1
|
Wei W, Du Y, Kang X, Liu Y, Liu Y, Guo Q, He Q, Wang J, Zhu S, Zhou JY, Bu W. Nanoscale Rhodium(I) Based Metal-Organic Framework Demonstrating Intense NIR-II Luminescence for Bioimaging. NANO LETTERS 2025; 25:4613-4620. [PMID: 40052801 DOI: 10.1021/acs.nanolett.5c00539] [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: 03/20/2025]
Abstract
Although luminescent metal-organic frameworks (MOFs) have been widely reported, rare examples were found to emit in the second near-infrared (NIR-II, 1000-1700 nm) window. In this work, two nanoscale rhodium(I)-based MOFs (Rh-1@SDS and Rh-1@DSPE-PEG) have been controllably constructed in the aqueous dispersions of sodium dodecyl sulfate (SDS) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-methoxy-poly(ethylene glycol) (DSPE-PEG), wherein micelle- and vesicle-like aggregates form, respectively, with high colloidal stability. The vesicular dispersion of Rh-1@DSPE-PEG exhibits intense NIR-II luminescence at 1125 (1245, shoulder) nm. Consequently, this nanoMOF was used as an NIR-II luminescence probe, indicative of high-resolution systemic and local vascular imaging, where the postoperative recovery process of flap transplantation was clearly visualized. Meanwhile, it also demonstrates superior tumor targeting in the NIR-II window. To the best of our knowledge, this research represents the first example of nanoMOFs having intense NIR-II luminescence and excellent imaging capabilities.
Collapse
Affiliation(s)
- Wenxuan Wei
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- School of Physical Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xiaomei Kang
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yilin Liu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yujia Liu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qian Guo
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Qun He
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun 130021, China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jin Yuan Zhou
- School of Physical Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Weifeng Bu
- Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
2
|
Liu Y, Song J, Guo Y, Li S, Yuan M, Tang J, Wang Y, Li M, Guo Y, Guo L. Synergistic therapy with celastrol-curcumin multifunctional nanomedicine: Anti-hepatocellular carcinoma and reduced hepatotoxicity. Int J Pharm 2025; 671:125289. [PMID: 39880142 DOI: 10.1016/j.ijpharm.2025.125289] [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: 10/16/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Hepatocellular carcinoma is one of the leading causes of cancer deaths globally and a key hindrance to extending life expectancy. Celastrol (CEL) demonstrates excellent antitumor activity, but faces challenges like low solubility and a narrow therapeutic window, limiting its clinical application. To address these limitations, drug combinations and nano-delivery systems have emerged as effective solutions. Curcumin (CUR), known for its antitumor and hepatoprotective effects, also exhibits good biocompatibility and the ability to mitigate drug-induced liver injury. Considering the complementary properties of CEL and CUR, including CEL's potent antitumor activity and CUR's hepatoprotective effects, we developed a novel self-assembling nanodrug delivery system (CCPN) for the co-loading of both compounds. CCPN nanoparticles were constructed through non-covalent interactions, including hydrogen bonding, π-π stacking, and electrostatic forces, which confer good stability and significantly enhance the solubility and bioavailability of CEL and CUR. Extensive in vitro and in vivo experiments demonstrated that CCPN effectively reduced CEL-induced hepatotoxicity in zebrafish and mouse models, exhibiting good biosafety. Additionally, CUR's fluorescence provides a unique advantage for real-time monitoring of drug distribution and release, facilitating the tracking of therapeutic progress. Furthermore, CCPN nanoparticles enhanced delivery efficiency in HepG2 cells, exhibiting superior anti-liver tumor outcomes, which are associated with the promotion of apoptosis in tumor cells. This study presents CCPN as a promising therapeutic strategy for hepatocellular carcinoma, integrating reduced hepatotoxicity, self-monitoring capabilities, and superior therapeutic efficacy.
Collapse
Affiliation(s)
- Yushi Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China; Department of Pharmacy, Chengdu Seventh People's Hospital, Chengdu 610213 China
| | - Jiawen Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yurou Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Meifeng Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137 China.
| |
Collapse
|
3
|
Cheng Y, Xu Q, Yu M, Dang C, Deng L, Chen H. Curcumin Nanoparticles-related Non-invasive Tumor Therapy, and Cardiotoxicity Relieve. Curr Med Chem 2025; 32:447-467. [PMID: 38918994 PMCID: PMC11826934 DOI: 10.2174/0109298673305616240610153554] [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: 02/22/2024] [Revised: 05/04/2024] [Accepted: 05/17/2024] [Indexed: 06/27/2024]
Abstract
Non-invasive antitumor therapy can treat tumor patients who cannot tolerate surgery or are unsuitable. However, tumor resistance to non-invasive antitumor therapy and cardiotoxicity caused by treatment seriously affect the quality of life and prognosis of patients. As a kind of polyphenol extracted from herbs, curcumin has many pharmacological effects, such as anti-inflammation, antioxidation, antitumor, etc. Curcumin plays the antitumor effect by directly promoting tumor cell death and reducing tumor cells' invasive ability. Curcumin exerts the therapeutic effect mainly by inhibiting the nuclear factor-κB (NF-κB) signal pathway, inhibiting the production of cyclooxygenase-2 (COX-2), promoting the expression of caspase-9, and directly inducing reactive oxygen species (ROS) production in tumor cells. Curcumin nanoparticles can solve curcumin's shortcomings, such as poor water solubility and high metabolic rate, and can be effectively used in antitumor therapy. Curcumin nanoparticles can improve the prognosis and quality of life of tumor patients by using as adjuvants to enhance the sensitivity of tumors to non-invasive therapy and reduce the side effects, especially cardiotoxicity. In this paper, we collect and analyze the literature of relevant databases. It is pointed out that future research on curcumin tends to alleviate the adverse reactions caused by treatment, which is of more significance to tumor patients.
Collapse
Affiliation(s)
- Yuhang Cheng
- Department of Cardiology, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, China
| | - Qian Xu
- Department of Cardiology, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, No. 411, Guogeli Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Miao Yu
- Department of Cardiology, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, China
| | - Chenwei Dang
- Department of Cardiology, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, China
| | - Limei Deng
- Department of Cardiology, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, No. 411, Guogeli Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| | - Huijun Chen
- Department of Cardiology, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, No. 411, Guogeli Road, Nangang District, Harbin, Heilongjiang Province, 150081, China
| |
Collapse
|
4
|
Guo QH, Jian LY, Hu Y, Wang S. A comprehensive and systematic review on Curcumin as a promising candidate for the inhibition of melanoma growth: From pre-clinical evidence to molecular mechanisms of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156073. [PMID: 39515103 DOI: 10.1016/j.phymed.2024.156073] [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: 04/24/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Melanoma, a highly malignant skin tumor, can develop systemic metastases during the early stage. Several studies of melanoma animal models indicate that curcumin, a natural plant extract, inhibits melanoma growth through various mechanisms. To evaluate the relationships among different experimental conditions, curcumin itself, its derivatives, and special formulations, it is necessary to conduct a systematic review and meta-analysis. PURPOSE This meta-analysis aims to evaluate the potential of Curcumin as a drug for inhibiting the growth of melanoma and to determine the optimal dosage range and treatment duration for Curcumin administration. METHODS A systematic search of studies published from inception to December 2023 was conducted across six databases (PubMed, Web of Science, Embase, China National Knowledge Infrastructure, Wanfang Data, and VIP). Methodological quality was assessed using SYRCLE's RoB tool. Study heterogeneity was assessed using Cochran's Q test and I2 statistics. Publication bias risk was evaluated using a funnel plot. All analyses were performed using R (version 4.3.3). Additionally, three-dimensional effect analysis and machine learning techniques were utilized to determine the optimal dosage range and treatment duration for Curcumin administration. RESULTS Forty studies involving 989 animals were included. The results demonstrated that, relative to the control group, administration of Curcumin resulted in a significant reduction in tumor volume. [SMD=-3.44; 95 % CI (-4.25, -2.63); P<0.01; I2 = 79 %] and tumor weight [SMD=-1.93; 95 % CI (-2.41, -1.45); P<0.01; I2 = 75 %]. Additionally, Curcumin demonstrated a significant capacity to decrease the number of lung tumor nodules and microangiogenesis, as well as to extend survival time, in animal models. The results from three-dimensional effect analysis and machine learning emphasize that the optimal dosage range for Curcumin is 25-50 mg/kg, with an intervention duration of 10-20 days. CONCLUSION Curcumin can inhibit the growth of melanoma, and the dose-response relationship is not linear. However, further large-scale animal and clinical studies are required to confirm these conclusions.
Collapse
Affiliation(s)
- Qi-Hao Guo
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Ling-Yan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Yihan Hu
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China.
| | - Shu Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China.
| |
Collapse
|
5
|
Wang S, Liu X, Wei D, Zhou H, Zhu J, Yu Q, Luo L, Dai X, Jiang Y, Yu L, Yang Y, Tan W. Polyvalent Aptamer Nanodrug Conjugates Enable Efficient Tumor Cuproptosis Therapy Through Copper Overload and Glutathione Depletion. J Am Chem Soc 2024; 146:30033-30045. [PMID: 39463177 DOI: 10.1021/jacs.4c06338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Cuproptosis, a recently identified form of copper-dependent cell death, shows promising tumor suppressive effects with minimal drug resistance. However, its therapeutic efficacy is hampered by its dependence on copper ions and the glutathione (GSH)-rich microenvironment in tumors. Here, we have developed polyvalent aptamer nanodrug conjugates (termed CuPEs@PApt) with a nucleosome-like structure to improve tumor cuproptosis therapy by exploiting mitochondrial copper overload and GSH depletion. Polyvalent aptamer (PApt), comprising polyvalent epithelial cell adhesion molecule aptamers for tumor targeting and repetitive PolyT sequences for copper chelation, facilitates efficient loading and targeted delivery of copper peroxide-Elesclomol nanodots (CuPEs). Upon internalization by tumor cells, Elesclomol released from CuPEs@PApt accumulates copper ions in mitochondria to initiate cuproptosis, while lysosomal degradation of CuP nanodots generates exogenous Cu2+ and H2O2, triggering a Fenton-like reaction for GSH depletion to enhance cuproptosis. In vitro and in vivo experiments confirm the efficacy of this strategy in inducing tumor cell cuproptosis and immunogenic cell death, the latter contributing to the activation of the antitumor immune response for synergistic tumor growth inhibition.
