1
|
Zhang L, Fan Y, Yang Z, Wong CY, Yang M. A novel reactive oxygen species nano-amplifier for tumor-targeted photoacoustic imaging and synergistic therapy. J Colloid Interface Sci 2025; 681:331-343. [PMID: 39612665 DOI: 10.1016/j.jcis.2024.11.183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Intracellular redox homeostasis and the type of exogenous Fenton reagent play crucial roles in determining the efficacy of chemodynamic therapy (CDT). Herein, we succeeded for the first time in preparing ultrasmall copper sulfide (CuS) nanodots (1-2 nm)-embedded hollow mesoporous organosilica nanoparticle (HMON), which served as an ideal nanocarrier to load both 3-amino-1,2,4-triazole (3-AT) and disulfiram (DSF) after folate-polyethylene glycol-silane (FA-PEG-Silane) modification. The as-prepared nanoplatform (3-AT/DSF@CuS/HMON-FA, denoted as ADCuSi-FA) was found to regulate intracellular redox homeostasis once internalized by 4T1 cells, showing rapid glutathione (GSH)-responsive 3-AT, DSF and Cu+ ions release. Specifically, 3-AT restrained the endogenous hydrogen peroxide (H2O2) consumption by suppressing catalase (CAT) activity, thereby augmenting hydroxyl radical (OH) generation via Cu+-based Fenton-like reaction. DSF, upon complexation with Cu2+, exhibited enhanced chemotherapeutic efficacy, while the by-product Cu+ ions further boosted the efficacy of CDT. Additionally, CuS nanodots enabled near-infrared-II (NIR-II) photothermal therapy (PTT) and facilitated photoacoustic (PA) imaging, with the ensuing hyperthermia expediting the CDT process. As expected, the tumor growth was dramatically inhibited with PTT/chemotherapy co-synergized CDT. This work offers an innovative paradigm for cooperative cancer treatment as well as new insights into the fabrication of biodegradable inorganic/organic hybrid materials.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China; Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China.
| | - Yadi Fan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Zhe Yang
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Chun-Yuen Wong
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China.
| |
Collapse
|
2
|
Chen W, Huang D, Wu R, Wen Y, Zhong Y, Guo J, Liu A, Lin L. A multi-functional integrated nanoplatform based on a tumor microenvironment-responsive PtAu/MnO 2 cascade nanoreactor with multi-enzymatic activities for multimodal synergistic tumor therapy. J Colloid Interface Sci 2025; 679:957-974. [PMID: 39486234 DOI: 10.1016/j.jcis.2024.10.160] [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: 07/11/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
The utilization or improvement of tumor microenvironment (TME) has become a breakthrough in emerging oncology therapies. To address the limited therapeutic efficacy of single modality, a multi-functional integrated nanoplatform based on a TME-responsive PtAu/MnO2 cascade nanoreactor with multi-enzymatic activities was developed for multimodal synergistic tumor therapy. Benefiting from the slightly acidic environment and high-level glutathione (GSH) in TME, PtAu/MnO2 cascade nanoreactor consumed GSH, followed by the reductive generation of manganese ion (Mn2+) and the release of PtAu nanoparticles (NPs). Then, the multimodal synergistic tumor therapy was activated as follows. First, GSH depletion inhibited the activity of glutathione peroxidase 4 and led to the accumulation of lipid peroxidation, thereby inducing tumor cell ferroptosis. Second, PtAu NPs exhibited catalase-like, glucose oxidase-like and nicotinamide adenine dinucleotide (NADH) oxidase-like activities, which generated oxygen for the cascade reaction to alleviate hypoxia and further depleted glucose, NADH and adenosine triphosphate, leading to the inhibition of tumor cell proliferation via starvation therapy. Third, the production of reactive oxygen species by the oxidase- and peroxidase-like activities of PtAu NPs and the Fenton-like reaction of Mn2+ simultaneously induced tumor cell apoptosis via chemodynamic therapy. Briefly, the in vitro and in vivo results confirmed that the multi-functional integrated nanoplatform based on a PtAu/MnO2 cascade nanoreactor with five nanozyme activities demonstrated outstanding biocompatibility and greater inhibition of tumor growth via synergistic ferroptosis/starvation therapy/apoptosis.
Collapse
Affiliation(s)
- Wenxin Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Dandan Huang
- Department of Pharmacy, Fujian Children's Hospital, Fuzhou, Fujian 350000, China
| | - Ruimei Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yujuan Wen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yu Zhong
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jianpeng Guo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Ailin Liu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Liqing Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
3
|
Gao W, Song Y, Wu F, Xu S, Liu B, Zeng L, Zheng E, Song H, Zhang Q. Tumor-Targeted Metal-Organic Framework for Improved Photodynamic Therapy and Inhibited Tumor Metastasis in Melanoma. ACS APPLIED MATERIALS & INTERFACES 2024; 16:69769-69788. [PMID: 39652639 DOI: 10.1021/acsami.4c18058] [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: 12/20/2024]
Abstract
Tumor hypoxia and elevated intracellular glutathione (GSH) levels significantly compromise the effectiveness of photodynamic therapy (PDT) in treating melanoma. In this study, we synthesized positively charged nanoparticles through a self-assembly method, incorporating photosensitizer verteporfin (VER), mitochondrial respiratory inhibitor atovaquone (ATO), and Fe3+. Subsequently, the nanoparticles were modified with sodium hyaluronate (HA) to obtain HA-ATO-Fe3+-VER nanoparticles (HAFV NPs). The fabricated HAFV NPs demonstrated excellent stability and in vitro Fenton reaction activity. HA facilitated the cellular internalization of HAFV NPs by targeting CD44 receptors, hence relieving tumor hypoxia through the disruption of the mitochondrial respiratory chain and involvement in the Fenton reaction. Simultaneously, ATO directly impeded the biosynthesis of GSH by diminishing ATP levels, while Fe3+ was supposed to oxidate GSH to GSSG, thereby doubly depleting GSH. The integration of these multiple mechanisms markedly enhanced the PDT efficacy of VER. Following intravenous administration, HAFV NPs preferentially accumulated in tumor tissues with minimal accumulation in the skin, demonstrating favorable biocompatibility in vivo. Furthermore, HAFV NPs effectively inhibited tumor growth and lung metastasis, which presents a promising strategy for melanoma treatment.
Collapse
Affiliation(s)
- Wenhao Gao
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Yutong Song
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Fei Wu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Shiting Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Bin Liu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lingjun Zeng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Enqin Zheng
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Hongtao Song
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Department of Pharmacy, The 900th Hospital of Joint Logistic Support Force PLA, Fuzhou 350025, China
| | - Qian Zhang
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
4
|
Han H, Wang S, Shahbazi MA, Du Y, Zuhorn IS, Li J, Chen J, Chen Y, Bártolo R, Cui W, Santos HA. Local glycolysis-modulating hydrogel microspheres for a combined anti-tumor and anti-metastasis strategy through metabolic trapping strategy. J Control Release 2024; 378:320-333. [PMID: 39689815 DOI: 10.1016/j.jconrel.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 12/12/2024] [Indexed: 12/19/2024]
Abstract
Anti-glycolysis is well-recognized for inhibition of tumor proliferation. However, tumor metabolic heterogeneity confers great challenges in the therapeutic efficacy of glycolysis inhibitors. Here, a metabolic trapping strategy was employed to avoid metabolism heterogeneity in tumors. Unlike usual glycolysis inhibition, the glycolysis level was first promoted. Then excessive metabolite of lactate was transformed into H2O2 and hydroxyl radical by lactate oxidase (LOX) and MIL-101 (Fe) nanoparticles (MF). Finally, the ATP production was inhibited, and the tumor was suppressed by the generation of toxic reactive oxygen species (ROS). We realized this strategy via methacrylated gelatin (GelMA) hydrogel microspheres, co-loaded with metformin (MET) and LOX@MF. The results showed that MET was completely released within 2 h, followed by most LOX@MF released within 72 h. LOX@MF and MET synergistically suppressed tumor proliferation and angiogenesis both in vitro and in vivo. Compared with control, the primary tumor volume was reduced by 75.7 %, and the average number of lung metastasis nodules decreased from 15.5 to 1.0. Regarding the metabolism, higher glycolytic enzymes expressions were observed initially, followed by lower lactate and vascular endothelial growth factor (VEGF), and finally elevated ROS levels. Overall, our study provides new insights to improve metabolism heterogeneity-limited metabolic cancer therapy.
Collapse
Affiliation(s)
- Huijie Han
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China; Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands; Department of Biology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124 P. R. China
| | - Shiqi Wang
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Inge S Zuhorn
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Jiachen Li
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Jie Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Yu Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Raquel Bártolo
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
5
|
Zhang X, An M, Zhang J, Zhao Y, Liu Y. Nano-medicine therapy reprogramming metabolic network of tumour microenvironment: new opportunity for cancer therapies. J Drug Target 2024; 32:241-257. [PMID: 38251656 DOI: 10.1080/1061186x.2024.2309565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 01/23/2024]
Abstract
Metabolic heterogeneity is one of the characteristics of tumour cells. In order to adapt to the tumour microenvironment of hypoxia, acidity and nutritional deficiency, tumour cells have undergone extensive metabolic reprogramming. Metabolites involved in tumour cell metabolism are also very different from normal cells, such as a large number of lactate and adenosine. Metabolites play an important role in regulating the whole tumour microenvironment. Taking metabolites as the target, it aims to change the metabolic pattern of tumour cells again, destroy the energy balance it maintains, activate the immune system, and finally kill tumour cells. In this paper, the regulatory effects of metabolites such as lactate, glutamine, arginine, tryptophan, fatty acids and adenosine were reviewed, and the related targeting strategies of nano-medicines were summarised, and the future therapeutic strategies of nano-drugs were discussed. The abnormality of tumour metabolites caused by tumour metabolic remodelling not only changes the energy and material supply of tumour, but also participates in the regulation of tumour-related signal pathways, which plays an important role in the survival, proliferation, invasion and metastasis of tumour cells. Regulating the availability of local metabolites is a new aspect that affects tumour progress. (The graphical abstract is by Figdraw).
