1
|
Wang T, Sun Y, Zeng D, Wang M, Zhang Y, Liu G, Chen X, Liu L. Integrated cell membrane encapsulated PQDs-TK quantum dot nanoclusters with ROS-responsive triggering for efficient and visualized DNA delivery. J Colloid Interface Sci 2025; 683:393-410. [PMID: 39740557 DOI: 10.1016/j.jcis.2024.12.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
With the unique photo-physical properties and strong bio-compatibility. Quantum dots (QDs) have sparked interest in biomedical fields such as imaging, biosensing and therapeutics. However, the low stability and insufficient tumor specificity have largely constrained their potential biomedical applications. Here, we reported a cell membrane encapsulated PQDs-TK quantum dots nanoclusters (CM-PQDs-TK) gene delivery system with ROS-responsive triggering for efficient and visualized DNA delivery. CM-PQDs-TK possessed excellent DNA loading capacity and protective ability. The particle size and morphology change suggested that CM-PQDs-TK displayed superior ROS responsiveness. As expected, CM-PQDs-TK had an obvious nanocluster structure, and the particle size was stabilized in the range of 200-300 nm. Compared with PQDs and PQDs-TK, HlM-PQDs-TK/DNA encapsulated by Hela cell membrane has higher uptake efficiency and transfection ability, reaching 67.14 % and 63.41 % in 293 T cells, and the DNA transfection efficiency could still reach 43.98 % even in cancer cells (Hela cells). Moreover, flow cytometry and fluorescence microscopy showedthat nanoclusters could effectively enter tumor cells, and the internal DNA could be effectively released. In this process, PQDs-TK responded to high ROS in tumor cells and greatly improved DNA delivery efficiency. After loading DNA, cell membrane encapsulation technology was used to enhance its biocompatibility and targeting further. This work was anticipated to provide an in-depth understanding of the important role of PEI quantum dots in the field of ROS-responsive intelligent gene carriers and laid a foundation for the design and preparation of novel quantum dots gene carriers.
Collapse
Affiliation(s)
- Tiange Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yanlin Sun
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Dong Zeng
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Mengying Wang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yajing Zhang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Gang Liu
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xin Chen
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Liang Liu
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
2
|
Li F, Zhou E, Wang M, Pan F, Zhou J, Yang M, Wang T, Li L, Wu L, Li Q. A novel sustainable photo/chemical magnetic nanomaterial effectively mitigates the allergenicity of phospholipase A2. Food Chem 2024; 461:140851. [PMID: 39167945 DOI: 10.1016/j.foodchem.2024.140851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/06/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
Reducing the allergenicity of edible insects is crucial for the comprehensive utilization of insect resources. Phospholipase A2 (PLA2) exists in various edible insects and mammalian tissues, which can cause serious allergic reactions. Herein, we constructed a magnetic nanocomposite with photo/chemical synergistic capability to mitigate the allergenicity of PLA2. The formation of prepared nanocomposite was systematically confirmed using various techniques. The nanocomposite exhibited uniform diameters, abundant functional groups, excellent magnetic capabilities. An effective photo/chemical method was established to reduce the allergenicity of PLA2 in vitro. The feasibility of the method was demonstrated through circular dichroism, fluorescence spectrum and IgE-binding analysis. The allergenicity and IgE-binding effect of PLA2 were significantly reduced due to conformational changes after nanomaterial treatment. These results demonstrate the sensitivity and effectiveness a strategy for reducing PLA2 allergenicity, providing a basis for development of nanomaterials to reduce the risk of novel food allergies in response to edible insect products.
Collapse
Affiliation(s)
- Fukai Li
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, PR China
| | - Enning Zhou
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, PR China
| | - Min Wang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, PR China
| | - Fei Pan
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, PR China
| | - Jian Zhou
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, PR China
| | - Mengrui Yang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, PR China
| | - Tongtong Wang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, PR China
| | - Liang Li
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100081, PR China
| | - Liming Wu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, PR China
| | - Qiangqiang Li
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100093, PR China.
| |
Collapse
|
3
|
Gao Y, Li A, Li Y, Guo H, He L, Li K, Shcharbin D, Shi X, Shen M. Dendrimer/Copper(II) Complex-Mediated siRNA Delivery Disrupts Lactate Metabolism to Reprogram the Local Immune Microenvironment against Tumor Growth and Metastasis. Biomacromolecules 2024; 25:7995-8005. [PMID: 39570391 DOI: 10.1021/acs.biomac.4c01249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Solid tumors reprogram metabolic pathways to meet their biosynthesis demands, resulting in elevated levels of metabolites in the tumor microenvironment (TME), including lactate. Excessive accumulation and active transportation of lactate within the TME drives tumor progression, metastasis, and immunosuppression. Interruption of TME lactate metabolism is expected to restore antitumor responses and sensitize tumor immunotherapy. Herein, we developed phenylboronic acid- and pyridine-modified poly(amidoamine) dendrimer/copper(II) (Cu(II)) complexes, namely, D-Cu complexes, to deliver monocarboxylate transporter 4 siRNA (siMCT4) and disrupt the tumor lactate shuttle. The D-Cu complexes are shown to have a Cu(II)-mediated chemodynamic effect and T1-weighted magnetic resonance imaging potential (r1 relaxivity = 1.19 mM-1 s-1), enabling effective siMCT4 delivery to inhibit lactate efflux within cancer cells. In combination with a CD11b immune agonist, the treatment of D-Cu/siMCT4 polyplexes in a mouse breast tumor model alleviates local TME immunosuppression, leading to excellent inhibition of both primary tumor growth and lung metastasis.
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Aiyu Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Yanying Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Honghua Guo
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201600, China
| | - Liangyu He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Kangan Li
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201600, China
| | - Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, Akademicheskaja 27, 220072 Minsk, Belarus
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
4
|
Salatin S, Azarfarin M, Farjami A, Hamidi S. The simultaneous use of nanovesicles and magnetic nanoparticles for cancer targeting and imaging. Ther Deliv 2024:1-15. [PMID: 39564978 DOI: 10.1080/20415990.2024.2426447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
Cancer is increasingly being recognized as a global health issue with considerable unmet medical need. Despite the rapid progression of anticancer pharmaceuticals, there are still significant challenges for the effective management of cancer. In many circumstances, cancer cells are difficult to detect and treat. Combination of nanovesicles (NVs) and magnetic nanoparticles (MNPs), referred as magnetic nanovesicles (MNVs), is now well recognized as a potential theranostic option for improving cancer treatment outcomes and reducing adverse effects. MNVs can be used for monitoring the long-term fate and functional benefits of cancer therapy. Moreover, MNV-mediated hyperthermia mechanism has been explored as a potential technique for triggering cancer cell death, and/or controlled release of laden cargo. In this review, we focus on the unique characteristics of MNVs as a promising avenue for targeted drug delivery, diagnosis, and treatments of cancer or tumor. Moreover, we discuss critical considerations related to the issues raised in this area, which will guide future research toward better anti-cancer therapeutics for clinical applications.