Collapse
Affiliation(s)
- Shuang Wang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China
| | - Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dali Wei
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huayuan Zhou
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiawei Zhu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qing Yu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Luo
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xinfeng Dai
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yiting Jiang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lu Yu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
6
|
Chen L, Liu M, Wang Y, Wei W, Li Y, Bai Y, Yu X, Jiao L, Wang M. TME-Activated MnO 2/Pt Nanoplatform of Hydroxyl Radical and Oxygen Generation to Synergistically Promote Radiotherapy and MR Imaging of Glioblastoma. Int J Nanomedicine 2024; 19:11055-11070. [PMID: 39502635 PMCID: PMC11537150 DOI: 10.2147/ijn.s474098] [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: 06/18/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Purpose Radiotherapy (RT) is currently recognized as an important treatment for glioblastoma (GBM), however, it is associated with several challenges. One of these challenges is the radioresistance caused by hypoxia, whereas the other is the low conversion efficiency of the strongly oxidized hydroxyl radical (•OH), which is produced by the decomposition of water due to high-energy X-ray radiation. These factors significantly limit the clinical effectiveness of radiotherapy. Results To address these limitations, we developed a highly stable and efficient nanoplatform (MnO2/Pt@BSA). Compared to MnO2@BSA, this platform demonstrates high stability, a high yield of oxygen (O2), enhanced production of •OH, and reduced clearance of •OH. The system exhibited increased O2 production in vitro and significantly improved oxygen production efficiency within 100 s at the Pt loading of 38.7%. Furthermore, compared with MnO2, the expression rate of hypoxia-inducible factor (HIF-1α) in glioma cells treated with MnO2/Pt decreased by half. Additionally, the system promotes •OH generation and consumes glutathione (GSH), thereby inhibiting the clearance of •OH and enhancing its therapeutic effect. Moreover, the degradation of the nanoplatform produces Mn2+, which serves as a magnetic resonance imaging (MRI) contrast agent with a T1-weighted enhancement effect at the tumor site. The nanoplatform exhibited excellent biocompatibility and performed multiple functions related to radiotherapy, with simpler components. In U87 tumor bearing mice model, we utilized MnO2/Pt nanocatalysis to enhance the therapeutic effect of radiotherapy on GBM. Conclusion This approach represents a novel and effective strategy for enhancing radiotherapy in gliomas, thereby advancing the field of catalytic radiotherapy and glioma treatment.
Collapse
Affiliation(s)
- Lijuan Chen
- Department of Medical Imaging, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Mingbo Liu
- Department of Radiotherapy, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Yunjuan Wang
- School of Life Sciences, Henan University, North Section of Jinming Avenue, Kaifeng, Henan, 475004, People’s Republic of China
- Institute of Biomedicine, Henan Academy of Sciences, Zhengzhou, Henan, 450046, People’s Republic of China
| | - Wei Wei
- Department of Medical Imaging, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Yaqiong Li
- Department of Pharmacy, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Yan Bai
- Department of Medical Imaging, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Xuan Yu
- Department of Medical Imaging, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
| | - Lei Jiao
- Institute of Molecular Metrology, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, Shandong, 266071, People’s Republic of China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People’s Hospital & the People’s Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People’s Republic of China
- Institute of Biomedicine, Henan Academy of Sciences, Zhengzhou, Henan, 450046, People’s Republic of China
| |
Collapse
|
7
|
Marinho MAG, da Silva Marques M, de Oliveira Vian C, de Moraes Vaz Batista Filgueira D, Horn AP. Photodynamic therapy with curcumin and near-infrared radiation as an antitumor strategy to glioblastoma cells. Toxicol In Vitro 2024; 100:105917. [PMID: 39142446 DOI: 10.1016/j.tiv.2024.105917] [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: 05/14/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Glioblastoma is a malignant neoplasm that develops in the central nervous system and is characterized by high rates of cell proliferation and invasion, presenting resistance to treatments and a poor prognosis. Photodynamic therapy (PDT) is a therapeutic modality that can be applied in oncological cases and stands out for being less invasive. Photosensitizers (PS) of natural origin gained prominence in PDT. Curcumin (CUR) is a natural compound that has been used in PDT, considered a promising PS. In this work, we evaluated the effects of PDT-mediated CUR and near-infrared radiation (NIR) in glioblastoma cells. Through trypan blue exclusion analysis, we chose the concentration of 5 μM of CUR and the dose of 2 J/cm2 of NIR that showed better responses in reducing the viable cell number in the C6 cell line and did not show cytotoxic/cytostatic effects in the HaCat cell line. Our results show that there is a positive interaction between CUR and NIR as a PDT model since there was an increase in ROS levels, a decrease in cell proliferation, increase in cytotoxicity with cell death by autophagy and necrosis, in addition to the presence of oxidative damage to proteins. These results suggest that the use of CUR and NIR is a promising strategy for the antitumor application of PDT.
Collapse
Affiliation(s)
- Marcelo Augusto Germani Marinho
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Cultura Celular, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil.
| | - Magno da Silva Marques
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| | - Camila de Oliveira Vian
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| | - Daza de Moraes Vaz Batista Filgueira
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Cultura Celular, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| | - Ana Paula Horn
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil; Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS 96210-900, Brazil
| |
Collapse
|
8
|
Qiao M, Zeng C, Liu C, Lei Z, Liu B, Xie H. The advancement of siRNA-based nanomedicine for tumor therapy. Nanomedicine (Lond) 2024; 19:1841-1862. [PMID: 39145477 PMCID: PMC11418284 DOI: 10.1080/17435889.2024.2377062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/03/2024] [Indexed: 08/16/2024] Open
Abstract
Small interfering RNA (siRNA) has been proved to be able to effectively down-regulate gene expression through the RNAi mechanism. Thus, siRNA-based drugs have become one of the hottest research directions due to their high efficiency and specificity. However, challenges such as instability, off-target effects and immune activation hinder their clinical application. This review explores the mechanisms of siRNA and the challenges in siRNA-based tumor therapy. It highlights the use of various nanomaterials - including lipid nanoparticles, polymeric nanoparticles and inorganic nanoparticles - as carriers for siRNA delivery in different therapeutic modalities. The application strategies of siRNA-based nanomedicine in chemotherapy, phototherapy and immunotherapy are discussed in detail, along with recent clinical advancements. Aiming to provide insights for future research and therapeutic approaches.
Collapse
Affiliation(s)
- Muchuan Qiao
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Chenlu Zeng
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Changqing Liu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Ziwei Lei
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Hailong Xie
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
9
|
Teng L, Li W, Shi Y, Qi F. The Efficacy of Curcumin Application to Melanoma in Mice: A Systematic Review and Meta-analysis. Ann Plast Surg 2024; 93:S75-S81. [PMID: 39101853 DOI: 10.1097/sap.0000000000003934] [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: 08/06/2024]
Abstract
OBJECTIVE Melanoma is a skin tumor that poses a serious threat to human health. Our study explores the effectiveness and safety of curcumin in the treatment of melanoma based on animal models, and providing evidence-based medical evidence for curcumin in the treatment of malignant melanoma. METHODS The study collected all randomized controlled trial data from the establishment of the database to October 2023 of curcumin for the treatment of melanoma in mice by searching PubMed, Embase, and the Cochrane Library. According to inclusion and exclusion criteria, data were extracted and quality assessment of included studies was performed by using the SYRCLE (Systematic Review Center for Laboratory animal Experimentation) animal experiment bias risk assessment tool. RevMan 5.4 and Stata 15.1 software were used for meta-analysis. RESULTS Eighteen randomized controlled trials were included in this study with a total of 185 mouse models, including 93 mice in the experimental group and 92 in the control group. The results of meta-analysis showed that the IC50 (inhibitory concentrations of 50%) in the experimental group is lower than that of the control group [standardized mean difference (SMD) = -4.68, 95% confidence interval (CI) (-7.30, -2.06), P < 0.01]; the tumor volume is significantly smaller than the control group [SMD = -3.10, 95% CI (-4.45, -1.75), P < 0.01]; the tumor weight is smaller than the control group [SMD = -3.01, 95% CI (-4.81, -1.21), P < 0.01]. However, there was no significant statistical difference in the apoptosis rate between the experimental group and the control group [SMD = 2.27, 95% CI (-1.39, 5.92), P < 0.01]. CONCLUSION Based on animal models for meta-analysis, curcumin can inhibit the growth and proliferation of melanoma in mice. Melanoma may be an effective method for treating melanoma. However, this result still requires further in-depth research.
Collapse
Affiliation(s)
- Liqun Teng
- From the Department of Burn and Plastic Surgery, Jinshan Hospital Fudan University, Shanghai
| | - Wei Li
- From the Department of Burn and Plastic Surgery, Jinshan Hospital Fudan University, Shanghai
| | - Yuedong Shi
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, Shanghai, People's Republic of China
| | - Fazhi Qi
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
10
|
Wang R, Hua S, Xing Y, Wang R, Wang H, Jiang T, Yu F. Organic dye-based photosensitizers for fluorescence imaging-guided cancer phototheranostics. Coord Chem Rev 2024; 513:215866. [DOI: 10.1016/j.ccr.2024.215866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
|
11
|
Qian C, Zhao G, Huo M, Su M, Hu X, Liu Q, Wang L. Tumor microenvironment-regulated drug delivery system combined with sonodynamic therapy for the synergistic treatment of breast cancer. RSC Adv 2024; 14:17612-17626. [PMID: 38828276 PMCID: PMC11141688 DOI: 10.1039/d4ra00539b] [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: 01/21/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Co-loading of sonosensitizers and chemotherapeutic drugs into nanocarriers can improve the biocompatibilities, stabilities, and targeting of drugs and reduce the adverse reactions of drugs, providing a robust platform to orchestrate the synergistic interplay between chemotherapy and sonodynamic therapy (SDT) in cancer treatment. In this regard, biodegradable manganese dioxide (MnO2) has attracted widespread attention because of its unique properties in the tumor microenvironment (TME). Accordingly, herein, MnO2 nanoshells with hollow mesoporous structures (H-MnO2) were etched to co-load hematoporphyrin monomethyl ether (HMME) and doxorubicin (DOX), and DOX/HMME-HMnO2@bovine serum albumin (BSA) obtained after simple BSA modification of DOX/HMME-HMnO2 exhibited excellent hydrophilicity and dispersibility. H-MnO2 rapidly degraded in the weakly acidic TME, releasing loaded HMME and DOX, and catalysed the decomposition of H2O2 abundantly present in TME, producing oxygen (O2) in situ, significantly increasing O2 concentration and downregulating the hypoxia-inducible factor 1α (HIF-1α). After irradiation of the tumor area with low-frequency ultrasound, the drug delivery efficiency of DOX/HMME-HMnO2@BSA substantially increased, and the excited HMME generated a large amount of reactive oxygen species (ROS), which caused irreversible damage to tumor cells. Moreover, the cell death rate exceeded 60% after synergistic SDT-chemotherapy. Therefore, the pH-responsive nanoshells designed in this study can realize drug accumulation in tumor regions by responding to TME and augment SDT-chemotherapy potency for breast cancer treatment by improving hypoxia in tumors. Thus, this study provides theoretical support for the development of multifunctional nanocarriers and scientific evidence for further exploration of safer and more efficient breast cancer treatments.
