1
|
Kwun MS, Lee DG. Ferroptosis-Like Death Induction in Saccharomyces cerevisiae by Gold Nanoparticles. J Microbiol Biotechnol 2025; 35:e2501029. [PMID: 40295204 PMCID: PMC12089944 DOI: 10.4014/jmb.2501.01029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 04/30/2025]
Abstract
Ferroptosis, a novel form of regulated cell death (RCD), has emerged as a promising therapeutic strategy for cancer treatment. While gold nanoparticles (AuNPs) are known to induce cell death and ferroptosis in combination with certain antibiotics, the mechanisms underlying ferroptosis in microorganisms remain poorly understood. This study aimed to investigate whether AuNPs induce ferroptosis-like cell death in the eukaryotic microbe Saccharomyces cerevisiae. Our findings revealed that AuNPs significantly reduced cell viability in S. cerevisiae, suggesting their ability to trigger cell death. Ferroptosis-related precursors, including intracellular iron overload and depletion of glutathione (GSH), were observed, leading to the inactivation of glutathione peroxidase (GPx). These changes were associated with the accumulation of reactive oxygen species (ROS) and lipid peroxidation, which amplified oxidative stress within the cells. Elevated ROS levels and lipid peroxidation further resulted in membrane rupture and the formation of 8-hydroxydeoxyguanosine, indicating DNA damage. Mitochondrial dysfunction, a hallmark of ferroptosis, was also evident. AuNP treatment caused mitochondrial membrane potential hyperpolarization and a reduction in mitochondrial membrane density. Unlike previously characterized forms of RCD, ferroptosis-like death in S. cerevisiae did not involve chromatin condensation, DNA fragmentation, or metacaspase activation. Finally, ferroptosis-like characteristics were confirmed using Liperfluo, a lipid ROS-specific probe. In conclusion, this study demonstrated that AuNPs can induce ferroptosis-like cell death in S. cerevisiae. These findings highlight the potential of AuNPs as antifungal agents and contribute to the broader understanding of ferroptosis in eukaryotic microbes.
Collapse
Affiliation(s)
- Min Seok Kwun
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dong Gun Lee
- School of Life Science, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
2
|
Liu H, Wang L, Jin H, Tao K, Zhu X, Zhang M, Hou Y, Liu S, Zhang H. FeOOH-Assisted Formation of Hybrid Polymer Nanospindles for Efficient Iron Delivery and Ferroptosis Tumor Therapy. Bioconjug Chem 2025; 36:464-475. [PMID: 40045454 DOI: 10.1021/acs.bioconjchem.4c00546] [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: 03/20/2025]
Abstract
Exogenous iron delivery using iron-containing nanomaterials is an alternative strategy for enhancing the efficacy in ferroptosis tumor therapy but limited by the problems of low iron content, low tumor enrichment, low cellular uptake, and uncontrolled release of iron ions. To solve the problems, an FeOOH-assisted approach is demonstrated to produce iron hybrid polymer nanospindles (IHPNSs) for efficient iron delivery and ferroptosis tumor therapy. The IHPNSs are prepared through the cohydrolysis of FeCl3·6H2O with aniline, pyrrole, or amino-pyrrole. On the one hand, the hydrolysis of Fe3+ generates FeOOH particles, which further act as the templates to form fusiform architectures. On the other hand, Fe3+ triggers the oxidative polymerization of aniline, pyrrole, or amino-pyrrole. The as-prepared polymers are capable of coordinating with excessive Fe3+ and locate on the FeOOH templates, thus producing Fe3+/polymer composite-coated FeOOH nanospindles. Systematic studies indicate that the one-dimension-like morphology facilitates tumor enrichment and cellular uptake of IHPNSs. Besides the high iron content of IHPNSs, the controlled release of Fe3+ stimulated by the overexpressed glutathione (GSH) in the tumor microenvironment is achieved. The released Fe3+ is further transformed to Fe2+ by scavenging GSH, which leads to excessive accumulation of reactive oxygen species and lipid peroxides and finally induces ferroptosis of tumor cells. As a proof of concept, the IHPNSs show good efficacy in the treatment of a rat model of bladder tumors in situ.
Collapse
Affiliation(s)
- Heng Liu
- Department of Urinary, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Lu Wang
- Department of Pediatric Dentistry, Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
| | - Hao Jin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Kepeng Tao
- Department of Urinary, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Xuanqi Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Mengsi Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yuchuan Hou
- Department of Urinary, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Shuwei Liu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
3
|
Xu W, Guan G, Yue R, Dong Z, Lei L, Kang H, Song G. Chemical Design of Magnetic Nanomaterials for Imaging and Ferroptosis-Based Cancer Therapy. Chem Rev 2025; 125:1897-1961. [PMID: 39951340 DOI: 10.1021/acs.chemrev.4c00546] [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: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of regulatory cell death, has garnered significant interest as a therapeutic target in cancer treatment due to its distinct characteristics, including lipid peroxide generation and redox imbalance. However, its clinical application in oncology is currently limited by issues such as suboptimal efficacy and potential off-target effects. The advent of nanotechnology has provided a new way for overcoming these challenges through the development of activatable magnetic nanoparticles (MNPs). These innovative MNPs are designed to improve the specificity and efficacy of ferroptosis induction. This Review delves into the chemical and biological principles guiding the design of MNPs for ferroptosis-based cancer therapies and imaging-guided therapies. It discusses the regulatory mechanisms and biological attributes of ferroptosis, the chemical composition of MNPs, their mechanism of action as ferroptosis inducers, and their integration with advanced imaging techniques for therapeutic monitoring. Additionally, we examine the convergence of ferroptosis with other therapeutic strategies, including chemodynamic therapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, within the context of nanomedicine strategies utilizing MNPs. This Review highlights the potential of these multifunctional MNPs to surpass the limitations of conventional treatments, envisioning a future of drug-resistance-free, precision diagnostics and ferroptosis-based therapies for treating recalcitrant cancers.
Collapse
Affiliation(s)
- Wei Xu
- School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Guoqiang Guan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, PR China
| | - Zhe Dong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Lingling Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, 12 Seoul 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| |
Collapse
|
4
|
Sun J, Gao S, Wei G, Yu S, Zhang S, Yang A, Lu W. A Near-Infrared-II Fluorescent Nanoprobe Offering Real-Time Tracking of Fenton-Like Reaction for Cancer Chemodynamic Theranostics. NANO LETTERS 2025; 25:343-352. [PMID: 39705211 DOI: 10.1021/acs.nanolett.4c05087] [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: 12/22/2024]
Abstract
Chemodynamic therapy (CDT) utilizing Fenton or Fenton-like reactions to generate cytotoxic hydroxyl radicals by metal ions has become a compelling strategy for cancer treatment. Visualizing intratumoral Fenton or Fenton-like reactions especially at a cellular level in real-time can directly monitor the process of CDT, which is not yet feasible. Here, we present a molecule BADA chelating Cu2+ to form Cu-BADA nanoparticles, exhibiting fluorescence quenching properties through intermolecular electron transfer. The nanoparticles are lit up owing to glutathione and acid dual activatable Fenton-like reaction and generation of near-infrared-II fluorescent o-quinones. Moreover, fluorescence vanishing correlated with the decreased intratumoral Cu concentration, thus enabling to track the "on-off" process of Fenton-like reaction specifically in the tumor. Compared to 660 nm-excitation, the o-quinones excited at 830 nm offer deeper tissue near-infrared-II fluorescence imaging with higher resolution. Our results demonstrate a fluorescence nanotheranostic agent for CDT capable of monitoring the spatiotemporal dynamics of Fenton-like reaction.
Collapse
Affiliation(s)
- Jingwen Sun
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
- Quzhou Fudan Institute, Quzhou, Zhejiang 324002, P.R. China
| | - Shuai Gao
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
| | - Guoguang Wei
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
| | - Sheng Yu
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
| | - Sihang Zhang
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
| | - Afeng Yang
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
| | - Wei Lu
- School of Pharmacy, Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P.R. China
- Quzhou Fudan Institute, Quzhou, Zhejiang 324002, P.R. China
| |
Collapse
|
5
|
Wang F, Wu Z, Zhang Y, Li M, Wei P, Yi T, Li J. Semiconducting polymer nanoprodrugs enable tumor-specific therapy via sono-activatable ferroptosis. Biomaterials 2025; 312:122722. [PMID: 39096841 DOI: 10.1016/j.biomaterials.2024.122722] [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: 04/15/2024] [Revised: 07/06/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
Ferroptosis, a recently identified form of cell death, holds promise for cancer therapy, but concerns persist regarding its uncontrolled actions and potential side effects. Here, we present a semiconducting polymer nanoprodrug (SPNpro) featuring an innovative ferroptosis prodrug (DHU-CBA7) to induce sono-activatable ferroptosis for tumor-specific therapy. DHU-CBA7 prodrug incorporate methylene blue, ferrocene and urea bond, which can selectively and specifically respond to singlet oxygen (1O2) to turn on ferroptosis action via rapidly cleaving the urea bonds. DHU-CBA7 prodrug and a semiconducting polymer are self-assembled with an amphiphilic polymer to construct SPNpro. Ultrasound irradiation of SPNpro leads to the production of 1O2 via sonodynamic therapy (SDT) of the semiconducting polymer, and the generated 1O2 activated DHU-CBA7 prodrug to achieve sono-activatable ferroptosis. Consequently, SPNpro combine SDT with the controlled ferroptosis to effectively cure 4T1 tumors covered by 2-cm tissue with a tumor inhibition efficacy as high as 100 %, and also completely restrain tumor metastases. This study introduces a novel sono-activatable prodrug strategy for regulating ferroptosis, allowing for precise cancer therapy.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Zhiting Wu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Yijing Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Meng Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Peng Wei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China.
| | - Tao Yi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China.
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
6
|
Bao Y, Li G, Li S, Zhou H, Yang Z, Wang Z, Yan R, Guo C, Jin Y. A novel nanomedicine integrating ferroptosis and photothermal therapy, well-suitable for PD-L1-mediated immune checkpoint blockade. Mater Today Bio 2024; 29:101346. [PMID: 39635320 PMCID: PMC11616610 DOI: 10.1016/j.mtbio.2024.101346] [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: 08/18/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Immunotherapy based on immune checkpoint blockade has emerged as a promising treatment strategy; however, the therapeutic efficacy is limited by the immunosuppressive microenvironment. Here, we developed a novel immune-activated nanoparticle (Fc-SS-Fe/Cu) to address the issue of insufficient immune infiltration. Specifically, the structure of Fc-SS-Fe/Cu collapsed in response to the tumor microenvironment, the ferrocene and disulfide bonds and the released Fe/Cu ions can effectively generate ·OH and deplete GSH to increase oxidative stress, thereby inducing ferroptosis. Withal, the positive feedback mechanisms of "laser-triggered mild-temperature photothermal therapy (PTT), PTT accelerated ferroptosis and LPO accumulation, LPO mediated HSPs down-regulated to promote PTT," effectively triggers immunogenic cell death (ICD) in tumor cells, significantly enhancing their immunogenicity. Moreover, the O2-generating ability induced by Fc-SS-Fe/Cu could reverse the hypoxic tumor microenvironment, and importantly, the expression of PD-L1 on tumor cell surfaces could be effectively downregulated by inhibiting the HIF-1α pathways, thereby augmenting the effect of anti-PD-L1 (αPD-L1) therapy. Therefore, this study provides valuable strategies into enhancing PD-L1-mediated ICB therapy.
