additionally, BSO suppresses the synthesis of intracellular reduced GSH and further amplifies the level of oxidative stress in cancer cells. Furthermore, PdAg nanozyme can be activated by near-infrared light to induce photothermal and photodynamic effects, causing a burst of ROS and simultaneously promoting cell apoptosis via provoking immunogenic cell death. The high-performance therapeutic effects of PBCH, based on the synergistic effect of aforementioned multiple oxidative damage and photothermal ablation, are validated in TNBC cells and animal models, declaring its potential as a safe and effective anti-tumor agent. The proposed approach offers new perspectives for precise and efficient treatment of TNBC.
Collapse
Affiliation(s)
- Li Zhao
- Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Yao Tong
- The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Jiawei Yin
- The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Hui Li
- Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Lutao Du
- The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, Shandong, 250033, China
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, 250033, China
| | - Juan Li
- The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Yanyan Jiang
- Liquid-Solid Structural Evolution & Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| |
Collapse
|
4
|
Jia X, Wang Y, Qiao Y, Jiang X, Li J. Nanomaterial-based regulation of redox metabolism for enhancing cancer therapy. Chem Soc Rev 2024; 53:11590-11656. [PMID: 39431683 DOI: 10.1039/d4cs00404c] [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: 10/22/2024]
Abstract
Altered redox metabolism is one of the hallmarks of tumor cells, which not only contributes to tumor proliferation, metastasis, and immune evasion, but also has great relevance to therapeutic resistance. Therefore, regulation of redox metabolism of tumor cells has been proposed as an attractive therapeutic strategy to inhibit tumor growth and reverse therapeutic resistance. In this respect, nanomedicines have exhibited significant therapeutic advantages as intensively reported in recent studies. In this review, we would like to summarize the latest advances in nanomaterial-assisted strategies for redox metabolic regulation therapy, with a focus on the regulation of redox metabolism-related metabolite levels, enzyme activity, and signaling pathways. In the end, future expectations and challenges of such emerging strategies have been discussed, hoping to enlighten and promote their further development for meeting the various demands of advanced cancer therapies. It is highly expected that these therapeutic strategies based on redox metabolism regulation will play a more important role in the field of nanomedicine.
Collapse
Affiliation(s)
- Xiaodan Jia
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Wang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yue Qiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiue Jiang
- Research Center for Analytical Science, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jinghong Li
- Beijing Institute of Life Science and Technology, Beijing 102206, P. R. China
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, P. R. China.
| |
Collapse
|
5
|
Zhen W, Germanas T, Weichselbaum RR, Lin W. Multifunctional Nanomaterials Mediate Cholesterol Depletion for Cancer Treatment. Angew Chem Int Ed Engl 2024; 63:e202412844. [PMID: 39146242 PMCID: PMC11534517 DOI: 10.1002/anie.202412844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
Cholesterol is an essential membrane component, and the metabolites from cholesterol play important biological functions to intricately support cancer progression and dampen immune responses. Preclinical and clinical studies have demonstrated the role of cholesterol metabolism regulation on inhibiting tumor growth, remodeling the immunosuppressive tumor microenvironment (TME), and enhancing anti-tumor immunity. In this minireview, we discuss complex cholesterol metabolism in tumors, its important role in cancer progression, and its influences on immune cells in the TME. We provide an overview of recent advances in cancer treatment through regulating cholesterol metabolism. We discuss the design of cholesterol-altering multifunctional nanomaterials to regulate oxidative stress, modulate immune checkpoints, manipulate mechanical stress responses, and alter cholesterol metabolic pathways. Additionally, we examine the interactions between cholesterol metabolism regulation and established cancer treatments with the aim of identifying efficient strategies to disrupt cholesterol metabolism and synergistic combination therapies for effective cancer treatment.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, The University of Chicago, Chicago, Illinois, 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois, 60637, United States