Collapse
Affiliation(s)
- Xiaojie Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Min An
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Juntao Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yumeng Zhao
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Hui Ethnic Medicine Modernization, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
6
|
Liu X, Liu S, Jin X, Liu H, Sun K, Wang X, Li M, Wang P, Chang Y, Wang T, Wang B, Yu XA. An encounter between metal ions and natural products: natural products-coordinated metal ions for the diagnosis and treatment of tumors. J Nanobiotechnology 2024; 22:726. [PMID: 39574109 PMCID: PMC11580416 DOI: 10.1186/s12951-024-02981-9] [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: 05/31/2024] [Accepted: 11/04/2024] [Indexed: 11/25/2024] Open
Abstract
Natural products-coordinated metal ions to form the nanomedicines are in the spotlight for cancer therapy. Some natural products could be coordinated with metal ions forming nanomedicines via simple and green environmental self-assembly, which not only improved the bioavailability of natural products, but also conferred multiple therapeutic modalities and multimodal imaging. On the one hand, in the weak acidity, glutathione (GSH) and hydrogen peroxide (H2O2) overexpression of tumor microenvironment (TME), such carrier-free nanomedicines could be further enhanced the therapeutic effect via optimizing the species of metal ions. On the other hand, nanomedicines could exert the precise treatment of tumor under the guidance of multiple imaging. Hence, this review summarized the research progress in recent years on the application of natural product-coordinated metal ions in cancer therapy. In addition, the prospects and challenges for the application of natural product-coordinated metal ions were discussed, especially how to improve targeting ability and stability and assess the safety of metal ions, so as to facilitate the clinical translation and application of natural product-coordinated metal ions nanomedicines.
Collapse
Affiliation(s)
- Xinyue Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Suyi Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xingyue Jin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Haifan Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Kunhui Sun
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiongqin Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Meifang Li
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Ping Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China
| | - Yanxu Chang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tiejie Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
| | - Bing Wang
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
| | - Xie-An Yu
- NMPA Key Laboratory for Bioequivalence Research of Generic Drug Evaluation, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Chinese Medicine, Shenzhen Institute for Drug Control, Shenzhen, 518057, China.
| |
Collapse
|
7
|
Sun Y, Qin L, Yang Y, Gao J, Zhang Y, Wang H, Wu Q, Xu B, Liu H. Zinc-Based ROS Amplifiers Trigger Cancer Chemodynamic/Ion Interference Therapy Through Self-Cascade Catalysis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402320. [PMID: 38881259 DOI: 10.1002/smll.202402320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/30/2024] [Indexed: 06/18/2024]
Abstract
Nanozyme-mediated chemodynamic therapy has emerged as a promising strategy due to its tumor specificity and controlled catalytic activity. However, the poor efficacy caused by low hydrogen peroxide (H2O2) levels in the tumor microenvironment (TME) poses challenges. Herein, an H2O2 self-supplying nanozyme is constructed through loading peroxide-like active platinum nanoparticles (Pt NPs) on zinc peroxide (ZnO2) (denoted as ZnO2@Pt). ZnO2 releases H2O2 in response to the acidic TME. Pt NPs catalyze the hydroxyl radical generation from H2O2 while reducing the mitigation of oxidative stress by glutathione, serving as a reactive oxygen (ROS) amplifier through self-cascade catalysis. In addition, Zn2+ released from ZnO2 interferes with tumor cell energy supply and metabolism, enabling ion interference therapy to synergize with chemodynamic therapy. In vitro studies demonstrate that ZnO2@Pt induces cellular oxidative stress injury through enhanced ROS generation and Zn2+ release, downregulating ATP and NAD+ levels. In vivo assessment of anticancer effects showed that ZnO2@Pt could generate ROS at tumor sites to induce apoptosis and downregulate energy supply pathways associated with glycolysis, resulting in an 89.7% reduction in tumor cell growth. This study presents a TME-responsive nanozyme capable of H2O2 self-supply and ion interference therapy, providing a paradigm for tumor-specific nanozyme design.
Collapse
Affiliation(s)
- Yun Sun
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Liting Qin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yuhan Yang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Jingzhe Gao
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Yudi Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Hongyu Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Qingyuan Wu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Bolong Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
8
|
Liang S, Wang J, Zhu W, Zhang L. Glutathione-responsive biodegradable nanohybrid for cancer photoacoustic imaging and gas-assisted photothermal therapy. Colloids Surf B Biointerfaces 2024; 245:114205. [PMID: 39241634 DOI: 10.1016/j.colsurfb.2024.114205] [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: 06/09/2024] [Revised: 08/25/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Photothermal therapy (PTT), particularly in the near-infrared-II (NIR-II) range, has attracted widespread attention over the past years. However, the accompanied inflammatory responses can result in undesirable side effects and contribute to treatment ineffectiveness. Herein, we introduced a novel biodegradable nanoplatform (CuS/HMON-PEG) capable of PTT and hydrogen sulfide (H2S) generation, aimed at modulating inflammation for improved cancer treatment outcomes. The embedded ultrasmall copper sulphide (CuS) nanodots (1-2 nm) possessed favorable photoacoustic imaging (PAI) and NIR-II photothermal capabilities, rendering CuS/HMON-PEG an ideal phototheranostic agent. Upon internalization by 4T1 cancer cells, the hollow mesoporous organosilica nanoparticle (HMON) component could react with the overproduced glutathione (GSH) to produce H2S. In addition to the anticipated photothermal tumor ablation and H2S-induced mitochondrial dysfunction, the anti-inflammatory regulation was also been demonstrated by the downregulation of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1beta (IL-1β). More importantly, the modulation of inflammation also promoted wound healing mediated by PTT. This work not only presents a H2S-based nanomodulator to boost NIR-II PTT but also provides insights into the construction of novel organic/inorganic hybrid nanosystems.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jingjing Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Li Zhang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China.
| |
Collapse
|
9
|
Li L, Chen Y, Zhang M, Li S, Feng S, He YQ, Zhang N, Liu Z, Liu M, Wang Q. A hydroxychloroquine platinum(IV) conjugate displaying potent antimetastatic activities by suppressing autophagy to improve the tumor microenvironment. Dalton Trans 2024; 53:13890-13905. [PMID: 39092626 DOI: 10.1039/d4dt01794c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Protective autophagy is a promising target for antitumor drug exploration. A hydroxychloroquine (HCQ) platinum(IV) complex with autophagy suppressing potency was developed, which displayed potent antitumor activities with a TGI rate of 44.2% against 4T1 tumors in vivo and exhibited a rather lower toxicity than cisplatin. Notably, it exhibited satisfactory antimetastatic activities toward lung pulmonary metastasis models with an inhibition rate of 49.6% and was obviously more potent than CDDP, which has an inhibition rate of 21.6%. Mechanism detection revealed that it caused serious DNA damage and upregulated the expression of γ-H2AX and p53. More importantly, the incorporation of an autophagy inhibitor HCQ endowed the platinum(IV) complex with potent autophagy impairing properties by perturbing the lysosomal function in tumor cells, which promoted apoptosis synergistically with DNA injury. Then, the impaired autophagy further led to the suppression of hypoxia and inflammation in the tumor microenvironment by downregulating ERK1/2, HIF-1α, iNOS, caspase1 and COX-2. Adaptive immune response was improved by inhibiting the immune checkpoint PD-L1 and further increasing CD4+ and CD8+ T cells in tumors. Then, tumor metastasis was effectively inhibited by restraining angiogenesis through inhibiting VEGFA, MMP-9, and CD34.
Collapse
Affiliation(s)
- Linming Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Yan Chen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Ming Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Suying Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Yan-Qin He
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| | - Meifeng Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, P. R. China.
| |
Collapse
|
10
|
Wang J, Liu Y, Cui T, Yang H, Lin L. Current progress in the regulation of endogenous molecules for enhanced chemodynamic therapy. Chem Sci 2024; 15:9915-9926. [PMID: 38966366 PMCID: PMC11220580 DOI: 10.1039/d4sc02129k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
Chemodynamic therapy (CDT) is a potential cancer treatment strategy, which relies on Fenton chemistry to transform hydrogen peroxide (H2O2) into highly cytotoxic reactive oxygen species (ROS) for tumor growth suppression. Although overproduced H2O2 in cancerous tissues makes CDT a feasible and specific tumor therapeutic modality, the treatment outcomes of traditional chemodynamic agents still fall short of expectations. Reprogramming cellular metabolism is one of the hallmarks of tumors, which not only supports unrestricted proliferative demands in cancer cells, but also mediates the resistance of tumor cells against many antitumor modalities. Recent discoveries have revealed that various cellular metabolites including H2O2, iron, lactate, glutathione, and lipids have distinct effects on CDT efficiency. In this perspective, we intend to provide a comprehensive summary of how different endogenous molecules impact Fenton chemistry for a deep understanding of mechanisms underlying endogenous regulation-enhanced CDT. Moreover, we point out the current challenges and offer our outlook on the future research directions in this field. We anticipate that exploring CDT through manipulating metabolism will yield significant advancements in tumor treatment.