Collapse
Affiliation(s)
- Yujun Bao
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Guanghao Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Siqi Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Haishui Zhou
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
| | - Ziqing Yang
- School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Zhiqiang Wang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Rui Yan
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Changhong Guo
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
| | - Yingxue Jin
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| |
Collapse
|
7
|
Dai X, Yang Y. Metal-organic frameworks: potential synergies with cold atmospheric plasmas for cancer control. J Mater Chem B 2024; 12:10770-10785. [PMID: 39350546 DOI: 10.1039/d4tb00968a] [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: 11/01/2024]
Abstract
Metal-organic frameworks (MOFs) have attracted increasing attention for cancer treatment due to their unique characteristics such as crystallized porous structures, high surface area, and diverse and modifiable chemical properties. Despite the plethora of reports on MOF-based onco-therapeutic designs, these nanocomposites have rarely been launched for clinical use, given, at least, one unavoidable concern, i.e., biosafety. Among the diverse possibilities that MOFs can be engaged for cancer treatment, one unignorable opportunity is how MOFs can be combined with other emerging anti-cancer approaches as one treatment modality to resolve issues of either one for surpassed treatment efficacy. Taking cold atmospheric plasmas (CAPs) as an example, this review delineates the unique features of MOFs and discusses the possible synergies they can create with CAPs for mutual benefits. By providing one example on how MOFs can help overcome the issues of other pre-clinical cancer treatment regimens, this review identifies one research niche that may thrive the field of plasma medicine and revolutionize the schema of MOFs for biological applications.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China.
| | - Yixuan Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China.
| |
Collapse
|
8
|
Wang Y, Chen Z, An X, Li J, Li J, Pei Z, Pei Y. A DNA damage-amplifying nanoagent for cancer treatment via two-way regulation of redox dyshomeostasis and downregulation of tetrahydrofolate. Int J Biol Macromol 2024; 277:134276. [PMID: 39084430 DOI: 10.1016/j.ijbiomac.2024.134276] [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: 04/22/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
DNA damage-based therapy is widely used in cancer treatment, yet its therapeutic efficacy is constrained by the redox homeostasis and DNA damage repair mechanisms of tumor cells. To address these limitations and enhance the efficacy of DNA damage-based therapy, HA-CuH@MTX, a copper-histidine metal-organic complex (CuH) loaded with methotrexate (MTX) and modified with hyaluronic acid (HA), was developed to amplify the DNA damage induced. In vitro experiments demonstrated that the presence of both Cu+ and Cu2+ in HA-CuH@MTX enables two-way regulated redox dyshomeostasis (RDH), achieved through Cu+-catalyzed generation of •OH and Cu2+-mediated consumption of glutathione, thereby facilitating efficient DNA oxidative damage. In addition, DNA damage repair is synergistically inhibited by impairing nucleotide synthesis via histidine metabolism and MTX downregulation of tetrahydrofolate, a crucial raw material in nucleotide synthesis. In vivo experiments with 4T1 tumor-bearing mice demonstrate 83.6 % inhibition of tumor growth by HA-CuH@MTX. This work provides a new strategy to amplify the DNA damage caused by DNA damage-based cancer therapies, and holds great potential for improving their therapeutic efficacy.
Collapse
Affiliation(s)
- Yi Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zelong Chen
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xingwang An
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jiahui Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jiaxuan Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhichao Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yuxin Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
9
|
Deng Y, Huang S, Jiang G, Zhou L, Nezamzadeh-Ejhieh A, Liu J, Zhou Z. Current status and prospects of MOFs loaded with H 2O 2-related substances for ferroptosis therapy. RSC Med Chem 2024; 15:2996-3016. [PMID: 39309362 PMCID: PMC11411616 DOI: 10.1039/d4md00261j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a programmed cell death mechanism characterized by the accumulation of iron (Fe)-dependent lipid peroxides within cells. Ferroptosis holds excellent promise in tumor therapy. Metal-organic frameworks (MOFs) offer unique advantages in tumor ferroptosis treatment due to their high porosity, excellent stability, high biocompatibility, and targeting capabilities. Inducing ferroptosis in tumor cells primarily involves the production of reactive oxygen species (ROS), like hydroxyl radicals (˙OH), through iron-mediated Fenton reactions. However, the intrinsic H2O2 levels in tumor cells are often insufficient to sustain prolonged consumption, limiting therapeutic efficacy if ˙OH production is inadequate. Therefore, catalyzing or supplementing the intracellular H2O2 levels in tumor cells is essential for inducing ferroptosis by nanoscale metal-organic frameworks. This article reviews the biological characteristics and molecular mechanisms of ferroptosis, introduces H2O2-related substances, and reviews MOF-based nanoscale strategies for enhancing intracellular H2O2 levels in tumor cells. Finally, the challenges and prospects of this approach are discussed, aiming to provide insights into improving the effectiveness of ferroptosis induced by MOFs.
Collapse
Affiliation(s)
- Yu Deng
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Sida Huang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Guanming Jiang
- Department of Oncology, Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital) 78 Wandao Road South Dongguan 523059 Guangdong China
| | - Luyi Zhou
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | | | - Jianqiang Liu
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| | - Zhikun Zhou
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials Dongguan 523808 China
| |
Collapse
|
10
|
Liu H, Xing F, Yu P, Shakya S, Peng K, Liu M, Xiang Z, Ritz U. Integrated design and application of stimuli-responsive metal-organic frameworks in biomedicine: current status and future perspectives. J Mater Chem B 2024; 12:8235-8266. [PMID: 39058314 DOI: 10.1039/d4tb00768a] [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: 07/28/2024]
Abstract
In recent years, metal-organic frameworks (MOFs) have garnered widespread attention due to their distinctive attributes, such as high surface area, tunable properties, biodegradability, extremely low density, high loading capacity, diverse chemical functionalities, thermal stability, well-defined pore sizes, and molecular dimensions. Increasingly, biomedical researchers have turned their focus towards their multifaceted development. Among these, stimuli-responsive MOFs, with their unique advantages, have captured greater interest from researchers. This review will delve into the merits and drawbacks of both endogenous and exogenous stimuli-responsive MOFs, along with their application directions. Furthermore, it will outline the characteristics of different synthesis routes of MOFs, exploring various design schemes and modification strategies and their impacts on the properties of MOF products, as well as how to control them. Additionally, we will survey different types of stimuli-responsive MOFs, discussing the significance of various MOF products reported in biomedical applications. We will categorically summarize different strategies such as anticancer therapy, antibacterial treatment, tissue repair, and biomedical imaging, as well as insights into the development of novel MOFs nanomaterials in the future. Finally, this review will conclude by summarizing the challenges in the development of stimuli-responsive MOFs in the field of biomedicine and providing prospects for future research endeavors.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Fei Xing
- Department of Pediatric Surgery, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Kun Peng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiang Xi, China
| | - Ming Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
- Department of Orthopedics, Sanya People's Hospital, 572000 Sanya, Hainan, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
11
|
Moore JM, Genna DT. Disulfide Bonds as Functional Tethers in Metal-Organic Frameworks. Chemistry 2024; 30:e202401713. [PMID: 38842480 DOI: 10.1002/chem.202401713] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
The functionality of metal-organic frameworks (MOFs) is often encoded by specific chemical moieties found within these architectures. As such, new techniques to install increasingly more complex functionalities in MOFs are regularly being reported in the literature. One such functional group is the disulfide bond. The redox behavior of this covalent linkage renders MOFs responsive to stimuli, often under reducing conditions. Here, we review examples in which disulfide-containing MOFs are deployed in applications including drug delivery, therapeutic ferroptosis, exfoliation, energy storage, sensing, and others.
Collapse
Affiliation(s)
- Jennifer M Moore
- Department of Chemical and Biological Sciences, Youngstown State University, 44555, Youngstown, Ohio, USA
| | - Douglas T Genna
- Department of Chemical and Biological Sciences, Youngstown State University, 44555, Youngstown, Ohio, USA
| |
Collapse
|
12
|
An N, Tang S, Wang Y, Luan J, Shi Y, Gao M, Guo C. FeP-Based Nanotheranostic Platform for Enhanced Phototherapy/Ferroptosis/Chemodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309940. [PMID: 38534030 DOI: 10.1002/smll.202309940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Ferroptosis is an iron-dependent and lipid peroxides (LPO)-overloaded programmed damage cell death, induced by glutathione (GSH) depletion and glutathione peroxide 4 (GPX4) inactivation. However, the inadequacy of endogenous iron and reactive oxygen species (ROS) restricts the efficacy of ferroptosis. To overcome this obstacle, a near-infrared photo-responsive FeP@PEG NPs is fabricated. Exogenous iron pool can enhance the effect of ferroptosis via the depletion of GSH and further regulate GPX4 inactivation. Generation of ·OH derived from the Fenton reaction is proved by increased accumulation of lipid peroxides. The heat generated by photothermal therapy and ROS generated by photodynamic therapy can enhance cell apoptosis under near-infrared (NIR-808 nm) irradiation, as evidenced by mitochondrial dysfunction and further accumulation of lipid peroxide content. FeP@PEG NPs can significantly inhibit the growth of several types of cancer cells in vitro and in vivo, which is validated by theoretical and experimental results. Meanwhile, FeP@PEG NPs show excellent T2-weighted magnetic resonance imaging (MRI) property. In summary, the FeP-based nanotheranostic platform for enhanced phototherapy/ferroptosis/chemodynamic therapy provides a reliable opportunity for clinical cancer theranostics.
Collapse
Affiliation(s)
- Na An
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Shuanglong Tang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yuwei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Jing Luan
- The HIT Center for Life Science, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Ying Shi
- Magnetic Resonance Department of the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Minghui Gao
- The HIT Center for Life Science, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Chongshen Guo
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
13
|
Tousian B, Khosravi AR, Ghasemi MH, Kadkhodaie M. Biomimetic functionalized metal organic frameworks as multifunctional agents: Paving the way for cancer vaccine advances. Mater Today Bio 2024; 27:101134. [PMID: 39027676 PMCID: PMC11255118 DOI: 10.1016/j.mtbio.2024.101134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Biomimetic functionalized metal-organic frameworks (Fn-MOFs) represent a cutting-edge approach in the realm of cancer vaccines. These multifunctional agents, inspired by biological systems, offer unprecedented opportunities for the development of next-generation cancer vaccines. The vast surface area, tunable pore size, and diverse chemistry of MOFs provide a versatile scaffold for the encapsulation and protection of antigenic components, crucial for vaccine stability and delivery. This work delves into the innovative design and application of Fn-MOFs, highlighting their role as carriers for immune enhancement and their potential to revolutionize vaccine delivery. By mimicking natural processes, Fn-MOFs, with their ability to be functionalized with a myriad of chemical and biological entities, exhibit superior biocompatibility and stimuli-responsive behavior and facilitate targeted delivery to tumor sites. This review encapsulates the latest advancements in Fn-MOF technology, from their synthesis and surface modification to their integration into stimuli-responsive and combination therapies. It underscores the significance of biomimetic approaches in overcoming current challenges in cancer vaccine development, such as antigen stability and immune evasion. By leveraging the biomimetic nature of Fn-MOFs, this work paves the way for innovative strategies in cancer vaccines, aiming to induce potent and long-lasting immune responses against malignancies.
Collapse
Affiliation(s)
- Bushra Tousian
- Department of Microbiology and Immunology, Veterinary Medicine Faculty, University of Tehran, PO Box 1419963111, Tehran, Iran
| | - Ali Reza Khosravi
- Department of Microbiology and Immunology, Veterinary Medicine Faculty, University of Tehran, PO Box 1419963111, Tehran, Iran
| | - Mohammad Hadi Ghasemi
- Applied Chemistry Research Group, ACECR-Tehran Organization, PO Box 13145-186, Tehran, Iran
| | - Majid Kadkhodaie
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
14
|
Deng Y, Wang D, Zhao W, Qiu G, Zhu X, Wang Q, Qin T, Tang J, Jiang J, Lin N, Wei L, Liu Y, Xie Y, Chen J, Deng L, Liu J. A Multifunctional Nanocatalytic Metal-Organic Framework as a Ferroptosis Amplifier for Mild Hyperthermia Photothermal Therapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0397. [PMID: 38952997 PMCID: PMC11214948 DOI: 10.34133/research.0397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/07/2024] [Indexed: 07/03/2024]
Abstract
Hyperthermia therapy is considered an effective anticancer strategy. However, high temperature can trigger an excessive inflammatory response, leading to tumor self-protection, immunosuppression, metastasis, and recurrence. To address this issue, we reported a multifunctional photothermal nanoplatform to achieve mild hyperthermia photothermal therapy (mild PTT) based on cisplatin (DDP) and a ferrocene metal-organic framework (MOF-Fc) nanocomposite, which can specifically enhance ferroptosis-triggered oxidative stress levels and synchronously amplify mild hyperthermia PTT-mediated anticancer responses. Both in vitro and in vivo antineoplastic results verify the superiority of mild PTT with DDP/MOF-Fc@HA. The combination of DDP and MOF-Fc exhibits Fenton catalytic activity and glutathione depletion capacity, magnifying mild hyperthermia effects via the radical oxygen species (ROS)-adenosine triphosphate (ATP)-HSP silencing pathway, with important implications for clinical hyperthermia therapy.
