1
|
Chen J, Li F, Luo WS, Zhu MF, Zhao NJ, Zhang ZH, Chen YF, Feng DX, Yang SY, Sun WJ. Therapeutic potential of Da Cheng Qi Decoction and its ingredients in regulating ferroptosis via the NOX2-GPX4 signaling pathway to alleviate and predict severe acute pancreatitis. Cell Signal 2025; 131:111733. [PMID: 40081545 DOI: 10.1016/j.cellsig.2025.111733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/27/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE This study aimed to elucidate the protective effects of Da Cheng Qi Decoction (DCQD) on severe acute pancreatitis (SAP) by targeting ferroptosis in pancreatic acinar cells and to establish a predictive signature and nomogram for acute pancreatitis (AP) risk assessment. METHODS We utilized microarray analysis to delineate gene expression patterns among 32 healthy controls and 87 AP patients stratified by severity. Employing SAP models and NOX2-deficient cells, we investigated the molecular underpinnings of ferroptosis. The impact of DCQD and the ferroptosis inhibitor Fer-1 on gene expression, oxidative stress, and inflammation was assessed. Machine learning algorithms identified differentially expressed genes (DEGs) sensitive to DCQD, SAP, and ferroptosis (DSNFGs), which were validated across multiple datasets. A predictive nomogram integrating DSNFGs was developed, and single-cell analysis provided a comprehensive view of the cellular dynamics. RESULTS The microarray analysis revealed upregulation of NOX2 and downregulation of GPX4 in AP, with expression patterns correlating with disease severity. DCQD ameliorated SAP-induced pancreatic acinar cell damage and ferroptosis by reducing inflammatory markers and enhancing GPX4 expression. NOX2 knockout mitigated ferroptosis in SAP models, suggesting a key role in the disease process. DCQD and Fer-1 differentially regulated the expression of ferroptosis-related genes, reduced reactive oxygen species (ROS) and high-mobility group box 1 (HMGB1) levels, and suppressed the inflammatory response in a SAP mouse model. The HPLC analysis of DCQD constituents indicated eight components (aloe-emodin, rhein, emodin, chrysophanol, naringin, hesperidin, magnolol, and honokiol) with the capacity to modulate ferroptosis. Venn analysis identified 48 DSNFGs, with a subset of five genes demonstrating significant predictive value. The developed nomogram, based on LASSO regression, showed high accuracy in validation cohorts. Single-cell RNA sequencing (scRNA-seq) and CellChat analysis uncovered heterogeneity and cell-cell communication networks in the pancreas during recovery from pancreatitis, implicating several signaling pathways. CONCLUSION DCQD and its eight ingredients exert its protective effect in SAP by inhibiting ferroptosis through the NOX2/GPX4 pathway. The DCQD-SAP-ferroptosis-related signature and nomogram offer a novel tool for AP risk assessment, prognosis prediction, and personalized therapeutic strategies in SAP management.
Collapse
Affiliation(s)
- Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand
| | - Fu Li
- Department of Hepatopancreatobiliary Surgery, Shuguang Hospital affliated to Shanghai University of Traditional Chinese Medincine, Shanghai 201203, China
| | - Wang-Sheng Luo
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421200, China
| | - Mei-Fang Zhu
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Neng-Jiang Zhao
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhi-Hai Zhang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Ya-Feng Chen
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Dian-Xu Feng
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China.
| | - Shu-Yu Yang
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China.
| | - Wen-Jie Sun
- Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China; Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China; Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
2
|
Ozcan M, Cicek C, Gok M. Exploring the GSTP1 inhibition potential of photosensitizer compounds for enhanced cancer treatment in photodynamic therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6963-6972. [PMID: 39702601 DOI: 10.1007/s00210-024-03726-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Photodynamic therapy (PDT) has gained considerable attention in cancer treatment due to its non-invasive nature and the ability of photosensitizers to generate reactive oxygen species upon light activation, leading to tumor destruction. Glutathione S-transferase P1 (GSTP1) is a key enzyme in chemotherapy resistance, often overexpressed in various cancers, and its inhibition of GSTP1 presents a promising strategy to enhance cancer treatment. This study is aimed at assessing the potential of prominent photosensitizers as GSTP1 inhibitors through molecular docking analysis to strengthen the efficacy of PDT. The photosensitizers were docked into the active site of GSTP1, and their binding affinities, inhibition constants (Ki), and molecular interactions were assessed. Among the tested photosensitizers, zinc phthalocyanine, hypericin, and temoporfin emerged as the top candidates, exhibiting binding energies of - 10.8, - 10.2, and - 9.8 kcal/mol, along with Ki values of 0.012, 0.033, and 0.064 µM, respectively. These compounds outperformed the reference inhibitor ethacrynic acid, which had a binding energy of - 6.6 kcal/mol and a Ki of 14.35 µM. These findings suggest that the dual action of these photosensitizers provides a promising strategy for combating cancer and overcoming treatment resistance.