| | - Tomas Germanas
- Department of Chemistry, The University of Chicago, Chicago, Illinois, 60637, United States
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois, 60637, United States
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, Illinois, 60637, United States
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois, 60637, United States
| |
Collapse
|
6
|
Romani A, Lodi G, Casciano F, Gonelli A, Secchiero P, Zauli G, Bortolini O, Valacchi G, Ragno D, Bondi A, Benedusi M, Esposito E, Voltan R. Enhanced Anti-Melanoma Activity of Nutlin-3a Delivered via Ethosomes: Targeting p53-Mediated Apoptosis in HT144 Cells. Cells 2024; 13:1678. [PMID: 39451196 PMCID: PMC11506859 DOI: 10.3390/cells13201678] [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: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
This study evaluated ethosomes as a novel nanodelivery system for nutlin-3a, a known MDM2 inhibitor and activator of the p53 pathway, to improve nutlin-3a's poor solubility, limiting its bio-distribution and therapeutic efficacy. The potential of nutlin-3a-loaded ethosomes was investigated on two in vitro models of melanoma: the HT144 cell line p53wild-type and the SK-MEL-28 cell line p53mutated. Nutlin-3a-loaded ethosomes were characterized for their physicochemical properties and used to treat melanoma cells at different concentrations, considering nutlin-3a solution and empty ethosomes as controls. The biological effects on cells were evaluated 24 and 48 h after treatment by analyzing the cell morphology and viability, cell cycle, and apoptosis rate using flow cytometry and the p53 pathway's activation via Western blotting. The results indicate that ethosomes are delivery systems able to maintain nutlin-3a's functionality and specific biological action, as evidenced by the molecular activation of the p53 pathway and the biological events leading to cell cycle block and apoptosis in p53wild-type cells. Nutlin-3a-loaded ethosomes induced morphological changes in the HT144 cell line, with evident apoptotic cells and a reduction in the number of viable cells of over 80%. Furthermore, nutlin-3a-loaded ethosomes successfully modulated two p53-regulated proteins involved in survival/apoptosis, with up to a 2.5-fold increase in membrane TRAIL-R2 and up to an 8.2-fold decrease in Notch-1 (Notch intracellular domain, NICD) protein expression. The expression of these molecules is known to be altered or dysfunctional in a large percentage of melanoma tumors. Notably, ethosomes, regardless of their nutlin-3a loading, exhibited the ability to reduce HT144 melanoma cellular migration, as assessed in real time using xCELLigence, likely due to the modification of lipid rafts, suggesting their potential antimetastatic properties. Overall, nutlin-3a delivery using ethosomes appears to be a significantly effective means for upregulating the p53 pathway and downregulating active Notch-1, while also taking advantage of their unexpected ability to reduce cellular migration. The findings of this study could pave the way for the development of specific nutlin-3a-loaded ethosome-based medicinal products for cutaneous use.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (P.S.)
| | - Giada Lodi
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (G.L.); (F.C.)
| | - Fabio Casciano
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (G.L.); (F.C.)
| | - Arianna Gonelli
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.G.); (O.B.); (G.V.)
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (P.S.)
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh 12329-8139, Saudi Arabia;
| | - Olga Bortolini
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.G.); (O.B.); (G.V.)
| | - Giuseppe Valacchi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.G.); (O.B.); (G.V.)
- Plants for Human Health Institute, Animal Sciences Department, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Daniele Ragno
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (D.R.); (A.B.)
| | - Agnese Bondi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (D.R.); (A.B.)
| | - Mascia Benedusi
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Elisabetta Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (D.R.); (A.B.)
| | - Rebecca Voltan
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (G.L.); (F.C.)