Collapse
Affiliation(s)
- Sara Salatin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Azarfarin
- School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Farjami
- Pharmaceutical and Food Control Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samin Hamidi
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Li LS, Chen PW, Zhao XJ, Cheng D, Liu BB, Tang XJ, Zhu WQ, Yang X, Zhao MX. Nuclear-targeted smart nanoplatforms featuring double-shell hollow mesoporous copper sulfide coated with manganese dioxide synergistically potentiate chemotherapy and immunotherapy in hepatocellular carcinoma cells. J Colloid Interface Sci 2024; 680:202-214. [PMID: 39504750 DOI: 10.1016/j.jcis.2024.11.003] [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: 09/06/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Smart nanoplatforms designed for nuclear-targeted delivery of chemotherapeutic agents to tumor sites are pivotal in advancing tumor treatment and immunotherapy. Herein, we introduced a novel nuclear-targeting double-shell smart nanoplatform (HMCuS/Pt/ICG@MnO2@9R-P201 (HMCPIM9P)), which synergistically enhances chemotherapy, photodynamic therapy (PDT), photothermal therapy (PTT), immunotherapy and chemodynamic therapy (CDT). The core of this nanoplatform consists of double-shell multifunctional nanoparticles (HMCuS@MnO2) that enable targeted delivery of the photosensitizer Indocyanine Green (ICG) and the chemotherapeutic agent cisplatin (Pt). By effectively consuming glutathione (GSH), these nanoparticles boost the chemotherapeutic efficacy of Pt. Additionally, the manganese ion (Mn2+) present activate the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) (cGAS-STING) pathway, bolstering adaptive immune responses against tumors and elevating the level of tumor-infiltrating CD8+ T cells. The incorporation of the hepatoma-targeting peptide (9R-P201 peptide) allows the system to exhibit FOXM1 receptor-mediated nuclear targeting properties specifically in hepatocellular carcinoma (HCC). Notably, when combined with near-infrared (NIR) light, HMCPIM9P demonstrated a remarkable tumor inhibition rate of 95.6 %, fostered a robust immune response, and significantly inhibited tumor growth and recurrence. Overall, the smart nanoplatform boasts active nuclear targeting capabilities, enabling the enrichment of chemotherapeutic agents at tumor sites, and holds great potential for synergistic applications in enhancing chemotherapy and immunotherapy for HCC.
Collapse
Affiliation(s)
- Lin-Song Li
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Peng-Wei Chen
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Xue-Jie Zhao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Dong Cheng
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Bang-Bang Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Xian-Jiao Tang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Wen-Qi Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Xiaojing Yang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
| | - Mei-Xia Zhao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, China; State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China; The Zhongzhou Laboratory for Integrative Biology, Henan University, Kaifeng, China.
| |
Collapse
|
6
|
Sueyoshi S, Vitor Silva J, Guizze F, Giarolla J. Dendrimers as drug delivery systems for oncotherapy: Current status of promising applications. Int J Pharm 2024; 663:124573. [PMID: 39134292 DOI: 10.1016/j.ijpharm.2024.124573] [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: 02/19/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Cancer affects millions of people worldwide, causing death and serious health problems. Despite significant investment in the development of new anticancer compounds, there are still several limitations that can still be found. Many compounds exhibit high levels of toxicity and low bioavailability. Therefore, it is urgent to design safer, more effective, and particularly more selective compounds for oncological treatment. Dendrimers are polymeric structures that have been shown to be potential drug nanocarriers to overcome physicochemical, pharmacokinetic, and indirect pharmacodynamic issues. Due to their versatility, they can be used in the design of nanovaccines, lipophilic complexes, amphiphilic complexes, smart nanocomplexes, and others. This work targets the use of dendrimers in oncological treatment and their importance and effectiveness as drug delivery systems for the development of new therapies. For this review, only publications from the last two years are considered in this review.
Collapse
Affiliation(s)
- Sophia Sueyoshi
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - João Vitor Silva
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - Felipe Guizze
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - Jeanine Giarolla
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Girma WM, Zhu Z, Guo Y, Xiao X, Wang Z, Mekuria SL, Hameed MMA, El-Newehy M, Guo R, Shen M, Shi X. Synthesis and Characterization of Copper-Crosslinked Carbon Dot Nanoassemblies for Efficient Macrophage Manipulation. Macromol Rapid Commun 2024:e2400511. [PMID: 39154350 DOI: 10.1002/marc.202400511] [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: 06/27/2024] [Revised: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Nanomedicines loaded in macrophages (MAs) can actively target tumors without dominantly relying on the enhanced permeability and retention (EPR) effect, making them effective for treating EPR-deficient malignancies. Herein, copper-crosslinked carbon dot clusters (CDCs) are synthesized with both photodynamic and chemodynamic functions to manipulate MAs, aiming to direct the MA-mediated tumor targeting. First, green fluorescent CDs (g-CDs) are prepared by a one-step hydrothermal method. Subsequently, the g-CDs are complexed with divalent copper ions to form copper-crosslinked CDCs (g-CDCs/Cu), which are incubated with MAs for their manipulation. Experimental results revealed that the prepared g-CDCs/Cu displayed good aqueous dispersibility and fluorescent emission properties. The nanoassemblies can be activated to deplete the overexpressed glutathione (GSH) and generate reactive oxygen species (ROS) in the presence of laser irradiation through the combined Cu-mediated chemodynamic therapy and CD-mediated photodynamic therapy. Furthermore, the ROS produced in MAs enabled polarization of MAs to antitumor M1 phenotype, suggesting the future potential use to reverse the immunosuppressive tumor microenvironment. These results obtained from the current study suggest a significant potential to develop g-CDCs/Cu for GSH depletion, ROS generation, and MA M1 polarization as a theransotic agent to tackle cancer.