Collapse
Affiliation(s)
- Mehmet Ozcan
- Department of Medical Biochemistry, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey.
| | - Cigdem Cicek
- Department of Medical Biochemistry, Faculty of Medicine, Yuksek Ihtisas University, Ankara, Turkey
| | - Muslum Gok
- Department of Medical Biochemistry, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
3
|
Xiao H, Raza F, Li K, Song J, Zafar H, Yang S, Su J, Qiu M. Cell membrane derived biomimetic nanomedicine for precision delivery of traditional Chinese medicine in cancer therapy. J Control Release 2025; 383:113829. [PMID: 40355044 DOI: 10.1016/j.jconrel.2025.113829] [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: 02/19/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
The rapidly developing modern nanotechnology has brought new vitality to the application of traditional Chinese medicine (TCM), improving the pharmacokinetics and bioavailability of unmodified natural drugs. However, synthetic materials inevitably introduce incompatibilities. This has led to focusing on biomimetic drug delivery systems (DDS) based on biologically derived cell membranes. This "top-down" approach to nanomedicine preparation is simple and effective, as the inherited cell membranes and cell surface substances can mimic nature when delivering drugs back into the body, interacting similarly to the source cells at the biological interface. The concept of biologically derived TCM and biomimetic membranes aligns well with nature, the human body, and medicine, thereby enhancing the in vivo compatibility of TCM. This review focused on the recent progress using biomimetic membranes for TCM in cancer therapy, emphasizing the effective integration of biomimetic nanomedicine and TCM in applications such as cancer diagnosis, imaging, precision treatment, and immunotherapy. The review also provided potential suggestions on the challenges and prospects in this field.
Collapse
Affiliation(s)
- Hang Xiao
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunwei Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinpu Song
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiqi Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
4
|
Lhaglham P, Jiramonai L, Jia Y, Huang B, Huang Y, Gao X, Zhang J, Liang XJ, Zhu M. Drug nanocrystals: Surface engineering and its applications in targeted delivery. iScience 2024; 27:111185. [PMID: 39555405 PMCID: PMC11564948 DOI: 10.1016/j.isci.2024.111185] [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: 11/19/2024] Open
Abstract
Drug nanocrystals have received significant attention in drug development due to their enhanced dissolution rate and improved water solubility, making them effective in overcoming issues related to drug hydrophobicity, thereby improving drug bioavailability and treatment effectiveness. Recent advances in preparation techniques have facilitated research on drug surface properties, leading to valuable surface engineering strategies. Surface modification can stabilize drug nanocrystals, making them suitable for versatile drug delivery platforms. Functionalized ligands further enhance the potential for targeted delivery, enabling precision medicine. This review focuses on the surface engineering of drug nanocrystals, discussing various preparation methods, surface ligand design strategies, and their applications in targeted drug delivery, especially for cancer treatments. Finally, challenges and future directions are also discussed to promote the development of drug nanocrystals. The surface engineering of drug nanocrystals promises new opportunities for treating complex and chronic diseases while broadening the application of drug delivery systems.
Collapse
Affiliation(s)
- Phattalapol Lhaglham
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Manufacturing Pharmacy, Faculty of Pharmacy, Mahidol University, Sri-ayudhya Road, Bangkok 10400, Thailand
| | - Luksika Jiramonai
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaru Jia
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Baoying Huang
- MHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, China CDC, Beijing 102206, China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xueyun Gao
- Center of Excellence for Environmental Safety and Biological Effects, Beijing Key Laboratory for Green Catalysis and Separation, Department of Chemistry, Beijing University of Technology, Beijing 100124, China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengliang Zhu
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Peng H, Jiang Q, Mao W, Hu Z, Wang Q, Yu Z, Zhang L, Wang X, Zhuang C, Mai J, Wang Z, Sun T. Fe-HCOF-PEG 2000 as a Hypoxia-Tolerant Photosensitizer to Trigger Ferroptosis and Enhance ROS-Based Cancer Therapy. Int J Nanomedicine 2024; 19:10165-10183. [PMID: 39399828 PMCID: PMC11468433 DOI: 10.2147/ijn.s479848] [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: 07/24/2024] [Accepted: 09/29/2024] [Indexed: 10/15/2024] Open
Abstract
Background The hypoxic tumor microenvironment and single mechanisms severely limit the photodynamic therapy (PDT) efficiency of covalent organic framework (COF) nanoparticles in cancer treatment. Purpose Here, we propose an iron-loaded, hydrophilic 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG2000)-modified hollow covalent organic framework (HCOF), Fe-HCOF-PEG2000, for use in hypoxic PDT and ferroptosis therapy owing to its type I and II photodynamic ability and iron nanoparticle loading property. Results Fe-HCOF-PEG2000 nanoparticles (Fe-HCOFs-PEG2000) with semiconducting polymers and microporous skeletons allow efficient photophysical properties. Moreover, the iron nanoparticles on Fe-HCOF-PEG2000 caused ferroptosis and further enhanced tumor elimination under normoxic and hypoxic conditions. DSPE-PEG2000 endowed Fe-HCOF-PEG2000 with hydrophilicity, allowing it to circulate and accumulate in organs rich in blood supply, especially tumors. 808 nm NIR activated Fe-HCOF-PEG2000 aggregated in tumors and significantly inhibited tumor growth under hypoxia. Conclusion To our knowledge, Fe-HCOF-PEG2000 is the leading combination of type I/II PDT and ferroptosis. The strong antitumor effects of this nanomaterial suggest prospects for clinical translation as a tumor nanotherapy drug.