| |
Collapse
|
7
|
Liu Y, Zhao H, Wang S, Niu R, Bi S, Han WK, Wang Y, Song S, Zhang H, Zhao Y. A Wurster-Type Covalent Organic Framework with Internal Electron Transfer-Enhanced Catalytic Capacity for Tumor Therapy. J Am Chem Soc 2024; 146:27345-27361. [PMID: 39316459 DOI: 10.1021/jacs.4c05555] [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: 09/26/2024]
Abstract
The low immunogenicity of tumors, along with the abnormal structural and biochemical barriers of tumor-associated vasculature, impedes the infiltration and function of effector T cells at the tumor site, severely inhibiting the efficacy of antitumor immunotherapy. In this study, a cobaloxime catalyst and STING agonist (MSA-2)-coloaded Wurster-type covalent organic framework (Co-TB COF-M) with internal electron transfer-enhanced catalytic capacity was developed as a COF-based immune activator. The covalently anchored cobaloxime adjusts the energy band structure of TB COF and provides it with good substrate adsorption sites, enabling it to act as an electron transmission bridge between the COF and substrate in proton reduction catalytic reactions. This property significantly enhances the sonodynamic catalytic performance. Under sono-irradiation, Co-TB COF-M can produce a substantial amount of reactive oxygen species (ROS) to induce Gasdermin D-mediated pro-inflammatory pyroptosis, thereby effectively enhancing the immunogenicity of tumors. Furthermore, MSA-2 is specifically released in response to ROS at the tumor site, minimizing the off-target side effects. More importantly, Co-TB COF-induced STING activation normalizes tumor vasculature and increases the expression of endothelial T cell adhesion molecules, which greatly enhance the infiltration and function of effector T cells. Thus, Co-TB COF-M as an immune activator could remold the tumor microenvironment, leading to increased infiltration and an improved function of T cells for immunotherapy.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Huan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Shihuai Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
- Yunnan Key Laboratory for Micro/Nano Materials & Technology, School of Materials and Energy, Yunnan University, Kunming, Yunnan 650091, P. R. China
| | - Rui Niu
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Shuai Bi
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Wang-Kang Han
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Hongjie Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
8
|
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
|
9
|
Zhen W, Fan Y, Germanas T, Tillman L, Li J, Blenko AL, Weichselbaum RR, Lin W. Digitonin-Loaded Nanoscale Metal-Organic Framework for Mitochondria-Targeted Radiotherapy-Radiodynamic Therapy and Disulfidptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2405494. [PMID: 39252688 DOI: 10.1002/adma.202405494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/18/2024] [Indexed: 09/11/2024]
Abstract
The efficacy of radiotherapy (RT) is limited by inefficient X-ray absorption and reactive oxygen species generation, upregulation of immunosuppressive factors, and a reducing tumor microenvironment (TME). Here, the design of a mitochondria-targeted and digitonin (Dig)-loaded nanoscale metal-organic framework, Th-Ir-DBB/Dig, is reported to overcome these limitations and elicit strong antitumor effects upon low-dose X-ray irradiation. Built from Th6O4(OH)4 secondary building units (SBUs) and photosensitizing Ir(DBB)(ppy)2 2+ (Ir-DBB, DBB = 4,4'-di(4-benzoato)-2,2'-bipyridine; ppy = 2-phenylpyridine) ligands, Th-Ir-DBB exhibits strong RT-radiodynamic therapy (RDT) effects via potent radiosensitization with high-Z SBUs for hydroxyl radical generation and efficient excitation of Ir-DBB ligands for singlet oxygen production. Th-Ir-DBB/Dig releases digitonin in acidic TMEs to trigger disulfidptosis of cancer cells and sensitize cancer cells to RT-RDT through glucose and glutathione depletion. The released digitonin simultaneously downregulates multiple immune checkpoints in cancer cells and T cells through cholesterol depletion. As a result, Th-Ir-DBB/dig plus X-ray irradiation induces strong antitumor immunity to effectively inhibit tumor growth in mouse models of colon and breast cancer.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Yingjie Fan
