1
|
Wang H, Deng X, Chen XZ, Ullah A. Multifunctional Temperature-Sensitive Lipid-Protein-Polymer Conjugates: Tailored Drug Delivery and Bioimaging. ACS APPLIED MATERIALS & INTERFACES 2024; 16:67411-67423. [PMID: 39576197 DOI: 10.1021/acsami.4c16258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
In this study, we introduce a protein-polymer bioconjugate comprising bovine serum albumin (BSA) and a lipid-based thermoresponsive block copolymer. These amphiphilic BSA-polymer conjugates can autonomously be organized into vesicular compartments for codelivery of glucose oxidase (GOx) and doxorubicin (DOX), demonstrating high drug loading content and remarkable antitumor activity via synergistic cancer therapy combining chemo-starvation strategies. Through the incorporation of a hydrophilic BSA block, the lower critical solution temperature (LCST) of the bioconjugates is tuned to around 40 °C, facilitating their targeted drug delivery to tumor cells. Consequently, these smart protein-polymer conjugates present greater promise compared to traditional drug delivery vehicles, particularly in the realm of anticancer therapy. Moreover, these bioconjugates displayed enhanced intracellular fluorescence intensity with increasing temperature, attributed to the clustering-triggered emission of the nonconventional chromophore moieties within poly(vinylcaprolactam) (PNVCL). The active aggregation-induced emission (AIE) characteristic and excellent biocompatibility suggest an opportunity to further apply these bioconjugates for biosensing and cellular imaging.
Collapse
Affiliation(s)
- Huiqi Wang
- Lipid Utilization Laboratories - Lipids/Materials Chemistry Group, Department of Agricultural, Food and Nutritional Science, 4-10 Agriculture/Forestry Centre, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Xiaoling Deng
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Aman Ullah
- Lipid Utilization Laboratories - Lipids/Materials Chemistry Group, Department of Agricultural, Food and Nutritional Science, 4-10 Agriculture/Forestry Centre, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| |
Collapse
|
2
|
Agrawal S, Singh GK, Tiwari S. Focused starvation of tumor cells using glucose oxidase: A comprehensive review. Int J Biol Macromol 2024; 281:136444. [PMID: 39389487 DOI: 10.1016/j.ijbiomac.2024.136444] [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: 06/12/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Starvation therapy targets the high metabolic demand of tumor cells. It primarily leans over the consumption of intracellular glucose and simultaneous blockade of alternative metabolic pathways. The strategy involves the use of glucose oxidase (GOx) for catalyzing the conversion of glucose into gluconic acid and hydrogen peroxide. Under these conditions, metabolic re-programming of tumor cells enables the utilization of substrates such as amino acids, fatty acids and lipids. This can be overcome by co-administration of chemo-, photo- and immuno-therapeutics together with glucose oxidase. Targeted delivery of glucose oxidase at tumor site can be enabled with the use of nanoformulations. In this review, we highlight that the outcomes of starvation therapy can be improved using rationally developed nano-formulations. It is possible to load synergistically acting bioactives in these formulations and deliver in site-specific manner and hence achieve the elimination of tumors cells with greater efficacy.
Collapse
Affiliation(s)
- Shivanshu Agrawal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India
| | - Gireesh K Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya 824236, India
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow 226002, India.