Collapse
Affiliation(s)
- Hui Peng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Center for Clinical Laboratory, General Hospital of the Yangtze River Shipping Wuhan Brain Hospital, Wuhan, Hubei, 430010, People’s Republic of China
| | - Qian Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Wenhao Mao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Zhonglan Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Qi Wang
- Department of Pharmacy, Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, 475000, People’s Republic of China
| | - Zhuo Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Li Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Xinyan Wang
- Department of Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Chunbo Zhuang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Jia Mai
- Department of Laboratory Medicine, West China Second Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Zhiyuan Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| |
Collapse
|
6
|
Zhang Y, Xiao B, Yuan S, Ding L, Pan Y, Jiang Y, Sun S, Ke X, Cai L, Jia L. Tryptanthrin targets GSTP1 to induce senescence and increases the susceptibility to apoptosis by senolytics in liver cancer cells. Redox Biol 2024; 76:103323. [PMID: 39180983 PMCID: PMC11388193 DOI: 10.1016/j.redox.2024.103323] [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: 07/13/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024] Open
Abstract
Targeting senescence has emerged as a promising strategy for liver cancer treatment. However, the lack of a safe agent capable of inducing complete senescence and being combined with senolytics poses a limitation. Here, we screened a natural product library and identified tryptanthrin (TRYP) as a potent inducer of cellular senescence in liver cancer cells both in vitro and in vivo. Mechanistically, Glutathione S-transferase P1 (GSTP1), a key regulator for redox homeostasis, was identified as a target protein for TRYP-induced senescence. TRYP directly bound to GSTP1 and inhibited its enzymatic activity, mediating reactive oxygen species (ROS) accumulation, followed by DNA damage response (DDR), consequently contributing to initiating primary senescence. Furthermore, TRYP triggered DNA damage-dependent activation of NF-κB pathway, which evoked senescence-associated secretory phenotype (SASP), thereby leading to senescence reinforcement. Importantly, TRYP exposed the vulnerability of tumor cells and sensitized senescent cells to apoptosis induced by senolytic agent ABT263, a Bcl2 inhibitor. Taken together, our findings reveal that TRYP induces cellular senescence via GSTP1/ROS/DDR/NF-κB/SASP axis, providing a novel potential application in synergizing with senolytic therapy in liver cancer.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Biying Xiao
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shuying Yuan
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Lele Ding
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yongfu Pan
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yanyu Jiang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Shenghao Sun
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Xisong Ke
- Center for Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Lili Cai
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| |
Collapse
|
7
|
Yi X, Wang Q, Zhang M, Shu Q, Zhu J. Ferroptosis: A novel therapeutic target of natural products against doxorubicin-induced cardiotoxicity. Biomed Pharmacother 2024; 178:117217. [PMID: 39079260 DOI: 10.1016/j.biopha.2024.117217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Doxorubicin (DOX), a commonly used chemotherapy drug, is hindered due to its tendency to induce cardiotoxicity (DIC). Ferroptosis, a novel mode of programmed cell death, has received substantial attention for its involvement in DIC. Recently, natural product-derived ferroptosis regulator emerged as a potential strategy for treating DIC. In this review, a comprehensive search was conducted across PubMed, Web of Science, Google Scholar, and ScienceDirect databases to gather relevant articles on the use of natural products for treating DIC in relation to ferroptosis. The available papers were carefully reviewed to summarize the therapeutic effects and underlying mechanisms of natural products in modulating ferroptosis for DIC treatment. It was found that ferroptosis plays an important role in DIC pathogenesis, with dysregulated expression of ferroptosis-related proteins strongly implicated in the condition. Natural products, such as flavonoids, polyphenols, terpenoids, and quinones can act as GPX4 activators, Nrf2 agonists, and lipid peroxidation inhibitors, thereby enhancing cell viability, attenuating myocardial fibrosis, improving cardiac function, and suppressing ferroptosis in both in vitro and in vivo models of DIC. This review demonstrates a strong correlation between DOX-induced cardiac ferroptosis and key proteins, such as GPX4, Keap1, Nrf2, AMPK, and HMOX1. Natural products are likely to exert therapeutic effects against DIC by modulating the activity of these proteins.
Collapse
Affiliation(s)
- Xiaojiao Yi
- Department of Pharmacy, Hangzhou Xixi Hospital, Hangzhou Sixth People's Hospital, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Qi Wang
- Department of Pharmacy, Hangzhou Xixi Hospital, Hangzhou Sixth People's Hospital, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Mengjie Zhang
- Department of Pharmacy, Hangzhou Xixi Hospital, Hangzhou Sixth People's Hospital, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Qi Shu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| | - Junfeng Zhu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
8
|
An N, Tang S, Wang Y, Luan J, Shi Y, Gao M, Guo C. FeP-Based Nanotheranostic Platform for Enhanced Phototherapy/Ferroptosis/Chemodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309940. [PMID: 38534030 DOI: 10.1002/smll.202309940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Ferroptosis is an iron-dependent and lipid peroxides (LPO)-overloaded programmed damage cell death, induced by glutathione (GSH) depletion and glutathione peroxide 4 (GPX4) inactivation. However, the inadequacy of endogenous iron and reactive oxygen species (ROS) restricts the efficacy of ferroptosis. To overcome this obstacle, a near-infrared photo-responsive FeP@PEG NPs is fabricated. Exogenous iron pool can enhance the effect of ferroptosis via the depletion of GSH and further regulate GPX4 inactivation. Generation of ·OH derived from the Fenton reaction is proved by increased accumulation of lipid peroxides. The heat generated by photothermal therapy and ROS generated by photodynamic therapy can enhance cell apoptosis under near-infrared (NIR-808 nm) irradiation, as evidenced by mitochondrial dysfunction and further accumulation of lipid peroxide content. FeP@PEG NPs can significantly inhibit the growth of several types of cancer cells in vitro and in vivo, which is validated by theoretical and experimental results. Meanwhile, FeP@PEG NPs show excellent T2-weighted magnetic resonance imaging (MRI) property. In summary, the FeP-based nanotheranostic platform for enhanced phototherapy/ferroptosis/chemodynamic therapy provides a reliable opportunity for clinical cancer theranostics.