Collapse
Affiliation(s)
- Chao Qian
- Shandong Provincial Hospital, Shandong University Jinan 250000 China
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Guoliang Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University Jinan 250014 China
| | - Mengping Huo
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Meixia Su
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Xuexue Hu
- School of Clinical Medicine, Shandong First Medical University Jinan 250117 China
| | - Qiang Liu
- Shandong Provincial Hospital, Shandong University Jinan 250000 China
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| | - Lei Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan 250021 China
| |
Collapse
|
12
|
Lu XX, Xue C, Dong JH, Zhang YZ, Gao F. Nanoplatform-based strategies for enhancing the lethality of current antitumor PDT. J Mater Chem B 2024; 12:3209-3225. [PMID: 38497405 DOI: 10.1039/d4tb00008k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Photodynamic therapy (PDT) exhibits great application prospects in future clinical oncology due to its spatiotemporal controllability and good biosafety. However, the antitumor efficacy of PDT is seriously hindered by many factors, including tumor hypoxia, limited light penetration ability, and strong defense mechanisms of tumors. Considering that it is difficult to completely solve the first two problems, enhancing the lethality of antitumor PDT has become a good idea to extend its clinical application. Herein, we summarize the nanoplatform-involved strategies to effectively amplify the tumoricidal capability of current PDT and then discuss the present bottlenecks and prospects of the nanoplatform-based PDT sensitization strategies in tumor therapy. We hope this review will provide some references for others to design high-performance PDT nanoplatforms for tumor therapy.
Collapse
Affiliation(s)
- Xin-Xin Lu
- 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.
| | - Jian-Hui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, 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.
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| |
Collapse
|
13
|
Cai X, Xu T, Ding R, Zhang D, Chen G, Zhao W, Hou J, Pan H, Zhang Q, Yin T. Oxygen self-supplying small size magnetic nanoenzymes for synergistic photodynamic and catalytic therapy of breast cancer. NANOSCALE 2024; 16:4095-4104. [PMID: 38333905 DOI: 10.1039/d3nr05289c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
In recent years, tumor catalytic therapy based on nanozymes has attracted widespread attention. However, its application is limited by the tumor hypoxic microenvironment (TME). In this study, we developed oxygen-supplying magnetic bead nanozymes that integrate hemoglobin and encapsulate the photosensitizer curcumin, demonstrating reactive oxygen species (ROS)-induced synergistic breast cancer therapy. Fe3O4 magnetic bead-mediated catalytic dynamic therapy (CDT) generates hydroxyl radicals (˙OH) through the Fenton reaction in the tumor microenvironment. The Hb-encapsulated Fe3O4 magnetic beads can be co-loaded with the photosensitizer/chemotherapeutic agent curcumin (cur), resulting in Fe3O4-Hb@cur. Under hypoxic conditions, oxygen molecules are released from Fe3O4-Hb@cur to overcome the TME hypoxia, resulting in comprehensive effects favoring anti-tumor responses. Upon near-infrared (NIR) irradiation, Fe3O4-Hb@cur activates the surrounding molecular oxygen to generate a certain amount of singlet oxygen (1O2), which is utilized for photodynamic therapy (PDT) in cancer treatment. Meanwhile, we validated that the O2 carried by Hb significantly enhances the intracellular ROS level, intensifying the catalytic therapy mediated by Fe3O4 magnetic beads and inflicting lethal damage to cancer cells, effectively inhibiting tumor growth. Therefore, significant in vivo synergistic therapeutic effects can be achieved through catalytic-photodynamic combination therapy.
Collapse
Affiliation(s)
- Xinyi Cai
- Dongguan Key Laboratory of Screening and Research of Anti-inflammatory Ingredients in Chinese Medicine, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Tiantian Xu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Rui Ding
- Dongguan Key Laboratory of Screening and Research of Anti-inflammatory Ingredients in Chinese Medicine, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Dou Zhang
- Dongguan Key Laboratory of Screening and Research of Anti-inflammatory Ingredients in Chinese Medicine, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Guiquan Chen
- Department of Gastroenterology, the Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan 523000, China
| | - Wenchang Zhao
- Dongguan Key Laboratory of Screening and Research of Anti-inflammatory Ingredients in Chinese Medicine, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Jiajie Hou
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qian Zhang
- Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai 200240, China
| | - Ting Yin
- Dongguan Key Laboratory of Screening and Research of Anti-inflammatory Ingredients in Chinese Medicine, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
14
|
Qannita RA, Alalami AI, Harb AA, Aleidi SM, Taneera J, Abu-Gharbieh E, El-Huneidi W, Saleh MA, Alzoubi KH, Semreen MH, Hudaib M, Bustanji Y. Targeting Hypoxia-Inducible Factor-1 (HIF-1) in Cancer: Emerging Therapeutic Strategies and Pathway Regulation. Pharmaceuticals (Basel) 2024; 17:195. [PMID: 38399410 PMCID: PMC10892333 DOI: 10.3390/ph17020195] [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: 12/17/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a key regulator for balancing oxygen in the cells. It is a transcription factor that regulates the expression of target genes involved in oxygen homeostasis in response to hypoxia. Recently, research has demonstrated the multiple roles of HIF-1 in the pathophysiology of various diseases, including cancer. It is a crucial mediator of the hypoxic response and regulator of oxygen metabolism, thus contributing to tumor development and progression. Studies showed that the expression of the HIF-1α subunit is significantly upregulated in cancer cells and promotes tumor survival by multiple mechanisms. In addition, HIF-1 has potential contributing roles in cancer progression, including cell division, survival, proliferation, angiogenesis, and metastasis. Moreover, HIF-1 has a role in regulating cellular metabolic pathways, particularly the anaerobic metabolism of glucose. Given its significant and potential roles in cancer development and progression, it has been an intriguing therapeutic target for cancer research. Several compounds targeting HIF-1-associated processes are now being used to treat different types of cancer. This review outlines emerging therapeutic strategies that target HIF-1 as well as the relevance and regulation of the HIF-1 pathways in cancer. Moreover, it addresses the employment of nanotechnology in developing these promising strategies.
Collapse
Affiliation(s)
- Reem A. Qannita
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ayah I. Alalami
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amani A. Harb
- Department of Basic Sciences, Faculty of Arts and Sciences, Al-Ahliyya Amman University, Amman 19111, Jordan;
| | - Shereen M. Aleidi
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| | - Jalal Taneera
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A. Saleh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Karem H. Alzoubi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad Hudaib
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (R.A.Q.); (A.I.A.); (J.T.); (E.A.-G.); (W.E.-H.); (M.A.S.); (K.H.A.); (M.H.S.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan; (S.M.A.); (M.H.)
| |
Collapse
|
15
|
Zandieh MA, Farahani MH, Daryab M, Motahari A, Gholami S, Salmani F, Karimi F, Samaei SS, Rezaee A, Rahmanian P, Khorrami R, Salimimoghadam S, Nabavi N, Zou R, Sethi G, Rashidi M, Hushmandi K. Stimuli-responsive (nano)architectures for phytochemical delivery in cancer therapy. Biomed Pharmacother 2023; 166:115283. [PMID: 37567073 DOI: 10.1016/j.biopha.2023.115283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
The use of phytochemicals for purpose of cancer therapy has been accelerated due to resistance of tumor cells to conventional chemotherapy drugs and therefore, monotherapy does not cause significant improvement in the prognosis and survival of patients. Therefore, administration of natural products alone or in combination with chemotherapy drugs due to various mechanisms of action has been suggested. However, cancer therapy using phytochemicals requires more attention because of poor bioavailability of compounds and lack of specific accumulation at tumor site. Hence, nanocarriers for specific delivery of phytochemicals in tumor therapy has been suggested. The pharmacokinetic profile of natural products and their therapeutic indices can be improved. The nanocarriers can improve potential of natural products in crossing over BBB and also, promote internalization in cancer cells through endocytosis. Moreover, (nano)platforms can deliver both natural and synthetic anti-cancer drugs in combination cancer therapy. The surface functionalization of nanostructures with ligands improves ability in internalization in tumor cells and improving cytotoxicity of natural compounds. Interestingly, stimuli-responsive nanostructures that respond to endogenous and exogenous stimuli have been employed for delivery of natural compounds in cancer therapy. The decrease in pH in tumor microenvironment causes degradation of bonds in nanostructures to release cargo and when changes in GSH levels occur, it also mediates drug release from nanocarriers. Moreover, enzymes in the tumor microenvironment such as MMP-2 can mediate drug release from nanocarriers and more progresses in targeted drug delivery obtained by application of nanoparticles that are responsive to exogenous stimulus including light.
Collapse
Affiliation(s)
- Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Melika Heydari Farahani
- Faculty of Veterinary Medicine, Islamic Azad University, Shahr-e kord Branch, Chaharmahal and Bakhtiari, Iran
| | - Mahshid Daryab
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Motahari
- Board-Certified in Veterinary Surgery, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Sarah Gholami
- Young Researcher and Elite Club, Islamic Azad University, Babol Branch, Babol, Iran
| | - Farshid Salmani
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Fatemeh Karimi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Seyedeh Setareh Samaei
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
16
|
Lara-Vega I, Vega-López A. Combinational photodynamic and photothermal - based therapies for melanoma in mouse models. Photodiagnosis Photodyn Ther 2023; 43:103596. [PMID: 37148952 DOI: 10.1016/j.pdpdt.2023.103596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/21/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Melanoma is a highly metastatic skin cancer with limited response to current therapies in advanced patients. To overcome resistance, novel treatments based on photodynamic and photothermal therapies (PDT and PTT, respectively) have been developed to treat melanoma in preclinical murine models. Despite success inhibiting implanted tumors' growth, there has been limited evaluation of their long-term effectiveness in preventing metastasis, recurrence, or improving survival rates. METHODS Combined and multidrug therapies based on PDT and/or PTT to treat cutaneous malignant melanoma in the preclinical mouse model were reviewed from 2016 onwards. PubMed® was the database in which the search was performed using mesh search algorithms resulting in fifty-one studies that comply with strict inclusion rules of screening. RESULTS B16 melanoma-bearing C57BLACK6 mice model was the most used to evaluate immunotherapies, chemotherapies, and targeted therapies in combination with PDT and/or PTT. Combined therapies demonstrated a synergistic effect, resulting in intense antitumor activity. The most extensively studied protocol for developing metastatic models involved the intravenous administration of malignant cells, with some combined therapies being tested. Furthermore, the review presents the composition of the nanostructures utilized for delivering the drugs and light-responsive agents and the treatment plans for each combined approach. CONCLUSIONS The identified mechanisms to simulate metastatic melanoma models and the therapeutic combinations may aid in evaluating the systemic protection of combined PDT and PTT-based therapies, particularly in conducting short-term preclinical experiments. Such simulations could have relevance to clinical studies.