Collapse
Affiliation(s)
- Ying Deng
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Duo Wang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School,
Southeast University, Nanjing, Jiangsu, China
| | - Wenhua Zhao
- Department of Oncology and Research Department, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Guanhua Qiu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoqi Zhu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Qin Wang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Tian Qin
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Jiali Tang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Jinghang Jiang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Ningjing Lin
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Lili Wei
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Yichen Liu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Yuan Xie
- Department of Oncology and Research Department, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Jie Chen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| | - Liu Deng
- Hunan Provincial Key Laboratory of Micro and Nano Materials Interface Science, College of Chemistry and Chemical Engineering,
Central South University, Changsha, Hunan, China
| | - Junjie Liu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital,
Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
15
|
Wang J, Wang Z, Li L, Wang M, Chang J, Gao M, Wang D, Li C. Ultra-small Janus nanoparticle-induced activation of ferroptosis for synergistic tumor immunotherapy. Acta Biomater 2024; 181:362-374. [PMID: 38663684 DOI: 10.1016/j.actbio.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/08/2024] [Accepted: 04/21/2024] [Indexed: 05/07/2024]
Abstract
Ferroptosis induced by lipid peroxide (LPO) accumulation is an effective cell death pathway for cancer therapy. However, how to effectively induce ferroptosis at tumor sites and improve its therapeutic effectiveness remains challenging. Here, MnFe2O4@NaGdF4@NLG919@HA (MGNH) nanocomplex with tumor-specific targeting and TME response is constructed to overcome immunosuppressive tumor microenvironment (TME) to potentiate the curative effect of ferroptosis by coupling the immune checkpoint indoleamine 2,3-dioxygenase (IDO) inhibitor, NLG919, and hyaluronic acid (HA) to novel ultra-small MnFe2O4@NaGdF4 (MG) nanoparticles with a Janus structure. Firstly, tumor site-precise delivery of MG and NLG919 is achieved with HA targeting. Secondly, MG acts as a magnetic resonance imaging contrast agent, which not only has a good photothermal effect to realize tumor photothermal therapy, but also depletes glutathione and catalyzes the production of reactive oxygen species from endogenous H2O2, which effectively promotes the accumulation of LPO and inhibits the expression of glutathione peroxidase 4, achieving enhanced ferroptosis. Thirdly, NLG919 inhibits the differentiation of Tregs by blocking the tryptophan/kynurenine immune escape pathway, thereby reversing immunosuppressive TME together with the Mn2+-activated cGAS-STING pathway. This work contributes new perspectives for the development of novel ultra-small Janus nanoparticles to reshape immunosuppressive TME and ferroptosis activation. STATEMENT OF SIGNIFICANCE: The Janus structured MnFe2O4@NaGdF4@NLG919@HA (MGNH) nanocomplex was synthesized, which can realize the precise delivery of T1/T2 contrast agents MnFe2O4@NaGdF4 (MG) and NLG919 at the tumor site under the ultra-small Janus structural characteristics and targeted molecule HA. The production of ROS, consumption of GSH, and photothermal properties of MGNH make it possible for CDT/PTT activated ferroptosis, and synergistically disrupt and reprogram tumor growth and immunosuppressive tumor microenvironment with NLG919 and Mn2+-mediated activation of cGAS-STING pathway, achieving CDT/PTT/immunotherapy activated by ferroptosis. Meanwhile, ultra-small structural properties of MGNH facilitate subsequent metabolic clearance by the body, allowing for the minimization of potential biotoxicity associated with its prolonged retention.
Collapse
Affiliation(s)
- Junrong Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Zhifang Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Lei Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Man Wang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Jiaying Chang
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Minghong Gao
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, College of Chemistry and Materials Sciences, Zhejiang Normal University, Jinhua 321004, PR China.
| | - Chunxia Li
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, Shandong 266237, PR China.
| |
Collapse
|
16
|
Su Y, Liu B, Wang B, Chan L, Xiong C, Lu L, Zhang X, Zhan M, He W. Progress and Challenges in Tumor Ferroptosis Treatment Strategies: A Comprehensive Review of Metal Complexes and Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310342. [PMID: 38221682 DOI: 10.1002/smll.202310342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/27/2023] [Indexed: 01/16/2024]
Abstract
Ferroptosis is a new form of regulated cell death featuring iron-dependent lipid peroxides accumulation to kill tumor cells. A growing body of evidence has shown the potential of ferroptosis-based cancer therapy in eradicating refractory malignancies that are resistant to apoptosis-based conventional therapies. In recent years, studies have reported a number of ferroptosis inducers that can increase the vulnerability of tumor cells to ferroptosis by regulating ferroptosis-related signaling pathways. Encouraged by the rapid development of ferroptosis-driven cancer therapies, interdisciplinary fields that combine ferroptosis, pharmaceutical chemistry, and nanotechnology are focused. First, the prerequisites and metabolic pathways for ferroptosis are briefly introduced. Then, in detail emerging ferroptosis inducers designed to boost ferroptosis-induced tumor therapy, including metal complexes, metal-based nanoparticles, and metal-free nanoparticles are summarized. Subsequently, the application of synergistic strategies that combine ferroptosis with apoptosis and other regulated cell death for cancer therapy, with emphasis on the use of both cuproptosis and ferroptosis to induce redox dysregulation in tumor and intracellular bimetallic copper/iron metabolism disorders during tumor treatment is discussed. Finally, challenges associated with clinical translation and potential future directions for potentiating cancer ferroptosis therapies are highlighted.
Collapse
Affiliation(s)
- Yanhong Su
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Bing Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Binghan Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Leung Chan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Chan Xiong
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Xuanjun Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
- MOE Frontiers Science Centre for Precision Oncology, University of Macau, Macau SAR, 999078, China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
| | - Weiling He
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, P. R. China
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China
| |
Collapse
|
17
|
Wang M, Yu A, Han W, Chen J, Lu C, Tu X. Self-assembled metal-phenolic nanocomplexes comprised of green tea catechin for tumor-specific ferroptosis. Mater Today Bio 2024; 26:101040. [PMID: 38590984 PMCID: PMC10999486 DOI: 10.1016/j.mtbio.2024.101040] [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: 02/08/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024] Open
Abstract
Ferroptosis, a newly discovered form of regulated cell death, has garnered significant attention in the field of tumor therapy. However, the presence of overexpressed glutathione (GSH) and insufficient levels of H2O2 in the tumor microenvironment (TME) hinders the occurrence of ferroptosis. In response to these challenges, here we have constructed the self-assembled nanocomplexes (FeE NPs) utilizing epigallocatechin-3-gallate (EGCG) from green tea polyphenols and metal ions (Fe3+) as components. After grafting PEG, the nanocomplexes (FeE@PEG NPs) exhibit good biocompatibility and synergistically enhanced tumor-inhibitory properties. FeE@PEG NPs can be disassembled by H2O2 in the TME, leading to the rapid release of Fe3+ and EGCG. The released Fe3+ produces large amounts of toxic •OH by the Fenton reactions while having minimal impact on normal cells. The generated •OH effectively induces lipid peroxidation, which leads to ferroptosis in tumor cells. Meanwhile, the released EGCG can autoxidize to produce H2O2, which further promotes the production of •OH radicals and increases lipid peroxide levels. Moreover, EGCG also depletes the high levels of intracellular GSH, leading to an intracellular redox imbalance and triggering ferroptosis. This study provides new insights into advancing anticancer ferroptosis through rational material design, offering promising avenues for future research.
Collapse
Affiliation(s)
- Min Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Aoling Yu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Wen Han
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Jingyi Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgery Research Institute of Fujian Province, Fuzhou, Fujian, 350001, China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Xiankun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Neurosurgery Research Institute of Fujian Province, Fuzhou, Fujian, 350001, China
| |
Collapse
|
18
|
Tripathy S, Haque S, Londhe S, Das S, Norbert CC, Chandra Y, Sreedhar B, Patra CR. ROS mediated Cu[Fe(CN) 5NO] nanoparticles for triple negative breast cancer: A detailed study in preclinical mouse model. BIOMATERIALS ADVANCES 2024; 160:213832. [PMID: 38547763 DOI: 10.1016/j.bioadv.2024.213832] [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: 01/27/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 05/04/2024]
Abstract
Triple negative breast cancer (TNBC) is an aggressive form of tumor, more prevalent in younger women resulting in poor survival rate (2nd in cancer deaths) because of its asymptomatic existence. The most popular and convenient approach for the treatment of TNBC is chemotherapy which is associated with several limitations. Considering the importance of nanotechnology in health care system, in the present manuscript, we have designed and developed a simple, efficient, cost effective, and ecofriendly method for the synthesis of copper nitroprusside analogue nanoparticles (Cu[Fe(CN)5NO] which is abbreviated as CuNPANP that may be the potential anti-cancer nanomedicine for the treatment of TNBC. Copper (present in CuNPANP) is used because of its affordability, nutritional value and various biomedical applications. The CuNPANP are thoroughly characterized using several analytical techniques. The in vitro cell viability (in normal cells) and the ex vivo hemolysis assay reveal the biocompatible nature of CuNPANP. The anti-cancer activity of the CuNPANP is established in TNBC cells (MDA-MB-231 and 4T1) through several in vitro assays along with plausible mechanisms. The intraperitoneal administration of CuNPANP in orthotopic breast tumor model by transplanting 4T1 cells into the mammary fat pad of BALB/c mouse significantly inhibits the growth of breast carcinoma as well as increases the survival time of tumor-bearing mice. These results altogether potentiate the anti-cancer efficacy of CuNPANP as a smart therapeutic nanomedicine for treating TNBC in near future after bio-safety evaluation in large animals.
Collapse
Affiliation(s)
- Sanchita Tripathy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Shagufta Haque
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Swapnali Londhe
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Sourav Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Caroline Celine Norbert
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
| | - Yogesh Chandra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Bojja Sreedhar
- Department of Analytical & Structural ChemistryCSIR-Indian Institute of Chemical Technology, Uppal Road,Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Kamala Nehru Nagar, Gaziabad 201002, U.P., India.
| |
Collapse
|
19
|
Zou W, Gao F, Meng Z, Cai X, Chen W, Zheng Y, Ying T, Wang L, Wu J. Lactic acid responsive sequential production of hydrogen peroxide and consumption of glutathione for enhanced ferroptosis tumor therapy. J Colloid Interface Sci 2024; 663:787-800. [PMID: 38442520 DOI: 10.1016/j.jcis.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ferroptosis is characterized by the lethal accumulation of lipid reactive oxygen species (ROS), which has great potential for tumor therapy. However, developing new ferroptosis-inducing strategies by combining nanomaterials with small molecule inducers is important. In this study, an enzyme-gated biodegradable natural-product delivery system based on lactate oxidase (LOD)-gated biodegradable iridium (Ir)-doped hollow mesoporous organosilica nanoparticles (HMONs) loaded with honokiol (HNK) (HNK@Ir-HMONs-LOD, HIHL) is designed to enhance ferroptosis in colon tumor therapy. After reaching the tumor microenvironment, the outer LOD dissociates and releases the HNK to induce ferroptosis. Moreover, the released dopant Ir4+ and disulfide-bridged organosilica frameworks deplete intracellular glutathione (GSH), which is followed by GSH-mediated Ir(IV)/Ir(III) conversion. This leads to the repression of glutathione peroxidase 4 (GPX4) activity and decomposition of intratumoral hydrogen peroxide (H2O2) into hydroxyl radicals (•OH) by Ir3+-mediated Fenton-like reactions. Moreover, LOD efficiently depletes lactic acid to facilitate the generation of H2O2 and boost the Fenton reaction, which in turn enhances ROS generation. With the synergistic effects of these cascade reactions and the release of HNK, notable ferroptosis efficacy was observed both in vitro and in vivo. This combination of natural product-induced and lactic acid-responsive sequential production of H2O2 as well as the consumption of glutathione may provide a new paradigm for achieving effective ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Weijuan Zou
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Feng Gao
- Department of Ultrasonic Imaging, the First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Zheying Meng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Xiaojun Cai
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Wu Chen
- Department of Ultrasonic Imaging, the First Hospital of Shanxi Medical University, Taiyuan, 030001, PR China
| | - Yuanyi Zheng
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Tao Ying
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| | - Longchen Wang
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| | - Jianrong Wu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| |
Collapse
|
20
|
Yan Z, Wu X, Tan W, Yan J, Zhou J, Chen S, Miao J, Cheng J, Shuai C, Deng Y. Single-Atom Cu Nanozyme-Loaded Bone Scaffolds for Ferroptosis-Synergized Mild Photothermal Therapy in Osteosarcoma Treatment. Adv Healthc Mater 2024; 13:e2304595. [PMID: 38424663 DOI: 10.1002/adhm.202304595] [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: 12/23/2023] [Revised: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The rapid multiplication of residual tumor cells and poor reconstruction quality of new bone are considered the major challenges in the postoperative treatment of osteosarcoma. It is a promising candidate for composite bone scaffold which combines photothermal therapy (PTT) and bone regeneration induction for the local treatment of osteosarcoma. However, it is inevitable to damage the normal tissues around the tumor due to the hyperthermia of PTT, while mild heat therapy shows a limited effect on antitumor treatment as the damage can be easily repaired by stress-induced heat shock proteins (HSP). This study reports a new type of single-atom Cu nanozyme-loaded bone scaffolds, which exhibit exceptional photothermal conversion properties as well as peroxidase and glutathione oxidase mimicking activities in vitro experiments. This leads to lipid peroxidation (LPO) and reactive oxygen species (ROS) upregulation, ultimately causing ferroptosis. The accumulation of LPO and ROS also contributes to HSP70 inactivation, maximizing PTT efficiency against tumors at an appropriate therapeutic temperature and minimizing the damage to surrounding normal tissues. Further, the bone scaffold promotes bone regeneration via a continuous release of bioactive ions (Ca2+, P5+, Si4+, and Cu2+). The results of in vivo experiments reveal that scaffolds inhibit tumor growth and promote bone repair.