Collapse
Affiliation(s)
- Na An
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Shuanglong Tang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yuwei Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Jing Luan
- The HIT Center for Life Science, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Ying Shi
- Magnetic Resonance Department of the First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Minghui Gao
- The HIT Center for Life Science, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Chongshen Guo
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
9
|
Wu X, Xin Y, Zhang H, Quan L, Ao Q. Biopolymer-Based Nanomedicine for Cancer Therapy: Opportunities and Challenges. Int J Nanomedicine 2024; 19:7415-7471. [PMID: 39071502 PMCID: PMC11278852 DOI: 10.2147/ijn.s460047] [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: 02/15/2024] [Accepted: 05/18/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer, as the foremost challenge among human diseases, has plagued medical professionals for many years. While there have been numerous treatment approaches in clinical practice, they often cause additional harm to patients. The emergence of nanotechnology has brought new directions for cancer treatment, which can deliver anticancer drugs specifically to tumor areas. This article first introduces the application scenarios of nanotherapies and treatment strategies of nanomedicine. Then, the noteworthy characteristics exhibited by biopolymer materials were described, which make biopolymers stand out in polymeric nanomedicine delivery. Next, we focus on summarizing the state-of-art studies of five categories of proteins (Albumin, Gelatin, Silk fibroin, Zein, Ferritin), nine varieties of polysaccharides (Chitosan, Starch, Hyaluronic acid, Dextran, cellulose, Fucoidan, Carrageenan, Lignin, Pectin) and liposomes in the field of anticancer drug delivery. Finally, we also provide a summary of the advantages and limitations of these biopolymers, discuss the prevailing impediments to their application, and discuss in detail the prospective research directions. This review not only helps readers understand the current development status of nano anticancer drug delivery systems based on biopolymers, but also is helpful for readers to understand the properties of various biopolymers and find suitable solutions in this field through comparative reading.
Collapse
Affiliation(s)
- Xixi Wu
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Yuan Xin
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Hengtong Zhang
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Liang Quan
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| |
Collapse
|
10
|
Prasad R, Jyothi VGS, Kommineni N, Bulusu RT, Mendes B, Lovell JF, Conde J. Biomimetic Ghost Nanomedicine-Based Optotheranostics for Cancer. NANO LETTERS 2024; 24:8217-8231. [PMID: 38848540 PMCID: PMC11247544 DOI: 10.1021/acs.nanolett.4c01534] [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: 03/31/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Theranostic medicine combines diagnostics and therapeutics, focusing on solid tumors at minimal doses. Optically activated photosensitizers are significant examples owing to their photophysical and chemical properties. Several optotheranostics have been tested that convert light to imaging signals, therapeutic radicals, and heat. Upon light exposure, conjugated photosensitizers kill tumor cells by producing reactive oxygen species and heat or by releasing cancer antigens. Despite clinical trials, these molecularly conjugated photosensitizers require protection from their surroundings and a localized direction for site-specific delivery during blood circulation. Therefore, cell membrane biomimetic ghosts have been proposed for precise and safe delivery of these optically active large molecules, which are clinically relevant because of their biocompatibility, long circulation time, bypass of immune cell recognition, and targeting ability. This review focuses on the role of biomimetic nanoparticles in the treatment and diagnosis of tumors through light-mediated diagnostics and therapy, providing insights into their preclinical and clinical status.