Collapse
Affiliation(s)
- Israel Lara-Vega
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, Mexico City C. P. 07738, Mexico
| | - Armando Vega-López
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio de Toxicología Ambiental, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, Mexico City C. P. 07738, Mexico.
| |
Collapse
|
17
|
Wu J, Shang J, An J, Chen W, Hong G, Hou H, Zheng WH, Song F, Peng X. Jointly Depleting Glutathione Based on Self-Assembled Nanomicelles for Enhancing Photodynamic Therapy. Chembiochem 2023; 24:e202300323. [PMID: 37169724 DOI: 10.1002/cbic.202300323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/13/2023]
Abstract
Photodynamic therapy (PDT) is one common ROS-generating therapeutic method with high tumor selectivity and low side effects. But the GSH-upregulation often alleviates its therapeutic efficiency. Here, we proposed a new strategy of jointly depleting GSH to enhance the therapeutic effect of PDT by preparing a nanomicelle by self-assembly method from GSH-activated photosensitizer DMT, curcumin, and amphiphilic polymer TPGS.
Collapse
Affiliation(s)
- Jingxi Wu
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jingjing Shang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Jing An
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Wenlong Chen
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Gaobo Hong
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Haoran Hou
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Wen-Heng Zheng
- Department of Interventional Therapy, Cancer Hospital of, Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, 110042, China
| | - Fengling Song
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao, 266237, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| |
Collapse
|
18
|
Afshari AR, Sanati M, Kesharwani P, Sahebkar A. Recent Advances in Curcumin-Based Combination Nanomedicines for Cancer Therapy. J Funct Biomater 2023; 14:408. [PMID: 37623653 PMCID: PMC10455605 DOI: 10.3390/jfb14080408] [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: 06/11/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Standard cancer chemotherapeutics often produce significant adverse effects and eventually lose their effectiveness due to the emergence of resistance mechanisms. As a result, patients with malignant tumors experience a poor quality of life and a short lifespan. Thus, combination medication regimens provide various advantages, including increased success rate, fewer side effects, and fewer occurrences of resistance. Curcumin (Cur), a potential phytochemical from turmeric, when coupled with traditional chemotherapeutics, has been established to improve the effectiveness of cancer treatment in clinical and preclinical investigations. Cur not only exerts multiple mechanisms resulting in apoptotic cancer cell death but also reduces the resistance to standard chemotherapy drugs, mainly through downregulating the multi-drug resistance (MDR) cargoes. Recent reports showed the beneficial outcomes of Cur combination with many chemotherapeutics in various malignancies. Nevertheless, owing to the limited bioavailability, devising co-delivery strategies for Cur and conventional pharmaceuticals appears to be required for clinical settings. This review summarized various Cur combinations with standard treatments as cancer therapeutics.
Collapse
Affiliation(s)
- Amir R. Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
- Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Karanlık CC, Karanlık G, Gok B, Budama-Kilinc Y, Kecel-Gunduz S, Erdoğmuş A. Exploring anticancer properties of novel Nano-Formulation of BODIPY Compound, Photophysicochemical, in vitro and in silico evaluations. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 301:122964. [PMID: 37302199 DOI: 10.1016/j.saa.2023.122964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/13/2023]
Abstract
A new BODIPY complex (C4) composed of meso- thienyl-pyridine substituted core unit diiodinated from 2- and 6- positions and distyryl moieties at 3- and 5- positions is synthesized. Nano-sized formulation of C4 is prepared by single emulsion method using poly(ε-caprolactone)(PCL) polymer. Encapsulation efficiency and loading capacity values of C4 loaded PCL nanoparticles (C4@PCL-NPs) are calculated and in vitro release profile of C4 is determined. The cytotoxicity and anti-cancer activity are conducted on the L929 and MCF-7 cell lines. Cellular uptake study is performed and interaction between C4@PCL-NPs and MCF-7 cell line is investigated. Anti-cancer activity of C4 is predicted with molecular docking studies and the inhibition property on EGFR, ERα, PR and mTOR are investigated for its anticancer properties. Molecular interactions, binding positions and docking score energies between C4 and EGFR, ERα, PR and mTOR targets are revealed using in silico methods. The druglikeness and pharmacokinetic properties of C4 are evaluated using the SwissADME and its bioavailability and toxicity profiles are assessed using the SwissADME, preADMET and pkCSM servers. In conclusion, the potential use of C4 as an anti-cancer agent is evaluated in vitro and in silico methods. Also, photophysicochemical properties are studied to investigate the potential of using Photodynamic Therapy (PDT). In photochemical studies, the calculated singlet oxygen quantum yield (ΦΔ) value was 0.73 for C4 and in photopysical studies, the calculated fluorescence quantum yield ΦF value was 0.19 for C4.
Collapse
Affiliation(s)
- Ceren Can Karanlık
- Department of Chemistry, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey.
| | - Gürkan Karanlık
- Department of Chemistry, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey.
| | - Bahar Gok
- Graduate School of Natural and Applied Science, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey.
| | - Yasemin Budama-Kilinc
- Department of Bioengineering, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey; Health Biotechnology Joint Research and Application Center of Excellence, 34220, Istanbul, Turkey.
| | | | - Ali Erdoğmuş
- Department of Chemistry, Yildiz Technical University, 34220 Esenler, Istanbul, Turkey; Health Biotechnology Joint Research and Application Center of Excellence, 34220, Istanbul, Turkey.
| |
Collapse
|
20
|
Li Z, Li Z, Wang J. Visualization of Phototherapy Evolution by Optical Imaging. Molecules 2023; 28:molecules28103992. [PMID: 37241733 DOI: 10.3390/molecules28103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is a non-invasive and effective approach used for cancer treatment, in which phototherapeutic agents are irradiated with an appropriate light source to produce cytotoxic reactive oxygen species (ROS) or heat to ablate cancer cells. Unfortunately, traditional phototherapy lacks a facile imaging method to monitor the therapeutic process and efficiency in real time, usually leading to severe side effects due to high levels of ROS and hyperthermia. To realize precise cancer treatment methods, it is highly desired to develop phototherapeutic agents possessing an imaging ability to evaluate the therapeutic process and efficacy in real time during cancer phototherapy. Recently, a series of self-reporting phototherapeutic agents were reported to monitor PDT and PTT processes by combining optical imaging technologies with phototherapy. Due to the real-time feedback provided by optical imaging technology, therapeutic responses or dynamic changes in the tumor microenvironment could be evaluated in a timely manner, thereby achieving personalized precision treatment and minimizing toxic side effects. In this review, we focus on the advances in the development of self-reporting phototherapeutic agents for a cancer phototherapy evaluation based on optical imaging technology to realize precision cancer treatments. Additionally, we propose the current challenges and future directions of self-reporting agents for precision medicine.
Collapse
Affiliation(s)
- Zhiheng Li
- College of Materials and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou 450002, China
| | - Zheng Li
- Wuhan Academy of Agricultural Sciences, Wuhan 430072, China
| | - Jie Wang
- The Key Lab of Health Chemistry & Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering & Materials Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
21
|
Zhang L, Lu H, Tang Y, Lu X, Zhang Z, Zhang Y, Liu Y, Wang C. Calcium-peroxide-mediated cascades of oxygen production and glutathione consumption induced efficient photodynamic and photothermal synergistic therapy. J Mater Chem B 2023; 11:2937-2945. [PMID: 36912360 DOI: 10.1039/d2tb02776c] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Photodynamic therapy (PDT) and photothermal therapy (PTT) are potent approaches to cancer treatment. However, the tumor microenvironment (TME) characterized by severe hypoxia and abundant glutathione (GSH) significantly reduces the effectiveness of PDT. In this study, we developed an oxidative stress amplifier CaO2/ICG@ZIF-8, which was capable of self-sufficient O2 delivery and GSH depletion to enhance PDT and PTT synergistic therapy. We utilized ZIF-8 as nanocarriers that when loaded with CaO2 and indocyanine green (ICG) form CaO2/ICG@ZIF-8 nanoparticles, which exhibit a uniform particle size distribution and a hydrated particle size of about 215 nm. CaO2 reacts with water under acidic conditions to produce O2 so CaO2/ICG@ZIF-8 has an excellent O2 supply capacity, which is essential for PDT. Moreover, CaO2/ICG@ZIF-8 also reacts with GSH to form glutathione disulfides (GSSH), enhancing the therapeutic outcome of PDT by preventing the consumption of local ractive oxygen species. Beyond that, CaO2/ICG@ZIF-8 can produce strong hyperthermia with a photothermal conversion efficiency of about 44%, which is exceedingly appropriate for PTT. Owing to its augmentation, PTT/PDT mediated by CaO2/ICG@ZIF-8 demonstrates intense tumor inhibitory effects in both in vitro and in vivo studies. Notably, the Zn and Ca generated by CaO2/ICG@ZIF-8 degradation are essential elements for the body, so CaO2/ICG@ZIF-8 shows favorable safety. Altogether, the research provides a promising PDT/PTT synergistic therapeutic strategy for cancer and may show more medical applications in the future.
Collapse
Affiliation(s)
- Lanfang Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| | - Hui Lu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| | - Yu Tang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| | - Xiaojie Lu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| | - Zhendong Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| | - Yan Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| | - Ying Liu
- Department of Pharmacy, Fuwai Central China Cardiovascular Hospital & Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, Henan, 451464, P. R. China.
| | - Chenhui Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 South Daxuecheng Road, Chongqing 401331, P. R. China.
| |
Collapse
|
22
|
Wang Y, Wang N, Du Y, Jiang X, Liu Y, Wang Y, Feng Y, Wang P, Meng S. Novel nanoparticles prepared from isothiocyanate derivatives for phototherapy of tumor. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B: BIOLOGY 2023; 242:112701. [PMID: 37003123 DOI: 10.1016/j.jphotobiol.2023.112701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/06/2023] [Accepted: 03/23/2023] [Indexed: 03/28/2023]
Abstract
Phototherapy is a new method to treat tumor, including photodynamic therapy (PDT) and photothermal therapy (PTT). However, the GSH in tumor cells could deplete ROS produced by photosensitizers, resulting in inadequate PDT. Isothiocyanate not only is a new type of anti-tumor drug, but also may combine with GSH, increasing the content of intracellular ROS and improving PDT effect. Here we synthesized a kind of water-soluble nanoparticles (BN NPs) parceling BODIPY-I-35 up with mPEG-ITC and lecithin. mPEG-ITC can react with GSH in tumor cells to reduce the consumption of ROS. BN NPs can be used as vectors to deliver drugs to tumor sites. Under 808 nm laser irradiation, BN NPs solution increased 13 °C within 10 min, indicating that BN NPs had superb photothermal performance. In vitro experiments, low dose BN NPs showed satisfactory PDT and PTT effects, and the cell viability of MCF-7 cell was only 13%. In vivo, BN NPs with excellent biocompatibility showed favorable phototherapy effect and tumor was effectively inhibited. Fluorescence imaging could present the long retention effect of BN NPs in tumor locations. In conclusion, the BN NPs showed the effect of enhancing phototherapy and provided a remarkable application prospect in the phototherapy of tumor cells.