Collapse
Affiliation(s)
- Wubshet Mekonnen Girma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
- Department of Chemistry, College of Natural Science, Wollo University, Dessie, 1000, Ethiopia
| | - Zewen Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Xianghao Xiao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Zhiqiang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Shewaye Lakew Mekuria
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Meera Moydeen Abdul Hameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| |
Collapse
|
8
|
Soozanipour A, Ejeian F, Razmjou A, Asadnia M, Nasr-Esfahani MH, Taheri-Kafrani A. Efficient PEGylated Dendrimer Nanoplatform for Codelivery of Hyaluronidase and Methotrexate: A New Frontier in Chemotherapeutic Efficacy and Tumor Penetration. ACS APPLIED NANO MATERIALS 2024; 7:17262-17277. [DOI: 10.1021/acsanm.4c01406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Asieh Soozanipour
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Fatemeh Ejeian
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan 81746, Iran
| | - Amir Razmjou
- Mineral Recovery Research Center (MRRC), School of Engineering, Edith Cowan University, Joondalup, Perth, WA 6027, Australia
| | - Mohsen Asadnia
- Department of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan 81746, Iran
| | - Asghar Taheri-Kafrani
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| |
Collapse
|
9
|
Huang H, Guo H, Liu J, Ni C, Xia L, Cao X, Xia J, Shi X, Guo R. Dendrimer/metal-phenolic nanocomplexes encapsulating CuO 2 for targeted magnetic resonance imaging and enhanced ferroptosis/cuproptosis/chemodynamic therapy by regulating the tumor microenvironment. Acta Biomater 2024; 183:252-263. [PMID: 38801869 DOI: 10.1016/j.actbio.2024.05.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
The combination of ferroptosis, cuproptosis, and chemodynamic therapy (CDT) would be a potential strategy for tumor diagnosis and enhanced treatment. However, the therapeutic effect was severely limited by the lack of specific delivery of catalytic ions and the low Fenton reaction efficiency in tumor microenvironment (TME) with excess glutathione, limited acidity and insufficient endogenous hydrogen peroxide. In this work, p-carboxybenzenesulfonamide (BS), a carbonic anhydrase IX (CA IX) inhibitor, was modified on the surface of generation-5 poly(amidoamine) dendrimer to load copper peroxide nanoparticles, which were complexed with iron (Fe)-tannic acid (TF) networks for targeted magnetic resonance (MR) imaging and enhanced ferroptosis/cuproptosis/CDT by regulating TME. The formed CuO2@G5-BS/TF nanocomplexes with an average size of 39.4 nm could be specifically accumulated at tumor site and effectively internalized by metastatic 4T1 cells via the specific interaction between BS and CA IX over-expressed on tumor cells. Meanwhile, the inhibition of CA IX activity could not only decrease the intracellular pH to accelerate Fe3+/Cu2+ release, H2O2 self-supply and Fenton reaction, but also suppress tumor metastasis by alleviating the extracellular acidity in TME. Moreover, the reduction of Fe3+/Cu2+ by intracellular glutathione (GSH) could further amplify ROS generation and enhance CDT efficacy, and the GSH depletion could in turn inhibit GPX-4 mediated antioxidant reaction to induce ferroptosis, resulting in effective therapeutic efficacy. In vivo experimental results demonstrated that CuO2@G5-BS/TF could provide better tumor MR imaging, effectively inhibit the growth and metastasis of 4T1 breast tumors, and be metabolized without significant systemic toxicity. Thus, CuO2@G5-BS/TF nanocomplexes provided a new approach for targeted MR imaging and enhanced ferroptosis/cuproptosis/CDT of triple-negative breast cancer. STATEMENT OF SIGNIFICANCE: Taking the advantage of dendrimer and metal-phenolic system, stable CuO2@G5-BS/TF nanocomplexes with an average size of 39.4 nm were synthesized to efficiently load Fe3+ and CuO2 nanoparticles for TNBC treatment and MR imaging. CuO2@G5-BS/TF nanocomplexes could target tumor cells overexpressing CAIX via the specific binding with BS, and the inhibition of CAIX activity could not only decrease the intracellular pH to accelerate Fe3+/Cu2+ release, H2O2 self-supply and Fenton reaction, but also suppress tumor metastasis by alleviating the extracellular acidity. The reduction of Fe3+/Cu2+ by intracellular GSH could further amplify ·OH generation, and the GSH depletion could in turn inhibit GPX-4 mediated antioxidant reaction to induce ferroptosis, resulting in effective therapeutic efficacy by enhanced ferroptosis/cuproptosis/CDT via tumor microenvironment regulation.
Collapse
Affiliation(s)
- Haoyu Huang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, PR China
| | - Honghua Guo
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, PR China
| | - Junjie Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, PR China
| | - Cheng Ni
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China
| | - Li Xia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, PR China
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China
| | - Jindong Xia
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, PR China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China.
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China.
| |
Collapse
|
10
|
Duan Y, Zhang W, Ouyang Y, Yang Q, Zhang Q, Zhao S, Chen C, Xu T, Zhang Q, Ran H, Liu H. Proton Sponge Nanocomposites for Synergistic Tumor Elimination via Autophagy Inhibition-Promoted Cell Apoptosis and Macrophage Repolarization-Enhanced Immune Response. ACS APPLIED MATERIALS & INTERFACES 2024; 16:17285-17299. [PMID: 38539044 DOI: 10.1021/acsami.4c01451] [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: 04/12/2024]
Abstract
Cytoprotective autophagy and an immunosuppressive tumor microenvironment (TME) are two positive promoters for tumor proliferation and metastasis that severely hinder therapeutic efficacy. Inhibiting autophagy and reconstructing TME toward macrophage activation simultaneously are of great promise for effective tumor elimination, yet are still a huge challenge. Herein, a kind of dendrimer-based proton sponge nanocomposites was designed and constructed for tumor chemo/chemodynamic/immunotherapy through autophagy inhibition-promoted cell apoptosis and macrophage repolarization-enhanced immune response. These obtained nanocomposites contain a proton sponge G5AcP dendrimer, a Fenton-like agent Cu(II), and chemical drug doxorubicin (DOX). When accumulated in tumor regions, G5AcP can act as an immunomodulator to realize deacidification-promoted macrophage repolarization toward antitumoral type, which then secretes inflammatory cytokines to activate T cells. They also regulate intracellular lysosomal pH to inhibit cytoprotective autophagy. The released Cu(II) and DOX can induce aggravated damage through a Fenton-like reaction and chemotherapeutic effect in this autophagy-inhibition condition. Tumor-associated antigens are released from these dying tumor cells to promote the maturity of dendritic cells, further activating T cells. Effective tumor elimination can be achieved by this dendrimer-based therapeutic strategy, providing significant guidance for the design of a promising antitumor nanomedicine.