Collapse
Affiliation(s)
- Rajendra Prasad
- School
of Biochemical Engineering, Indian Institute
of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Vaskuri G. S. Jyothi
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center (UTHSC), Memphis, Tennessee 38163, United States
| | - Nagavendra Kommineni
- Center
for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ravi Teja Bulusu
- Department
of Pharmaceutical Sciences, Florida A&M
University, Tallahassee, Florida 32307, United States
| | - Bárbara
B. Mendes
- NOVA
Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon 1169-056, Portugal
- ToxOmics,
NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, 1169-056, Portugal
| | - Jonathan F. Lovell
- Department
of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - João Conde
- NOVA
Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon 1169-056, Portugal
- ToxOmics,
NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, 1169-056, Portugal
| |
Collapse
|
11
|
Nkune NW, Abrahamse H. The phototoxic effect of a gold-antibody-based nanocarrier of phthalocyanine on melanoma monolayers and tumour spheroids. RSC Adv 2024; 14:19490-19504. [PMID: 38895533 PMCID: PMC11184583 DOI: 10.1039/d4ra03858d] [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: 05/25/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024] Open
Abstract
In recent years, photodynamic therapy (PDT) has garnered significant attention in cancer treatment due to its increased potency and non-invasiveness compared to conventional therapies. Active-targeted delivery of photosensitizers (PSs) is a mainstay strategy to significantly reduce its off-target toxicity and enhance its phototoxic efficacy. The anti-melanoma inhibitory activity (MIA) antibody is a targeting biomolecule that can be integrated into a nanocarrier system to actively target melanoma cells due to its specific binding to MIA antigens that are highly expressed on the surface of melanoma cells. Gold nanoparticles (AuNPs) are excellent nanocarriers due to their ability to encapsulate a variety of therapeutics, such as PSs, and their ability to bind with targeting moieties for improved bioavailability in cancer cells. Hence, we designed a nanobioconjugate (NBC) composed of zinc phthalocyanine tetrasulfonic acid (ZnPcS4), AuNPs and anti-MIA Ab to improve ZnPcS4 bioavailability and phototoxicity in two and three-dimensional tumour models. In summary, we demonstrated that this nanobioconjugate showed significant inhibitory effects on both melanoma models due to increased ROS yields and bioavailability of the melanoma cells compared to free ZnPcS4.
Collapse
Affiliation(s)
- Nkune Williams Nkune
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg P.O. Box 17011 Doornfontein 2028 South Africa +27-11-559-655
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg P.O. Box 17011 Doornfontein 2028 South Africa +27-11-559-655
| |
Collapse
|
12
|
Wang X, Feng JH, Zeng CM, Zhang ZS, Cao FL, Zhang WH, Chen JX, Young DJ. [Fe IIICl(TMPPH 2)][Fe IIICl 4] 2: A Stand-Alone Molecular Nanomedicine That Induces High Cytotoxicity by Ferroptosis. Molecules 2024; 29:2495. [PMID: 38893373 PMCID: PMC11173869 DOI: 10.3390/molecules29112495] [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: 04/28/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Developing clinically meaningful nanomedicines for cancer therapy requires the drugs to be effective, safe, simple, cheap, and easy to store. In the present work, we report that a simple cationic Fe(III)-rich salt of [FeIIICl(TMPPH2)][FeIIICl4]2 (Fe-TMPP) exhibits a superior anticancer performance on a broad spectrum of cancer cell lines, including breast, colorectal cancer, liver, pancreatic, prostate, and gastric cancers, with half maximal inhibitory concentration (IC50) values in the range of 0.098-3.97 μM (0.066-2.68 μg mL-1), comparable to the best-reported medicines. Fe-TMPP can form stand-alone nanoparticles in water without the need for extra surface modification or organic-solvent-assisted antisolvent precipitation. Critically, Fe-TMPP is TME-responsive (TME = tumor microenvironment), and can only elicit its function in the TME with overexpressed H2O2, converting H2O2 to the cytotoxic •OH to oxidize the phospholipid of the cancer cell membrane, causing ferroptosis, a programmed cell death process of cancer cells.
Collapse
Affiliation(s)
- Xiao Wang
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University, Suzhou 215123, China; (X.W.); (C.-M.Z.); (Z.-S.Z.); (F.-L.C.)
| | - Jia-Hao Feng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China;
| | - Chun-Mei Zeng
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University, Suzhou 215123, China; (X.W.); (C.-M.Z.); (Z.-S.Z.); (F.-L.C.)
| | - Ze-Sheng Zhang
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University, Suzhou 215123, China; (X.W.); (C.-M.Z.); (Z.-S.Z.); (F.-L.C.)
| | - Feng-Lin Cao
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University, Suzhou 215123, China; (X.W.); (C.-M.Z.); (Z.-S.Z.); (F.-L.C.)
| | - Wen-Hua Zhang
- College of Chemistry, Chemical Engineering, and Materials Science, Soochow University, Suzhou 215123, China; (X.W.); (C.-M.Z.); (Z.-S.Z.); (F.-L.C.)
| | - Jin-Xiang Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China;
| | - David J. Young
- Glasgow College UESTC, University of Electronic Science and Technology of China, Chengdu 611731, China;
| |
Collapse
|
13
|
Ai S, Li Y, Zheng H, Zhang M, Tao J, Liu W, Peng L, Wang Z, Wang Y. Collision of herbal medicine and nanotechnology: a bibliometric analysis of herbal nanoparticles from 2004 to 2023. J Nanobiotechnology 2024; 22:140. [PMID: 38556857 PMCID: PMC10983666 DOI: 10.1186/s12951-024-02426-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Herbal nanoparticles are made from natural herbs/medicinal plants, their extracts, or a combination with other nanoparticle carriers. Compared to traditional herbs, herbal nanoparticles lead to improved bioavailability, enhanced stability, and reduced toxicity. Previous research indicates that herbal medicine nanomaterials are rapidly advancing and making significant progress; however, bibliometric analysis and knowledge mapping for herbal nanoparticles are currently lacking. We performed a bibliometric analysis by retrieving publications related to herbal nanoparticles from the Web of Science Core Collection (WoSCC) database spanning from 2004 to 2023. Data processing was performed using the R package Bibliometrix, VOSviewers, and CiteSpace. RESULTS In total, 1876 articles related to herbal nanoparticles were identified, originating from various countries, with China being the primary contributing country. The number of publications in this field increases annually. Beijing University of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, and Saveetha University in India are prominent research institutions in this domain. The Journal "International Journal of Nanomedicine" has the highest number of publications. The number of authors of these publications reached 8234, with Yan Zhao, Yue Zhang, and Huihua Qu being the most prolific authors and Yan Zhao being the most frequently cited author. "Traditional Chinese medicine," "drug delivery," and "green synthesis" are the main research focal points. Themes such as "green synthesis," "curcumin," "wound healing," "drug delivery," and "carbon dots" may represent emerging research areas. CONCLUSIONS Our study findings assist in identifying the latest research frontiers and hot topics, providing valuable references for scholars investigating the role of nanotechnology in herbal medicine.