Collapse
|
23
|
Yi M, Xiong B, Li Y, Guo W, Huang Y, Lu B. Manipulate tumor hypoxia for improved photodynamic therapy using nanomaterials. Eur J Med Chem 2023; 247:115084. [PMID: 36599230 DOI: 10.1016/j.ejmech.2022.115084] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023]
Abstract
Due to its low adverse effects, minimal invasiveness, and outstanding patient compliance, photodynamic therapy (PDT) has drawn a great deal of interest, which is achieved through incomplete reduction of O2 by a photosensitizer under light illumination that produces amounts of reactive oxygen species (ROS). However, tumor hypoxia significantly hinders the therapeutic effect of PDT so that tumor cells cannot be eliminated, which results in tumor cells proliferating, invading, and metastasizing. Additionally, O2 consumption during PDT exacerbates hypoxia in tumors, leading to several adverse events after PDT treatment. In recent years, various investigations have focused on conquering or using tumor hypoxia by nanomaterials to amplify PDT efficacy, which is summarized in this review. This comprehensive review's objective is to present novel viewpoints on the advancement of oxygenation nanomaterials in this promising field, which is motivated by hypoxia-associated anti-tumor therapy.
Collapse
Affiliation(s)
- Mengqi Yi
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Bei Xiong
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Yuyang Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Wei Guo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Yunhan Huang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Bo Lu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
24
|
Huang L, Hu S, Fu YN, Wan Y, Li G, Wang X. Multicomponent carrier-free nanodrugs for cancer treatment. J Mater Chem B 2022; 10:9735-9754. [PMID: 36444567 DOI: 10.1039/d2tb02025d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nanocarriers can be used to deliver insoluble anticancer drugs to optimize therapeutic efficacy. However, the potential toxicity of nanocarriers cannot be ignored. Carrier-free nanodrugs are emerging safe drug delivery systems, which are composed of multiple components, such as drugs, bioactive molecules and functional ingredients, avoiding the usage of inert carrier materials and offering advantages that include high drug loading, low toxicity, synergistic therapy, versatile design, and easy surface functionalization. Therefore, how to design multicomponent carrier-free nanodrugs is becoming a priority. In this review, the common strategies for rapid construction of multicomponent carrier-free nanodrugs are briefly explored from the perspective of methodology. The properties of organic-organic, organic-inorganic and inorganic-inorganic multiple carrier-free nanosystems are analyzed according to wettability and in-depth understanding is provided. Further advances in the applications of multiple carrier-free nanodrugs are outlined in anticipation of grasping the intrinsic nature for the design and development of carrier-free nanodrugs.
Collapse
Affiliation(s)
- Lifei Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Shuyang Hu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ya-Nan Fu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Yan Wan
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
25
|
Jiang Y, Fan M, Yang Z, Liu X, Xu Z, Liu S, Feng G, Tang S, Li Z, Zhang Y, Chen S, Yang C, Law WC, Dong B, Xu G, Yong KT. Recent advances in nanotechnology approaches for non-viral gene therapy. Biomater Sci 2022; 10:6862-6892. [PMID: 36222758 DOI: 10.1039/d2bm01001a] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene therapy has shown great potential in the treatment of many diseases by downregulating the expression of certain genes. The development of gene vectors as a vehicle for gene therapy has greatly facilitated the widespread clinical application of nucleic acid materials (DNA, mRNA, siRNA, and miRNA). Currently, both viral and non-viral vectors are used as delivery systems of nucleic acid materials for gene therapy. However, viral vector-based gene therapy has several limitations, including immunogenicity and carcinogenesis caused by the exogenous viral vectors. To address these issues, non-viral nanocarrier-based gene therapy has been explored for superior performance with enhanced gene stability, high treatment efficiency, improved tumor-targeting, and better biocompatibility. In this review, we discuss various non-viral vector-mediated gene therapy approaches using multifunctional biodegradable or non-biodegradable nanocarriers, including polymer-based nanoparticles, lipid-based nanoparticles, carbon nanotubes, gold nanoparticles (AuNPs), quantum dots (QDs), silica nanoparticles, metal-based nanoparticles and two-dimensional nanocarriers. Various strategies to construct non-viral nanocarriers based on their delivery efficiency of targeted genes will be introduced. Subsequently, we discuss the cellular uptake pathways of non-viral nanocarriers. In addition, multifunctional gene therapy based on non-viral nanocarriers is summarized, in which the gene therapy can be combined with other treatments, such as photothermal therapy (PTT), photodynamic therapy (PDT), immunotherapy and chemotherapy. We also provide a comprehensive discussion of the biological toxicity and safety of non-viral vector-based gene therapy. Finally, the present limitations and challenges of non-viral nanocarriers for gene therapy in future clinical research are discussed, to promote wider clinical applications of non-viral vector-based gene therapy.
Collapse
Affiliation(s)
- Yihang Jiang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Miaozhuang Fan
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Zhenxu Yang
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Xiaochen Liu
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shikang Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Gang Feng
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shuo Tang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Zhengzheng Li
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Yibin Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Shilin Chen
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong, China
| | - Biqin Dong
- Guangdong Provincial Key Laboratory of Durability for Marine Civil Engineering, College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia. .,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia.,The Biophotonics and Mechanobioengineering Laboratory, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
26
|
Zheng K, Zhou D, Wu L, Li J, Zhao B, Zhang S, He R, Xiao L, Zoya I, Yu L, Zhang Y, Li Y, Gao J, Li K. K. ZHENG ET AL.Gold-nanoparticle-based multistage drug delivery system for antitumor therapy. Drug Deliv 2022; 29:3186-3196. [PMID: 36226475 PMCID: PMC9578448 DOI: 10.1080/10717544.2022.2128469] [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: 07/29/2022] [Revised: 09/18/2022] [Accepted: 09/18/2022] [Indexed: 11/19/2022] Open
Abstract
Nanoparticles can promote the accumulation of drugs in tumors. However, they find limited clinical applications because they cannot easily penetrate the stroma of cancer tissues, and it is difficult to control drug release. We developed a multiresponse multistage drug-delivery nanogel with improved tumor permeability and responsiveness to the tumor microenvironment for the controlled delivery of anticancer agents. For this purpose, ∼100 nm multistage drug delivery nanogels with pH, redox, near-infrared stimulation, and enzyme responsiveness were grown in situ using 20 nm gold nanoparticles (AuNPs) via an emulsion-aiding crosslinking technique with cysteine crosslinker. An alginate cysteine AuNP (ACA) nanocarrier can efficiently load the cationic drug doxorubicin (DOX) to produce a multistage drug delivery nanocarrier (DOX@ACA). DOX@ACA can maintain the slow release of DOX and reduce its toxicity. In cancer tissues, the high pH and reductase microenvironment combined with the in vitro delivery of alginate and near-infrared light drove drug release. The developed nanoparticles effectively inhibited cancer cells, and in vivo evaluations showed that they effectively enhanced antitumor activity while having negligible in vivo toxicity to major organs.
Collapse
Affiliation(s)
- Kaikai Zheng
- Department of Oncology, Shanghai Fourth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
| | - Dong Zhou
- Department of Oncology, Shanghai Fourth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jian Li
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
| | - Bing Zhao
- Department of Oncology, Shanghai Fourth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shihao Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Ruiying He
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
| | - Lan Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove Campus, Brisbane, Queensland, Australia
- The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Brisbane, Queensland, Australia
| | - Iqbal Zoya
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Li Yu
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
- Department of Trauma Orthopedics and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuhong Zhang
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
| | - Yulin Li
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Jie Gao
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, China
| | - Kaichun Li
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory for the Synthesis and Application of Organic Functional Molecules of Ministry of Education, Key Laboratory for the Green Preparation and Application of Functional Materials of Ministry of Education, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
| |
Collapse
|
27
|
Zeng Q, Li X, Xie S, Xing D, Zhang T. Specific disruption of glutathione-defense system with activatable single molecule-assembled nanoprodrug for boosted photodynamic/chemotherapy eradication of drug-resistant tumors. Biomaterials 2022; 290:121867. [DOI: 10.1016/j.biomaterials.2022.121867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/02/2022]
|
28
|
Li P, Wang D, Hu J, Yang X. The role of imaging in targeted delivery of nanomedicine for cancer therapy. Adv Drug Deliv Rev 2022; 189:114447. [PMID: 35863515 DOI: 10.1016/j.addr.2022.114447] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/27/2022] [Accepted: 07/06/2022] [Indexed: 01/24/2023]
Abstract
Nanomedicines overcome the pharmacokinetic limitations of traditional drug formulations and have promising prospect in cancer treatment. However, nanomedicine delivery in vivo is still facing challenges from the complex physiological environment. For the purpose of effective tumor therapy, they should be designed to guarantee the five features principle, including long blood circulation, efficient tumor accumulation, deep matrix penetration, enhanced cell internalization and accurate drug release. To ensure the excellent performance of the designed nanomedicine, it would be better to monitor the drug delivery process as well as the therapeutic effects by real-time imaging. In this review, we summarize strategies in developing nanomedicines for efficiently meeting the five features of drug delivery, and the role of several imaging modalities (fluorescent imaging (FL), magnetic resonance imaging (MRI), computed tomography (CT), photoacoustic imaging (PAI), positron emission tomography (PET), and electron microscopy) in tracing drug delivery and therapeutic effect in vivo based on five features principle.