Collapse
Affiliation(s)
- Yifan Duan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Yi Ouyang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Qiang Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Qiuye Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Sheng Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Chunmei Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Ting Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Qun Zhang
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Hui Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing 400715, China
- Office of Clinical Trial of Drug, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, Guangdong, China
| |
Collapse
|
11
|
Zhao L, Tan L, Wu Q, Fu C, Ren X, Ren J, Wang Z, Zhang J, Meng X. A two-stage exacerbated hypoxia nanoengineering strategy induced amplifying activation of tirapazamine for microwave hyperthermia-chemotherapy of breast cancer. J Colloid Interface Sci 2024; 659:178-190. [PMID: 38163404 DOI: 10.1016/j.jcis.2023.12.149] [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: 10/18/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Microwave hyperthermia (MH) is an emerging treatment for solid tumors, such as breast cancer, due to its advantages of minimally invasive and deep tissue penetration. However, MH induced tumor hypoxia is still an obstacle to breast tumor treatment failure. Therefore, an original nanoengineering strategy was proposed to exacerbate hypoxia in two stages, thereby amplifying the efficiency of activating tirapazamine (TPZ). And a novel microwave-sensitized nanomaterial (GdEuMOF@TPZ, GEMT) is designed. GdEuMOF (GEM) nanoparticles are certified excellent microwave (MW) sensitization performance, thus improving tumor selectivity to achieve MH. Meanwhile MW can aggravate the generation of thrombus and caused local circulatory disturbance of tumor, resulting in the Stage I exacerbated hypoxia environment passively. Due to tumor heterogeneity and uneven hypoxia, GEMT nanoparticles under microwave could actively deplete residual oxygen through the chemical reaction, exacerbating hypoxia level more evenly, thus forming the Stage II of exacerbated hypoxia environment. Consequently, a two-stage exacerbated hypoxia GEMT nanoparticles realize amplifying activation of TPZ, significantly enhance the efficacy of microwave hyperthermia and chemotherapy, and effectively inhibit breast cancer. This research provides insights into the development of progressive nanoengineering strategies for effective breast tumor therapy.
Collapse
Affiliation(s)
- Lirong Zhao
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Longfei Tan
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Qiong Wu
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Changhui Fu
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xiangling Ren
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jun Ren
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Zhen Wang
- Laboratory Medicine Center, Allergy center, Department of Transfusion medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Jingjie Zhang
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xianwei Meng
- Key Laboratory of Cryogenic Science and Technology, Technical Institute of Physics and Chemistry, Laboratory of Controllable Preparation and Application of Nanomaterials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
12
|
Shi Q, Zhang W, Zhou Y, Huang S, Yu J, Yang M, Zhang Z, Ma J, Luo J, Rao S, Lu D, Peng S, Cao Y, Liu L, Yan Z. Hypoxia-activated cascade nanovaccine for synergistic chemoembolization-immune therapy of hepatocellular carcinoma. Biomaterials 2024; 306:122480. [PMID: 38271787 DOI: 10.1016/j.biomaterials.2024.122480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/25/2023] [Accepted: 01/20/2024] [Indexed: 01/27/2024]
Abstract
In this work, a promising treatment strategy for triggering robust antitumor immune responses in transarterial chemoembolization of hepatocellular carcinoma (HCC) is presented. The zeolitic imidazolate framework nanoparticles loaded with hypoxia-activated prodrug tirapazamine and immune adjuvant resiquimod facilitated in situ generation of nanovaccine via a facile approach. The nanovaccine can strengthen the ability of killing the liver cancer cells under hypoxic environment, while was capable of improving immunogenic tumor microenvironment and triggering strong antitumor immune responses by increasing the primary and distant intratumoral infiltration of immune cells such as cytotoxic T cells. Moreover, a porous microcarrier, approved by FDA as pharmaceutical excipient, was designed to achieve safe and effective delivery of the nanovaccine via transarterial therapy in rabbit orthotopic VX2 liver cancer model. The microcarrier exhibited the characteristics of excellent drug loading and occlusion of peripheral artery. The collaborative delivery of the microcarrier and nanovaccine demonstrated an exciting inhibitory effect on solid tumors and tumor metastases, which provided a great potential as novel combination therapy for HCC interventional therapy.
Collapse
Affiliation(s)
- Qin Shi
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yongjie Zhou
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Songjiang Huang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiaze Yu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Minjie Yang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zihan Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingqin Ma
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jianjun Luo
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shengxiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong, 519000, China
| | - Yongbin Cao
- State Key Laboratory of Genetic Engineering and MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China; Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Lingxiao Liu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Zhiping Yan
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institution of Medical Imaging, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Zhang C, Guo Y, Shen M, Shi X. Dendrimer‐Based Nanodrugs for Chemodynamic Therapy of Tumors. ADVANCED NANOBIOMED RESEARCH 2024; 4. [DOI: 10.1002/anbr.202300149] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
To realize precise tumor treatment, chemodynamic therapy (CDT) that utilizes metal element to trigger Fenton or Fenton‐like reaction to generate cytotoxic hydroxyl radicals in tumor region has been widely investigated. Recently, the dendrimers featured with abundant surface functional groups and excellent biocompatibility are regarded as promising carriers of metal elements for tumor delivery. Much effort has been devoted to design dendrimer‐based nanodrugs for CDT and CDT‐involved synergistic therapy of tumors. Herein, the recent advances in the construction of dendrimer‐based nanodrugs (in most cases, poly(amidoamine) dendrimers) for CDT, CDT/chemotherapy, CDT/phototherapy, CDT/gene therapy, or CDT‐involved multimodal therapy are reviewed. Furthermore, the future perspectives with regard to the development of dendrimer‐based nanodrugs for CDT‐involved tumor treatment are also briefly discussed.