Collapse
Affiliation(s)
- Jun Wang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Yina Liu
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Tingting Cui
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore Singapore 119074 Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore Singapore 117597 Singapore
| | - Huanghao Yang
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| | - Lisen Lin
- New Cornerstone Science Laboratory, MOE Key Laboratory for Analytical Science of Food Safety and Biology, Engineering Technology Research Center on Reagent and Instrument for Rapid Detection of Product Quality and Food Safety in Fujian Province, College of Chemistry, Fuzhou University Fuzhou 350108 China
| |
Collapse
|
11
|
Huang S, Xu Z, Zhi W, Li Y, Hu Y, Zhao F, Zhu X, Miao M, Jia Y. pH/GSH dual-responsive nanoparticle for auto-amplified tumor therapy of breast cancer. J Nanobiotechnology 2024; 22:324. [PMID: 38858692 PMCID: PMC11163783 DOI: 10.1186/s12951-024-02588-0] [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: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
Breast cancer remains a malignancy that poses a serious threat to human health worldwide. Chemotherapy is one of the most widely effective cancer treatments in clinical practice, but it has some drawbacks such as poor targeting, high toxicity, numerous side effects, and susceptibility to drug resistance. For auto-amplified tumor therapy, a nanoparticle designated GDTF is prepared by wrapping gambogic acid (GA)-loaded dendritic porous silica nanoparticles (DPSNs) with a tannic acid (TA)-Fe(III) coating layer. GDTF possesses the properties of near-infrared (NIR)-enhanced and pH/glutathione (GSH) dual-responsive drug release, photothermal conversion, GSH depletion and hydroxyl radical (·OH) production. When GDTF is exposed to NIR laser irradiation, it can effectively inhibit cell proliferation and tumor growth both in vitro and in vivo with limited toxicity. This may be due to the synergistic effect of enhanced tumor accumulation, and elevated reactive oxygen species (ROS) production, GSH depletion, and TrxR activity reduction. This study highlights the enormous potential of auto-amplified tumor therapy.
Collapse
Affiliation(s)
- Shengnan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China.
| | - Zhiling Xu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Weiwei Zhi
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Yijing Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China
| | - Fengqin Zhao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China
| | - Xiali Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| | - Yongyan Jia
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| |
Collapse
|
12
|
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
|
13
|
Wang C, Zhou H, Kurboniyon MS, Tang Y, Cai Z, Ning S, Zhang L, Liang X. Chemodynamic PtMn Nanocubes for Effective Photothermal ROS Storm a Key Anti-Tumor Therapy in-vivo. Int J Nanomedicine 2024; 19:5045-5056. [PMID: 38832334 PMCID: PMC11146616 DOI: 10.2147/ijn.s455936] [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: 01/25/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
Background Chemodynamic therapy (CDT) is a new treatment approach that is triggered by endogenous stimuli in specific intracellular conditions for generating hydroxyl radicals. However, the efficiency of CDT is severely limited by Fenton reaction agents and harsh reaction conditions. Methods Bimetallic PtMn nanocubes were rationally designed and simply synthesized through a one-step high-temperature pyrolysis process by controlling both the nucleation process and the subsequent crystal growth stage. The polyethylene glycol was modified to enhance biocompatibility. Results Benefiting from the alloying of Pt nanocubes with Mn doping, the structure of the electron cloud has changed, resulting in different degrees of the shift in electron binding energy, resulting in the increasing of Fenton reaction activity. The PtMn nanocubes could catalyze endogenous hydrogen peroxide to toxic hydroxyl radicals in mild acid. Meanwhile, the intrinsic glutathione (GSH) depletion activity of PtMn nanocubes consumed GSH with the assistance of Mn3+/Mn2+. Upon 808 nm laser irradiation, mild temperature due to the surface plasmon resonance effect of Pt metal can also enhance the Fenton reaction. Conclusion PtMn nanocubes can not only destroy the antioxidant system via efficient reactive oxygen species generation and continuous GSH consumption but also propose the photothermal effect of noble metal for enhanced Fenton reaction activity.
Collapse
Affiliation(s)
- Chen Wang
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Hongmei Zhou
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | | | - Yanping Tang
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Zhengmin Cai
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Shufang Ning
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Litu Zhang
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| | - Xinqiang Liang
- Department of Research & Guangxi Cancer Molecular Medicine Engineering Research Center & Guangxi Key Laboratory of Basic and Translational Research for Colorectal Cancer, Guangxi Medical University Cancer Hospital, Nanning, People’s Republic of China
| |
Collapse
|
14
|
Li L, Yue T, Feng J, Zhang Y, Hou J, Wang Y. Recent progress in lactate oxidase-based drug delivery systems for enhanced cancer therapy. NANOSCALE 2024; 16:8739-8758. [PMID: 38602362 DOI: 10.1039/d3nr05952a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Lactate oxidase (LOX) is a natural enzyme that efficiently consumes lactate. In the presence of oxygen, LOX can catalyse the formation of pyruvate and hydrogen peroxide (H2O2) from lactate. This process led to acidity alleviation, hypoxia, and a further increase in oxidative stress, alleviating the immunosuppressive state of the tumour microenvironment (TME). However, the high cost of LOX preparation and purification, poor stability, and systemic toxicity limited its application in tumour therapy. Therefore, the rational application of drug delivery systems can protect LOX from the organism's environment and maintain its catalytic activity. This paper reviews various LOX-based drug-carrying systems, including inorganic nanocarriers, organic nanocarriers, and inorganic-organic hybrid nanocarriers, as well as other non-nanocarriers, which have been used for tumour therapy in recent years. In addition, this area's challenges and potential for the future are highlighted.
Collapse
Affiliation(s)
- Lu Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Jie Feng
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Yujun Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Jun Hou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, Sichuan, China.
| |
Collapse
|
15
|
Zhou J, Hu Y, Cao Y, Ding S, Zeng L, Zhang Y, Cao M, Duan G, Zhang X, Bian XW, Tian G. A Lactate-Depleting metal organic framework-based nanocatalyst reinforces intratumoral T cell response to boost anti-PD1 immunotherapy. J Colloid Interface Sci 2024; 660:869-884. [PMID: 38277843 DOI: 10.1016/j.jcis.2024.01.129] [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: 10/20/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
Infiltration and activation of intratumoral T lymphocytes are critical for immune checkpoint blockade (ICB) therapy. Unfortunately, the low tumor immunogenicity and immunosuppressive tumor microenvironment (TME) induced by tumor metabolic reprogramming cooperatively hinder the ICB efficacy. Herein, we engineered a lactate-depleting MOF-based catalytic nanoplatform (LOX@ZIF-8@MPN), encapsulating lactate oxidase (LOX) within zeolitic imidazolate framework-8 (ZIF-8) coupled with a coating of metal polyphenol network (MPN) to reinforce T cell response based on a "two birds with one stone" strategy. LOX could catalyze the degradation of the immunosuppressive lactate to promote vascular normalization, facilitating T cell infiltration. On the other hand, hydrogen peroxide (H2O2) produced during lactate depletion can be transformed into anti-tumor hydroxyl radical (•OH) by the autocatalytic MPN-based Fenton nanosystem to trigger immunogenic cell death (ICD), which largely improved the tumor immunogenicity. The combination of ICD and vascular normalization presents a better synergistic immunopotentiation with anti-PD1, inducing robust anti-tumor immunity in primary tumors and recurrent malignancies. Collectively, our results demonstrate that the concurrent depletion of lactate to reverse the immunosuppressive TME and utilization of the by-product from lactate degradation via cascade catalysis promotes T cell response and thus improves the effectiveness of ICB therapy.
Collapse
Affiliation(s)
- Jingrong Zhou
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China.
| | - Yunping Hu
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China; Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, PR China
| | - Yuhua Cao
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China; Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, PR China
| | - Shuaishuai Ding
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China; Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, PR China
| | - Lijuan Zeng
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China
| | - Yu Zhang
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China
| | - Mianfu Cao
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China
| | - Guangjie Duan
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China
| | - Xiao Zhang
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China; Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, PR China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China; Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, PR China.
| | - Gan Tian
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, PR China; Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, PR China.
| |
Collapse
|
16
|
Hou DY, Zhang NY, Wang L, Lv MY, Li XP, Zhang P, Wang YZ, Shen L, Wu XH, Fu B, Guo PY, Wang ZQ, Cheng DB, Wang H, Xu W. Inducing mitochondriopathy-like damages by transformable nucleopeptide nanoparticles for targeted therapy of bladder cancer. Natl Sci Rev 2024; 11:nwae028. [PMID: 38425424 PMCID: PMC10903983 DOI: 10.1093/nsr/nwae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/02/2024] [Accepted: 01/19/2024] [Indexed: 03/02/2024] Open
Abstract
Mitochondriopathy inspired adenosine triphosphate (ATP) depletions have been recognized as a powerful way for controlling tumor growth. Nevertheless, selective sequestration or exhaustion of ATP under complex biological environments remains a prodigious challenge. Harnessing the advantages of in vivo self-assembled nanomaterials, we designed an Intracellular ATP Sequestration (IAS) system to specifically construct nanofibrous nanostructures on the surface of tumor nuclei with exposed ATP binding sites, leading to highly efficient suppression of bladder cancer by induction of mitochondriopathy-like damages. Briefly, the reported transformable nucleopeptide (NLS-FF-T) self-assembled into nuclear-targeted nanoparticles with ATP binding sites encapsulated inside under aqueous conditions. By interaction with KPNA2, the NLS-FF-T transformed into a nanofibrous-based ATP trapper on the surface of tumor nuclei, which prevented the production of intracellular energy. As a result, multiple bladder tumor cell lines (T24, EJ and RT-112) revealed that the half-maximal inhibitory concentration (IC50) of NLS-FF-T was reduced by approximately 4-fold when compared to NLS-T. Following intravenous administration, NLS-FF-T was found to be dose-dependently accumulated at the tumor site of T24 xenograft mice. More significantly, this IAS system exhibited an extremely antitumor efficacy according to the deterioration of T24 tumors and simultaneously prolonged the overall survival of T24 orthotopic xenograft mice. Together, our findings clearly demonstrated the therapeutic advantages of intracellular ATP sequestration-induced mitochondriopathy-like damages, which provides a potential treatment strategy for malignancies.