Collapse
Affiliation(s)
- Puze Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dongdong Wang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
29
|
Du Y, Han J, Jin F, Du Y. Recent Strategies to Address Hypoxic Tumor Environments in Photodynamic Therapy. Pharmaceutics 2022; 14:pharmaceutics14091763. [PMID: 36145513 PMCID: PMC9505114 DOI: 10.3390/pharmaceutics14091763] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Photodynamic therapy (PDT) has become a promising method of cancer treatment due to its unique properties, such as noninvasiveness and low toxicity. The efficacy of PDT is, however, significantly reduced by the hypoxia tumor environments, because PDT involves the generation of reactive oxygen species (ROS), which requires the great consumption of oxygen. Moreover, the consumption of oxygen caused by PDT would further exacerbate the hypoxia condition, which leads to angiogenesis, invasion of tumors to other parts, and metastasis. Therefore, many research studies have been conducted to design nanoplatforms that can alleviate tumor hypoxia and enhance PDT. Herein, the recent progress on strategies for overcoming tumor hypoxia is reviewed, including the direct transport of oxygen to the tumor site by O2 carriers, the in situ generation of oxygen by decomposition of oxygen-containing compounds, reduced O2 consumption, as well as the regulation of tumor microenvironments. Limitations and future perspectives of these technologies to improve PDT are also discussed.
Collapse
|
30
|
Yu L, Jin Y, Song M, Zhao Y, Zhang H. When Natural Compounds Meet Nanotechnology: Nature-Inspired Nanomedicines for Cancer Immunotherapy. Pharmaceutics 2022; 14:1589. [PMID: 36015215 PMCID: PMC9412684 DOI: 10.3390/pharmaceutics14081589] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Recent significant strides of natural compounds in immunomodulation have highlighted their great potential against cancer. Despite many attempts being made for cancer immunotherapy, the biomedical application of natural compounds encounters a bottleneck because of their unclear mechanisms, low solubility and bioavailability, and limited efficacy. Herein, we summarize the immune regulatory mechanisms of different natural compounds at each step of the cancer-immunity cycle and highlight their anti-tumor potential and current limitations. We then propose and present various drug delivery strategies based on nanotechnology, including traditional nanoparticles (NPs)-based delivery strategies (lipid-based NPs, micelles, and polysaccharide/peptide/protein-based NPs) and novel delivery strategies (cell-derived NPs and carrier-free NPs), thus providing solutions to break through existing bottlenecks. Furthermore, representative applications of nature-inspired nanomedicines are also emphasized in detail with the advantages and disadvantages discussed. Finally, the challenges and prospects of natural compounds for cancer immunotherapy are provided, hopefully, to facilitate their far-reaching development toward clinical translation.
Collapse
Affiliation(s)
- Linna Yu
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Mingjie Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Yu Zhao
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| |
Collapse
|
31
|
Ratkaj I, Mušković M, Malatesti N. Targeting Microenvironment of Melanoma and Head and Neck Cancers
in Photodynamic Therapy. Curr Med Chem 2022; 29:3261-3299. [DOI: 10.2174/0929867328666210709113032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 11/22/2022]
Abstract
Background:
Photodynamic therapy (PDT), in comparison to other skin cancers,
is still far less effective for melanoma, due to the strong absorbance and the role of
melanin in cytoprotection. The tumour microenvironment (TME) has a significant role in
tumour progression, and the hypoxic TME is one of the main reasons for melanoma progression
to metastasis and its resistance to PDT. Hypoxia is also a feature of solid tumours
in the head and neck region that indicates negative prognosis.
Objective:
The aim of this study was to individuate and describe systematically the main
strategies in targeting the TME, especially hypoxia, in PDT against melanoma and head
and neck cancers (HNC), and assess the current success in their application.
Methods:
PubMed was used for searching, in MEDLINE and other databases, for the
most recent publications on PDT against melanoma and HNC in combination with the
TME targeting and hypoxia.
Results:
In PDT for melanoma and HNC, it is very important to control hypoxia levels,
and amongst the different approaches, oxygen self-supply systems are often applied. Vascular
targeting is promising, but to improve it, optimal drug-light interval, and formulation
to increase the accumulation of the photosensitiser in the tumour vasculature, have to
be established. On the other side, the use of angiogenesis inhibitors, such as those interfering
with VEGF signalling, is somewhat less successful than expected and needs to be
further investigated.
Conclusion:
The combination of PDT with immunotherapy by using multifunctional nanoparticles
continues to develop and seems to be the most promising for achieving a
complete and lasting antitumour effect.
Collapse
Affiliation(s)
- Ivana Ratkaj
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Martina Mušković
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Nela Malatesti
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
32
|
Chen C, Wu C, Yu J, Zhu X, Wu Y, Liu J, Zhang Y. Photodynamic-based combinatorial cancer therapy strategies: Tuning the properties of nanoplatform according to oncotherapy needs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214495] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Cao J, Zheng M, Sun Z, Li Z, Qi X, Shen S. One-Step Fabrication of Multifunctional PLGA-HMME-DTX@MnO2 Nanoparticles for Enhanced Chemo-Sonodynamic Antitumor Treatment. Int J Nanomedicine 2022; 17:2577-2591. [PMID: 35698563 PMCID: PMC9188410 DOI: 10.2147/ijn.s365570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Sonodynamic therapy (SDT) and its synergistic cancer therapy derivatives, such as combined chemotherapy-SDT (chemo-SDT), are promising approaches for tumor treatment. However, the main drawbacks restricting their applications are hypoxia in tumors and the reducing microenvironment or high glutathione (GSH) levels. Methods In this study, a hybrid metal MnO2 was deposited onto nanoparticles fabricated using poly(lactic-co-glycolic acid) (PLGA), carrying docetaxel (DTX) and the sonosensitizer hematoporphyrin monomethyl ether (HMME) (PHD@MnO2) via a one-step flash nanoprecipitation (FNP) method. Characterization and in vitro and in vivo experiments were conducted to explore the chemo-SDT effect of PHD@MnO2 and evaluate the synergetic antitumor treatment of this nanosystem. Results When low-power ultrasound is applied, the acquired PHD@MnO2, whether in solution or in MCF-7 cells, generated ROS more efficiently than other groups without MnO2 or those treated via monotherapy. Specifically, GSH-depletion was observed when MnO2 was introduced into the system. PHD@MnO2 presented good biocompatibility and biosafety in vitro and in vivo. These results indicated that the PHD@MnO2 nanoparticles overcame hypoxia in tumor tissue and suppressed the expression of hypoxia-inducible factor 1 alpha (HIF-1α), achieving enhanced chemo-SDT. Conclusion This study provides a paradigm that rationally engineered multifunctional metal-hybrid nanoparticles can serve as an effective platform for augmenting the antitumor therapeutic efficiency of chemo-SDT.
Collapse
Affiliation(s)
- Jin Cao
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, People’s Republic of China
| | - Mingxue Zheng
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, People’s Republic of China
| | - Zhenyan Sun
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, People’s Republic of China
| | - Zhiye Li
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, People’s Republic of China
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, People’s Republic of China
| | - Song Shen
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, People’s Republic of China
- Correspondence: Song Shen; Xueyong Qi, School of Pharmacy, Jiangsu University, 301 Xuefu Road, 212013, Zhenjiang, Jiangsu, People’s Republic of China, Tel +86-0511-88795939, Email ;
| |
Collapse
|
34
|
Acharya S, Misra R. Hypoxia responsive phytonanotheranostics: A novel paradigm towards fighting cancer. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 42:102549. [PMID: 35301157 DOI: 10.1016/j.nano.2022.102549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/22/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
Hypoxia enhances tumor aggressiveness, thereby reducing the efficacy of anticancer therapies. Phytomedicine, which is nowadays considered as the new panacea owing to its dynamic physiological properties, is often plagued by shortcomings. Incorporating these wonder drugs in nanoparticles (phytonanomedicine) for hypoxia therapy is a new prospect in the direction of cancer management. Similarly, the concept of phytonanotheranostics for the precise tumor lesion detection and treatment monitoring in the hypoxic scenario is going on a rampant speed. In the same line, smart nanoparticles which step in for "on-demand" drug release based on internal or external stimuli are also being explored as a new tool for cancer management. However, studies regarding these smart and tailor-made nanotheranostics in the hypoxic tumor microenvironment are very limited. The present review is an attempt to collate these smart stimuli-responsive phytonanotherapeutics in one place for initiating future research in this upcoming field for better cancer treatment.
Collapse
Affiliation(s)
- Sarbari Acharya
- School of Applied Sciences, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, India.
| | - Ranjita Misra
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India.
| |
Collapse
|
35
|
Huang X, Han L, Wang R, Zhu W, Zhang N, Qu W, Liu W, Liu F, Feng F, Xue J. Dual-responsive nanosystem based on TGF-β blockade and immunogenic chemotherapy for effective chemoimmunotherapy. Drug Deliv 2022; 29:1358-1369. [PMID: 35506467 PMCID: PMC9090387 DOI: 10.1080/10717544.2022.2069877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The antitumor immune response induced by chemotherapy has attracted considerable attention. However, the immunosuppressive tumor microenvironment hinders the immune activation effect of cancer chemotherapy. TGF-β plays a key role in driving tumor immunosuppression and can prevent effective antitumor immune response through multiple roles. In this study, a dual-responsive prodrug micelle (PAOL) is designed to co-deliver LY2109761 (a TGF-β receptor I/II inhibitor) and oxaliplatin (OXA, a conventional chemotherapy) to remodel tumor microenvironment and trigger immunogenic cell death (ICD) to induce antitumor immunity response. Under hypoxia tumor environment, the polyethylene glycol shell of the micelle cleavages, along with the release of LY2109761 and OXA prodrug. Cytotoxic effect of OXA is then activated by glutathione-mediated reduction in tumor cells and the activated OXA significantly enhances tumor immunogenicity and promotes intratumoral accumulation of cytotoxic T lymphocytes. Meanwhile, TGF-β blockade through LY2109761 reprograms tumor microenvironment by correcting the immunosuppressive state and regulating tumor extracellular matrix, which further maintaining OXA induced immune response. Therefore, due to the capability of boosting tumor-specific antitumor immunity, the bifunctional micelle presents markedly synergistic antitumor efficacies and provides a potent therapeutic strategy for chemoimmunotherapy of solid tumors.