Collapse
Affiliation(s)
- Zuyun Yan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Xin Wu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jinpeng Yan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, 410017, P. R. China
| | - Jun Zhou
- Medical Science Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Shijie Chen
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jinglei Miao
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Jun Cheng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Cijun Shuai
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
21
|
Li X, Lin H, Hu J, Fang J, Liu H, Fu C, Zhao K. A redox homeostasis disruptor based on a biodegradable nanoplatform for ultrasound (US) imaging-guided high-performance ferroptosis therapy of tumors. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2024; 25:2351354. [PMID: 38800054 PMCID: PMC11123443 DOI: 10.1080/14686996.2024.2351354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
The synergistic disruption of intracellular redox homeostasis through the combination of ferroptosis/gas therapy shows promise in enhancing the antitumor efficacy. However, the development of an optimal delivery system encounters significant challenges, including effective storage, precise delivery, and controlled release of therapeutic gas. In this study, we propose the utilization of a redox homeostasis disruptor that is selectively activated by the tumor microenvironment (TME), in conjunction with our newly developed nanoplatforms (MC@HMOS@Au@RGD), for highly efficient ferroptosis therapy of tumors. The TME-triggered degradation of HMOS initiates the release of MC and AuNPs from the MC@HMOS@Au@RGD nanoplatform. The released MC subsequently reacts with endogenous hydrogen peroxide (H2O2) and H+ to enable the on-demand release of CO gas, leading to mitochondrial damage. Simultaneously, the released AuNPs exhibit GOx-like activity, catalyzing glucose to generate gluconic acid and H2O2. This process not only promotes the decomposition of MnCO to enhance CO production but also enhances the Fenton-like reaction between Mn2+ and H2O2, generating ROS through the modulation of the H+ and H2O2-enriched TME. Moreover, the generation of CO bubbles enables the monitoring of the ferroptosis treatment process through ultrasound (US) imaging. The efficacy of our prepared MC@HMOS@Au@RGD disruptors in ferroptosis therapy is validated through both in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Xia Li
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huijian Lin
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianbo Hu
- Medical Imaging Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiajin Fang
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Science Experiment Center, Guangdong Huayan Biomedical Technology Centre, Guangzhou, China
| | - Hongsheng Liu
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Science Experiment Center, Guangdong Huayan Biomedical Technology Centre, Guangzhou, China
| | - Can Fu
- Functional Examination Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kewei Zhao
- Laboratory Department, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Vilchis-Landeros MM, Vázquez-Meza H, Vázquez-Carrada M, Uribe-Ramírez D, Matuz-Mares D. Antioxidant Enzymes and Their Potential Use in Breast Cancer Treatment. Int J Mol Sci 2024; 25:5675. [PMID: 38891864 PMCID: PMC11171593 DOI: 10.3390/ijms25115675] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
According to the World Health Organization (WHO), breast cancer (BC) is the deadliest and the most common type of cancer worldwide in women. Several factors associated with BC exert their effects by modulating the state of stress. They can induce genetic mutations or alterations in cell growth, encouraging neoplastic development and the production of reactive oxygen species (ROS). ROS are able to activate many signal transduction pathways, producing an inflammatory environment that leads to the suppression of programmed cell death and the promotion of tumor proliferation, angiogenesis, and metastasis; these effects promote the development and progression of malignant neoplasms. However, cells have both non-enzymatic and enzymatic antioxidant systems that protect them by neutralizing the harmful effects of ROS. In this sense, antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), thioredoxin reductase (TrxR), and peroxiredoxin (Prx) protect the body from diseases caused by oxidative damage. In this review, we will discuss mechanisms through which some enzymatic antioxidants inhibit or promote carcinogenesis, as well as the new therapeutic proposals developed to complement traditional treatments.
Collapse
Affiliation(s)
- María Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Héctor Vázquez-Meza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Melissa Vázquez-Carrada
- Institute of Microbiology, Cluster of Excellence on Plant Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Daniel Uribe-Ramírez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, Mexico City C.P. 07738, Mexico;
| | - Deyamira Matuz-Mares
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| |
Collapse
|
23
|
Xue P, Zhuang H, Bai T, Zeng X, Deng J, Shao S, Yan S. Iron (II)-based metal-organic framework nanozyme for boosting tumor ferroptosis through inhibiting DNA damage repair and system Xc . J Nanobiotechnology 2024; 22:228. [PMID: 38715049 PMCID: PMC11077818 DOI: 10.1186/s12951-024-02508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Development of ferroptosis-inducible nanoplatforms with high efficiency and specificity is highly needed and challenging in tumor ferrotherapy. Here, we demonstrate highly effective tumor ferrotherapy using iron (II)-based metal-organic framework (FessMOF) nanoparticles, assembled from disulfide bonds and ferrous ions. The as-prepared FessMOF nanoparticles exhibit peroxidase-like activity and pH/glutathione-dependent degradability, which enables tumor-responsive catalytic therapy and glutathione depletion by the thiol/disulfide exchange to suppress glutathione peroxidase 4, respectively. Upon PEGylation and Actinomycin D (ActD) loading, the resulting FessMOF/ActD-PEG nanoplatform induces marked DNA damage and lipid peroxidation. Concurrently, we found that ActD can inhibit Xc- system and elicit ferritinophagy, which further boosts the ferrotherapeutic efficacy of the FessMOF/ActD-PEG. In vivo experiments demonstrate that our fabricated nanoplatform presents excellent biocompatibility and a high tumor inhibition rate of 91.89%.
Collapse
Affiliation(s)
- Panpan Xue
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Huilan Zhuang
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Tingjie Bai
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Xuemei Zeng
- Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, 1 Keji Road, Fuzhou, 350117, PR China.
| | - Jinpeng Deng
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Sijie Shao
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Shuangqian Yan
- The Straits Institute of Flexible Electronics (SIFE, Future Technologies), The Straits Laboratory of Flexible Electronics (SLoFE), Fujian Normal University, Fuzhou, Fujian, 350117, China.
| |
Collapse
|
24
|
Sheikh A, Kesharwani P, Almalki WH, Almujri SS, Dai L, Chen ZS, Sahebkar A, Gao F. Understanding the Novel Approach of Nanoferroptosis for Cancer Therapy. NANO-MICRO LETTERS 2024; 16:188. [PMID: 38698113 PMCID: PMC11065855 DOI: 10.1007/s40820-024-01399-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/16/2024] [Indexed: 05/05/2024]
Abstract
As a new form of regulated cell death, ferroptosis has unraveled the unsolicited theory of intrinsic apoptosis resistance by cancer cells. The molecular mechanism of ferroptosis depends on the induction of oxidative stress through excessive reactive oxygen species accumulation and glutathione depletion to damage the structural integrity of cells. Due to their high loading and structural tunability, nanocarriers can escort the delivery of ferro-therapeutics to the desired site through enhanced permeation or retention effect or by active targeting. This review shed light on the necessity of iron in cancer cell growth and the fascinating features of ferroptosis in regulating the cell cycle and metastasis. Additionally, we discussed the effect of ferroptosis-mediated therapy using nanoplatforms and their chemical basis in overcoming the barriers to cancer therapy.
Collapse
Affiliation(s)
- Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, 61421, Asir-Abha, Saudi Arabia
| | - Linxin Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, People's Republic of China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, 11439, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, People's Republic of China.
| |
Collapse
|
25
|
Song Y, Xu X, Wang Z, Zhao Y. Metal-Organic Framework-Based Nanomedicines for Ferroptotic Cancer Therapy. Adv Healthc Mater 2024; 13:e2303533. [PMID: 38221753 DOI: 10.1002/adhm.202303533] [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: 10/15/2023] [Revised: 11/28/2023] [Indexed: 01/16/2024]
Abstract
As an iron-dependent, non-apoptosis, regulated cell death (RCD) modality, ferroptosis has gained growing attention for cancer therapy. With the development of nanomaterials in the biomedical field, ferroptotic cancer nanomedicine is extensively investigated. Amongst various nanomaterials, metal-organic frameworks (MOFs) are hybridized porous materials consisting of metal ions or clusters bridged by organic linkers. The superior properties of MOFs, such as high porosity and cargo loading, ease of surface modification, and good biocompatibility, make them appealing in inducing or sensitizing ferroptotic cell death. There are remarkable achievements in the field of MOF-based ferroptosis cancer therapy. However, this topic is not reviewed. This review will introduce the fundamentals of MOF and ferroptosis machinery, summarize the recent progress of MOF-based ferroptotic anticancer drug delivery, discuss the benefits and problems of MOFs as vehicles and sensitizers for cancer ferroptosis, and provide the perspective on future research direction on this promising field.
Collapse
Affiliation(s)
- Yue Song
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Tianjin University, Tianjin, 300072, China
| | - Xinran Xu
- Department of Obstetrics, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University Affiliated Maternity Hospital, Tianjin, 300100, China
| | - Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Tianjin University, Tianjin, 300072, China
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
26
|
Huang M, Teng Q, Cao F, Huang J, Pang J. Ferroptosis and ferroptosis-inducing nanomedicine as a promising weapon in combination therapy of prostate cancer. Biomater Sci 2024; 12:1617-1629. [PMID: 38379396 DOI: 10.1039/d3bm01894f] [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: 02/22/2024]
Abstract
Incidence and mortality of prostate cancer (PCa) rank in the top five among male tumors. However, single treatment modalities are often restricted due to biochemical recurrence and drug resistance, necessitating the development of new approaches for the combination treatment of castration-resistant and neuroendocrine PCa. Ferroptosis is characterized by the accumulation of iron-overload-mediated lipid peroxidation and has shown promising outcomes in anticancer treatment, prompting us to present a review reporting the application of ferroptosis in the treatment of PCa. First, the process and mechanism of ferroptosis are briefly reviewed. Second, research advances combining ferroptosis-inducing agents and clinical treatment regimens, which exhibit a "two-pronged approach" effect, are further summarized. Finally, the recent progress on ferroptosis-inducing nanomaterials for combination anticancer therapy is presented. This review is expected to provide novel insights into ferroptosis-based combination treatment in drug-resistant PCa.
Collapse
Affiliation(s)
- Mengjun Huang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Qiliang Teng
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Fei Cao
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Jinsheng Huang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
27
|
Wang Y, Chen Z, Li J, Wen Y, Li J, Lv Y, Pei Z, Pei Y. A Paramagnetic Metal-Organic Framework Enhances Mild Magnetic Hyperthermia Therapy by Downregulating Heat Shock Proteins and Promoting Ferroptosis via Aggravation of Two-Way Regulated Redox Dyshomeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306178. [PMID: 38161219 PMCID: PMC10953551 DOI: 10.1002/advs.202306178] [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: 08/31/2023] [Revised: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Mild magnetic hyperthermia therapy (MMHT) holds great potential in treating deep-seated tumors, but its efficacy is impaired by the upregulation of heat shock proteins (HSPs) during the treatment process. Herein, Lac-FcMOF, a lactose derivative (Lac-NH2 ) modified paramagnetic metal-organic framework (FcMOF) with magnetic hyperthermia property and thermal stability, has been developed to enhance MMHT therapeutic efficacy. In vitro studies showed that Lac-FcMOF aggravates two-way regulated redox dyshomeostasis (RDH) via magnetothermal-accelerated ferricenium ions-mediated consumption of glutathione and ferrocene-catalyzed generation of ∙OH to induce oxidative damage and inhibit heat shock protein 70 (HSP70) synthesis, thus significantly enhancing the anti-cancer efficacy of MMHT. Aggravated RDH promotes glutathione peroxidase 4 inactivation and lipid peroxidation to promote ferroptosis, which further synergizes with MMHT. H22-tumor-bearing mice treated with Lac-FcMOF under alternating magnetic field (AMF) demonstrated a 90.4% inhibition of tumor growth. This work therefore provides a new strategy for the simple construction of a magnetic hyperthermia agent that enables efficient MMHT by downregulating HSPs and promoting ferroptosis through the aggravation of two-way regulated RDH.