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Tomas Germanas
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Langston Tillman
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Jinhong Li
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Abigail L Blenko
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, IL, 60637, USA
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
10
|
Bu JW, Wang ZG, Liu HY, Liu SL. Metal nanozymes modulation of reactive oxygen species as promising strategies for cancer therapy. Int J Pharm 2024; 662:124453. [PMID: 39013531 DOI: 10.1016/j.ijpharm.2024.124453] [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: 03/11/2024] [Revised: 06/11/2024] [Accepted: 07/07/2024] [Indexed: 07/18/2024]
Abstract
Nanozymes, nanostructured materials emulating natural enzyme activities, exhibit potential in catalyzing reactive oxygen species (ROS) production for cancer treatment. By facilitating oxidative reactions, elevating ROS levels, and influencing the tumor microenvironment (TME), nanozymes foster the eradication of cancer cells. Noteworthy are their superior stability, ease of preservation, and cost-effectiveness compared to natural enzymes, rendering them invaluable for medical applications. This comprehensive review intricately explores the interplay between ROS and tumor therapy, with a focused examination of metal-based nanozyme strategies mitigating tumor hypoxia. It provides nuanced insights into diverse catalytic processes, mechanisms, and surface modifications of various metal nanozymes, shedding light on their role in intra-tumoral ROS generation and applications in antioxidant therapy. The review concludes by delineating specific potential prospects and challenges associated with the burgeoning use of metal nanozymes in future tumor therapies.
Collapse
Affiliation(s)
- Jin-Wei Bu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| | - Zhi-Gang Wang
- College of Chemistry and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Hao-Yang Liu
- College of Chemistry and School of Medicine, Nankai University, Tianjin 300071, P. R. China.
| | - Shu-Lin Liu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China; College of Chemistry and School of Medicine, Nankai University, Tianjin 300071, P. R. China.
| |
Collapse
|
11
|
Zhang W, Tian X, Li X. Fabrication of Nanocatalytic Medicine from Self-Assembling Peptides Containing an ATCUN-Like Copper-Binding Motif for Anticancer Therapy. Chembiochem 2024; 25:e202400216. [PMID: 38801354 DOI: 10.1002/cbic.202400216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 05/29/2024]
Abstract
Development of nanomaterials with multiple enzymatic activities via a facile approach receives growing interests in recent years. Although peptide self-assembling provides an effective approach for the construction of biomimetic materials in recent years, fabrication of artificial enzymes from self-assembling peptides with multiple catalytic activities for anticancer therapy is still a challenge. Here, we report a simple method to prepare nanocatalysts with multienzyme-like activities from self-assembling peptides containing ATCUN copper-binding motifs. With the aid of the coordination interactions between the ATCUN motif and Cu(II) ions, these peptides could perform supramolecular self-assembly to form nanomaterials with biomimetic peroxidase, ascorbate oxidase and glutathione peroxidase activities. Moreover, these trienzyme-like effects can elevate oxidative stress levels and suppress the antioxidative capability of cancer cells, which synergistically induce the apoptosis of cancer cells. Because of the high biocompatibility, catalytic activities and drug encapsulation properties, this self-assembled peptide provides a biomimetic platform for the development of new nanocatalytic medicines for multimodal synergistic cancer therapies.