Collapse
Affiliation(s)
- Xiaoxian Huang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Lingfei Han
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Ruyi Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Wanfang Zhu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Ning Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Wei Qu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Wenyuan Liu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China.,Zhejiang Center for Safety Study of Drug Substances (Industrial Technology Innovation Platform), Hangzhou, China
| | - Fulei Liu
- Tumor Precise Intervention and Translational Medicine Laboratory, Taian City Central Hospital, Taian, China.,Pharmaceutical Department, Taian City Central Hospital, Taian, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China.,Jiangsu Food and Pharmaceutical Science College, Huaian, China
| | - Jingwei Xue
- Tumor Precise Intervention and Translational Medicine Laboratory, Taian City Central Hospital, Taian, China
| |
Collapse
|
36
|
Marinho MAG, Marques MDS, Cordeiro MF, de Moraes Vaz Batista Filgueira D, Horn AP. Combination of Curcumin and Photodynamic Therapy Based on the Use of Red Light or Near-Infrared Radiation in Cancer: a Systematic Review. Anticancer Agents Med Chem 2022; 22:2985-2997. [PMID: 35469576 DOI: 10.2174/1871520622666220425093657] [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: 10/06/2021] [Revised: 01/13/2022] [Accepted: 02/18/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT) is a therapeutic intervention that can be applied to the treatment of cancer. The interaction between a photosensitizer (PS), ideal wavelength radiation and tissue molecular oxygen, triggers a series of photochemical reactions that are responsible for the production of reactive oxygen species. These highly reactive species can decrease proliferation and induce tumor cell death. The search for PS of natural origin extracted from plants becomes relevant, as they have photoactivation capacity, preferentially targeting tumor cells and because they do not present any or little toxicity to healthy cells. OBJECTIVE Our work aimed to carry out a qualitative systematic review to investigate the effects of curcumin (CUR), a molecule considered as PS of natural origin, on PDT, using red light or near infrared radiation, in tumor models. METHODS A systematic search was performed in three databases (PubMed, Scopus, and Web of Science) using the PICOT method, retrieving a total of 1,373 occurrences. At the end of the peer screening, using inclusion, exclusion, and eligibility criteria, 25 eligible articles were included in this systematic review. RESULTS CUR, whether in its free state, associated with metal complexes or other PS, and in a nanocarrier system, was considered a relevant PS for PDT using red light or near-infrared against tumoral models in vitro and in vivo, acting by increasing cytotoxicity, inhibiting proliferation, inducing cell death mainly by apoptosis, and changing oxidative parameters. CONCLUSION The results found in this systematic review suggest the potential use of CUR as a PS of natural origin to be applied in PDT against many neoplasms, encouraging further search in the field of PDT against cancer and serving as an investigative basis for upcoming pre-clinical and clinical applications.
Collapse
Affiliation(s)
- Marcelo Augusto Germani Marinho
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil.,Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil.,Laboratório de Cultura Celular, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil
| | - Magno da Silva Marques
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil.,Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil
| | - Marcos Freitas Cordeiro
- Programa de Pós-Graduação em Biociências e Saúde, Universidade do Oeste de Santa Catarina-UNOESC, Joaçaba, SC, 89600-000, Brasil
| | - Daza de Moraes Vaz Batista Filgueira
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil.,Laboratório de Cultura Celular, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil
| | - Ana Paula Horn
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil.,Laboratório de Neurociências, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande-FURG, Rio Grande, RS, 96210-900, Brasil
| |
Collapse
|
37
|
Wang Y, Huo J, Li S, Huang R, Fan D, Cheng H, Wan B, Du Y, He H, Zhang G. Self-Rectifiable and Hypoxia-Assisted Chemo-Photodynamic Nanoinhibitor for Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10092-10101. [PMID: 35170301 DOI: 10.1021/acsami.1c23121] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) can eradicate cancer cells under light irradiation, mainly because of reactive singlet oxygen (1O2) being transformed from intratumoral oxygen. Nonetheless, the consumption of oxygen during PDT results in serious hypoxic conditions and an elevated hypoxia-inducing factor-1α (HIF-1α) level that hamper further photodynamic efficacy and induce tumor metastasis. To address this problem, we developed hypoxia-assisted NP-co-encapsulating Ce6 (photosensitizer) and YC-1 (HIF-1α inhibitor) as a self-rectifiable nanoinhibitor for synergistic antitumor treatment. PDT-aggravated intracellular hypoxic stress facilitated NP dissociation to release the drug (YC-1), which achieved tumor killing and HIF-1α inhibition to further enhance the therapeutic effect of PDT and prevent tumor metastasis. Besides, in vivo studies revealed that the HC/PI@YC-1 NPs afforded synergistic anticancer efficacy with minimal toxicity. Therefore, this study provides a prospective approach against PDT drawbacks and combination cancer therapy.
Collapse
Affiliation(s)
- Yanan Wang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Jian Huo
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Shuang Li
- Department of Pathology, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou 450003, China
| | - Ran Huang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Daopeng Fan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hanghang Cheng
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Bo Wan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yongkun Du
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hua He
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Gaiping Zhang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
38
|
Veselov VV, Nosyrev AE, Jicsinszky L, Alyautdin RN, Cravotto G. Targeted Delivery Methods for Anticancer Drugs. Cancers (Basel) 2022; 14:622. [PMID: 35158888 PMCID: PMC8833699 DOI: 10.3390/cancers14030622] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Several drug-delivery systems have been reported on and often successfully applied in cancer therapy. Cell-targeted delivery can reduce the overall toxicity of cytotoxic drugs and increase their effectiveness and selectivity. Besides traditional liposomal and micellar formulations, various nanocarrier systems have recently become the focus of developmental interest. This review discusses the preparation and targeting techniques as well as the properties of several liposome-, micelle-, solid-lipid nanoparticle-, dendrimer-, gold-, and magnetic-nanoparticle-based delivery systems. Approaches for targeted drug delivery and systems for drug release under a range of stimuli are also discussed.
Collapse
Affiliation(s)
- Valery V. Veselov
- Center of Bioanalytical Investigation and Molecular Design, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia; (V.V.V.); (A.E.N.)
| | - Alexander E. Nosyrev
- Center of Bioanalytical Investigation and Molecular Design, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia; (V.V.V.); (A.E.N.)
| | - László Jicsinszky
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy;
| | - Renad N. Alyautdin
- Department of Pharmacology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Giancarlo Cravotto
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia
| |
Collapse
|
39
|
Zhang X, He C, Xiang G. Engineering nanomedicines to inhibit hypoxia-inducible Factor-1 for cancer therapy. Cancer Lett 2022; 530:110-127. [PMID: 35041892 DOI: 10.1016/j.canlet.2022.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/18/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1), an essential promoter of tumor progression, has attracted increasing attention as a therapeutic target. In addition to hypoxic cellular conditions, HIF-1 activation can be triggered by cancer treatment, which causes drug tolerance and therapeutic failure. To date, a series of effective strategies have been explored to suppress HIF-1 function, including silencing the HIF-1α gene, inhibiting HIF-1α protein translation, degrading HIF-1α protein, and inhibiting HIF-1 transcription. Furthermore, nanoparticle-based drug delivery systems have been widely developed to improve the stability and pharmacokinetics of HIF-1 inhibitors or achieve HIF-1-targeted combination therapies as a nanoplatform. In this review, we summarize the current literature on nanomedicines targeting HIF-1 to combat cancer and discuss their potential for future development.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuanchuan He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
40
|
Wang N, Wang Y, Shi R, Lin Y, Jiang X, Feng Y, Meng S. The photodynamic/photothermal synergistic therapeutic effect of BODIPY-I-35 liposomes with urea. Photodiagnosis Photodyn Ther 2022; 37:102723. [PMID: 35032702 DOI: 10.1016/j.pdpdt.2022.102723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 01/01/2023]
Abstract
Phototherapy is a new treatment means for cancer which can reduce the side effects of traditional cancer treatments to humans. Urea is a naturally occurring metabolite in the human body. Some studies have shown that it can inhibit the proliferation of tumor cells and cause oxidative stress. In order to explore the application of urea in enhancing the phototherapy effect, we synthesized a new structure photosensitizer (BODIPY-I-35) with good phototherapeutic effect and encapsulated it in liposomes. Compared with free BODIPY-I-35, water-soluble nanoliposomes (LipoBOD) produced a huge redshift (> 122 nm) of fluorescence emission in solution. When LipoBOD was irradiated with 808 nm laser (1 W/cm2) for 10 min, the temperature contrast increased by 20 °C, which was 4 times higher than free BODIPY-I-35. Confocal microscopy showed appreciable accumulation of LipoBOD in HeLa cells. In addition, when LipoBOD was incubated with urea in HeLa cells, we found that urea not only obviously enhanced the production of ROS, but also increased the apoptosis of HeLa cells. The synergistic effect of LipoBOD (20 μg/mL, at BODIPY-I-35-eq) with urea (250 mM) showed significantly higher phototoxicity than LipoBOD alone. Low dose can reduce the cell viability to 10%. Therefore, we have obtained an effective method of using urea to enhance the phototherapy effect.
Collapse
Affiliation(s)
- Ning Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| | - Yuguang Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| | - Ruijie Shi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| | - Yanxin Lin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| | - Xu Jiang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| | - Yaqing Feng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| | - Shuxian Meng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300050, PR China.
| |
Collapse
|
41
|
Wen X, Liu N, Ren J, Jiao X, Lv J, Akhtar MH, Qi H, Zhu J, Yu C, Li Y. In situ synthesis of a functional ZIF-8 nanocomposite for synergistic photodynamic–chemotherapy and pH and NIR-stimulated drug release. NEW J CHEM 2022. [DOI: 10.1039/d2nj00082b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
UCNP/TiO2/Cur@ZIF-8 can be used as a NIR and pH-responsive photodynamic-chemo candidate for tumor therapy.
Collapse
Affiliation(s)
- Xin Wen
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun 130022, China
| | - Ning Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jia Ren
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiaorui Jiao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Junying Lv
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Mahmood Hassan Akhtar
- Department of chemistry, National University of Technology, I-12 IJP Road, Islamabad, Pakistan
| | - Hong Qi
- Tumor Hospital of Jilin Province, Changchun 130061, China
| | - Jianwei Zhu
- Zhongshan Institute of Changchun University of Science and Technology, China
| | - Cong Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- University of Science and Technology of China, Hefei 230026, China
| | - Yunhui Li
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun 130022, China
- Zhongshan Institute of Changchun University of Science and Technology, China
| |
Collapse
|
42
|
Su Y, Lu H, Li Q, Shao Z, Wang S, Quan Y, Zeng Y, Zheng Y. Driving co-precipitation of hydrophobic drugs in water by conjugating alkyl chains. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
43
|
Wan Y, Fu LH, Li C, Lin J, Huang P. Conquering the Hypoxia Limitation for Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103978. [PMID: 34580926 DOI: 10.1002/adma.202103978] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/11/2021] [Indexed: 06/13/2023]
Abstract
Photodynamic therapy (PDT) has aroused great research interest in recent years owing to its high spatiotemporal selectivity, minimal invasiveness, and low systemic toxicity. However, due to the hypoxic nature characteristic of many solid tumors, PDT is frequently limited in therapeutic effect. Moreover, the consumption of O2 during PDT may further aggravate the tumor hypoxic condition, which promotes tumor proliferation, metastasis, and invasion resulting in poor prognosis of treatment. Therefore, numerous efforts have been made to increase the O2 content in tumor with the goal of enhancing PDT efficacy. Herein, these strategies developed in past decade are comprehensively reviewed to alleviate tumor hypoxia, including 1) delivering exogenous O2 to tumor directly, 2) generating O2 in situ, 3) reducing tumor cellular O2 consumption by inhibiting respiration, 4) regulating the TME, (e.g., normalizing tumor vasculature or disrupting tumor extracellular matrix), and 5) inhibiting the hypoxia-inducible factor 1 (HIF-1) signaling pathway to relieve tumor hypoxia. Additionally, the O2 -independent Type-I PDT is also discussed as an alternative strategy. By reviewing recent progress, it is hoped that this review will provide innovative perspectives in new nanomaterials designed to combat hypoxia and avoid the associated limitation of PDT.