Collapse
Affiliation(s)
- Da-Yong Hou
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Ni-Yuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Lu Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Mei-Yu Lv
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
| | - Xiang-Peng Li
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Peng Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Yue-Ze Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering & Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, Wuhan 430070, China
| | - Xiu-Hai Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Bo Fu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Peng-Yu Guo
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Zi-Qi Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, Wuhan 430070, China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Wanhai Xu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin 150001, China
| |
Collapse
|
17
|
Wu GL, Tan X, Yang Q. Recent Advances on NIR-II Light-Enhanced Chemodynamic Therapy. Adv Healthc Mater 2024; 13:e2303451. [PMID: 37983596 DOI: 10.1002/adhm.202303451] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Chemodynamic therapy (CDT) is a particular oncological therapeutic strategy by generates the highly toxic hydroxyl radical (•OH) from the dismutation of endogenous hydrogen peroxide (H2O2) via Fenton or Fenton-like reactions. However, single CDT therapies have been limited by unsatisfactory efficacy. Enhanced chemodynamic therapy (ECDT) triggered by near-infrared (NIR) is a novel therapeutic modality based on light energy to improve the efficiency of Fenton or Fenton-like reactions. However, the limited penetration and imaging capability of the visible (400-650 nm) and traditional NIR-I region (650-900 nm) light-amplified CDT restrict the prospects for its clinical application. Combined with the high penetration/high precision imaging characteristics of the second near-infrared (NIR-II,) nanoplatform, it is expected to kill deep tumors efficiently while imaging the treatment process in real-time, and more notably, the NIR-II region radiation with wavelengths above 1000 nm can minimize the irradiation damage to normal tissues. Such NIR-II ECDT nanoplatforms have greatly improved the effectiveness of CDT therapy and demonstrated extraordinary potential for clinical applications. Accordingly, various strategies have been explored in the past years to improve the efficiency of NIR-II Enhanced CDT. In this review, the mechanisms and strategies used to improve the performance of NIR-II-enhanced CDT are outlined.
Collapse
Affiliation(s)
- Gui-Long Wu
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaofeng Tan
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China
| | - Qinglai Yang
- Center for Molecular Imaging Probe, Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
18
|
Gao F, Xue C, Dong J, Lu X, Yang N, Ou C, Mou X, Zhang YZ, Dong X. Tumor Microenvironment-Induced Drug Depository for Persistent Antitumor Chemotherapy and Immune Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307736. [PMID: 38009506 DOI: 10.1002/smll.202307736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Indexed: 11/29/2023]
Abstract
Herein, a drug-loading nanosystem that can in situ form drug depository for persistent antitumor chemotherapy and immune regulation is designed and built. The system (DOX@MIL-LOX@AL) is fabricated by packaging alginate on the surface of Doxorubicin (DOX) and lactate oxidase (LOX) loaded MIL-101(Fe)-NH2 nanoparticle, which can easily aggregate in the tumor microenvironment through the cross-linking with intratumoral Ca2+. Benefiting from the tumor retention ability, the fast-formed drug depository will continuously release DOX and Fe ions through the ATP-triggered slow degradation, thus realizing persistent antitumor chemotherapy and immune regulation. Meanwhile, LOX in the non-aggregated nanoparticles is able to convert the lactic acid to H2O2, which will be subsequently decomposed into ·OH by Fe ions to further enhance the DOX-induced immunogenic death effect of tumor cells. Together, with the effective consumption of immunosuppressive lactic acid, long-term chemotherapy, and oxidation therapy, DOX@MIL-LOX@AL can execute high-performance antitumor chemotherapy and immune activation with only one subcutaneous administration.
Collapse
Affiliation(s)
- Fan Gao
- Jiangsu Key Laboratory of Atmospheric Environment Monitoring and Pollution Control, Collaborative Innovation Center of Atmospheric Environment and Equipment Technology, School of Environmental Science and Engineering, Nanjing University of Information Science & Technology, Nanjing, 210044, China
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Jianhui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Xinxin Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Nan Yang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Changjin Ou
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Xiaozhou Mou
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Cancer Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yi-Zhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, China
| |
Collapse
|
19
|
Zhang Z, Liang X, Yang X, Liu Y, Zhou X, Li C. Advances in Nanodelivery Systems Based on Metabolism Reprogramming Strategies for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6689-6708. [PMID: 38302434 DOI: 10.1021/acsami.3c15686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Tumor development and metastasis are closely related to the complexity of the metabolism network. Recently, metabolism reprogramming strategies have attracted much attention in tumor metabolism therapy. Although there is preliminary success of metabolism therapy agents, their therapeutic effects have been restricted by the effective reaching of the tumor sites of drugs. Nanodelivery systems with unique physical properties and elaborate designs can specifically deliver to the tumors. In this review, we first summarize the research progress of nanodelivery systems based on tumor metabolism reprogramming strategies to enhance therapies by depleting glucose, inhibiting glycolysis, depleting lactic acid, inhibiting lipid metabolism, depleting glutamine and glutathione, and disrupting metal metabolisms combined with other therapies, including chemotherapy, radiotherapy, photodynamic therapy, etc. We further discuss in detail the advantages of nanodelivery systems based on tumor metabolism reprogramming strategies for tumor therapy. As well as the opportunities and challenges for integrating nanodelivery systems into tumor metabolism therapy, we analyze the outlook for these emerging areas. This review is expected to improve our understanding of modulating tumor metabolisms for enhanced therapy.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiangyu Zhou
- Department of Thyroid and Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
20
|
Nie W, Jiang A, Ou X, Zhou J, Li Z, Liang C, Huang LL, Wu G, Xie HY. Metal-polyphenol "prison" attenuated bacterial outer membrane vesicle for chemodynamics promoted in situ tumor vaccines. Biomaterials 2024; 304:122396. [PMID: 38043464 DOI: 10.1016/j.biomaterials.2023.122396] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 12/05/2023]
Abstract
As natural adjuvants, the bacterial outer membrane vesicles (OMV) hold great potential in cancer vaccines. However, the inherent immunotoxicity of OMV and the rarity of tumor-specific antigens seriously hamper the clinical translation of OMV-based cancer vaccines. Herein, metal-phenolic networks (MPNs) are used to attenuate the toxicity of OMV, meanwhile, provide tumor antigens via the chemodynamic effect induced immunogenic cell death (ICD). Specifically, MPNs are assembled on the OMV surface through the coordination reaction between ferric ions and tannic acid. The iron-based "prison" is locally collapsed in the tumor microenvironment (TME) with both low pH and high ATP features, and thus the systemic toxicity of OMV is significantly attenuated. The released ferric ions in TME promote the ICD of cancer cells through Fenton reaction and then the generation of abundant tumor antigens, which can be used to fabricate in-situ vaccines by converging with OMV. Together with the immunomodulatory effect of OMV, potent tumor repression on a bilateral tumor model is achieved with good biosafety.
Collapse
Affiliation(s)
- Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
| | - Anqi Jiang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
| | - Xu Ou
- School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
| | - Jiaxin Zhou
- School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
| | - Zijin Li
- School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
| | - Li-Li Huang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, PR China
| | - Guanghao Wu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, PR China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
21
|
Cheng Q, Shi X, Li Q, Wang L, Wang Z. Current Advances on Nanomaterials Interfering with Lactate Metabolism for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305662. [PMID: 37941489 PMCID: PMC10797484 DOI: 10.1002/advs.202305662] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/15/2023] [Indexed: 11/10/2023]
Abstract
Increasing numbers of studies have shown that tumor cells prefer fermentative glycolysis over oxidative phosphorylation to provide a vast amount of energy for fast proliferation even under oxygen-sufficient conditions. This metabolic alteration not only favors tumor cell progression and metastasis but also increases lactate accumulation in solid tumors. In addition to serving as a byproduct of glycolytic tumor cells, lactate also plays a central role in the construction of acidic and immunosuppressive tumor microenvironment, resulting in therapeutic tolerance. Recently, targeted drug delivery and inherent therapeutic properties of nanomaterials have attracted great attention, and research on modulating lactate metabolism based on nanomaterials to enhance antitumor therapy has exploded. In this review, the advanced tumor therapy strategies based on nanomaterials that interfere with lactate metabolism are discussed, including inhibiting lactate anabolism, promoting lactate catabolism, and disrupting the "lactate shuttle". Furthermore, recent advances in combining lactate metabolism modulation with other therapies, including chemotherapy, immunotherapy, photothermal therapy, and reactive oxygen species-related therapies, etc., which have achieved cooperatively enhanced therapeutic outcomes, are summarized. Finally, foreseeable challenges and prospective developments are also reviewed for the future development of this field.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Xiao‐Lei Shi
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Qi‐Lin Li
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Lin Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Research Center for Tissue Engineering and Regenerative MedicineUnion HospitalHuazhong University of Science and TechnologyWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| | - Zheng Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchWuhan430022China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhongUniversity of Science and TechnologyWuhan430022China
| |
Collapse
|
22
|
Li Z, Cui J. Targeting the lactic acid metabolic pathway for antitumor therapy. Mol Ther Oncolytics 2023; 31:100740. [PMID: 38033399 PMCID: PMC10682057 DOI: 10.1016/j.omto.2023.100740] [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] [Indexed: 12/02/2023] Open
Abstract
Lactic acid is one of the most abundant products of cellular metabolism and has historically been considered a cell-damaging metabolic product. However, as research has deepened, the beneficial effects of lactic acid on tumor cells and the tumor microenvironment have received increasing attention from the oncology community. Lactic acid can not only provide tumor cells with energy but also act as a messenger molecule that promotes tumor growth and progression and protects tumor cells from immune cells and killing by radiation and chemotherapy. Thus, the inhibition of tumor cell lactic acid metabolism has emerged as a novel antitumor treatment strategy that can also effectively enhance the efficacy of conventional antitumor therapies. In this review, we classify the currently available therapies targeting lactic acid metabolism and examine their prospects for clinical application.