Collapse
Affiliation(s)
- Sinan Ai
- China-Japan Friendship Hospital, Beijing, China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huijuan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Meiling Zhang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiayin Tao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Beijing, China.
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, Beijing, China.
| | - Zhen Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Yaoxian Wang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| |
Collapse
|
14
|
Li X, Liu Y, Wu L, Zhao J. Molecular Nanoarchitectonics of Natural Photosensitizers and Their Derivatives Nanostructures for Improved Photodynamic Therapy. ChemMedChem 2024; 19:e202300599. [PMID: 38069595 DOI: 10.1002/cmdc.202300599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/01/2023] [Indexed: 01/25/2024]
Abstract
Natural photosensitizers (PSs) and their derivatives have drawn ever-increasing attention in photodynamic therapy (PDT) for their wild range of sources, desirable biocompatibility, and good photosensitivity. Nevertheless, many factors such as poor solubility, high body clearance rate, limited tumor targeting ability, and short excitation wavelengths severely hinder their applications in efficient PDT. In recent years, fabricating nanostructures by utilizing molecular assembly technique is proposed to solve these problems. This technique is easy to put into effect, and the assembled nanostructures could improve the physical properties of the PSs so as to meet the requirement of PDT. In this concept, we focus on the construction of natural PSs and their derivatives nanostructures through molecular assembly technique to enhance PDT efficacy (Figure 1). Furthermore, current challenges and future perspectives of natural PSs and their derivatives for efficient PDT are discussed.
Collapse
Affiliation(s)
- Xiaochen Li
- Shaanxi University of Chinese Medicine, 712046, Xianyang, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, 100029, Beijing, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Yilin Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS, Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, 100029, Beijing, China
- Key Laboratory of Health Cultivation of Beijing, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Jie Zhao
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS, Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
| |
Collapse
|
15
|
Xu Y, Yang L, Wang C, Sun W, Zheng Y, Ou B, Wu L, Shi L, Lin X, Chen W. Ferroptosis boosted oral cancer photodynamic therapy by carrier-free Sorafenib-Ce6 self-assembly nanoparticles. J Control Release 2024; 366:798-811. [PMID: 38184236 DOI: 10.1016/j.jconrel.2023.12.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Oral cancer is a disease with high morbidity and mortality worldwide and greatly impacts the quality of life, especially in patients with advanced stages. Photodynamic therapy (PDT) is one of the most effective clinical treatments for oral cancers. However, most clinically applied photosensitizers have several deficiencies, including oxygen dependence, poor aqueous solubility, and a lack of tumor-targeting ability. Herein, the carrier-free multifunctional Sorafenib (Sor), chlorin e6 (Ce6), and Fe3+ self-assembly co-delivery nanoparticles (Sor-Ce6 NPs) were constructed via combining a ferroptosis inducer Sor and a photosensitizer Ce6 for synergetic therapy. The as-synthesized Sor-Ce6 NPs presented excellent colloidal stability and water dispersity with good in vivo tumor-targeting ability. More significantly, the low dose of Sor-Ce6 NPs had little dark toxicity but produced significantly enhanced ROS and supplied O2 sustainably to increase phototoxicity through ferroptosis pathway. Notably, the Sor-Ce6 NPs showed significantly higher in vitro and in vivo anti-tumor efficacy than the Sor/Ce6 mixture due to the improvement of cellular uptake and the incorporation of foreign Fe ions in the system, which also confer the T1 magnetic resonance-guided imaging ability to the formed Sor-Ce6 NPs. Our study demonstrates a promising self-assembled strategy for overcoming hypoxia-related PDT resistance for oral cancer treatment.