Collapse
Affiliation(s)
- Caiyun Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P. R. China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P. R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P. R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P. R. China
| |
Collapse
|
14
|
Shcharbin D, Zhogla V, Abashkin V, Gao Y, Majoral JP, Mignani S, Shen M, Bryszewska M, Shi X. Recent advances in multifunctional dendrimer-based complexes for cancer treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1951. [PMID: 38456205 DOI: 10.1002/wnan.1951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/29/2024] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
The application of nanotechnology in biological and medical fields have resulted in the creation of new devices, supramolecular systems, structures, complexes, and composites. Dendrimers are relatively new nanotechnological polymers with unique features; they are globular in shape, with a topological structure formed by monomeric subunit branches diverging to the sides from the central nucleus. This review analyzes the main features of dendrimers and their applications in biology and medicine regarding cancer treatment. Dendrimers have applications that include drug and gene carriers, antioxidant agents, imaging agents, and adjuvants, but importantly, dendrimers can create complex nanosized constructions that combine features such as drug/gene carriers and imaging agents. Dendrimer-based nanosystems include different metals that enhance oxidative stress, polyethylene glycol to provide biosafety, an imaging agent (a fluorescent, radioactive, magnetic resonance imaging probe), a drug or/and nucleic acid that provides a single or dual action on cells or tissues. One of major benefit of dendrimers is their easy release from the body (in contrast to metal nanoparticles, fullerenes, and carbon nanotubes), allowing the creation of biosafe constructions. Some dendrimers are already clinically approved and are being used as drugs, but many nanocomplexes are currently being studied for clinical practice. In summary, dendrimers are very useful tool in the creation of complex nanoconstructions for personalized nanomedicine. This article is categorized under: Diagnostic Tools > Diagnostic Nanodevices Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dzmitry Shcharbin
- Institute of Biophysics and Cell Engineering of NASB, Minsk, Belarus
| | - Viktoria Zhogla
- Institute of Biophysics and Cell Engineering of NASB, Minsk, Belarus
| | - Viktar Abashkin
- Institute of Biophysics and Cell Engineering of NASB, Minsk, Belarus
| | - Yue Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, Toulouse, France
- Université Toulouse, Toulouse, France
| | - Serge Mignani
- Centro de Química da Madeira (CQM), MMRG, Universidade da Madeira, Campus Universitário da Penteada, Funchal, Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, China
- Centro de Química da Madeira (CQM), MMRG, Universidade da Madeira, Campus Universitário da Penteada, Funchal, Portugal
| |
Collapse
|
15
|
Gao Y, Ouyang Z, Shen S, Yu H, Jia B, Wang H, Shen M, Shi X. Manganese Dioxide-Entrapping Dendrimers Co-Deliver Protein and Nucleotide for Magnetic Resonance Imaging-Guided Chemodynamic/Starvation/Immune Therapy of Tumors. ACS NANO 2023; 17:23889-23902. [PMID: 38006397 DOI: 10.1021/acsnano.3c08174] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Development of a nanoscale drug delivery system that can simultaneously exert efficient tumor therapeutic efficacy while creating the desired antitumor immune responses is still challenging. Herein, we report the use of a manganese dioxide (MnO2)-entrapping dendrimer nanocarrier to codeliver glucose oxidase (GOx) and cyclic GMP-AMP (cGAMP), an agonist of the stimulator of interferon genes (STING) for improved tumor chemodynamic/starvation/immune therapy. Methoxy poly(ethylene glycol) (mPEG)- and phenylboronic acid (PBA)-modified generation 5 (G5) poly(amidoamine) dendrimers were first synthesized and then entrapped with MnO2 nanoparticles (NPs) to generate the hybrid MnO2@G5-mPEG-PBA (MGPP) NPs. The created MGPP NPs with an MnO2 core size of 2.8 nm display efficient glutathione depletion ability, and a favorable Mn2+ release profile under a tumor microenvironment mimetic condition to enable Fenton-like reaction and T1-weighted magnetic resonance (MR) imaging. We show that the MGPP-mediated GOx delivery facilitates enhanced chemodynamic/starvation therapy of cancer cells in vitro, and further codelivery of cGAMP can effectively trigger immunogenic cell death (ICD) to strongly promote the maturation of dendritic cells. In a bilateral mouse colorectal tumor model, the dendrimer delivery nanosystem elicits a potent antitumor performance with a strong abscopal effect, greatly improving the overall mouse survival rate. Importantly, the dendrimer-mediated codelivery not only allows the coordination of Mn2+ with GOx and cGAMP for respective chemodynamic/starvation-triggered ICD and augmented STING activation to boost systemic antitumor immune responses, but also enables T1-weighted tumor MR imaging, potentially serving as a promising nanoplatform for enhanced antitumor therapy with desired immune responses.
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Zhijun Ouyang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Siyan Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Hongwei Yu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Bingyang Jia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| |
Collapse
|
16
|
Xie P, Liu P. Formulation of DOX-dimer with bi-functionalized chitooligosaccharide for tumor-specific self-boosted drug release and synergistic chemo/chemodynamic therapy. Carbohydr Polym 2023; 320:121210. [PMID: 37659811 DOI: 10.1016/j.carbpol.2023.121210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/15/2023] [Indexed: 09/04/2023]
Abstract
The toxic side effects and possible drug resistance of the chemotherapeutics hinder their antitumor efficacy. Here, a pH/reactive oxygen species (ROS) dual-triggered nanodrug was developed for the tumor-specific self-boosted drug release and synergistic chemo/chemodynamic therapy, by formulating ROS-cleavable doxorubicin (DOX)-based dimer (DOX-TK-DOX) with bi-functionalized chitooligosaccharide (COS-Fc-TK) with ferrocenecarboxylic acid (Fc) and thioketal (TK). The resultant DOX-TK-DOX/COS-Fc-TK nanoparticles with a high DOX content of 39.70 % showed tumor-specific self-boosted drug release, which was triggered by highly toxic OH generated via Fc-catalyzed Fenton reaction of the endogenous H2O2 in tumor intracellular microenvironment. As a result, a synergistic chemo/chemodynamic therapy with combination index (CI) of 0.94 was achieved for selective treatment of tumors.
Collapse
Affiliation(s)
- Pengwei Xie
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
17
|
Du C, Wang C, Jiang SH, Zheng X, Li Z, Yao Y, Ding Y, Chen T, Yi H. pH/GSH dual-responsive supramolecular nanomedicine for hypoxia-activated combination therapy. Biomater Sci 2023; 11:5674-5679. [PMID: 37439102 DOI: 10.1039/d3bm00519d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Moderate oxygen (O2) supply and uneven distribution of oxygen at the tumor site usually hinder the therapeutic efficacy of hypoxia-activated prodrugs. In this report, we designed a ferrocene-containing supramolecular nanomedicine (PFC/GOD-TPZ) with the PEG corona and disulfide-bond cross-linked core to co-encapsulate 4-di-N-oxide tirapazamine (TPZ) and glucose oxidase (GOD). The PEG corona of PFC/GOD-TPZ could be weakly acidic tumor pH-responsively detached for an enhanced cellular internalization, while the disulfide-bond cross-linked core could be cleavaged by intracellular glutathione (GSH) to present a GSH-triggered drug-release behavior. Subsequently, the cascade reactions, including catalytic reactions among the released GOD, glucose, and O2 to generate H2O2 and the subsequent Fenton reaction between ferrocene and H2O2, occurred. With the depletion of O2, the non-toxic TPZ was activated and converted into the cytotoxic therapeutic agent benzotriazinyl (BTZ) radical under the exacerbated hypoxic microenvironment. Collectively, the PFC/GOD-TPZ provides a promising strategy for effective combination therapy of GOD-mediated starvation therapy, chemodynamic therapy (CDT), and hypoxia-activated chemotherapy (CT).
Collapse
Affiliation(s)
- Chang Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chenwei Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, P. R. China.
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiangqin Zheng
- Department of Gynecology Oncology, Fujian Provincial Maternity and Children's Hospital, Fujian Provincial Key Gynecology Clinical Specialty, The Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China.
| | - Zelong Li
- Department of Gynecology Oncology, Fujian Provincial Maternity and Children's Hospital, Fujian Provincial Key Gynecology Clinical Specialty, The Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China.