Collapse
Affiliation(s)
- Zhi Li
- Cancer Center, First Hospital of Jilin University, Changchun 130021, China
| | - Jiuwei Cui
- Cancer Center, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
23
|
Zeng S, Liu X, Kafuti YS, Kim H, Wang J, Peng X, Li H, Yoon J. Fluorescent dyes based on rhodamine derivatives for bioimaging and therapeutics: recent progress, challenges, and prospects. Chem Soc Rev 2023; 52:5607-5651. [PMID: 37485842 DOI: 10.1039/d2cs00799a] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Since their inception, rhodamine dyes have been extensively applied in biotechnology as fluorescent markers or for the detection of biomolecules owing to their good optical physical properties. Accordingly, they have emerged as a powerful tool for the visualization of living systems. In addition to fluorescence bioimaging, the molecular design of rhodamine derivatives with disease therapeutic functions (e.g., cancer and bacterial infection) has recently attracted increased research attention, which is significantly important for the construction of molecular libraries for diagnostic and therapeutic integration. However, reviews focusing on integrated design strategies for rhodamine dye-based diagnosis and treatment and their wide application in disease treatment are extremely rare. In this review, first, a brief history of the development of rhodamine fluorescent dyes, the transformation of rhodamine fluorescent dyes from bioimaging to disease therapy, and the concept of optics-based diagnosis and treatment integration and its significance to human development are presented. Next, a systematic review of several excellent rhodamine-based derivatives for bioimaging, as well as for disease diagnosis and treatment, is presented. Finally, the challenges in practical integration of rhodamine-based diagnostic and treatment dyes and the future outlook of clinical translation are also discussed.
Collapse
Affiliation(s)
- Shuang Zeng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China.
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian 116024, China
| | - Xiaosheng Liu
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian 116024, China
| | - Yves S Kafuti
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian 116024, China
| | - Heejeong Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea.
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China.
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian 116024, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China.
| | - Haidong Li
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China.
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Hi-tech Zone, Dalian 116024, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning 110042, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
24
|
Yang Z, Yang C, Yang D, Zhang Y, Yang Q, Qu F, Guo W. l-Arginine-Modified CoWO 4 /FeWO 4 S-Scheme Heterojunction Enhances Ferroptosis against Solid Tumor. Adv Healthc Mater 2023; 12:e2203092. [PMID: 36907173 DOI: 10.1002/adhm.202203092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/27/2023] [Indexed: 03/13/2023]
Abstract
Ferroptosis has recently attracted much attention as an anti-tumor therapy. Evidence suggests that ferroptosis can induce oxidative stress and accumulation of lethal lipid peroxides in cancer cells, leading to cell damage. However, unsuitable pH, H2 O2 levels, and high glutathione (GSH) expression in the tumor microenvironment hinder the development of ferroptosis-mediated therapy. In this study, an l-arginine (l-arg)-modified CoWO4 /FeWO4 (CFW) S-scheme heterojunction is strategically designed and constructed for ultrasound (US)-triggered sonodynamic- and gas therapy-induced ferroptosis. CFW not only has excellent Fenton-catalytic activity, outstanding GSH consumption capacity, and excellent ability to overcome tumor hypoxia, but its S-scheme heterostructure can also avoid the rapid combination of electron (e) and hole (h+ ) pairs, thereby enhancing the sonodynamic effects. As a precursor of nitric oxide (NO), l-arg is modified on the surface of CFW (CFW@l-arg) to achieve controlled NO release under US irradiation, thereby enhancing ferroptosis. In addition, poly(allylamine hydrochloride) is further modified on the surface of CFW@l-arg to stabilize l-arg and achieve controllable NO release. Both in vitro and in vivo results demonstrate that such a multifunctional therapeutic nanoplatform can achieve high therapeutic efficacy through sonodynamic and gas therapy-enhanced ferroptosis. This designed oncotherapy nanoplatform provides new inspiration for ferroptosis-mediated therapy.
Collapse
Affiliation(s)
- Zhuoran Yang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Chunyu Yang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Dan Yang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Ye Zhang
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Qingzhu Yang
- College of Life Science and Agriculture Forestry, Qiqihar University, Qiqihar, 161006, China
| | - Fengyu Qu
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Wei Guo
- Key Laboratory of Photochemical Biomaterials and Energy Storage Materials, Heilongjiang Province and College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| |
Collapse
|
25
|
Jiang S, Chen X, Lin J, Huang P. Lactate-Oxidase-Instructed Cancer Diagnosis and Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207951. [PMID: 36353879 DOI: 10.1002/adma.202207951] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/15/2022] [Indexed: 05/12/2023]
Abstract
Lactate oxidase (LOx) has attracted extensive interest in cancer diagnosis and therapy in recent years owing to its specific catalysis on l-lactate; its catalytic process consumes oxygen (O2 ) and generates a large amount of hydrogen peroxide (H2 O2 ) and pyruvate. Given high levels of lactate in tumor tissues and its tight correlation with tumor growth, metastasis, and recurrence, LOx-based biosensors including H2 O2 -based, O2 -based, pH-sensitive, and electrochemical have been designed for cancer diagnosis, and various LOx-based cancer therapy strategies including lactate-depletion-based metabolic cancer therapy/immunotherapy, hypoxia-activated chemotherapy, H2 O2 -based chemodynamic therapy, and multimodal synergistic cancer therapy have also been developed. In this review, the lactate-specific catalytic properties of LOx are introduced, and the recent advances on LOx-instructed cancer diagnostic or therapeutic platforms and corresponding biological applications are summarized. Additionally, the challenges and potential of LOx-based nanomedicines are highlighted.
Collapse
Affiliation(s)
- Shanshan Jiang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Xin Chen
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| |
Collapse
|
26
|
Cheng C, Jiang W, Luo Y, Wan L, Guo X, Xie Z, Tang R, Huang T, Wang J, Du C, Wang Z, Ran H, Li P, Zhou Z, Ren J. NIR Activated Multimodal Therapeutics Based on Metal-Phenolic Networks-Functionalized Nanoplatform for Combating against Multidrug Resistance and Metastasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206174. [PMID: 36651135 DOI: 10.1002/smll.202206174] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Multidrug resistance (MDR) and metastasis in cancer have become increasingly serious problems since antitumor efficiency is greatly restricted by a single therapeutic modality and the insensitive tumor microenvironment (TME). Herein, metal-phenolic network-functionalized nanoparticles (t-P@TFP NPs) are designed to realize multiple therapeutic modalities and reshape the TME from insensitive to sensitive under multimodal imaging monitoring. After a single irradiation, a near-infrared laser-activated multistage reaction occurs. t-P@TFP NPs trigger the phase transition of perfluoropentane (PFP) to release tannic acid (TA)/ferric ion (Fe3+ )-coated paclitaxel (PTX) and cause hyperthermia in the tumor region to efficiently kill cancer cells. Additionally, PTX is released after the disassembly of the TA-Fe3+ film by the abundant adenosine triphosphate (ATP) in the malignant tumor, which concurrently inhibits ATP-dependent drug efflux to improve sensitivity to chemotherapeutic agents. Furthermore, hyperthermia-induced immunogenic cell death (ICD) transforms "cold" tumors into "hot" tumors with the assistance of PD-1/PD-L1 blockade to evoke antitumor immunogenicity. This work carefully reveals the mechanisms underlying the abilities of these multifunctional NPs, providing new insights into combating the proliferation and metastasis of multidrug-resistant tumors.
Collapse
Affiliation(s)
- Chen Cheng
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
- Department of Ultrasound, Bishan Hospital of Chongqing, Bishan hospital of Chongqing medical university, Chongqing, 402760, P. R. China
| | - Weixi Jiang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Yuanli Luo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Li Wan
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Xun Guo
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Zhuoyan Xie
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Rui Tang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Tong Huang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Jingxue Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Chier Du
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Zhigang Wang
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Haitao Ran
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Pan Li
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Zhiyi Zhou
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
- Department of General Practice, Chongqing General Hospital, Chongqing, 401147, P. R. China
| | - Jianli Ren
- Department of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| |
Collapse
|
27
|
Chen J, Zhu Y, Wu C, Shi J. Engineering lactate-modulating nanomedicines for cancer therapy. Chem Soc Rev 2023; 52:973-1000. [PMID: 36597879 DOI: 10.1039/d2cs00479h] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lactate in tumors has long been considered "metabolic junk" derived from the glycolysis of cancer cells and utilized only as a biomarker of malignancy, but is presently believed to be a pivotal regulator of tumor development, maintenance and metastasis. Indeed, tumor lactate can be a "fuel" for energy supply and functions as a signaling molecule, which actively contributes to tumor progression, angiogenesis, immunosuppression, therapeutic resistance, etc., thus providing promising opportunities for cancer treatment. However, the current approaches for regulating lactate homeostasis with available agents are still challenging, which is mainly due to the short half-life, low bioavailability and poor specificity of these agents and their unsatisfactory therapeutic outcomes. In recent years, lactate modulation nanomedicines have emerged as a charming and efficient strategy for fighting cancer, which play important roles in optimizing the delivery of lactate-modulating agents for more precise and effective modulation and treatment. Integrating specific lactate-modulating functions in diverse therapeutic nanomedicines may overcome the intrinsic restrictions of different therapeutic modalities by remodeling the pathological microenvironment for achieving enhanced cancer therapy. In this review, the most recent advances in the engineering of functional nanomedicines that can modulate tumor lactate for cancer therapy are summarized and discussed, and the fundamental mechanisms by which lactate modulation benefits various therapeutics are elucidated. Finally, the challenges and perspectives of this emerging strategy in the anti-tumor field are highlighted.