| |
Collapse
|
3
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [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: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
4
|
Cai M, Fu T, Zhu R, Hu P, Kong J, Liao S, Du Y, Zhang Y, Qu C, Dong X, Yin X, Ni J. An iron-based metal-organic framework nanoplatform for enhanced ferroptosis and oridonin delivery as a comprehensive antitumor strategy. Acta Pharm Sin B 2024; 14:4073-4086. [PMID: 39309488 PMCID: PMC11413704 DOI: 10.1016/j.apsb.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a recently discovered pathway for regulated cell death pathway. However, its efficacy is affected by limited iron content and intracellular ion homeostasis. Here, we designed a metal-organic framework (MOF)-based nanoplatform that incorporates calcium peroxide (CaO2) and oridonin (ORI). This platform can improve the tumor microenvironment and disrupt intracellular iron homeostasis, thereby enhancing ferroptosis therapy. Fused cell membranes (FM) were used to modify nanoparticles (ORI@CaO2@Fe-TCPP, NPs) to produce FM@ORI@CaO2@Fe-TCPP (FM@NPs). The encapsulated ORI inhibited the HSPB1/PCBP1/IREB2 and FSP1/COQ10 pathways simultaneously, working in tandem with Fe3+ to induce ferroptosis. Photodynamic therapy (PDT) guided by porphyrin (TCPP) significantly enhanced ferroptosis through excessive accumulation of reactive oxygen species (ROS). This self-amplifying strategy promoted robust ferroptosis, which could work synergistically with FM-mediated immunotherapy. In vivo experiments showed that FM@NPs inhibited 91.57% of melanoma cells within six days, a rate 5.6 times higher than chemotherapy alone. FM@NPs were biodegraded and directly eliminated in the urine or faeces without substantial toxicity. Thus, this study demonstrated that combining immunotherapy with efficient ferroptosis induction through nanotechnology is a feasible and promising strategy for melanoma treatment.
Collapse
Affiliation(s)
- Mengru Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tingting Fu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rongyue Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Panxiang Hu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiahui Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shilang Liao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuji Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yongqiang Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhai Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoxv Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
5
|
Torres-Herrero B, Armenia I, Ortiz C, de la Fuente JM, Betancor L, Grazú V. Opportunities for nanomaterials in enzyme therapy. J Control Release 2024; 372:619-647. [PMID: 38909702 DOI: 10.1016/j.jconrel.2024.06.035] [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: 03/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
In recent years, enzyme therapy strategies have rapidly evolved to catalyze essential biochemical reactions with therapeutic potential. These approaches hold particular promise in addressing rare genetic disorders, cancer treatment, neurodegenerative conditions, wound healing, inflammation management, and infectious disease control, among others. There are several primary reasons for the utilization of enzymes as therapeutics: their substrate specificity, their biological compatibility, and their ability to generate a high number of product molecules per enzyme unit. These features have encouraged their application in enzyme replacement therapy where the enzyme serves as the therapeutic agent to rectify abnormal metabolic and physiological processes, enzyme prodrug therapy where the enzyme initiates a clinical effect by activating prodrugs, and enzyme dynamic or starving therapy where the enzyme acts upon host substrate molecules. Currently, there are >20 commercialized products based on therapeutic enzymes, but approval rates are considerably lower than other biologicals. This has stimulated nanobiotechnology in the last years to develop nanoparticle-based solutions that integrate therapeutic enzymes. This approach aims to enhance stability, prevent rapid clearance, reduce immunogenicity, and even enable spatio-temporal activation of the therapeutic catalyst. This comprehensive review delves into emerging trends in the application of therapeutic enzymes, with a particular emphasis on the synergistic opportunities presented by incorporating enzymes into nanomaterials. Such integration holds the promise of enhancing existing therapies or even paving the way for innovative nanotherapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Torres-Herrero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Cecilia Ortiz
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Jesús Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lorena Betancor
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
6
|
Wang Q, Ge L, Guo J, Zhang H, Chen T, Lian F, Li L, Xu Y, Xu J, Chen N, Zhang Y, Ruan Z, Xiao J, Zhang H, Yang L. Acid Neutralization by Composite Lysine Nanoparticles for Spinal Cord Injury Recovery through Mitigating Mitochondrial Dysfunction. ACS Biomater Sci Eng 2024; 10:4480-4495. [PMID: 38885615 DOI: 10.1021/acsbiomaterials.4c00612] [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] [Indexed: 06/20/2024]
Abstract