Collapse
Affiliation(s)
- Yi Wang
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Zelong Chen
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Jiahui Li
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Yafei Wen
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Jiaxuan Li
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Yinghua Lv
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Zhichao Pei
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| | - Yuxin Pei
- College of Chemistry and PharmacyNorthwest A&F UniversityYanglingShaanxi712100P. R. China
| |
Collapse
|
28
|
Li B, Ashrafizadeh M, Jiao T. Biomedical application of metal-organic frameworks (MOFs) in cancer therapy: Stimuli-responsive and biomimetic nanocomposites in targeted delivery, phototherapy and diagnosis. Int J Biol Macromol 2024; 260:129391. [PMID: 38242413 DOI: 10.1016/j.ijbiomac.2024.129391] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
The nanotechnology is an interdisciplinary field that has become a hot topic in cancer therapy. Metal-organic frameworks (MOFs) are porous materials and hybrid composites consisted of organic linkers and metal cations. Despite the wide application of MOFs in other fields, the potential of MOFs for purpose of cancer therapy has been revealed by the recent studies. High surface area and porosity, significant drug loading and encapsulation efficiency are among the benefits of using MOFs in drug delivery. MOFs can deliver genes/drugs with selective targeting of tumor cells that can be achieved through functionalization with ligands. The photosensitizers and photo-responsive nanostructures including carbon dots and gold nanoparticles can be loaded in/on MOFs to cause phototherapy-mediated tumor ablation. The immunogenic cell death induction and increased infiltration of cytotoxic CD8+ and CD4+ T cells can be accelerated by MOF platforms in providing immunotherapy of tumor cells. The stimuli-responsive MOF platforms responsive to pH, redox, enzyme and ion can accelerate release of therapeutics in tumor site. Moreover, MOF nanocomposites can be modified ligands and green polymers to improve their selectivity and biocompatibility for cancer therapy. The application of MOFs for the detection of cancer-related biomarkers can participate in the early diagnosis of patients.
Collapse
Affiliation(s)
- Beixu Li
- School of Policing Studies, Shanghai University of Political Science and Law, Shanghai 201701, China; Shanghai Fenglin Forensic Center, Shanghai 200231, China; State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Department of Pathology, University of Maryland, Baltimore, MD 21201, USA
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, 155 North Nanjing St, Shenyang 110001, China.
| |
Collapse
|
29
|
Su Y, Lv M, Huang Z, An N, Chen Y, Wang H, Li Z, Wu S, Ye F, Shen J, Li A. Defect engineering to tailor structure-activity relationship in biodegradable nanozymes for tumor therapy by dual-channel death strategies. J Control Release 2024; 367:557-571. [PMID: 38301929 DOI: 10.1016/j.jconrel.2024.01.066] [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: 11/11/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Pursuing biodegradable nanozymes capable of equipping structure-activity relationship provides new perspectives for tumor-specific therapy. A rapidly degradable nanozymes can address biosecurity concerns. However, it may also reduce the functional stability required for sustaining therapeutic activity. Herein, the defect engineering strategy is employed to fabricate Pt-doping MoOx (PMO) redox nanozymes with rapidly degradable characteristics, and then the PLGA-assembled PMO (PLGA@PMO) by microfluidics chip can settle the conflict between sustaining therapeutic activity and rapid degradability. Density functional theory describes that Pt-doping enables PMO nanozymes to exhibit an excellent multienzyme-mimicking catalytic activity originating from synergistic catalysis center construction with the interaction of Pt substitution and oxygen vacancy defects. The peroxidase- (POD), oxidase- (OXD), glutathione peroxidase- (GSH-Px), and catalase- (CAT) mimicking activities can induce robust ROS output and endogenous glutathione depletion under tumor microenvironment (TME) response, thereby causing ferroptosis in tumor cells by the accumulation of lipid peroxide and inactivation of glutathione peroxidase 4. Due to the activated surface plasmon resonance effect, the PMO nanozymes can cause hyperthermia-induced apoptosis through 1064 nm laser irradiation, and augment multienzyme-mimicking catalytic activity. This work represents a potential biological application for the development of therapeutic strategy for dual-channel death via hyperthermia-augmented enzyme-mimicking nanocatalytic therapy.
Collapse
Affiliation(s)
- Yutian Su
- School of Chemistry and Chemical Engineering, MOE Key Laboratory of High Performance Polymer Materials and Technology, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou 510120, China
| | - Mengdi Lv
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, Nanjing Normal University, Nanjing 210046, China
| | - Zheng Huang
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Nannan An
- School of Chemistry and Chemical Engineering, MOE Key Laboratory of High Performance Polymer Materials and Technology, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Yi Chen
- School of Chemistry and Chemical Engineering, MOE Key Laboratory of High Performance Polymer Materials and Technology, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China
| | - Haoru Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou 510120, China
| | - Zhengtu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou 510120, China
| | - Shishan Wu
- School of Chemistry and Chemical Engineering, MOE Key Laboratory of High Performance Polymer Materials and Technology, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China.
| | - Feng Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, National Center for Respiratory Medicine, Guangzhou 510120, China
| | - Jian Shen
- School of Chemistry and Chemical Engineering, MOE Key Laboratory of High Performance Polymer Materials and Technology, Nanjing University, 163 Xianlin Avenue, Qixia District, Nanjing 210023, China; National and Local Joint Engineering Research Center of Biomedical Functional Materials, Nanjing Normal University, Nanjing 210046, China
| | - Ao Li
- Department of Ultrasound, Jiangsu Province People's Hospital, Nanjing Medical University First Affiliated Hospital, Nanjing 210029, China
| |
Collapse
|
30
|
Liu J, Li Z, Peng S, Tang J, Zhang D, Ye Y. ONOO - Activatable Fluorescent Sulfur Dioxide Donor for a More Accurate Assessment of Cell Ferroptosis. Anal Chem 2024; 96:2041-2051. [PMID: 38270108 DOI: 10.1021/acs.analchem.3c04565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Ferroptosis is critical in the treatment of tumor therapies. Thus, monitoring reactive oxygen species (ROS) is of great significance for accurate assessment in ferroptosis without any interference. However, current probes for monitoring ROS during ferroptosis suffer from a drawback in that the probes consume ROS during detection, which inhibits the ferroptosis process and thus affects the accuracy and effectiveness of monitoring the process of ferroptosis. Herein, a new fluorescent donor probe, TFMU-SO2D, with the combination of the moiety of the SO2 donor is designed and synthesized by introducing the aryl boronate moieties that could give it the ability to effectively recognize ONOO-. The released SO2 could consume excess glutathione and regulate oxidative stress by elevating ROS levels, which would offset the ROS depletion by TFMU-SO2D and ensure accuracy in monitoring the ferroptosis process. The experimental results demonstrated that TFMU-SO2D possessed satisfactory performance for monitoring ONOO- as well as simultaneously releasing SO2 in oxidative stress stimulated by monensin and ferroptosis stimulated by erastin and RSL3. Additionally, the capability of SO2 synergized with ferroptosis to inhibit the viability of cancer cells was demonstrated by the CCK8 assay, which may be due to the fact that SO2 can potentiate ferroptosis cell death by increasing the ROS level. Overall, these combined results indicated that TFMU-SO2D possesses the excellent ability to precisely monitor ONOO- during ferroptosis without interference, which is significant for accurately accessing ferroptosis, cancer treatment, and drug development.
Collapse
Affiliation(s)
- Jianfei Liu
- Phosphorus Chemical Engineering Research Center of Henan Province, the College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Zipeng Li
- Phosphorus Chemical Engineering Research Center of Henan Province, the College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Shuxin Peng
- Phosphorus Chemical Engineering Research Center of Henan Province, the College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Jun Tang
- School of Chemical and Materials Engineering, Xinxiang University, Xinxiang 453003, China
| | - Di Zhang
- Henan Key Laboratory of Grain Quality and Safety Testing, Institute of Quality and Safety for Agro-products, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yong Ye
- Phosphorus Chemical Engineering Research Center of Henan Province, the College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
31
|
Ko MJ, Yoo W, Min S, Zhang YS, Joo J, Kang H, Kim DH. Photonic control of image-guided ferroptosis cancer nanomedicine. Coord Chem Rev 2024; 500:215532. [PMID: 38645709 PMCID: PMC11027759 DOI: 10.1016/j.ccr.2023.215532] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Photonic nanomaterials, characterized by their remarkable photonic tunability, empower a diverse range of applications, including cutting-edge advances in cancer nanomedicine. Recently, ferroptosis has emerged as a promising alternative strategy for effectively killing cancer cells with minimizing therapeutic resistance. Novel design of photonic nanomaterials that can integrate photoresponsive-ferroptosis inducers, -diagnostic imaging, and -synergistic components provide significant benefits to effectively trigger local ferroptosis. This review provides a comprehensive overview of recent advancements in photonic nanomaterials for image-guided ferroptosis cancer nanomedicine, offering insights into their strengths, constraints, and their potential as a future paradigm in cancer treatment.
Collapse
Affiliation(s)
- Min Jun Ko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Woojung Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital Harvard Medical School, Cambridge, MA 02139, USA
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
32
|
Guan Y, Lei H, Xing C, Yan B, Lin B, Yang X, Huang H, Kang Y, Pang J. Multimodal Nanoplatform with ROS Amplification to Overcome Multidrug Resistance in Prostate Cancer via Targeting P-Glycoprotein and Ferroptosis. Adv Healthc Mater 2024; 13:e2301345. [PMID: 37855250 DOI: 10.1002/adhm.202301345] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Chemotherapy remains the most essential treatment for prostate cancer, but multidrug resistance (MDR) contributes to chemotherapy failure and tumor-related deaths. The overexpression of P-glycoprotein (P-gp) is one of the main mechanisms behind MDR. Here, this work reports a multimodal nanoplatform with a reactive oxygen species (ROS) cascade for gas therapy/ferroptosis/chemotherapy in reversing MDR. The nanoplatform disassembles when responding to intracellular ROS and exerts three main functions: First, nitric oxide (NO) targeted delivery can reverse MDR by downregulating P-gp expression and inhibiting mitochondrial function. Second, ferrocene-induced ferroptosis breaks the redox balance in the tumor intracellular microenvironment and synergistically acts against the tumor. Third, the release of paclitaxel (PTX) is precisely controlled in situ in the tumor for chemotherapy that avoids damage to normal tissues. Excitingly, this multimodal nanoplatform is a promising weapon for reversing MDR and may provide a pioneering paradigm for synergetic cancer therapy.
Collapse
Affiliation(s)
- Yupeng Guan
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Hanqi Lei
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Chengyuan Xing
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Binyuan Yan
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Bingbiao Lin
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Xiangwei Yang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Hai Huang
- Department of Urology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, P. R. China
| | - Yang Kang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P. R. China
| |
Collapse
|
33
|
Farzipour S, Zefrei FJ, Bahadorikhalili S, Alvandi M, Salari A, Shaghaghi Z. Nanotechnology Utilizing Ferroptosis Inducers in Cancer Treatment. Anticancer Agents Med Chem 2024; 24:571-589. [PMID: 38275050 DOI: 10.2174/0118715206278427231215111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 01/27/2024]
Abstract
Current cancer treatment options have presented numerous challenges in terms of reaching high efficacy. As a result, an immediate step must be taken to create novel therapies that can achieve more than satisfying outcomes in the fight against tumors. Ferroptosis, an emerging form of regulated cell death (RCD) that is reliant on iron and reactive oxygen species, has garnered significant attention in the field of cancer therapy. Ferroptosis has been reported to be induced by a variety of small molecule compounds known as ferroptosis inducers (FINs), as well as several licensed chemotherapy medicines. These compounds' low solubility, systemic toxicity, and limited capacity to target tumors are some of the significant limitations that have hindered their clinical effectiveness. A novel cancer therapy paradigm has been created by the hypothesis that ferroptosis induced by nanoparticles has superior preclinical properties to that induced by small drugs and can overcome apoptosis resistance. Knowing the different ideas behind the preparation of nanomaterials that target ferroptosis can be very helpful in generating new ideas. Simultaneously, more improvement in nanomaterial design is needed to make them appropriate for therapeutic treatment. This paper first discusses the fundamentals of nanomedicine-based ferroptosis to highlight the potential and characteristics of ferroptosis in the context of cancer treatment. The latest study on nanomedicine applications for ferroptosis-based anticancer therapy is then highlighted.