Collapse
Affiliation(s)
- Jiajie Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yufang Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.,Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200331, P. R. China
| |
Collapse
|
28
|
Self-intensified synergy of a versatile biomimetic nanozyme and doxorubicin on electrospun fibers to inhibit postsurgical tumor recurrence and metastasis. Biomaterials 2023; 293:121942. [PMID: 36512863 DOI: 10.1016/j.biomaterials.2022.121942] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/26/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Tumor-positive resection margins after surgery can result in tumor recurrence and metastasis. Although adjuvant postoperative radiotherapy and chemotherapy have been adopted in clinical practice, they lack efficacy and result in unavoidable side effects. Herein, a self-intensified in-situ therapy approach using electrospun fibers loaded with a biomimetic nanozyme and doxorubicin (DOX) is developed. The fabricated PEG-coated zeolite imidazole framework-67 (PZIF67) is demonstrated as a versatile nanozyme triggering reactions in cancer cells based on endogenous H2O2 and •O2-. The PZIF67-generated •OH induces reactive oxygen species (ROS) overload, implementing chemodynamic therapy (CDT). The O2 produced by PZIF67 inhibits the expression of hypoxia-up-regulated proteins, thereby suppressing tumor progression. PZIF67 also catalyzes the degradation of glutathione, further disturbing the intracellular redox homeostasis and enhancing CDT. Furthermore, the introduced DOX not only kills cancer cells individually, but also replenishes the continuously consumed substrates for PZIF67-catalyzed reactions. The PZIF67-weakened drug resistance strengthens the cytotoxicity of DOX. The combined application of PZIF67 and DOX also suppresses metastasis-associated genes. Both in vitro and in vivo results demonstrate that the self-intensified synergy of PZIF67 and DOX on electrospun fibers efficiently prevents postsurgical tumor recurrence and metastasis, offering a feasible therapeutic regimen for operable malignant tumors.
Collapse
|
29
|
Zhao J, Tian Z, Zhao S, Feng D, Guo Z, Wen L, Zhu Y, Xu F, Zhu J, Ma S, Hu J, Jiang T, Qu Y, Chen D, Liu L. Insights into the Effect of Catalytic Intratumoral Lactate Depletion on Metabolic Reprogramming and Immune Activation for Antitumoral Activity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204808. [PMID: 36479819 PMCID: PMC9896070 DOI: 10.1002/advs.202204808] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/24/2022] [Indexed: 06/17/2023]
Abstract
Lactate, a characteristic metabolite of the tumor microenvironment (TME), drives immunosuppression and promotes tumor progression. Material-engineered strategies for intratumoral lactate modulations demonstrate their promise for tumor immunotherapy. However, understanding of the inherent interconnections of material-enabled lactate regulation, metabolism, and immunity in the TME is scarce. To address this issue, urchin-like catalysts of the encapsulated Gd-doped CeO2 , syrosingopine, and lactate oxidase are used in ZIF-8 (USL, where U, S, and L represent the urchin-like Gd-doped CeO2 @ZIF-8, syrosingopine, and lactate oxidase, respectively) and orthotopic tumor models. The instructive relationships of intratumoral lactate depletion, metabolic reprogramming, and immune activation for catalytic immunotherapy of tumors is illustrated. The catalysts efficiently oxidize intratumoral lactate and significantly promote tumor cell apoptosis by in situ-generated ·OH, thereby reducing glucose supply and inducing mitochondrial damage via lactate depletion, thus reprogramming glycometabolism. Subsequently, such catalytic metabolic reprogramming evokes both local and systemic antitumor immunity by activating M1-polarizaed macrophages and CD8+ T cells, leading to potent antitumor immunity. This study provides valuable mechanistic insights into material-interfered tumor therapy through intratumoral lactate depletion and consequential connection with metabolic reprogramming and immunity remodeling, which is thought to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Junlong Zhao
- Department of GastroenterologyDaping HospitalArmy Medical UniversityChongqing400032P. R. China
- State Key Laboratory of Cancer BiologyDepartment of Medical Genetics and Development BiologyFourth Military Medical UniversityXi'an710032P. R. China
- Present address:
Department of GastroenterologyChongqing Key Laboratory of Digestive MalignanciesDaping Hospital, Army Medical University (Third Military Medical University)Chongqing400042P. R. China
| | - Zhimin Tian
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information TechnologySchool of Chemistry and Chemical EngineeringNorthwestern Polytechnical UniversityXi'an710072P. R. China
- Xi'an People's Hospital (Xi'an Fouth Hospital)Shaanxi Eye HospitalAffiliated Guangren HospitalSchool of MedicineXi'an Jiaotong UniversityXi'an710004P. R. China
| | - Shoujie Zhao
- Department of General SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Dayun Feng
- Department of SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Zhixiong Guo
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information TechnologySchool of Chemistry and Chemical EngineeringNorthwestern Polytechnical UniversityXi'an710072P. R. China
| | - Liangzhi Wen
- Department of GastroenterologyDaping HospitalArmy Medical UniversityChongqing400032P. R. China
| | - Yejing Zhu
- Department of General SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Fenghua Xu
- Department of GastroenterologyDaping HospitalArmy Medical UniversityChongqing400032P. R. China
| | - Jun Zhu
- Department of SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Shouzheng Ma
- Department of SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Jie Hu
- Department of SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Tao Jiang
- Department of SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| | - Yongquan Qu
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information TechnologySchool of Chemistry and Chemical EngineeringNorthwestern Polytechnical UniversityXi'an710072P. R. China
| | - Dongfeng Chen
- Department of GastroenterologyDaping HospitalArmy Medical UniversityChongqing400032P. R. China
| | - Lei Liu
- Department of GastroenterologyDaping HospitalArmy Medical UniversityChongqing400032P. R. China
- Department of General SurgeryTangdu HospitalFourth Military Medical UniversityXi'an710038P. R. China
| |
Collapse
|
30
|
Oxygen-carrying nanoplatform to reprogram tumor immunosuppressive microenvironment and enhance photothermal-immunotherapy. Mater Today Bio 2023; 19:100555. [PMID: 36793322 PMCID: PMC9922928 DOI: 10.1016/j.mtbio.2023.100555] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/19/2023] Open
Abstract
Immunotherapy shows great promise on treating tumors. However, insufficient antigen exposure and immunosuppressive tumor microenvironment (TME) caused by hypoxia impose a serial of constraints on the therapeutic efficacy. In this study, we developed an oxygen-carrying nanoplatform loaded with perfluorooctyl bromide (PFOB, a second-generation of perfluorocarbon-based blood substitute), IR780 (a photosensitizer) and imiquimod (R837, an immune adjuvant) to reprogram immunosuppressive TME and reinforce photothermal-immunotherapy. The obtained oxygen-carrying nanoplatforms (abbreviated as IR-R@LIP/PFOB) show highly efficient oxygen release behavior and excellent hyperthermia performance upon laser irradiation, thus achieving the attenuation of the inherent tumor hypoxia and the exposure of tumor associated antigens in situ, and transforming the immunosuppressive TME to an immunosupportive one. We found that the photothermal therapy of IR-R@LIP/PFOB together with anti-programmed cell death protein-1 (anti-PD-1) would elicit a robust antitumor immunity by increasing the tumor-infiltrating frequencies of cytotoxic CD8+ T cells and tumoricidal M1-phenotype macrophages, while reducing immunosuppressive M2-phenotype macrophages and regulatory T cells (Tregs). This study presents these oxygen-carrying IR-R@LIP/PFOB nanoplatforms are potent in removing some negative impacts of immunosuppressive TME caused by hypoxia, and suppressing tumor growth by initiating antitumor immune responses, especially in combination with anti-PD-1 immunotherapy.
Collapse
|
31
|
Choi H, Yeo M, Kang Y, Kim HJ, Park SG, Jang E, Park SH, Kim E, Kang S. Lactate oxidase/catalase-displaying nanoparticles efficiently consume lactate in the tumor microenvironment to effectively suppress tumor growth. J Nanobiotechnology 2023; 21:5. [PMID: 36597089 PMCID: PMC9811728 DOI: 10.1186/s12951-022-01762-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
The aggressive proliferation of tumor cells often requires increased glucose uptake and excessive anaerobic glycolysis, leading to the massive production and secretion of lactate to form a unique tumor microenvironment (TME). Therefore, regulating appropriate lactate levels in the TME would be a promising approach to control tumor cell proliferation and immune suppression. To effectively consume lactate in the TME, lactate oxidase (LOX) and catalase (CAT) were displayed onto Aquifex aeolicus lumazine synthase protein nanoparticles (AaLS) to form either AaLS/LOX or AaLS/LOX/CAT. These complexes successfully consumed lactate produced by CT26 murine colon carcinoma cells under both normoxic and hypoxic conditions. Specifically, AaLS/LOX generated a large amount of H2O2 with complete lactate consumption to induce drastic necrotic cell death regardless of culture condition. However, AaLS/LOX/CAT generated residual H2O2, leading to necrotic cell death only under hypoxic condition similar to the TME. While the local administration of AaLS/LOX to the tumor site resulted in mice death, that of AaLS/LOX/CAT significantly suppressed tumor growth without any severe side effects. AaLS/LOX/CAT effectively consumed lactate to produce adequate amounts of H2O2 which sufficiently suppress tumor growth and adequately modulate the TME, transforming environments that are favorable to tumor suppressive neutrophils but adverse to tumor-supportive tumor-associated macrophages. Collectively, these findings showed that the modular functionalization of protein nanoparticles with multiple metabolic enzymes may offer the opportunity to develop new enzyme complex-based therapeutic tools that can modulate the TME by controlling cancer metabolism.