Collapse
Affiliation(s)
- Yingying Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Liu Yang
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Chengyan Wang
- Laboratory Animal Center, Fujian Medical University, Fuzhou 350122, China
| | - Weiming Sun
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yijing Zheng
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Beiwei Ou
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lixian Wu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xi Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou 350122, China.
| | - Wei Chen
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
16
|
Luo H, Ji X, Zhang M, Ren Y, Tan R, Jiang H, Wu X. Aloe-emodin: Progress in Pharmacological Activity, Safety, and Pharmaceutical Formulation Applications. Mini Rev Med Chem 2024; 24:1784-1798. [PMID: 38639277 DOI: 10.2174/0113895575298364240409064833] [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: 02/04/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Aloe-emodin (AE) is an anthraquinone derivative and a biologically active component sourced from various plants, including Rheum palmatum L. and Aloe vera. Known chemically as 1,8-dihydroxy-3-hydroxymethyl-anthraquinone, AE has a rich history in traditional medicine and is esteemed for its accessibility, safety, affordability, and effectiveness. AE boasts multiple biochemical and pharmacological properties, such as strong antibacterial, antioxidant, and antitumor effects. Despite its array of benefits, AE's identity as an anthraquinone derivative raises concerns about its potential for liver and kidney toxicity. Nevertheless, AE is considered a promising drug candidate due to its significant bioactivities and cost efficiency. Recent research has highlighted that nanoformulated AE may enhance drug delivery, biocompatibility, and pharmacological benefits, offering a novel approach to drug design. This review delves into AE's pharmacological impacts, mechanisms, pharmacokinetics, and safety profile, incorporating insights from studies on its nanoformulations. The goal is to outline the burgeoning research in this area and to support the ongoing development and utilization of AE-based therapies.
Collapse
Affiliation(s)
- Haimeng Luo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaoyun Ji
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Mengyu Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yaoyao Ren
- School of Chemistry, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Rui Tan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Hezhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaoqing Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
17
|
Huang Y, Li X, Zhang Z, Xiong L, Wang Y, Wen Y. Photodynamic Therapy Combined with Ferroptosis Is a Synergistic Antitumor Therapy Strategy. Cancers (Basel) 2023; 15:5043. [PMID: 37894410 PMCID: PMC10604985 DOI: 10.3390/cancers15205043] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Ferroptosis is a programmed death mode that regulates redox homeostasis in cells, and recent studies suggest that it is a promising mode of tumor cell death. Ferroptosis is regulated by iron metabolism, lipid metabolism, and intracellular reducing substances, which is the mechanism basis of its combination with photodynamic therapy (PDT). PDT generates reactive oxygen species (ROS) and 1O2 through type I and type II photochemical reactions, and subsequently induces ferroptosis through the Fenton reaction and the peroxidation of cell membrane lipids. PDT kills tumor cells by generating excessive cytotoxic ROS. Due to the limited laser depth and photosensitizer enrichment, the systemic treatment effect of PDT is not good. Combining PDT with ferroptosis can compensate for these shortcomings. Nanoparticles constructed by photosensitizers and ferroptosis agonists are widely used in the field of combination therapy, and their targeting and biological safety can be improved through modification. These nanoparticles not only directly kill tumor cells but also further exert the synergistic effect of PDT and ferroptosis by activating antitumor immunity, improving the hypoxia microenvironment, and inhibiting the tumor angiogenesis. Ferroptosis-agonist-induced chemotherapy and PDT-induced ablation also have good clinical application prospects. In this review, we summarize the current research progress on PDT and ferroptosis and how PDT and ferroptosis promote each other.
Collapse
Affiliation(s)
- Yunpeng Huang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Xiaoyu Li
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China;
| | - Zijian Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yongxiang Wang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| | - Yu Wen
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.H.); (Z.Z.); (L.X.)
| |
Collapse
|
18
|
Chen Z, Wang W, Abdul Razak SR, Han T, Ahmad NH, Li X. Ferroptosis as a potential target for cancer therapy. Cell Death Dis 2023; 14:460. [PMID: 37488128 PMCID: PMC10366218 DOI: 10.1038/s41419-023-05930-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/24/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023]
Abstract
Ferroptosis is a recently discovered essential type of cell death that is mainly characterized by iron overload and lipid peroxidation. Emerging evidence suggests that ferroptosis is a double-edged sword in human cancer. However, the precise underlying molecular mechanisms and their differential roles in tumorigenesis are unclear. Therefore, in this review, we summarize and briefly present the key pathways of ferroptosis, paying special attention to the regulation of ferroptosis as well as its dual role as an oncogenic and as a tumor suppressor event in various human cancers. Moreover, multiple pharmacological ferroptosis activators are summarized, and the prospect of targeting ferroptosis in cancer therapy is further elucidated.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Pulau Pinang, Malaysia
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Weilong Wang
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, China
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Pulau Pinang, Malaysia
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Siti Razila Abdul Razak
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Pulau Pinang, Malaysia
| | - Tao Han
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, China
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Nor Hazwani Ahmad
- Department of Biomedical Science, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam 13200, Kepala Batas, Pulau Pinang, Malaysia.
| | - Xiumin Li
- Department of Gastroenterology, the Third Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Tumor Molecular Therapy Medicine, Xinxiang, 453003, Henan Province, China.