Collapse
Affiliation(s)
- Wenhui Zhang
- College of chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xin Tian
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China
| | - Xinming Li
- College of chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| |
Collapse
|
12
|
Wang J, Liu Y, Cui T, Yang H, Lin L. Current progress in the regulation of endogenous molecules for enhanced chemodynamic therapy. Chem Sci 2024; 15:9915-9926. [PMID: 38966366 PMCID: PMC11220580 DOI: 10.1039/d4sc02129k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Chemodynamic therapy (CDT) is a potential cancer treatment strategy, which relies on Fenton chemistry to transform hydrogen peroxide (H2O2) into highly cytotoxic reactive oxygen species (ROS) for tumor growth suppression. Although overproduced H2O2 in cancerous tissues makes CDT a feasible and specific tumor therapeutic modality, the treatment outcomes of traditional chemodynamic agents still fall short of expectations. Reprogramming cellular metabolism is one of the hallmarks of tumors, which not only supports unrestricted proliferative demands in cancer cells, but also mediates the resistance of tumor cells against many antitumor modalities. Recent discoveries have revealed that various cellular metabolites including H2O2, iron, lactate, glutathione, and lipids have distinct effects on CDT efficiency. In this perspective, we intend to provide a comprehensive summary of how different endogenous molecules impact Fenton chemistry for a deep understanding of mechanisms underlying endogenous regulation-enhanced CDT. Moreover, we point out the current challenges and offer our outlook on the future research directions in this field. We anticipate that exploring CDT through manipulating metabolism will yield significant advancements in tumor treatment.
Collapse
Affiliation(s)
- Jun Wang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Yina Liu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Tingting Cui
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore Singapore 119074 Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore Singapore 117597 Singapore
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Lisen Lin
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| |
Collapse
|
13
|
Ye B, Hu W, Yu G, Yang H, Gao B, Ji J, Mao Z, Huang F, Wang W, Ding Y. A Cascade-Amplified Pyroptosis Inducer: Optimizing Oxidative Stress Microenvironment by Self-Supplying Reactive Nitrogen Species Enables Potent Cancer Immunotherapy. ACS NANO 2024; 18:16967-16981. [PMID: 38888082 DOI: 10.1021/acsnano.4c03172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Selective generation of sufficient pyroptosis inducers at the tumor site without external stimulation holds immense significance for a longer duration of immunotherapy. Here, we report a cascade-amplified pyroptosis inducer CSCCPT/SNAP that utilizes reactive nitrogen species (RNS), self-supplied from the diffusion-controlled reaction between reactive oxygen species (ROS) and nitric oxide (NO) to potentiate pyroptosis and immunotherapy, while both endogenous mitochondrial ROS stimulated by released camptothecin and released NO initiate pyroptosis. Mechanistically, cascade amplification of the antitumor immune response is prompted by the cooperation of ROS and NO and enhanced by RNS with a long lifetime, which could be used as a pyroptosis trigger to effectively compensate for the inherent drawbacks of ROS, resulting in long-lasting pyroptosis for favoring immunotherapy. Tumor growth is efficiently inhibited in mouse melanoma tumors through the facilitation of reactive oxygen/nitrogen species (RONS)-NO synergy. In summary, our therapeutic approach utilizes supramolecular engineering and nanotechnology to integrate ROS producers and NO donors of tumor-specific stimulus responses into a system that guarantees synchronous generation of these two reactive species to elicit pyroptosis-evoked immune response, while using self-supplied RNS as a pyroptosis amplifier. RONS-NO synergy achieves enhanced and sustained pyroptosis and antitumor immune responses for robust cancer immunotherapy.