Collapse
Affiliation(s)
- Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali Zefrei
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahadorikhalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, 43007, Tarragona, Spain
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Shaghaghi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
34
|
Su G, Xu H, Zhou F, Gong X, Tan S, He Y. Boosting Reactive Oxygen Species Generation with a Dual-Catalytic Nanomedicine for Enhanced Tumor Nanocatalytic Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:59175-59188. [PMID: 38095444 DOI: 10.1021/acsami.3c13882] [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: 12/28/2023]
Abstract
Generating lethal reactive oxygen species (ROS) within tumors by nanocatalytic medicines is an advanced strategy for tumor-specific therapy in recent years. Nevertheless, the low yield of ROS restrains its therapeutic efficiency. Herein, a dual-catalytic nanomedicine based on tumor microenvironment (TME)-responsive liposomal nanosystem co-delivering CuO2 and dihydroartemisinin (DHA) (LIPSe@CuO2&DHA) is developed to boost ROS generation against tumor. The liposomal nanosystem can degrade in the ROS-overexpressed TME and liberate CuO2 and DHA to initiate Cu-based dual-catalytic ROS generation. Serving as generators of H2O2 and Cu2+, CuO2 can self-produce plenty of toxic hydroxyl radicals via Fenton-like reaction in the acidic TME. Meanwhile, the released Cu2+ can catalyze DHA to generate cytotoxic C-centered radicals. Together, the self-supplied H2O2 and Cu-based dual-catalytic reaction greatly increase the intratumoral level of lethal ROS. Importantly, Cu2+ can decrease the GSH-mediated scavenging effect on the produced ROS via a redox reaction and undergo a Cu2+-to-Cu+ conversion to enhance the Fenton-like reaction, further guaranteeing the high efficiency of ROS generation. Resultantly, LIPSe@CuO2&DHA induces remarkable cancer cell death and tumor growth inhibition, which may present a promising nanocatalytic medicine for cancer therapy.
Collapse
Affiliation(s)
- Guoting Su
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| | - Hui Xu
- Institute of Super-Microstructure and Ultrafast Process in Advanced Materials, School of Physics and Electronics, Central South University, Changsha 410083, Hunan, China
| | - FangFang Zhou
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Xiyu Gong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Yongju He
- School of Materials Science and Engineering, Central South University, Changsha 410083, Hunan, China
| |
Collapse
|
35
|
Ding X, Wang T, Bai S, Yang M, Peng N, Qiu T, Liu Y. A Dual Heat Shock Protein Down-Regulation Strategy Using PDA/Cu/ICG/R Controlled by NIR "Switch" Enhances Mild-Photothermal Therapy Effect. Adv Healthc Mater 2023; 12:e2300929. [PMID: 37300324 DOI: 10.1002/adhm.202300929] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/06/2023] [Indexed: 06/12/2023]
Abstract
The purpose of this study is to down-regulate heat shock proteins and improve the mild photothermal therapy (mild-PTT) effect of polydopamine (PDA) by preparing the nanosystem of Cu2+ and indocyanine green (ICG)-loaded PDA nanospheres with surface modification of integrin-targeted cyclic peptide (cRGD) (PDA/Cu/ICG/R), which can limit ATP synthesis through the double mitochondrial destruction pathway. In vitro and in vivo experiments using PDA/Cu/ICG/R irradiated with an NIR laser demonstrate that when NIR is "OFF," Cu2+ can undergo Fenton-like reaction in tumor cells, producing a large amount of hydroxyl radicals (·OH), which leads to oxidative stress in cells. This oxidative stress can cause mitochondrial oxidative phosphorylation dysfunction, resulting in limited ATP synthesis. When NIR is "ON," mild-PTT can accelerate Cu2+ to produce ·OH. Simultaneously, NIR can activate ICG to produce reactive oxygen species (ROS) storm, amplify intracellular oxidative stress, and continuously damage mitochondria. The biodegradability of PDA greatly reduces the risk of toxicity caused by long-term retention of PDA/Cu/ICG/R in organisms. Finally, the improvement of the mild-PTT effect of PDA is successfully achieved through the double mitochondrial destruction pathway of Cu2+ and ICG controlled by NIR "switch."
Collapse
Affiliation(s)
- Xin Ding
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Shiwei Bai
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Mian Yang
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
| | - Na Peng
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
- Belt and Road Joint Laboratory on Measurement and Control Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
| | - Yi Liu
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province & Institute of Advanced Materials and Nanotechnology, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, 430081, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan, 430023, P. R. China
| |
Collapse
|
36
|
Qiao L, Li X, Wei C, Li Z, Han S, Cheng D. A chemodynamic nanoenzyme with highly efficient Fenton reaction for cancer therapy. Biomed Mater 2023; 18:055024. [PMID: 37567189 DOI: 10.1088/1748-605x/acef87] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/10/2023] [Indexed: 08/13/2023]
Abstract
Chemodynamic therapy (CDT) is a rising technology for cancer therapy by converting intracellular hydrogen peroxide (H2O2) into hydroxyl radical (•OH) via transition-metal-containing nanoparticles (NPs) catalysis reaction (i.e. Fenton reaction) to kill tumor cells. Highly efficient Fenton reaction and favorable delivery of the catalytic NPs 'nanoenzyme' are the key for successful treatment of cancer. In this work, we developed a novel nanoenzyme MnFe2O4@GFP forin vitroandin vivoantitumor therapy. A new MnFe2O4nanoparticle containing two transition-metal-element Fe and Mn was synthesized for enhanced Fenton reaction and used to co-deliver protein with high biocompatibility through post-modification with dopamine polymerization, green fluorescent protein adsorption, and PEG coating. The enrichment of H2O2and glutathione (GSH) in tumor tissue provided a favorable microenvironment forin situgeneration of toxic free radicals. Fe3+and GSH triggered a redox reaction to produce Fe2+, which in turn catalyzed H2O2into •OH, with the consumption of antioxidant GSH. By combining Fe3+with another catalyzer, the catalytic efficiency of the nanoenzyme were greatly improved. Consequently, the nanoenzyme showed efficient antitumor ability bothin vitroandin vivo. Thus, the multifunctional CDT nanoenzyme platform shows great promising for antitumor therapy through the combination of catalyzers Fe3+and Mn2+and codelivery of protein cargo.
Collapse
Affiliation(s)
- Lihong Qiao
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
- Department of Obstetrics and Gynecology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, People's Republic of China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, People's Republic of China
| | - Xiaoxia Li
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Chuanqi Wei
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| | - Zhongjun Li
- Department of Obstetrics and Gynecology, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, People's Republic of China
- Dongguan Key Laboratory of Major Diseases in Obstetrics and Gynecology, Dongguan, People's Republic of China
| | - Shisong Han
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
- Zhuhai Institute of Translational Medicine, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, People's Republic of China
| | - Du Cheng
- Key Laboratory for Polymeric Composite & Functional Materials of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| |
Collapse
|
37
|
Rao Z, Xia Y, Jia Q, Zhu Y, Wang L, Liu G, Liu X, Yang P, Ning P, Zhang R, Zhang X, Qiao C, Wang Z. Iron-based metal-organic framework co-loaded with buthionine sulfoximine and oxaliplatin for enhanced cancer chemo-ferrotherapy via sustainable glutathione elimination. J Nanobiotechnology 2023; 21:265. [PMID: 37563614 PMCID: PMC10416514 DOI: 10.1186/s12951-023-01998-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND Emerging ferroptosis-driven therapies based on nanotechnology function either by increasing intracellular iron level or suppressing glutathione peroxidase 4 (GPX4) activity. Nevertheless, the therapeutic strategy of simultaneous iron delivery and GPX4 inhibition remains challenging and has significant scope for improvement. Moreover, current nanomedicine studies mainly use disulfide-thiol exchange to deplete glutathione (GSH) for GPX4 inactivation, which is unsatisfactory because of the compensatory effect of continuous GSH synthesis. METHODS In this study, we design a two-in-one ferroptosis-inducing nanoplatform using iron-based metal-organic framework (MOF) that combines iron supply and GPX4 deactivation by loading the small molecule buthionine sulfoxide amine (BSO) to block de novo GSH biosynthesis, which can achieve sustainable GSH elimination and dual ferroptosis amplification. A coated lipid bilayer (L) can increase the stability of the nanoparticles and a modified tumor-homing peptide comprising arginine-glycine-aspartic acid (RGD/R) can achieve tumor-specific therapies. Moreover, as a decrease in GSH can alleviate resistance of cancer cells to chemotherapy drugs, oxaliplatin (OXA) was also loaded to obtain BSO&OXA@MOF-LR for enhanced cancer chemo-ferrotherapy in vivo. RESULTS BSO&OXA@MOF-LR shows a robust tumor suppression effect and significantly improved the survival rate in 4T1 tumor xenograft mice, indicating a combined effect of dual amplified ferroptosis and GSH elimination sensitized apoptosis. CONCLUSION BSO&OXA@MOF-LR is proven to be an efficient ferroptosis/apoptosis hybrid anti-cancer agent. This study is of great significance for the clinical development of novel drugs based on ferroptosis and apoptosis for enhanced cancer chemo-ferrotherapy.
Collapse
Grants
- Nos. 32101147, 82272159, 91959124, and 32071406 National Natural Science Foundation of China
- Nos. 32101147, 82272159, 91959124, and 32071406 National Natural Science Foundation of China
- Nos. 32101147, 82272159, 91959124, and 32071406 National Natural Science Foundation of China
- 2023-YBSF-362 Key Research and Development Projects of Shaanxi Province
- No. 2022TQ0249 China Postdoctoral Science Foundation
- Nos. QTZX22068, QTZX22070 Fundamental Research Funds for the Central Universities
- Nos. 2022YFB3203800, 2017YFC1309100 and 2017YFA0205200 National Key Research and Development Program of China
- Nos. 2022YFB3203800, 2017YFC1309100 and 2017YFA0205200 National Key Research and Development Program of China
- No. 2023A1515030207 Basic and Applied Basic Research Foundation of Guangdong Province
- 2022TD-52 Innovation Capability Support Program of Shaanxi
- No. CBSKL2022ZDKF14 the Open Project Program of the State Key Laboratory of Cancer Biology
Collapse
Affiliation(s)
- Zhiping Rao
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Yutian Xia
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Qian Jia
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Yutong Zhu
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Lexuan Wang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Guohuan Liu
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Xuelan Liu
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Peng Yang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Pengbo Ning
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Ruili Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Xianghan Zhang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China
| | - Chaoqiang Qiao
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China.
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China.
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, 710126, Shaanxi, China.
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, 710071, Shaanxi, China.
| |
Collapse
|
38
|
Hou G, Qian J, Wang Y, Xu W, Guo M, Li Z, Wang J, Suo A. Hydrazide/Metal/Indocyanine Green Coordinated Nanoplatform for Potentiating Reciprocal Ferroptosis and Immunity against Melanoma. ACS APPLIED MATERIALS & INTERFACES 2023; 15:37143-37156. [PMID: 37498789 DOI: 10.1021/acsami.3c05580] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Ferroptosis holds great potential in cancer treatment, but its efficacy is severely limited by a low Fenton reaction efficacy. Meanwhile, the interactive relationship between Ferroptosis and the PD-1 blockade is still vague. Herein, a hydrazide/Cu/Fe/indocyanine green coordinated nanoplatform (TCFI) is constructed by a hydrazide-metal-sulfonate coordination process. The TCFI nanoplatform exhibits Fenton-/catalase-/glutathione oxidase-like triple activities and accordingly can trigger lipid peroxidation, relieve hypoxia, and downregulate the glutathione/glutathione peroxidase 4 axis, thus achieving positively and negatively dually enhanced Ferroptosis in B16F10 cancer cells. Under near-infrared laser irradiation, the TCFI nanoplatform induces robust immunogenic cancer cell death by elevating the intracellular reactive oxygen species level through synergistic photodynamic therapy/Ferroptosis, which significantly potentiates CD8+ T cell infiltration into tumors and interferon-γ secretion. Moreover, upregulated interferon-γ efficiently inhibits system xc- activity and sensitizes cancer cells to Ferroptosis. Interestingly, the PD-1 blockade may strengthen the reciprocal process. The combination of the TCFI nanoplatform and αPD-1 can eliminate primary tumors and inhibit distant tumor growth, lung metastasis, and tumor recurrence. This study presents a simple and novel coordination strategy to fabricate tumor microenvironment-responsive nanodrugs and highlights the enhancement effect of photodynamic therapy on reciprocal Ferroptosis and antitumor immunity.