Collapse
Affiliation(s)
- Hyukjun Choi
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Mirae Yeo
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Yujin Kang
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Hyo Jeong Kim
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Seong Guk Park
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Eunjung Jang
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Sung Ho Park
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Eunhee Kim
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| | - Sebyung Kang
- grid.42687.3f0000 0004 0381 814XDepartment of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919 South Korea
| |
Collapse
|
32
|
Liu R, Shi D, Guo L, Xiao S, Shang M, Sun X, Meng D, Zhao Y, Wang X, Li J. Ultrasound-Targeted Microbubble Disruption with Key Nanodroplets for Effective Ferroptosis in Triple-Negative Breast Cancer Using Animal Model. Int J Nanomedicine 2023; 18:2037-2052. [PMID: 37155504 PMCID: PMC10122866 DOI: 10.2147/ijn.s400495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is known to be the most aggressive form of breast cancer. Due to its high recurrence and mortality rates, the treatment of TNBC is a significant challenge for the medical community. Besides, ferroptosis is an emerging regulatory cell death that may provide new insights into the treatment of TNBC. As a central inhibitor of the ferroptosis process, the selenoenzyme glutathione peroxidase 4 (GPX4) is its classical therapeutic target. However, inhibition of GPX4 expression is quite detrimental to normal tissues. Ultrasound contrast agents, as an emerging visualization precision treatment, may provide a solution to the existing problem. Methods In this study, nanodroplets (NDs) carrying simvastatin (SIM) were constructed using the homogeneous/emulsification method. Then, the characterization of SIM-NDs was systematically evaluated. Meanwhile, in this study, the ability of SIM-NDs combined with ultrasound-targeted microbubble disruption (UTMD) to initiate ferroptosis and its respective mechanisms of ferroptosis induction were verified. Finally, the antitumor activity of SIM-NDs was investigated in vitro and in vivo using MDA-MB-231 cells and TNBC animal models. Results SIM-NDs exhibited excellent pH- and ultrasound-responsive drug release and noticeable ultrasonographic imaging ability, also showing good biocompatibility and biosafety. UTMD could promote increased intracellular reactive oxygen species and consume intracellular glutathione. However, SIM-NDs were efficiently internalized into cells under ultrasound irradiation, followed by the rapid release of SIM, which inhibited intracellular mevalonate production, and synergistically downregulated GPX4 expression, thereby promoting ferroptosis. Moreover, this combined treatment demonstrated strong antitumor ability in vitro and in vivo. Conclusion The combination of UTMD and SIM-NDs presents a promising avenue for harnessing ferroptosis in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Dong Meng
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Correspondence: Jie Li, Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China, Email
| |
Collapse
|
33
|
Zhang Y, Li M, Zhang X, Zhang P, Liu Z, Feng M, Ren G, Liu J. Tumor microenvironment-activated Nb2C quantum dots/lactate oxidase nanocatalyst mediates lactate consumption and macrophage repolarization for enhanced chemodynamic therapy. Colloids Surf B Biointerfaces 2022; 221:113005. [DOI: 10.1016/j.colsurfb.2022.113005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
|
34
|
Xiao Y, Chen P, Lei S, Bai F, Fu L, Lin J, Huang P. Biocatalytic Depletion of Tumorigenic Energy Sources Driven by Cascade Reactions for Efficient Antitumor Therapy. Angew Chem Int Ed Engl 2022; 61:e202204584. [DOI: 10.1002/anie.202204584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Ya‐Ping Xiao
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Peng‐Hang Chen
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Shan Lei
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Fang Bai
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Lian‐Hua Fu
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| |
Collapse
|
35
|
Luo S, Qin S, Oudeng G, Zhang L. Iron-Based Hollow Nanoplatforms for Cancer Imaging and Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3023. [PMID: 36080059 PMCID: PMC9457987 DOI: 10.3390/nano12173023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 05/27/2023]
Abstract
Over the past decade, iron (Fe)-based hollow nanoplatforms (Fe-HNPs) have attracted increasing attention for cancer theranostics, due to their high safety and superior diagnostic/therapeutic features. Specifically, Fe-involved components can serve as magnetic resonance imaging (MRI) contrast agents (CAs) and Fenton-like/photothermal/magnetic hyperthermia (MTH) therapy agents, while the cavities are able to load various small molecules (e.g., fluorescent dyes, chemotherapeutic drugs, photosensitizers, etc.) to allow multifunctional all-in-one theranostics. In this review, the recent advances of Fe-HNPs for cancer imaging and treatment are summarized. Firstly, the use of Fe-HNPs in single T1-weighted MRI and T2-weighted MRI, T1-/T2-weighted dual-modal MRI as well as other dual-modal imaging modalities are presented. Secondly, diverse Fe-HNPs, including hollow iron oxide (IO) nanoparticles (NPs), hollow matrix-supported IO NPs, hollow Fe-complex NPs and hollow Prussian blue (PB) NPs are described for MRI-guided therapies. Lastly, the potential clinical obstacles and implications for future research of these hollow Fe-based nanotheranostics are discussed.
Collapse
Affiliation(s)
- Shun Luo
- Key Laboratory for Photoelectronic Technology and Application, Guizhou University, Guiyang 550025, China
| | - Shuijie Qin
- Key Laboratory for Photoelectronic Technology and Application, Guizhou University, Guiyang 550025, China
| | - Gerile Oudeng
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Futian, Shenzhen 518038, China
| | - Li Zhang
- School of Science, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
| |
Collapse
|
36
|
Feng W, Shi W, Wang Z, Cui Y, Shao X, Liu S, Rong L, Liu Y, Zhang H. Enhancing Tumor Therapy of Fe(III)-Shikonin Supramolecular Nanomedicine via Triple Ferroptosis Amplification. ACS APPLIED MATERIALS & INTERFACES 2022; 14:37540-37552. [PMID: 35944147 DOI: 10.1021/acsami.2c11130] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ferroptosis has been considered as a promising pathway to overcome apoptosis-induced tumor chemoresistance. However, the antitumor efficacy of ferroptosis-inducing agents is still limited because of the complexity and diversity of tumor microenvironments. Herein, we demonstrate a triple ferroptosis amplification strategy for tumor therapy by associating iron-based nanocarriers, ferroptosis molecular drugs, and H2O2-producing enzymes. Fe(III)-Shikonin (FeShik) metal-polyphenol-coordinated networks are employed to load a ferroptosis inducer of sorafenib (SRF) inside and glucose oxidase (GOx) outside, thus producing SRF@FeShik-GOx supramolecular nanomedicines (SNs). After delivering into glutathione (GSH)-overexpressed tumor cells, FeShik will disassemble and release Fe2+ to induce cell death via ferroptosis. At the same time, GOx executes its catalytic activity to produce an acid environment and plenty of H2O2 for stimulating •OH generation via the Fenton reaction. Moreover, SRF will suppress the biosynthesis of GSH by inhibiting system Xc-, further deactivating the enzymatic activity of glutathione peroxidase 4 (GPX4). Up-regulation of the oxidative stress level and down-regulation of GPX4 expression can dramatically accelerate the accumulation of lethal lipid peroxides, leading to ferroptosis amplification of tumor cells. The current strategy that utilizes ferroptosis-inducing agents as both nanocarriers and cargoes provides a pathway to enhance the efficacy of ferroptosis-based tumor therapy.
Collapse
Affiliation(s)
- Wenjie Feng
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wanrui Shi
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yanqi Cui
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xinxin Shao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Sciences, Jilin University, Changchun 130012, P. R. China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Li Rong
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
37
|
Ren M, Zheng X, Gao H, Jiang A, Yao Y, He W. Nanomedicines Targeting Metabolism in the Tumor Microenvironment. Front Bioeng Biotechnol 2022; 10:943906. [PMID: 35992338 PMCID: PMC9388847 DOI: 10.3389/fbioe.2022.943906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer cells reprogram their metabolism to meet their growing demand for bioenergy and biosynthesis. The metabolic profile of cancer cells usually includes dysregulation of main nutritional metabolic pathways and the production of metabolites, which leads to a tumor microenvironment (TME) having the characteristics of acidity, hypoxic, and/or nutrient depletion. Therapies targeting metabolism have become an active and revolutionary research topic for anti-cancer drug development. The differential metabolic vulnerabilities between tumor cells and other cells within TME provide nanotechnology a therapeutic window of anti-cancer. In this review, we present the metabolic characteristics of intrinsic cancer cells and TME and summarize representative strategies of nanoparticles in metabolism-regulating anti-cancer therapy. Then, we put forward the challenges and opportunities of using nanoparticles in this emerging field.