Collapse
Affiliation(s)
- Yilin Wan
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Chunying Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
44
|
Zhou X, Huang JQ, Liu LS, Deng FA, Liu YB, Li YM, Chen AL, Yu XY, Li SY, Cheng H. Self-Remedied Nanomedicine for Surmounting the Achilles' Heel of Photodynamic Tumor Therapy. ACS APPLIED BIO MATERIALS 2021; 4:8023-8032. [PMID: 35006783 DOI: 10.1021/acsabm.1c00938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Oxygen-dependent photodynamic therapy (PDT) could exacerbate tumor hypoxia to induce the upregulation of hypoxia-inducible factor-1α (HIF-1α), which would promote tumor growth and metastasis. In this paper, a self-remedied nanomedicine is developed based on a photosensitizer and a HIF-1α inhibitor to surmount the Achilles' heel of PDT for enhanced antitumor efficacy. Specifically, the nanomedicine (designated as CYC-1) is prepared by the self-assembly of chlorine e6 (Ce6) and 3-(5'-hydroxy-methyl-2'-furyl)-1-benzylindazole (YC-1) through π-π stacking and hydrophobic interactions. Of special note, carrier-free CYC-1 holds an extremely high drug loading rate and avoids excipient-triggered adverse reactions. Intravenously administered CYC-1 prefers to accumulate in the tumor tissue for effective cellular uptake. More importantly, it is verified that CYC-1 is capable of inhibiting the HIF-1α activity, thereby improving its PDT efficacy on tumor suppression. Besides, CYC-1 has the overwhelming superiority in restraining tumor proliferation over the combined administration of Ce6 and YC-1, which highlights the advantage of this self-remedied strategy in drug delivery and tumor therapy. This study sheds light on the development of self-delivery nanomedicine for efficient PDT against malignancies.
Collapse
Affiliation(s)
- Xiang Zhou
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Jia-Qi Huang
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Ling-Shan Liu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - Fu-An Deng
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yi-Bin Liu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yan-Mei Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| | - A-Li Chen
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Xi-Yong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Shi-Ying Li
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
45
|
Jin ZY, Fatima H, Zhang Y, Shao Z, Chen XJ. Recent Advances in Bio‐Compatible Oxygen Singlet Generation and Its Tumor Treatment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zheng Yang Jin
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Hira Fatima
- Western Australia School of Mines: Minerals Energy and Chemical Engineering (WASM‐MECE) Curtin University Perth Western Australia 6102 Australia
| | - Yue Zhang
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Zongping Shao
- Western Australia School of Mines: Minerals Energy and Chemical Engineering (WASM‐MECE) Curtin University Perth Western Australia 6102 Australia
- State Key Laboratory of Materials‐Oriented Chemical Engineering College of Chemical Engineering Nanjing Tech University Nanjing Jiangsu 211816 P. R. China
| | - Xiang Jian Chen
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| |
Collapse
|
46
|
Sun Y, Zhou Z, Yang S, Yang H. Modulating hypoxia inducible factor-1 by nanomaterials for effective cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1766. [PMID: 34713633 DOI: 10.1002/wnan.1766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
Hypoxia, which is induced by abnormal tumor growth when it outstrips its oxygen supply, is a major character of cancer. The reaction of cells against hypoxia is mainly concentrated on the hypoxia-induced transcription factors (HIFs), especially HIF-1, which remain stabilized during hypoxia. Additionally, the oxygen-independent mechanism of regulating HIF-1 acts a vital part in different stages of tumor progression as well as chemo-/radio-/PDT resistance, resulting in poor curative effects and prognosis. In this review, we will outline the up-to-date information about how HIF-1 interferes with tumor metastasis and therapy resistance, followed by a detailed introduction of motivating techniques based on various nanomaterials to interfere with HIF signaling for effective cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yun Sun
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Zhiguo Zhou
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Shiping Yang
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Hong Yang
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| |
Collapse
|
47
|
Nanotheranostic agents for neurodegenerative diseases. Emerg Top Life Sci 2021; 4:645-675. [PMID: 33320185 DOI: 10.1042/etls20190141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases (NDDs), including Alzheimer's disease (AD) and Parkinson's disease (PD), affect the ageing population worldwide and while severely impairing the quality of life of millions, they also cause a massive economic burden to countries with progressively ageing populations. Parallel with the search for biomarkers for early detection and prediction, the pursuit for therapeutic approaches has become growingly intensive in recent years. Various prospective therapeutic approaches have been explored with an emphasis on early prevention and protection, including, but not limited to, gene therapy, stem cell therapy, immunotherapy and radiotherapy. Many pharmacological interventions have proved to be promising novel avenues, but successful applications are often hampered by the poor delivery of the therapeutics across the blood-brain-barrier (BBB). To overcome this challenge, nanoparticle (NP)-mediated drug delivery has been considered as a promising option, as NP-based drug delivery systems can be functionalized to target specific cell surface receptors and to achieve controlled and long-term release of therapeutics to the target tissue. The usefulness of NPs for loading and delivering of drugs has been extensively studied in the context of NDDs, and their biological efficacy has been demonstrated in numerous preclinical animal models. Efforts have also been made towards the development of NPs which can be used for targeting the BBB and various cell types in the brain. The main focus of this review is to briefly discuss the advantages of functionalized NPs as promising theranostic agents for the diagnosis and therapy of NDDs. We also summarize the results of diverse studies that specifically investigated the usage of different NPs for the treatment of NDDs, with a specific emphasis on AD and PD, and the associated pathophysiological changes. Finally, we offer perspectives on the existing challenges of using NPs as theranostic agents and possible futuristic approaches to improve them.
Collapse
|
48
|
Niu B, Liao K, Zhou Y, Wen T, Quan G, Pan X, Wu C. Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials 2021; 277:121110. [PMID: 34482088 DOI: 10.1016/j.biomaterials.2021.121110] [Citation(s) in RCA: 558] [Impact Index Per Article: 139.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 01/17/2023]
Abstract
Glutathione (GSH) is an important member of cellular antioxidative system. In cancer cells, a high level of GSH is indispensable to scavenge excessive reactive oxygen species (ROS) and detoxify xenobiotics, which make it a potential target for cancer therapy. Plenty of studies have shown that loss of intracellular GSH makes cancer cells more susceptible to oxidative stress and chemotherapeutic agents. GSH depletion has been proved to improve the therapeutic efficacy of ROS-based therapy (photodynamic therapy, sonodynamic therapy, and chemodynamic therapy), ferroptosis, and chemotherapy. In this review, various strategies for GSH depletion used in cancer therapy are comprehensively summarized and discussed. First, the functions of GSH in cancer cells are analyzed to elucidate the necessity of GSH depletion in cancer therapy. Then, the synthesis and metabolism of GSH are briefly introduced to bring up some crucial targets for GSH modulation. Finally, different approaches to GSH depletion in the literature are classified and discussed in detail according to their mechanisms. Particularly, functional materials with GSH-consuming ability based on nanotechnology are elaborated due to their unique advantages and potentials. This review presents the ingenious application of GSH-depleting strategy in cancer therapy for improving the outcomes of various therapeutic regimens, which may provide useful guidance for designing intelligent drug delivery system.
Collapse
Affiliation(s)
- Boyi Niu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Kaixin Liao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yixian Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ting Wen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
49
|
Liu Y, Wu W, Wang Y, Han S, Yuan Y, Huang J, Shuai X, Peng Z. Recent development of gene therapy for pancreatic cancer using non-viral nanovectors. Biomater Sci 2021; 9:6673-6690. [PMID: 34378568 DOI: 10.1039/d1bm00748c] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pancreatic cancer (PC), characterized by its dense desmoplastic stroma and hypovascularity, is one of the most lethal cancers with a poor prognosis in the world. Traditional treatments such as chemotherapy, radiotherapy, and targeted therapy show little benefit in the survival rate in patients with advanced PC due to the poor penetration and resistance of drugs, low radiosensitivity, or severe side effects. Gene therapy can modify the morbific and drug-resistant genes as well as insert the tumor-suppressing genes, which has been shown to have great potential in PC treatment. The development of safe non-viral vectors for the highly efficient delivery of nucleic acids is essential for effective gene therapy, and has been attracting much attention. In this review, we first summarized the PC-promoting genes and gene therapies using plasmid DNA, mRNA, miRNA/siRNA-based RNA interference technology, and genome editing technology. Second, the commonly used non-viral nanovector and theranostic gene delivery nanosystem, especially the tumor microenvironment-sensitive delivery nanosystem and the cell/tumor-penetrating delivery nanosystem, were introduced. Third, a combination of non-viral nanovector-based gene therapy and other therapies, such as immunotherapy, chemotherapy, photothermal therapy (PTT), and photodynamic therapy (PDT), for PDAC treatment was discussed. Finally, a number of clinical trials have demonstrated the proof-of-principle that gene therapy or the combination of gene therapy and chemotherapy using non-viral vectors can inhibit the progression of PC. Although most of the non-viral vector-based gene therapies and their combination therapy are still under preclinical research, the development of genetics, molecular biology, and novel vectors would promote the clinical transformation of gene therapy.
Collapse
Affiliation(s)
- Yu Liu
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Wei Wu
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yiyao Wang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Shisong Han
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Yuanyuan Yuan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinsheng Huang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510275, China.
| | - Zhao Peng
- Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
50
|
Wu C, Liu Q, Wang Y, Xie Z, Huang H, Li N, Wei X, Yang G, Li T, Yang H, Li S, Qin X, Liu Y. Tirapazamine encapsulated hyaluronic acid nanomicelles realized targeted and efficient photo-bioreductive cascading cancer therapy. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.02.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|