Collapse
Affiliation(s)
- Binglin Ye
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang 310009, China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Wenting Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Bingqiang Gao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang 310009, China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang 310027, China
- Zhejiang-Israel Joint Laboratory of Self-Assembling Functional Materials, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, Zhejiang 311215, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang 310009, China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang 310009, China
- The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang 310009, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
14
|
Wu L, Lin H, Cao X, Tong Q, Yang F, Miao Y, Ye D, Fan Q. Bioorthogonal Cu Single-Atom Nanozyme for Synergistic Nanocatalytic Therapy, Photothermal Therapy, Cuproptosis and Immunotherapy. Angew Chem Int Ed Engl 2024; 63:e202405937. [PMID: 38654446 DOI: 10.1002/anie.202405937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Single-atom nanozymes (SAzymes) with atomically dispersed active sites are potential substitutes for natural enzymes. A systematic study of its multiple functions can in-depth understand SAzymes's nature, which remains elusive. Here, we develop a novel ultrafast synthesis of sputtered SAzymes by in situ bombarding-embedding technique. Using this method, sputtered copper (Cu) SAzymes (CuSA) is developed with unreported unique planar Cu-C3 coordinated configuration. To enhance the tumor-specific targeting, we employ a bioorthogonal approach to engineer CuSA, denoted as CuSACO. CuSACO not only exhibits minimal off-target toxicity but also possesses exceptional ultrahigh catalase-, oxidase-, peroxidase-like multienzyme activities, resulting in reactive oxygen species (ROS) storm generation for effective tumor destruction. Surprisingly, CuSACO can release Cu ions in the presence of glutathione (GSH) to induce cuproptosis, enhancing the tumor treatment efficacy. Notably, CuSACO's remarkable photothermal properties enables precise photothermal therapy (PTT) on tumors. This, combined with nanozyme catalytic activities, cuproptosis and immunotherapy, efficiently inhibiting the growth of orthotopic breast tumors and gliomas, and lung metastasis. Our research highlights the potential of CuSACO as an innovative strategy to utilize multiple mechanism to enhance tumor therapeutic efficacy, broadening the exploration and development of enzyme-like behavior and physiological mechanism of action of SAzymes.
Collapse
Affiliation(s)
- Luyan Wu
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Huihui Lin
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), Singapore, 627833, Singapore
- Department of Chemistry, National University of Singapore, Singapore, 117549, Singapore
| | - Xiang Cao
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), School of Chemistry and Life Sciences, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Qiang Tong
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Fangqi Yang
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Yinxing Miao
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Deju Ye
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Quli Fan
- State Key Laboratory for Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), School of Materials Science and Engineering, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| |
Collapse
|
15
|
Liu Y, Niu R, Zhang X, Zhang B, Chen X, Guo J, Song S, Wang Y, Zhang H, Zhao Y. Metal-Organic Framework-Based Nanovaccine for Relieving Immunosuppressive Tumors via Hindering Efferocytosis of Macrophages and Promoting Pyroptosis and Cuproptosis of Cancer Cells. ACS NANO 2024; 18:12386-12400. [PMID: 38699808 DOI: 10.1021/acsnano.4c01518] [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: 05/05/2024]
Abstract
Current cancer vaccines face challenges due to an immunosuppressive tumor microenvironment and their limited ability to produce an effective immune response. To address the above limitations, we develop a 3-(2-spiroadamantyl)-4-methoxy-4-(3-phosphoryloxy)-phenyl-1,2-dioxetane (alkaline phosphatase substrate) and XMD8-92 (extracellular signal-regulated kinase 5 inhibitor)-codelivered copper-tetrahydroxybenzoquinone (Cu-THBQ/AX) nanosized metal-organic framework to in situ-generate therapeutic vaccination. Once inside the early endosome, the alkaline phosphatase overexpressed in the tumor cells' membrane activates the in situ type I photodynamic effect of Cu-THBQ/AX for generating •O2-, and the Cu-THBQ/AX catalyzes O2 and H2O2 to •O2- and •OH via semiquinone radical catalysis and Fenton-like reactions. This surge of ROS in early endosomes triggers caspase-3-mediated proinflammatory pyroptosis via activating phospholipase C. Meanwhile, Cu-THBQ/AX can also induce the oligomerization of dihydrolipoamide S-acetyltransferase to trigger tumor cell cuproptosis. The production of •OH could also trigger the release of XMD8-92 for effectively inhibiting the efferocytosis of macrophages to convert immunosuppressive apoptosis of cancer cells into proinflammatory secondary necrosis. The simultaneous induction of pyroptosis, cuproptosis, and secondary necrosis effectively converts the tumor microenvironment from "cold" to "hot" conditions, making it an effective antigen pool. This transformation successfully activates the antitumor immune response, inhibiting tumor growth and metastasis.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Rui Niu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiaodong Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Bin Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Xiaokai Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Jingjing Guo
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