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, P. R. China.
| | - Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, P. R. China.
| | - Tingting Chen
- School of Chemistry and Chemical Engineering, Nantong University, Nantong 226019, P. R. China.
| | - Huan Yi
- Department of Gynecology Oncology, Fujian Provincial Maternity and Children's Hospital, Fujian Provincial Key Gynecology Clinical Specialty, The Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China.
| |
Collapse
|
18
|
Ni C, Ouyang Z, Li G, Liu J, Cao X, Zheng L, Shi X, Guo R. A tumor microenvironment-responsive core-shell tecto dendrimer nanoplatform for magnetic resonance imaging-guided and cuproptosis-promoted chemo-chemodynamic therapy. Acta Biomater 2023; 164:474-486. [PMID: 37040813 DOI: 10.1016/j.actbio.2023.04.003] [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: 12/31/2022] [Revised: 03/25/2023] [Accepted: 04/04/2023] [Indexed: 04/13/2023]
Abstract
Theranostic nanoplatforms for combination tumor therapy have gained lots of attention recently due to the optimized therapeutic efficiency and simultaneous diagnosis performance. Herein, a novel tumor microenvironment (TME)-responsive core-shell tecto dendrimer (CSTD) was assembled by phenylboronic acid- and mannose-modified poly(amidoamine) dendrimers via the phenylboronic ester bonds that are responsive to low pH and reactive oxygen species (ROS), and efficiently loaded with copper ions and chemotherapeutic drug disulfiram (DSF) for tumor-targeted magnetic resonance (MR) imaging and cuproptosis-promoted chemo-chemodynamic therapy. The formed CSTD-Cu(II)@DSF could be specifically taken up by MCF-7 breast cancer cells, accumulated to the tumor model after circulation, and released drugs in response to the weakly acidic TME with overexpressed ROS. The enriched intracellular Cu(II) ions could induce the oligomerization of lipoylated proteins and proteotoxic stress for cuproptosis, and lipid peroxidation for chemodynamic therapy as well. Moreover, the CSTD-Cu(II)@DSF could cause the dysfunction of mitochondria and arrest the cell cycle at the G2/M phase, leading to enhanced DSF-mediated cell apoptosis. As a result, CSTD-Cu(II)@DSF could effectively inhibit the growth of MCF-7 tumors by a combination therapy strategy integrating chemotherapy with cuproptosis and chemodynamic therapy. Lastly, the CSTD-Cu(II)@DSF also displays Cu(II)-associated r1 relaxivity, allowing for T1-weighted real-time MR imaging of tumors in vivo. The developed tumor-targeted and TME-responsive CSTD-based nanomedicine formulation may be developed for accurate diagnosis and synergistic treatment of other cancer types. STATEMENT OF SIGNIFICANCE: Constructing an effective nanoplatform for the combination of therapeutic effects and real-time tumor imaging remains a challenge. In this study, we reported for the first time an all-in-one tumor-targeted and tumor microenvironment (TME) responsive nanoplatform based on core-shell tecto dendrimer (CSTD) for the cuproptosis-promoted chemo-chemodynamic therapy and enhanced MR imaging. The efficient loading, selective tumor-targeting, and TME-responsive release of Cu(II) and disulfiram could enhance the intracellular accumulation of drugs, induce cuproptosis of cancer cells, and amplify the synergistic chemo-chemodynamic therapeutic effect, resulting in enhanced MR imaging and accelerated tumor eradication. This study sheds new light on the development of theranostic nanoplatforms for early accurate diagnosis and effective treatment of cancers.
Collapse
Affiliation(s)
- Cheng Ni
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Zhijun Ouyang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Gaoming Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Junjie Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, China
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Linfeng Zheng
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People's Republic of China.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China.
| |
Collapse
|
19
|
Liang J, Zhang W, Wang J, Li W, Ge F, Jin W, Tao Y. Development of the Cu/ZIF-8 MOF Acid-Sensitive Nanocatalytic Platform Capable of Chemo/Chemodynamic Therapy with Improved Anti-Tumor Efficacy. ACS OMEGA 2023; 8:19402-19412. [PMID: 37305251 PMCID: PMC10249029 DOI: 10.1021/acsomega.3c00269] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/11/2023] [Indexed: 06/13/2023]
Abstract
Recently, the combination of chemotherapy and chemodynamic therapy (CDT) has become a desirable strategy in the treatment of cancer. However, a satisfactory therapeutic outcome is often difficult to achieve due to the deficiency of endogenous H2O2 and O2 in the tumor microenvironment. In this study, a CaO2@DOX@Cu/ZIF-8 nanocomposite was prepared as a novel nanocatalytic platform to enable the combination of chemotherapy and CDT in cancer cells. The anticancer drug doxorubicin hydrochloride (DOX) was loaded onto calcium peroxide (CaO2) nanoparticles (NPs) to form CaO2@DOX, which was then encapsulated in a copper zeolitic imidazole ester MOF (Cu/ZIF-8) to form CaO2@DOX@Cu/ZIF-8 NPs. In the mildly acidic tumor microenvironment, CaO2@DOX@Cu/ZIF-8 NPs rapidly disintegrated, releasing CaO2, which reacted with water to generate H2O2 and O2 in the tumor microenvironment. The ability of CaO2@DOX@Cu/ZIF-8 NPs to combine chemotherapy and CDT was assessed by conducting cytotoxicity, living dead staining, cellular uptakes, H&E staining, and TUNEL assays in vitro and in vivo. The combination of chemotherapy and CDT of CaO2@DOX@Cu/ZIF-8 NPs had a more favorable tumor suppression effect than the nanomaterial precursors, which were not capable of the combined chemotherapy/CDT.