- Xinxiang Key Laboratory for Molecular Therapy of Cancer, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China.
| |
Collapse
|
19
|
Wang H, Qiao C, Guan Q, Wei M, Li Z. Nanoparticle-mediated synergistic anticancer effect of ferroptosis and photodynamic therapy: Novel insights and perspectives. Asian J Pharm Sci 2023; 18:100829. [PMID: 37588992 PMCID: PMC10425855 DOI: 10.1016/j.ajps.2023.100829] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/05/2023] [Accepted: 07/02/2023] [Indexed: 08/18/2023] Open
Abstract
Current antitumor monotherapy has many limitations, highlighting the need for novel synergistic anticancer strategies. Ferroptosis is an iron-dependent form of nonapoptotic cell death that plays a pivotal regulatory role in tumorigenesis and treatment. Photodynamic therapy (PDT) causes irreversible chemical damage to target lesions and is widely used in antitumor therapy. However, PDT's effectiveness is usually hindered by several obstacles, such as hypoxia, excess glutathione (GSH), and tumor resistance. Ferroptosis improves the anticancer efficacy of PDT by increasing oxygen and reactive oxygen species (ROS) or reducing GSH levels, and PDT also enhances ferroptosis induction due to the ROS effect in the tumor microenvironment (TME). Strategies based on nanoparticles (NPs) can subtly exploit the potential synergy of ferroptosis and PDT. This review explores recent advances and current challenges in the landscape of the underlying mechanisms regulating ferroptosis and PDT, as well as nano delivery system-mediated synergistic anticancer activity. These include polymers, biomimetic materials, metal organic frameworks (MOFs), inorganics, and carrier-free NPs. Finally, we highlight future perspectives of this novel emerging paradigm in targeted cancer therapies.
Collapse
Affiliation(s)
- Haiying Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chu Qiao
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qiutong Guan
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zhenhua Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
20
|
Avancini G, Menilli L, Visentin A, Milani C, Mastrotto F, Moret F. Mesenchymal Stem Cell Membrane-Coated TPCS 2a-Loaded Nanoparticles for Breast Cancer Photodynamic Therapy. Pharmaceutics 2023; 15:1654. [PMID: 37376102 PMCID: PMC10302938 DOI: 10.3390/pharmaceutics15061654] [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: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Despite substantial improvements in breast cancer (BC) treatment there is still an urgent need to find alternative treatment options to improve the outcomes for patients with advanced-stage disease. Photodynamic therapy (PDT) is gaining a lot of attention as a BC therapeutic option because of its selectivity and low off-target effects. However, the hydrophobicity of photosensitizers (PSs) impairs their solubility and limits the circulation in the bloodstream, thus representing a major challenge. The use of polymeric nanoparticles (NPs) to encapsulate the PS may represent a valuable strategy to overcome these issues. Herein, we developed a novel biomimetic PDT nanoplatform (NPs) based on a polymeric core of poly(lactic-co-glycolic)acid (PLGA) loaded with the PS meso-tetraphenylchlorin disulfonate (TPCS2a). TPCS2a@NPs of 98.89 ± 18.56 nm with an encapsulation efficiency percentage (EE%) of 81.9 ± 7.92% were obtained and coated with mesenchymal stem cells-derived plasma membranes (mMSCs) (mMSC-TPCS2a@NPs, size of 139.31 ± 12.94 nm). The mMSC coating armed NPs with biomimetic features to impart long circulation times and tumor-homing capabilities. In vitro, biomimetic mMSC-TPCS2a@NPs showed a decrease in macrophage uptake of 54% to 70%, depending on the conditions applied, as compared to uncoated TPCS2a@NPs. Both NP formulations efficiently accumulated in MCF7 and MDA-MB-231 BC cells, while the uptake was significantly lower in normal breast epithelial MCF10A cells with respect to tumor cells. Moreover, encapsulation of TPCS2a in mMSC-TPCS2a@NPs effectively prevents its aggregation, ensuring efficient singlet oxygen (1O2) production after red light irradiation, which resulted in a considerable in vitro anticancer effect in both BC cell monolayers (IC50 < 0.15 µM) and three-dimensional spheroids.
Collapse
Affiliation(s)
- Greta Avancini
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Luca Menilli
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Adele Visentin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Celeste Milani
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Francesca Moret
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| |
Collapse
|
21
|
Tan X, Huang X, Niu B, Guo X, Lei X, Qu B. Targeting GSTP1-dependent ferroptosis in lung cancer radiotherapy: Existing evidence and future directions. Front Mol Biosci 2022; 9:1102158. [PMID: 36589232 PMCID: PMC9800622 DOI: 10.3389/fmolb.2022.1102158] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Radiotherapy is applied in about 70% patients with tumors, yet radioresistance of tumor cells remains a challenge that limits the efficacy of radiotherapy. Ferroptosis, an iron-dependent lipid peroxidation regulated cell death, is involved in the development of a variety of tumors. Interestingly, there is evidence that ferroptosis inducers in tumor treatment can significantly improve radiotherapy sensitivity. In addition, related studies show that Glutathione S-transferase P1 (GSTP1) is closely related to the development of ferroptosis. The potential mechanism of targeting GSTP1 to inhibit tumor cells from evading ferroptosis leading to radioresistance has been proposed in this review, which implies that GSTP1 may play a key role in radiosensitization of lung cancer via ferroptosis pathway.
Collapse
Affiliation(s)
- Xin Tan
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China
| | - Xiang Huang
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Baolong Niu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xingdong Guo
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China
| | - Xiao Lei
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Xiao Lei, ; Baolin Qu,
| | - Baolin Qu
- Department of Radiation Oncology, The First Medical Center of Chinese PLA General Hospital, Beijing, China,*Correspondence: Xiao Lei, ; Baolin Qu,
| |
Collapse
|