16
|
Du C, Guo X, Qiu X, Jiang W, Wang X, An H, Wang J, Luo Y, Du Q, Wang R, Cheng C, Guo Y, Teng H, Ran H, Wang Z, Li P, Zhou Z, Ren J. Self-Reinforced Bimetallic Mito-Jammer for Ca 2+ Overload-Mediated Cascade Mitochondrial Damage for Cancer Cuproptosis Sensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306031. [PMID: 38342617 PMCID: PMC11022715 DOI: 10.1002/advs.202306031] [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: 08/24/2023] [Revised: 01/21/2024] [Indexed: 02/13/2024]
Abstract
Overproduction of reactive oxygen species (ROS), metal ion accumulation, and tricarboxylic acid cycle collapse are crucial factors in mitochondria-mediated cell death. However, the highly adaptive nature and damage-repair capabilities of malignant tumors strongly limit the efficacy of treatments based on a single treatment mode. To address this challenge, a self-reinforced bimetallic Mito-Jammer is developed by incorporating doxorubicin (DOX) and calcium peroxide (CaO2) into hyaluronic acid (HA) -modified metal-organic frameworks (MOF). After cellular, Mito-Jammer dissociates into CaO2 and Cu2+ in the tumor microenvironment. The exposed CaO2 further yields hydrogen peroxide (H2O2) and Ca2+ in a weakly acidic environment to strengthen the Cu2+-based Fenton-like reaction. Furthermore, the combination of chemodynamic therapy and Ca2+ overload exacerbates ROS storms and mitochondrial damage, resulting in the downregulation of intracellular adenosine triphosphate (ATP) levels and blocking of Cu-ATPase to sensitize cuproptosis. This multilevel interaction strategy also activates robust immunogenic cell death and suppresses tumor metastasis simultaneously. This study presents a multivariate model for revolutionizing mitochondria damage, relying on the continuous retention of bimetallic ions to boost cuproptosis/immunotherapy in cancer.
Collapse
Affiliation(s)
- Chier Du
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xun Guo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaoling Qiu
- Department of Intensive Care Unitthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Weixi Jiang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaoting Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hongjin An
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Jingxue Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Yuanli Luo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Qianying Du
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Ruoyao Wang
- Department of Breast and Thyroid SurgerySecond Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Chen Cheng
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Yuan Guo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hua Teng
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Haitao Ran
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Zhigang Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Pan Li
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Zhiyi Zhou
- Department of General PracticeChongqing General HospitalChongqing400010P. R. China
| | - Jianli Ren
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imagingthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| |
Collapse
|
17
|
Liu Y, Zhao H, Zhao Y. Designing Efficient Single Metal Atom Biocatalysts at the Atomic Structure Level. Angew Chem Int Ed Engl 2024; 63:e202315933. [PMID: 38206594 DOI: 10.1002/anie.202315933] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 01/12/2024]
Abstract
Various nanomaterials as biocatalysts could be custom-designed and modified to precisely match the specific microenvironment of diseases, showing a promise in achieving effective therapy outcomes. Compared to conventional biocatalysts, single metal atom catalysts (SMACs) with maximized atom utilization through well-defined structures offer enhanced catalytic activity and selectivity. Currently, there is still a gap in a comprehensive overview of the connection between structures and biocatalytic mechanisms of SMACs. Therefore, it is crucial to deeply investigate the role of SMACs in biocatalysis from the atomic structure level and to elucidate their potential mechanisms in biocatalytic processes. In this minireview, we summarize catalysis regulation methods of SMACs at the atomic structure level, focusing on the optimization of catalytic active sites, coordination environment, and active site-support interactions, and briefly discuss biocatalytic mechanisms for biomedical applications.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Huan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
18
|
Da J, Di X, Xie Y, Li J, Zhang L, Liu Y. Recent advances in nanomedicine for metabolism-targeted cancer therapy. Chem Commun (Camb) 2024; 60:2442-2461. [PMID: 38321983 DOI: 10.1039/d3cc05858a] [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: 02/08/2024]
Abstract
Metabolism denotes the sum of biochemical reactions that maintain cellular function. Different from most normal differentiated cells, cancer cells adopt altered metabolic pathways to support malignant properties. Typically, almost all cancer cells need a large number of proteins, lipids, nucleotides, and energy in the form of ATP to support rapid division. Therefore, targeting tumour metabolism has been suggested as a generic and effective therapy strategy. With the rapid development of nanotechnology, nanomedicine promises to have a revolutionary impact on clinical cancer therapy due to many merits such as targeting, improved bioavailability, controllable drug release, and potentially personalized treatment compared to conventional drugs. This review comprehensively elucidates recent advances of nanomedicine in targeting important metabolites such as glucose, glutamine, lactate, cholesterol, and nucleotide for effective cancer therapy. Furthermore, the challenges and future development in this area are also discussed.