Collapse
|
20
|
Yu J, Wang L, Xie X, Zhu W, Lei Z, Lv L, Yu H, Xu J, Ren J. Multifunctional Nanoparticles Codelivering Doxorubicin and Amorphous Calcium Carbonate Preloaded with Indocyanine Green for Enhanced Chemo-Photothermal Cancer Therapy. Int J Nanomedicine 2023; 18:323-337. [PMID: 36700147 PMCID: PMC9869790 DOI: 10.2147/ijn.s394896] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/07/2023] [Indexed: 01/20/2023] Open
Abstract
Background Multifunctional stimuli-responsive nanoparticles with photothermal-chemotherapy provided a powerful tool for improving the accuracy and efficiency in the treatment of malignant tumors. Methods Herein, photosensitizer indocyanine green (ICG)-loaded amorphous calcium-carbonate (ICG@) nanoparticle was prepared by a gas diffusion reaction. Doxorubicin (DOX) and ICG@ were simultaneously encapsulated into poly(lactic-co-glycolic acid)-ss-chondroitin sulfate A (PSC) nanoparticles by a film hydration method. The obtained PSC/ICG@+DOX hybrid nanoparticles were characterized and evaluated by Fourier transform infrared spectroscopy (FTIR), dynamic light scattering (DLS), transmission electron microscopy (TEM), and differential scanning calorimetry (DSC). The cellular uptake and cytotoxicity of PSC/ICG@+DOX nanoparticles were analyzed by confocal laser scanning microscopy (CLSM) and MTT assay in 4T1 cells. In vivo antitumor activity of the nanoparticles was evaluated in 4T1-bearing Balb/c mice. Results PSC/ICG@+DOX nanoparticles were nearly spherical in shape by TEM observation, and the diameter was 407 nm determined by DLS. Owing to calcium carbonate and disulfide bond linked copolymer, PSC/ICG@+DOX nanoparticles exhibited pH and reduction-sensitive drug release. Further, PSC/ICG@+DOX nanoparticles showed an effective photothermal effect under near-infrared (NIR) laser irradiation, and improved cellular uptake and cytotoxicity in breast cancer 4T1 cells. Importantly, PSC/ICG@+DOX nanoparticles demonstrated the most effective suppression of tumor growth in orthotopic 4T1-bearing mice among the treatment groups. In contrast with single chemotherapy or photothermal therapy, chemo-photothermal treatment by PSC/ICG@+DOX nanoparticles synergistically inhibited the growth of 4T1 cells. Conclusion This study demonstrated that PSC/ICG@+DOX nanoparticles with active targeting and stimuli-sensitivity would be a promising strategy to enhance chemo-photothermal cancer therapy.
Collapse
Affiliation(s)
- Jingmou Yu
- Huzhou Key Laboratory of Medical and Environmental Applications Technologies, School of Life Sciences, Huzhou University, Huzhou, People’s Republic of China,Jiangxi Provincial Laboratory Laboratory of System Biomedicine, Jiujiang University, Jiujiang, People’s Republic of China,School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China,Correspondence: Jingmou Yu; Jin Ren, Email ;
| | - Liangliang Wang
- Affiliated Hospital of Jiujiang University, Jiujiang, People’s Republic of China
| | - Xin Xie
- Jiangxi Provincial Laboratory Laboratory of System Biomedicine, Jiujiang University, Jiujiang, People’s Republic of China
| | - Wenjing Zhu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Zhineng Lei
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Linghui Lv
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Hongling Yu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Jing Xu
- Affiliated Hospital of Jiujiang University, Jiujiang, People’s Republic of China
| | - Jin Ren
- Jiangxi Provincial Laboratory Laboratory of System Biomedicine, Jiujiang University, Jiujiang, People’s Republic of China,School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| |
Collapse
|
21
|
Shen M, Chen C, Guo Q, Wang Q, Liao J, Wang L, Yu J, Xue M, Duan Y, Zhang J. Systemic Delivery of mPEG-Masked Trispecific T-Cell Nanoengagers in Synergy with STING Agonists Overcomes Immunotherapy Resistance in TNBC and Generates a Vaccination Effect. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203523. [PMID: 36089659 PMCID: PMC9661824 DOI: 10.1002/advs.202203523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/13/2022] [Indexed: 06/15/2023]
Abstract
T-cell engagers (TCEs) represent a breakthrough in hematological malignancy treatment but are vulnerable to antigen escape and lack a vaccination effect. The "immunologically cold" solid tumor presents substantial challenges due to intratumor heterogeneity and an immunosuppressive tumor microenvironment (TME). Here, a methoxy poly(ethylene glycol) (mPEG)-masked CD44×PD-L1/CD3 trispecific T-cell nanoengager loaded with the STING agonist c-di-AMP (CDA) (PmTriTNE@CDA) for the treatment of triple-negative breast cancer (TNBC) is rationally designed. PmTriTNE@CDA shows tumor-specific accumulation and is preferentially unmasked in response to a weakly acidic TME to prevent on-target off-tumor toxicity. The unmasked CD44×PD-L1/CD3 trispecific T-cell nanoengager (TriTNE) targets dual tumor-associated antigens (TAAs) to redirect CD8+ T cells for heterogeneous TNBC lysis while achieving PD-L1 blockade. PmTriTNE synergized with CDA to transform the cold tumor into a hot tumor, eradicate the large established TNBC tumor, and induce protective immune memory in a 4T1 orthotopic tumor model without causing obvious toxicity. PmTriTNE@CDA shows potent efficacy in cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models. This study serves as a proof-of-concept demonstration of a nanobased TCEs strategy to expand therapeutic combinations that previously could not be achieved due to systemic toxicity with the aim of overcoming TNBC heterogeneity and immunotherapy resistance.
Collapse
Affiliation(s)
- Ming Shen
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
- Shanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai200032China
| | - Chuanrong Chen
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
- Department of OncologyYijishan Hospital of Wannan Medical CollegeWuhu240001China
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related GenesRenji HospitalSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Quan Wang
- State Key Laboratory of Oncogenes and Related GenesRenji HospitalSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Jinghan Liao
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
| | - Liting Wang
- State Key Laboratory of Oncogenes and Related GenesRenji HospitalSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200127China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
| | - Man Xue
- Shanghai Institute for Biomedical and Pharmaceutical TechnologiesShanghai200032China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
| | - Jiali Zhang
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
| |
Collapse
|
22
|
Li M, Zhang W, Xu X, Liu G, Dong M, Sun K, Zhang P. Nanosystems for chemodynamic based combination therapy: Strategies and recent advances. Front Pharmacol 2022; 13:1065438. [PMID: 36386143 PMCID: PMC9651923 DOI: 10.3389/fphar.2022.1065438] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
Chemodynamic therapy (CDT), a newly developed approach for cancer treatment, can convert hydrogen peroxide (H2O2) into toxic hydroxyl radicals (•OH) by using Fenton/Fenton-like reaction to kill tumor cells. However, due to the complexity of the intracellular environment of tumor cells, the therapeutic efficacy of CDT was severely restricted. Recently, combination therapy strategies have become popular approaches for tumor treatment, and there are numerous studies have demonstrated that the CDT-based combination strategies can significantly improve the anti-tumor efficiency of CDT. In this review, we outline some of the recent progress in cancer chemodynamic therapy from 2020, and discuss the progress in the design of nanosystems for CDT synergistic combination therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Peng Zhang