Collapse
Affiliation(s)
- Mengdi Ren
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoqiang Zheng
- Institute for Stem Cell and Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huan Gao
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aimin Jiang
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yu Yao
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yu Yao, ; Wangxiao He,
| | - Wangxiao He
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yu Yao, ; Wangxiao He,
| |
Collapse
|
38
|
Zhang J, Liang C, Wei Z, Yang W, Ge W, Qu X, Si W, Wang W, Mou X, Dong X. TME-triggered MnSiO 3@Met@GOx nanosystem for ATP dual-inhibited starvation/chemodynamic synergistic therapy. Biomaterials 2022; 287:121682. [PMID: 35870264 DOI: 10.1016/j.biomaterials.2022.121682] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 12/14/2022]
Abstract
Adenosine triphosphate (ATP) is an essential substance for maintaining tumor cell survival and proliferation. Inhibiting the ATP-producing pathways has emerged as a promising cancer treatment strategy. However, the antitumor efficiency of ATP inhibitors is compromised by the inter-compensation of multiple ATP-producing pathways in tumor cells and biological barriers in the complex tumor microenvironment (TME). Herein, we developed metformin (Met) and glucose oxidase (GOx) co-loaded manganese silicon nanoplatform MnSiO3@Met@GOx (MMG) for TME-responsive ATP dual inhibited starvation/chemodynamic synergistic therapy. Under the mildly acidic conditions in TME, MMG was decomposed, releasing Met and GOx for effective ATP suppression by inhibiting oxidative phosphorylation (OXPHOS) and aerobic glycolysis pathways, respectively. Meanwhile, GOx-catalyzed glucose oxidation increased tumor acidity and hydrogen peroxide (H2O2) concentration in tumors, which not only accelerated MMG decomposition and drug release but also promoted manganese ions-mediated Fenton-like reaction. In vitro and in vivo experiments further demonstrated the effectiveness and biosafety of MMG-based synergistic therapy. This study provides a novel strategy for tumor treatment based on tumor metabolism regulation.
Collapse
Affiliation(s)
- Jiayao Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Chen Liang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Ziye Wei
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Wanlan Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Wei Ge
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Xinyu Qu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China
| | - Weili Si
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China.
| | - Wenjun Wang
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng, 252059, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, China; School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, China.
| |
Collapse
|
39
|
Xiao YP, Chen PH, Lei S, Bai F, Fu LH, Lin J, Huang P. Biocatalytic Depletion of Tumorigenic Energy Sources Driven by Cascade Reactions for Efficient Antitumor Therapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Shan Lei
- Shenzhen University School of Medicine CHINA
| | - Fang Bai
- Shenzhen University School of Medicine CHINA
| | - Lian-Hua Fu
- Shenzhen University School of Medicine CHINA
| | - Jing Lin
- Shenzhen University School of Medicine CHINA
| | - Peng Huang
- Shenzhen University 3688 Nanhai Ave, Nanshan 518060 Shenzhen CHINA
| |
Collapse
|
40
|
Tian H, Zhou L, Wang Y, Nice EC, Huang C, Zhang H. A targeted nanomodulator capable of manipulating tumor microenvironment against metastasis. J Control Release 2022; 348:590-600. [PMID: 35716882 DOI: 10.1016/j.jconrel.2022.06.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 02/08/2023]
Abstract
Recently, lactate has been considered as an alternative direct energy substance to glucose for tumor proliferation and metastasis. Meanwhile, mitochondria, as important energy-supplying organelles, are also closely related to tumor progression. Consequently, a new research direction for lactate comprises lactate deprivation coupled with mitochondria-targeted phototherapy to achieve a safer and more effective strategy against tumor metastasis. Herein, linoleic acid-conjugated hyaluronic acid (HL), disulfide bond-rich nanovehicle (mesoporous silica, MOS), mitochondria-targeted IR780 (M780) and lactate oxidase (LOD) are rationally designed as a specific-targeting metabolism nanomodulator (HL/MOS@M780&LOD NPs), fulfilling the task of simultaneous depriving cells of lactate and damaging mitochondria to prevent tumor metastasis. Interestingly, M780-mediated photodynamic therapy (PDT) and LOD-mediated starvation therapy can effectively exacerbate the hypoxia state of tumor cells, thereby increasing the free iron levels to activate ferroptosis. On one hand, pyruvic acid and H2O2 generated by LOD-mediated lactate metabolism can provide powerful conditions for iron-catalyzed ferroptosis. On the other, the depleted GSH and increased reactive oxygen species (ROS) can oxidize linoleic acid into lipid peroxides (LPO) to further augment ferroptosis. The designed nanomodulator therefore shows great promise for fighting tumor metastasis by manipulating energy metabolism and the hypoxia microenvironment.
Collapse
Affiliation(s)
- Hailong Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yu Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China.
| |
Collapse
|
41
|
Liu Y, Kong L, Li H, Yuan R, Chai Y. Electrochemical Aptamer Biosensor Based on ATP-Induced 2D DNA Structure Switching for Rapid and Ultrasensitive Detection of ATP. Anal Chem 2022; 94:6819-6826. [PMID: 35471959 DOI: 10.1021/acs.analchem.2c00613] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Herein, a two-dimensional (2D) DNA structure with multiple ATP aptamers was elegantly designed to establish an electrochemical biosensor for rapid and sensitive detection of ATP based on ATP-induced structure switching. Concretely, the prepared 2D DNA structure containing numerous ATP aptamers as ATP-specific toehold switches could not only immobilize a large number of methylene blue (MB) for generating a remarkable electrochemical signal, but also greatly increase the local concentration of ATP aptamers to obviously enhance the capture efficiency of ATP. Once the target ATP interacted with the toehold switches, the 2D DNA structure could be sharply collapsed to trigger the burst release of MB from the electrode surface, ultimately resulting in a significantly decreased electrochemical signal for ultrasensitive detection of target ATP over a short period of time. Impressively, by dexterously adjusting the length of the ATP-specific toehold switches to 15-base, optimization of the binding affinity between ATP and the toehold switches was achieved for cutting down the detection time to 30 min and achieving a low detection limit of 0.3 pM, which addressed the shortcoming of time-consuming and poor sensitivity in the previous sensors with a small quantity of ATP aptamers and deficient binding affinity to ATP. Consequently, this strategy opened a promising avenue for ultrasensitive and rapid detection of various biomolecules in biomedical application and disease diagnosis.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, People's Republic of China
| | - Lingqi Kong
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, People's Republic of China
| | - Hao Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, People's Republic of China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, People's Republic of China
| | - Yaqin Chai
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
42
|
Zhu G, Chi H, Liu M, Yin Y, Diao H, Liu Z, Guo Z, Xu W, Xu J, Cui C, Xing XJ, Ma K. Multifunctional "ball-rod" Janus nanoparticles boosting Fenton reaction for ferroptosis therapy of non-small cell lung cancer. J Colloid Interface Sci 2022; 621:12-23. [PMID: 35447518 DOI: 10.1016/j.jcis.2022.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 01/13/2023]
Abstract
Ferroptosis is a newly found cell death mechanism, which could bypass apoptosis and reverse multidrug resistance of tumors. However, efficient induction of tumor ferroptosis remains a challenge. In this study, multifunctional "ball-rod" Janus nanoparticles (FTG/L&SMD) were constructed for non-small cell lung cancer (NSCLC) ferroptosis treatment. Protected by tannic acid (TA), FTG/L&SMD maintains long-term function in blood circulation, while modification by 2, 3-dimethylmaleic anhydride (DMMA) confers the FTG/L&SMD with pH-responsive charge reversal. Glucose oxidase (GOD) on FTG/L&SMD catalyzes glucose to produce H2O2. Then, iron ion converts H2O2 to highly active hydroxyl radicals (OH•) via Fenton reaction, leading to lethal lipid peroxidation (LPO) accumulation. Meanwhile, TA reduces Fe3+ to Fe2+ to boost Fenton reaction cycle. Sor down-regulated glutathione peroxidase 4 (GPX4) expression in another pathway to induce ferroptosis synergistically. In vitro studies have shown that compared with sorafenib (Sor), FTG/L&SMD not only has more efficient tumor targeting and higher cytotoxicity, but also inhibits tumor migration. In vivo antitumor therapy experiments demonstrate that FTG/L&SMD inhibits tumor growth efficiently, and its toxicity is negligible. In general, FTG/L&SMD can initiate Fenton reaction cycle and reinforced ferroptosis to kill tumor cells, which is a promising anti-tumor nano-drug for NSCLC.
Collapse
Affiliation(s)
- Guang Zhu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Hao Chi
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Meichen Liu
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Yalin Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - He Diao
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zicheng Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Zhaoming Guo
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Weiping Xu
- School of Ocean Science and Technology, Dalian University of Technology, Panjin 124221, China
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China; Panjin Institute of Industrial Technology, Dalian University of Technology, Panjin 124221, China
| | - Changhao Cui
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| | - Xiao-Jin Xing
- Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| | - Kun Ma
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
43
|
Zhang L, Forgham H, Shen A, Qiao R, Guo B. Recent Advances in Single Fe-Based Nanoagents for Photothermal-Chemodynamic Cancer Therapy. BIOSENSORS 2022; 12:86. [PMID: 35200346 PMCID: PMC8869282 DOI: 10.3390/bios12020086] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Monomodal cancer therapies are often unsatisfactory, leading to suboptimal treatment effects that result in either an inability to stop growth and metastasis or prevent relapse. Thus, synergistic strategies that combine different therapeutic modalities to improve performance have become the new research trend. In this regard, the integration of photothermal therapy (PTT) with chemodynamic therapy (CDT), especially PTT/CDT in the second near-infrared (NIR-II) biowindow, has been demonstrated to be a highly efficient and relatively safe concept. With the rapid development of nanotechnology, nanoparticles can be designed from specific elements, such as Fe, that are equipped with both PTT and CDT therapeutic functions. In this review, we provide an update on the recent advances in Fe-based nanoplatforms for combined PTT/CDT. The perspectives on further improvement of the curative efficiency are described, highlighting the important scientific obstacles that require resolution in order to reach greater heights of clinical success. We hope this review will inspire the interest of researchers in developing novel Fe-based nanomedicines for multifunctional theranostics.
Collapse
Affiliation(s)
- Li Zhang
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen 518055, China;
| | - Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; (H.F.); (A.S.)
| | - Ao Shen
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; (H.F.); (A.S.)
| | - Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; (H.F.); (A.S.)
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen 518055, China;
| |
Collapse
|