Collapse
Affiliation(s)
- Jun Da
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China.
| | - XinJia Di
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China.
| | - YuQi Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China.
| | - JiLi Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China.
| | - LiLi Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China.
| | - YanLan Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
19
|
Liu Y, Niu R, Zhao H, Wang Y, Song S, Zhang H, Zhao Y. Single-Site Nanozymes with a Highly Conjugated Coordination Structure for Antitumor Immunotherapy via Cuproptosis and Cascade-Enhanced T Lymphocyte Activity. J Am Chem Soc 2024; 146:3675-3688. [PMID: 38305736 DOI: 10.1021/jacs.3c08622] [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: 02/03/2024]
Abstract
The extracellular matrix (ECM) in the tumor microenvironment (TME) and upregulated immune checkpoints (ICs) on antitumor immune cells impede the infiltration and killing effect of T cells, creating an immunosuppressive TME. Herein, a cholesterol oxidase (CHO) and lysyl oxidase inhibitor (LOX-IN-3) co-delivery copper-dibenzo-[g,p]chrysene-2,3,6,7,10,11,14,15-octaol single-site nanozyme (Cu-DBCO/CL) was developed. The conjugated organic ligand and well-distributed Cu-O4 sites endow Cu-DBCO with unique redox capabilities, enabling it to catalyze O2 and H2O2 to ·O2- and ·OH. This surge of reactive oxygen species (ROS) leads to impaired mitochondrial function and insufficient ATP supply, impacting the function of copper-transporting ATPase-1 and causing dihydrolipoamide S-acetyltransferase oligomerization-mediated cuproptosis. Moreover, multiple ROS storms and glutathione peroxidase 4 depletion also induce lipid peroxidation and trigger ferroptosis. Simultaneously, the ROS-triggered release of LOX-IN-3 reshapes the ECM by inhibiting lysyl oxidase activity and further enhances the infiltration of cytotoxic T lymphocytes (CD8+ T cells). CHO-triggered cholesterol depletion not only increases ·OH generation but also downregulates the expression of ICs such as PD-1 and TIM-3, restoring the antitumor activity of tumor-infiltrating CD8+ T cells. Therefore, Cu-DBCO/CL exhibits efficient properties in activating a potent antitumor immune response by cascade-enhanced CD8+ T cell viability. More importantly, ECM remodeling and cholesterol depletion could suppress the metastasis and proliferation of the tumor cells. In short, this immune nanoremodeler can greatly enhance the infiltration and antitumor activity of T cells by enhancing tumor immunogenicity, remodeling ECM, and downregulating ICs, thus achieving effective inhibition of tumor growth and metastasis.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Rui Niu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
| | - Huan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| | - Yinghui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
| | - Shuyan Song
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, People's Republic of China
- Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, 637371, Singapore
| |
Collapse
|