- *Correspondence: Kaoxiang Sun, ; Peng Zhang,
| |
Collapse
|
23
|
Achieving a “all in one” Fe/Tm-MOFs with controllable photothermal and catalytic performance for imaging-guided multi-modal synergetic therapy. J Colloid Interface Sci 2022; 623:124-134. [DOI: 10.1016/j.jcis.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/20/2022]
|
24
|
Deng R, Wang Y, Bu Y, Wu H. BNIP3 mediates the different adaptive responses of fibroblast-like synovial cells to hypoxia in patients with osteoarthritis and rheumatoid arthritis. Mol Med 2022; 28:64. [PMID: 35690741 PMCID: PMC9188199 DOI: 10.1186/s10020-022-00490-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/25/2022] [Indexed: 01/01/2023] Open
Abstract
Background Hypoxia is one of the important characteristics of synovial microenvironment in rheumatoid arthritis (RA), and plays an important role in synovial hyperplasia. In terms of cell survival, fibroblast-like synovial cells (FLSs) are relatively affected by hypoxia. In contrast, fibroblast-like synovial cells from patients with RA (RA-FLSs) are particularly resistant to hypoxia-induced cell death. The purpose of this study was to evaluate whether fibroblast-like synovial cells in patients with osteoarthritis (OA-FLSs) and RA-FLSs have the same adaptation to hypoxia. Methods CCK-8, flow cytometry and BrdU were used to detect the proliferation of OA-FLSs and RA-FLSs under different oxygen concentrations. Apoptosis was detected by AV/PI, TUNEL and Western blot, mitophagy was observed by electron microscope, laser confocal microscope and Western blot, the state of mitochondria was detected by ROS and mitochondrial membrane potential by flow cytometry, BNIP3 and HIF-1α were detected by Western blot and RT-qPCR. The silencing of BNIP3 was achieved by stealth RNA system technology. Results After hypoxia, the survival rate of OA-FLSs decreased, while the proliferation activity of RA-FLSs further increased. Hypoxia induced an increase in apoptosis and inhibition of mitophagy in OA-FLSs, but not in RA-FLSs. Hypoxia led to a more lasting adaptive response. RA-FLSs displayed a more significant increase in the expression of genes transcriptionally regulated by HIF-1α. Interestingly, they showed higher BNIP3 expression than OA-FLSs, and showed stronger mitophagy and proliferation activities. BNIP3 siRNA experiment confirmed the potential role of BNIP3 in the survival of RA-FLSs. Inhibition of BNIP3 resulted in the decrease of cell proliferation, mitophagy and the increase of apoptosis. Conclusion In summary, RA-FLSs maintained intracellular redox balance through mitophagy to promote cell survival under hypoxia. The mitophagy of OA-FLSs was too little to maintain the redox balance of mitochondria, resulting in apoptosis. The difference of mitophagy between OA-FLSs and RA-FLSs under hypoxia is mediated by the level of BNIP3 expression.
Collapse
Affiliation(s)
- Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China. .,Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China. .,Anhui Province Key Laboratory of Research &, Development of Chinese Medicine, Hefei, 230012, China. .,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
25
|
Zhang Y, Lou J, Williams GR, Ye Y, Ren D, Shi A, Wu J, Chen W, Zhu LM. Cu2+-Chelating Mesoporous Silica Nanoparticles for Synergistic Chemotherapy/Chemodynamic Therapy. Pharmaceutics 2022; 14:pharmaceutics14061200. [PMID: 35745773 PMCID: PMC9229203 DOI: 10.3390/pharmaceutics14061200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/22/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
In this study, a pH-responsive controlled-release mesoporous silica nanoparticle (MSN) formulation was developed. The MSNs were functionalized with a histidine (His)-tagged targeting peptide (B3int) through an amide bond, and loaded with an anticancer drug (cisplatin (CP)) and a lysosomal destabilization mediator (chloroquine (CQ)). Cu2+ was then used to seal the pores of the MSNs via chelation with the His-tag. The resultant nanoparticles showed pH-responsive drug release, and could effectively target tumor cells via the targeting effect of B3int. The presence of CP and Cu2+ permits reactive oxygen species to be generated inside cells; thus, the chemotherapeutic effect of CP is augmented by chemodynamic therapy. In vitro and in vivo experiments showed that the nanoparticles are able to effectively kill tumor cells. An in vivo cancer model revealed that the nanoparticles increase apoptosis in tumor cells, and thereby diminish the tumor volume. No off-target toxicity was noted. It thus appears that the functionalized MSNs developed in this work have great potential for targeted, synergistic anticancer therapies.
Collapse
Affiliation(s)
- Yanyan Zhang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (Y.Z.); (J.L.); (Y.Y.); (D.R.)
| | - Jiadong Lou
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (Y.Z.); (J.L.); (Y.Y.); (D.R.)
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
| | - Yuhan Ye
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (Y.Z.); (J.L.); (Y.Y.); (D.R.)
| | - Dandan Ren
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (Y.Z.); (J.L.); (Y.Y.); (D.R.)
| | - Anhua Shi
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China; (A.S.); (J.W.)
| | - Junzi Wu
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming 650500, China; (A.S.); (J.W.)
| | - Wenling Chen
- School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
- Correspondence: (W.C.); (L.-M.Z.)
| | - Li-Min Zhu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; (Y.Z.); (J.L.); (Y.Y.); (D.R.)
- Correspondence: (W.C.); (L.-M.Z.)
| |
Collapse
|
26
|
Ferrocene-based multifunctional nanoparticles for combined chemo/chemodynamic/photothermal therapy. J Colloid Interface Sci 2022; 626:719-728. [DOI: 10.1016/j.jcis.2022.06.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/26/2022] [Accepted: 06/23/2022] [Indexed: 11/21/2022]
|
27
|
Zhang H, Zhang M, Zhang X, Gao Y, Ma Y, Chen H, Wan J, Li C, Wang F, Sun X. Enhanced postoperative cancer therapy by iron-based hydrogels. Biomater Res 2022; 26:19. [PMID: 35606838 PMCID: PMC9125885 DOI: 10.1186/s40824-022-00268-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/11/2022] [Indexed: 12/13/2022] Open
Abstract
AbstractSurgical resection is a widely used method for the treatment of solid tumor cancers. However, the inhibition of tumor recurrence and metastasis are the main challenges of postoperative tumor therapy. Traditional intravenous or oral administration have poor chemotherapeutics bioavailability and undesirable systemic toxicity. Polymeric hydrogels with a three-dimensional network structure enable on-site delivery and controlled release of therapeutic drugs with reduced systemic toxicity and have been widely developed for postoperative adjuvant tumor therapy. Among them, because of the simple synthesis, good biocompatibility, biodegradability, injectability, and multifunctionality, iron-based hydrogels have received extensive attention. This review has summarized the general synthesis methods and construction principles of iron-based hydrogels, highlighted the latest progress of iron-based hydrogels in postoperative tumor therapy, including chemotherapy, photothermal therapy, photodynamic therapy, chemo-dynamic therapy, and magnetothermal-chemical combined therapy, etc. In addition, the challenges towards clinical application of iron-based hydrogels have also been discussed. This review is expected to show researchers broad perspectives of novel postoperative tumor therapy strategy and provide new ideas in the design and application of novel iron-based hydrogels to advance this sub field in cancer nanomedicine.
Collapse
|