Collapse
Affiliation(s)
- Guanghui Hou
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an710049, P. R. China
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou215123, China
| | - Junmin Qian
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an710049, P. R. China
| | - Yaping Wang
- Department of Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an710061, P. R. China
| | - Weijun Xu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an710049, P. R. China
| | - Min Guo
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an710049, P. R. China
| | - Zhi Li
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an710049, P. R. China
| | - Jinlei Wang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an710049, P. R. China
| | - Aili Suo
- Department of Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an710061, P. R. China
| |
Collapse
|
39
|
Zhang L, Cui S, Ding N, Zhang J, Cui E, Xiang Q, Zhou Z, Sun B, Wang Y, Hong H, Ma Y, Yang D. Tumor-Associated Macrophages Regulating a Polymer Nanoplatform for Synergistic Treatment of Breast Tumors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:34527-34539. [PMID: 37462215 DOI: 10.1021/acsami.3c05497] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Tumor-associated macrophages (TAMs) play a critical role in tumor progression and metastasis. Modulation of TAM polarization is one of the most effective strategies to change the immunosuppressive tumor microenvironment (TME). In this study, organic polymer nanoparticles (CPHT) were prepared using hyaluronic acid (HA)-conjugated disulfide-bonded polyethylene imide (PEIS) as a carrier through a self-assembly strategy. These nanoparticles were modified by transferrin (Tf) and loaded with chlorin e6 (Ce6). The results showed that CPHT had good dispersion with a particle size of about 30 nm. CPHT gradually disintegrated under the exposure with a high concentration of glutathione (GSH) in tumor cells, proving the possibility for the controlled release of Ce6 and photodynamic therapy. An in vitro test showed that the uptake of CPHT in tumor cells was mediated by both HA and Tf, indicating the active tumor-targeting capacity of CPHT. CPHT significantly downregulated the ratio of CD206/CD86 and triggered the upregulation of immune factors such as TNF-α and iNOS, suggesting the repolarization of TAMs. We also found that CPHT effectively induced ferroptosis in tumor cells through lipid peroxide accumulation, GSH depletion, and downregulation of lipid peroxidase (GPX4) expression. Animal experiments confirmed that CPHT not only effectively inhibited the growth of tumors in situ but also significantly decelerated the growth of the distal tumor. Elevated levels of CD86 and IFN-γ and decreased expression of CD206 were observed at the tumor sites post CPHT treatment. These results confirmed the value of CPHT as a multifunctional nanoplatform that can tune the TME and provide new hope for tumor treatment.
Collapse
Affiliation(s)
- Li Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Shuai Cui
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Ning Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Jing Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Enna Cui
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Qian Xiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Zhenghao Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Bo Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Yinan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210093, PR China
| | - Yunsu Ma
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
- Jiangsu Yuanlong Hospital Management Co. LTD, Xuzhou, Jiangsu 221000, PR China
| | - Dongzhi Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| |
Collapse
|
40
|
Turrina C, Klassen A, Milani D, Rojas-González DM, Ledinski G, Auer D, Sartori B, Cvirn G, Mela P, Berensmeier S, Schwaminger SP. Superparamagnetic iron oxide nanoparticles for their application in the human body: Influence of the surface. Heliyon 2023; 9:e16487. [PMID: 37274707 PMCID: PMC10238907 DOI: 10.1016/j.heliyon.2023.e16487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Iron oxide nanoparticles (IONs) are of great interest in nanomedicine for imaging, drug delivery, or for hyperthermia treatment. Although many research groups have focused on the synthesis and application of IONs in nanomedicine, little is known about the influence of the surface properties on the particles' behavior in the human body. This study analyzes the impact of surface coatings (dextran, polyvinyl alcohol, polylactide-co-glycolide) on the nanoparticles' cytocompatibility, agglomeration, degradation, and the resulting oxidative stress induced by the particle degradation. All particles, including bare IONs (BIONs), are highly cytocompatible (>70%) and show no significant toxicity towards smooth muscle cells. Small-angle X-ray scattering profiles visualize the aggregation behavior of nanoparticles and yield primary particle sizes of around 20 nm for the investigated nanoparticles. A combined experimental setup of dynamic light scattering and phenanthroline assay was used to analyze the long-term agglomeration and degradation profile of IONs in simulated body fluids, allowing fast screening of multiple candidates. All particles degraded in simulated endosomal and lysosomal fluid, confirming the pH-dependent dissolution. The degradation rate decreased with the shrinking size of particles leading to a plateau. The fastest Fe2+ release could be measured for the polyvinyl-coated IONs. The analytical setup is ideal for a quick preclinical study of IONs, giving often neglected yet crucial information about the behavior and toxicity of nanoparticles in the human body. Moreover, this study allows for the development and evaluation of novel ferroptosis-inducing agents.
Collapse
Affiliation(s)
- Chiara Turrina
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Germany
| | - Anna Klassen
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Germany
| | - Davide Milani
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Germany
| | - Diana M. Rojas-González
- Chair of Medical Materials and Implants, TUM School of Engineering and Design, Munich Institute of Biomedical Engineering, Technical University of Munich, Germany
| | - Gerhard Ledinski
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Doris Auer
- Division of Medical Physics and Biophysics, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Barbara Sartori
- Institute of Inorganic Chemistry, Graz University of Technology, Stremayrgasse 9/IV, Graz, 8010, Austria
| | - Gerhard Cvirn
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Petra Mela
- Chair of Medical Materials and Implants, TUM School of Engineering and Design, Munich Institute of Biomedical Engineering, Technical University of Munich, Germany
| | - Sonja Berensmeier
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Germany
| | - Sebastian P. Schwaminger
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Germany
- Division of Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Austria
- BioTechMed-Graz, Austria
| |
Collapse
|
41
|
Zhu R, Cai M, Fu T, Yin D, Peng H, Liao S, Du Y, Kong J, Ni J, Yin X. Fe-Based Metal Organic Frameworks (Fe-MOFs) for Bio-Related Applications. Pharmaceutics 2023; 15:1599. [PMID: 37376050 DOI: 10.3390/pharmaceutics15061599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Metal-organic frameworks (MOFs) are porous materials composed of metal ions and organic ligands. Due to their large surface area, easy modification, and good biocompatibility, MOFs are often used in bio-related fields. Fe-based metal-organic frameworks (Fe-MOFs), as important types of MOF, are favored by biomedical researchers for their advantages, such as low toxicity, good stability, high drug-loading capacity, and flexible structure. Fe-MOFs are diverse and widely used. Many new Fe-MOFs have appeared in recent years, with new modification methods and innovative design ideas, leading to the transformation of Fe-MOFs from single-mode therapy to multi-mode therapy. In this paper, the therapeutic principles, classification, characteristics, preparation methods, surface modification, and applications of Fe-MOFs in recent years are reviewed to understand the development trends and existing problems in Fe-MOFs, with the view to provide new ideas and directions for future research.
Collapse
Affiliation(s)
- Rongyue Zhu
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mengru Cai
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tingting Fu
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongge Yin
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hulinyue Peng
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shilang Liao
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuji Du
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiahui Kong
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
42
|
Naletova I, Tomasello B, Attanasio F, Pleshkan VV. Prospects for the Use of Metal-Based Nanoparticles as Adjuvants for Local Cancer Immunotherapy. Pharmaceutics 2023; 15:1346. [PMID: 37242588 PMCID: PMC10222518 DOI: 10.3390/pharmaceutics15051346] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Immunotherapy is among the most effective approaches for treating cancer. One of the key aspects for successful immunotherapy is to achieve a strong and stable antitumor immune response. Modern immune checkpoint therapy demonstrates that cancer can be defeated. However, it also points out the weaknesses of immunotherapy, as not all tumors respond to therapy and the co-administration of different immunomodulators may be severely limited due to their systemic toxicity. Nevertheless, there is an established way through which to increase the immunogenicity of immunotherapy-by the use of adjuvants. These enhance the immune response without inducing such severe adverse effects. One of the most well-known and studied adjuvant strategies to improve immunotherapy efficacy is the use of metal-based compounds, in more modern implementation-metal-based nanoparticles (MNPs), which are exogenous agents that act as danger signals. Adding innate immune activation to the main action of an immunomodulator makes it capable of eliciting a robust anti-cancer immune response. The use of an adjuvant has the peculiarity of a local administration of the drug, which positively affects its safety. In this review, we will consider the use of MNPs as low-toxicity adjuvants for cancer immunotherapy, which could provide an abscopal effect when administered locally.
Collapse
Affiliation(s)
- Irina Naletova
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, V.le Andrea Doria 6, 95125 Catania, Italy
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| |
Collapse
|
43
|
He H, Du L, Xue H, An Y, Zeng K, Huang H, He Y, Zhang C, Wu J, Shuai X. Triple Tumor Microenvironment-Responsive Ferroptosis Pathways Induced by Manganese-Based Imageable Nanoenzymes for Enhanced Breast Cancer Theranostics. SMALL METHODS 2023:e2300230. [PMID: 37096886 DOI: 10.1002/smtd.202300230] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Previous studies have found that activated CD8+ T cells secrete elevated levels of interferon-gamma (IFN-γ) to trigger ferroptosis in tumor cells. However, IFN-γ-mediated ferroptosis is induced at low levels in tumor cells because of the limited IFN-γ secreted by CD8+ T cells in the immunosuppressive tumor microenvironment. Recent studies have shown that manganese ion can activate the cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) synthase/stimulator of interferon genes (cGAS-STING) pathway and support adaptive immune responses against tumors, which enhances the level of tumor-infiltrating CD8+ T cells. Therefore, tumor microenvironment-responsive Mn-based nanoenzymes (Mn-based NEs) that activated the cGAS-STING pathway are designed to amplify immune-driven ferroptosis. The multifunctional all-in-one nanoplatform is simply and mildly synthesized by the coordination between Mn3+ ions and 3,3'-dithiodipropionic acid. After intracellular delivery, each component of Mn-based NEs exerts its function. That is, glutathione is depleted through disulfide-thiol exchange and redox pair of Mn3+ /Mn2+ , a hydroxyl radical (·OH) is generated via the Fenton-like reaction to cause ferroptosis, and Mn2+ augments cGAS-STING activity to boost immune-driven ferroptosis. In addition, ferroptosis amplifies Mn2+ -induced immunogenic cell death and initiates the antitumor immune "closed loop" along with immune-driven ferroptosis. Notably, this multifunctional nanoplatform is effective in killing both primary and distant tumors.
Collapse
Affiliation(s)
- Haozhe He
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Lihua Du
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Hongman Xue
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yongcheng An
- Laboratory of Interventional Radiology, Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Kejing Zeng
- Department of Endocrinology, Department of Diabetes and Obesity Reversal Research Centre, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Huaping Huang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jun Wu
- RNA Biomedical Institute, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511400, China
- Department of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| |
Collapse
|
44
|
Tkachenko A, Onishchenko A, Myasoedov V, Yefimova S, Havranek O. Assessing regulated cell death modalities as an efficient tool for in vitro nanotoxicity screening: a review. Nanotoxicology 2023; 17:218-248. [PMID: 37083543 DOI: 10.1080/17435390.2023.2203239] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Nanomedicine is a fast-growing field of nanotechnology. One of the major obstacles for a wider use of nanomaterials for medical application is the lack of standardized toxicity screening protocols for assessing the safety of newly synthesized nanomaterials. In this review, we focus on less frequently studied nanomaterials-induced regulated cell death (RCD) modalities, including eryptosis, necroptosis, pyroptosis, and ferroptosis, as a tool for in vitro nanomaterials safety evaluation. We summarize the latest insights into the mechanisms that mediate these RCDs in response to nanomaterials exposure. Comprehensive data from reviewed studies suggest that ROS (reactive oxygen species) overproduction and ROS-mediated pathways play a central role in nanomaterials-induced RCDs activation. On the other hand, studies also suggest that individual properties of nanomaterials, including size, shape, or surface charge, could determine specific toxicity pathways with consequent RCD induction as well. We anticipate that the evaluation of RCDs can become one of the mechanism-based screening methods in nanotoxicology. In addition to the toxicity assessment, evaluation of necroptosis-, pyroptosis-, and ferroptosis-promoting capacity of nanomaterials could simultaneously provide useful information for specific medical applications as could be their anti-tumor potential. Moreover, a detailed understanding of molecular mechanisms driving nanomaterials-mediated induction of immunogenic RCDs will substantially aid novel anti-tumor nanodrugs development.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Anatolii Onishchenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Valeriy Myasoedov
- Department of Medical Biology, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Svetlana Yefimova
- Institute for Scintillation Materials, National Academy of Sciences of Ukraine, Kharkiv, Ukraine
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
45
|
Lee J, Roh JL. Targeting GPX4 in human cancer: Implications of ferroptosis induction for tackling cancer resilience. Cancer Lett 2023; 559:216119. [PMID: 36893895 DOI: 10.1016/j.canlet.2023.216119] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/25/2023] [Accepted: 03/07/2023] [Indexed: 03/09/2023]
Abstract
Cancer metabolic alterations have been emphasized to protect cancer cells from cell death. The metabolic reprogramming toward a mesenchymal state makes cancer cells resistant to therapy but vulnerable to ferroptosis induction. Ferroptosis is a new form of regulated cell death based on the iron-dependent accumulation of excessive lipid peroxidation. Glutathione peroxidase 4 (GPX4) is the core regulator of ferroptosis by detoxifying cellular lipid peroxidation using glutathione as a cofactor. GPX4 synthesis requires selenium incorporation into the selenoprotein through isopentenylation and selenocysteine tRNA maturation. GPX4 synthesis and expression can be regulated by multiple levels of its transcription, translation, posttranslational modifications, and epigenetic modifications. Targeting GPX4 in cancer may be a promising strategy for effectively inducing ferroptosis and killing therapy-resistant cancer. Several pharmacological therapeutics targeting GPX4 have been developed constantly to activate ferroptosis induction in cancer. The potential therapeutic index of GPX4 inhibitors remains to be tested with thorough examinations of their safety and adverse effects in vivo and clinical trials. Many papers have been published continuously in recent years, requiring state-of-the-art updates in targeting GPX4 in cancer. Herein, we summarize targeting the GPX4 pathway in human cancer, which leads to implications of ferroptosis induction for tackling cancer resilience.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Seongnam, Republic of Korea.
| |
Collapse
|
46
|
Xie Z, Zhou Q, Qiu C, Zhu D, Li K, Huang H. Inaugurating a novel adjuvant therapy in urological cancers: Ferroptosis. CANCER PATHOGENESIS AND THERAPY 2023; 1:127-140. [PMID: 38328400 PMCID: PMC10846326 DOI: 10.1016/j.cpt.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 02/09/2024]
Abstract
Ferroptosis, a distinctive form of programmed cell death, is involved in numerous diseases with specific characteristics, including certain cell morphology, functions, biochemistry, and genetics, that differ from other forms of programmed cell death, such as apoptosis. Many studies have explored ferroptosis and its associated mechanisms, drugs, and clinical applications in diseases such as kidney injury, stroke, ischemia-reperfusion injury, and prostate cancer. In this review, we summarize the regulatory mechanisms of some ferroptosis inducers, such as enzalutamide and erastin. These are current research focuses and have already been studied extensively. In summary, this review focuses on the use of ferroptosis induction as a therapeutic strategy for treating tumors of the urinary system.
Collapse
Affiliation(s)
- Zhaoxiang Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Cheng Qiu
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Dingjun Zhu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Kaiwen Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China
| |
Collapse
|
47
|
Fu F, Wang W, Wu L, Wang W, Huang Z, Huang Y, Wu C, Pan X. Inhalable Biomineralized Liposomes for Cyclic Ca 2+-Burst-Centered Endoplasmic Reticulum Stress Enhanced Lung Cancer Ferroptosis Therapy. ACS NANO 2023; 17:5486-5502. [PMID: 36883602 DOI: 10.1021/acsnano.2c10830] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Lung cancer with the highest mortality poses a great threat to human health. Ferroptosis therapy has recently been raised as a promising strategy for lung cancer treatment by boosting the reactive species (ROS) production and lipid peroxidation (LPO) accumulation intracellularly. However, the insufficient intracellular ROS level and the unsatisfactory drug accumulation in lung cancer lesions hamper the efficacy of ferroptosis therapy. Here, an inhalable biomineralized liposome LDM co-loaded with dihydroartemisinin (DHA) and pH-responsive calcium phosphate (CaP) was constructed as a ferroptosis nanoinducer for achieving Ca2+-burst-centered endoplasmic reticulum (ER) stress enhanced lung cancer ferroptosis therapy. Equipped with excellent nebulization properties, about 6.80-fold higher lung lesions drug accumulation than intravenous injection made the proposed inhalable LDM an ideal nanoplatform for lung cancer treatment. The Fenton-like reaction mediated by DHA with peroxide bridge structure could contribute to intracellular ROS production and induce ferroptosis. Assisted by DHA-mediated sarco-/endoplasmic reticulum calcium ATPase (SERCA) inhibition, the initial Ca2+ burst caused by CaP shell degradation triggered the Ca2+-mediated intense ER stress and subsequently induced mitochondria dysfunction to further boost ROS accumulation, which strengthens ferroptosis. The second Ca2+ burst occurred as a result of Ca2+ influx through ferroptotic pores on cell membranes, thus sequentially constructing the lethal "Ca2+ burst-ER stress-ferroptosis" cycle. Consequently, the Ca2+-burst-centered ER stress enhanced ferroptosis process was confirmed as a cell swelling and cell membrane disruption process driven by notable intracellular ROS and LPO accumulation. The proposed LDM showed an encouraging lung retention property and extraordinary antitumor ability in an orthotropic lung tumor murine model. In conclusion, the constructed ferroptosis nanoinducer could be a potential tailored nanoplatform for nebulization-based pulmonary delivery and underscore the application of Ca2+-burst-centered ER stress enhanced lung cancer ferroptosis therapy.
Collapse
Affiliation(s)
- Fangqin Fu
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Linjing Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China
| | - Wenhua Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, P. R. China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, P. R. China
| |
Collapse
|
48
|
Pei Z, Lei H, Cheng L. Bioactive inorganic nanomaterials for cancer theranostics. Chem Soc Rev 2023; 52:2031-2081. [PMID: 36633202 DOI: 10.1039/d2cs00352j] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bioactive materials are a special class of biomaterials that can react in vivo to induce a biological response or regulate biological functions, thus achieving a better curative effect than traditional inert biomaterials. For cancer theranostics, compared with organic or polymer nanomaterials, inorganic nanomaterials possess unique physical and chemical properties, have stronger mechanical stability on the basis of maintaining certain bioactivity, and are easy to be compounded with various carriers (polymer carriers, biological carriers, etc.), so as to achieve specific antitumor efficacy. After entering the nanoscale, due to the nano-size effect, high specific surface area and special nanostructures, inorganic nanomaterials exhibit unique biological effects, which significantly influence the interaction with biological organisms. Therefore, the research and applications of bioactive inorganic nanomaterials in cancer theranostics have attracted wide attention. In this review, we mainly summarize the recent progress of bioactive inorganic nanomaterials in cancer theranostics, and also introduce the definition, synthesis and modification strategies of bioactive inorganic nanomaterials. Thereafter, the applications of bioactive inorganic nanomaterials in tumor imaging and antitumor therapy, including tumor microenvironment (TME) regulation, catalytic therapy, gas therapy, regulatory cell death and immunotherapy, are discussed. Finally, the biosafety and challenges of bioactive inorganic nanomaterials are also mentioned, and their future development opportunities are prospected. This review highlights the bioapplication of bioactive inorganic nanomaterials.
Collapse
Affiliation(s)
- Zifan Pei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Huali Lei
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| |
Collapse
|
49
|
Chen Y, Yao Z, Liu P, Hu Q, Huang Y, Ping L, Zhang F, Tang H, Wan T, Ping Y, Li B. A self-assembly nano-prodrug for triple-negative breast cancer combined treatment by ferroptosis therapy and chemotherapy. Acta Biomater 2023; 159:275-288. [PMID: 36709836 DOI: 10.1016/j.actbio.2023.01.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023]
Abstract
Chemotherapeutics have been recommended as the standard protocol for inoperable patients with triple-negative breast cancer (TNBC) at advanced stage, yet limited success has been achieved in prolonging survival rates by this monotherapy. A major reason for this failure is the chemo-resistance from traditional apoptotic pathways resulting in poor therapeutic effect. Ferroptosis has become a powerful modality of no-apoptotic cell death, which can effectively evade chemo-resistance in apoptotic pathways. Herein, we propose an active-targeting small-molecular self-assembly nano-prodrug for co-delivery of chemotherapeutics (CPT), Ferrocene (Fc) and GPX4 inhibitor (RSL3) to overcome the chemo-resistance from traditional apoptotic pathways. In this nano-prodrug, the disulfide linkage not only serves as a GSH-responsive trigger, but also exhibits a stable self-assembly behavior that forms nanoparticle. Interestingly, the RSL3 can be loaded during this self-assembly process that forms a three-components nano-prodrug. In tumor environment, the high GSH level can disassemble the nano-prodrug to trigger the release of the parent drug, which can improve the therapeutic effect by synergistic effects of ferroptosis and apoptosis. In different TNBC mice models, the nano-prodrug is encapsulated into RGD-modified phospholipid micelles (DSPE-PEG2000-RGD) and exhibits high anti-tumor and anti-metastasis efficacy, especially in orthotopic models. The application of ferroptosis to assist the enhancement of chemotherapeutics may serve as a promising strategy for TNBC treatment. STATEMENT OF SIGNIFICANCE: Chemotherapeutics have been recommended as the standard of care for palliative and adjuvant treatment in patients with triple-negative breast cancer (TNBC), yet limited success has been achieved in prolonging the overall survival of patients by this monotherapy. A major reason for this failure is the chemo-resistance from traditional apoptotic pathways resulting in poor therapeutic effect. Thus, the co-delivery of the apoptosis and ferroptosis drug may overcome or evade the resistance in chemotherapy-induced apoptotic pathways and provide a promising strategy to combat TNBC. In this work, we developed a small-molecular self-assembly nano-prodrug for co-delivery of chemotherapeutics (CPT), Ferrocene (Fc) and ferroptosis resistance inhibitor (RSL3), which could overcome the chemo-resistance and improve the therapeutic effect by synergistic effects of ferroptosis and apoptosis.
Collapse
Affiliation(s)
- Yuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhuo Yao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Peilian Liu
- School of Chemistry and Chemical Engineering, Key Laboratory of Clean Energy Materials Chemistry of Guangdong Higher Education Institutes, Lingnan Normal University, Zhanjiang, Guangdong 524048, China.
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Yong Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Li Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China
| | - Honglin Tang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine Zhejiang University, Hangzhou 310058, China
| | - Tao Wan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China.
| | - Bowen Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
50
|
Elmehrath S, Nguyen HL, Karam SM, Amin A, Greish YE. BioMOF-Based Anti-Cancer Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:953. [PMID: 36903831 PMCID: PMC10005089 DOI: 10.3390/nano13050953] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/19/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
A variety of nanomaterials have been developed specifically for biomedical applications, such as drug delivery in cancer treatment. These materials involve both synthetic and natural nanoparticles and nanofibers of varying dimensions. The efficacy of a drug delivery system (DDS) depends on its biocompatibility, intrinsic high surface area, high interconnected porosity, and chemical functionality. Recent advances in metal-organic framework (MOF) nanostructures have led to the achievement of these desirable features. MOFs consist of metal ions and organic linkers that are assembled in different geometries and can be produced in 0, 1, 2, or 3 dimensions. The defining features of MOFs are their outstanding surface area, interconnected porosity, and variable chemical functionality, which enable an endless range of modalities for loading drugs into their hierarchical structures. MOFs, coupled with biocompatibility requisites, are now regarded as highly successful DDSs for the treatment of diverse diseases. This review aims to present the development and applications of DDSs based on chemically-functionalized MOF nanostructures in the context of cancer treatment. A concise overview of the structure, synthesis, and mode of action of MOF-DDS is provided.
Collapse
Affiliation(s)
- Sandy Elmehrath
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Ha L. Nguyen
- Department of Chemistry University of California—Berkeley, Kavli Energy Nanoscience Institute at UC Berkeley, and Berkeley Global Science Institute, Berkeley, CA 94720, USA
- Joint UAEU−UC Berkeley Laboratories for Materials Innovations, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Sherif M. Karam
- Department of Anatomy, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Amr Amin
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Department of Biology, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Yaser E. Greish
- Department of Chemistry, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Joint UAEU−UC Berkeley Laboratories for Materials Innovations, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| |
Collapse
|