After spinal cord injury (SCI), significant alterations in the tissue microenvironment lead to mitochondrial dysfunction, inducing apoptosis and inhibiting the remodeling of neural circuits, thereby impeding recovery. Although previous studies have demonstrated a marked decrease in pH at the injury site, creating an acidic microenvironment, the impact of improving this acidic microenvironment on SCI recovery has not been investigated. This study prepared a lysine@hollow mesoporous silica nanoparticle/gelatin methacrylate (GelMA) (L@H/G) composite hydrogel. The L@H/G composite hydrogel was demonstrated to release lysine and efficiently improve the acidic microenvironment slowly. Significantly, the composite hydrogel reduced cell apoptosis, promoted nerve regeneration, inhibited glial scar formation, and ultimately enhanced motor function recovery in mice with SCI. Mechanistically, the L@H/G hydrogel improved the mitochondrial tricarboxylic acid (TCA) cycle and fatty acid metabolism, restoring energy supply and facilitating mitochondrial function recovery. To the best of our knowledge, this is the first report confirming that improving the acidic microenvironment could promote SCI repair, providing a potential therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Qiuchen Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang 315302, China
| | - Lu Ge
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiali Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haijuan Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Tianling Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Feifei Lian
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lei Li
- Science and Teaching Affairs Section, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315040, China
| | - Yun Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jinyu Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Nuo Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhanwei Ruan
- Department of Emergency, The Third Affiliated Hospital, Wenzhou Medical University, No. 108 Wansong Road, Ruian, Wenzhou, Zhejiang 325200, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang 315302, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang 315302, China
- Central Laboratory, Wenzhou Medical University, Lishui People's Hospital, Lishui, Zhejiang 323000, China
| | - Liangliang Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang 315302, China
| |
Collapse
|
7
|
Li S, Zhao Y, Ma W, Wang D, Liu H, Wang W, Peng D, Yu CY, Wei H. A multivalent polyphenol-metal-nanoplatform for cascade amplified chemo-chemodynamic therapy. Acta Biomater 2024; 173:389-402. [PMID: 37967695 DOI: 10.1016/j.actbio.2023.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/17/2023]
Abstract
Chemodynamic therapy (CDT), as an emerging therapeutic strategy, kills cancer cells by converting intracellular hydrogen peroxide (H2O2) into cytotoxic oxidizing hydroxyl radicals (⋅OH). However, the therapeutic efficiency of CDT is compromised due to the insufficient endogenous H2O2 and metal catalysts in tumor cells. The use of multivalent polyphenols with multiple hydroxyl functions provides a facile yet robust means for efficient CDT augmentation. For this purpose, we reported herein the construction of polyphenol-metal nanoparticles (NPs) via a phenol-metal coordination strategy. The uniqueness of this study is the preparation of only one polymer construct with multivalency that can afford various supramolecular interactions for simultaneous "one-pot" loading of different therapeutic species, i.e., doxorubicin (DOX), glucose oxidases (GOD), and Fe3+ and further co-self-assembly into a stabilized nanomedicine for cascade amplified chemo-chemodynamic therapy. Specifically, the tumor intracellular acidic pH-triggered DOX release could serve for chemotherapy as well as enhance the intracellular H2O2 level. Together with the extra H2O2 and gluconic acid produced by the GOD-triggered glucose consumption, DOX@POAD-Fe@GOD NPs promoted Fe3+participation in the Fe-mediated Fenton reaction for cascade amplified chemo-chemodynamic therapy. Notably, this formulation displayed a greater anti-tumor effect with a tumor inhibition ratio 1.6-fold higher than that of free DOX in a BALB/c mice model bearing 4T1 tumors. Overall, the multivalent polyphenol-metal nanoplatform developed herein integrates chemotherapy, starvation therapy, and CDT for synergistic enhanced anticancer efficiency, which shows great potential for clinical translations. STATEMENT OF SIGNIFICANCE: Chemodynamic therapy (CDT) generally suffers from compromised therapeutic efficiency due to insufficient endogenous H2O2 and metal catalysts in tumor cells. To develop a facile yet robust strategy for efficient CDT augmentation, we reported herein construction of a multivalent polyphenol-metal nanoplatform, DOX@POAD-Fe@GOD nanoparticles (NPs) via a phenol-metal coordination strategy. This nanoplatform integrates multiple supramolecular dynamic interactions not only for simultaneously safe encapsulation of doxorubicin (DOX), Fe3+, and glucose oxidases (GOD), but also for cascade amplified chemo-chemodynamic therapy. Specifically, the intracellular acidic pH-triggered dissociation of DOX@POAD-Fe@GOD NPs promoted the release of Fe3+, DOX, and GOD for significantly increased ROS levels that can accelerate Fenton reactions for cascaded chemotherapy, starvation therapy, and CDT with amplified antitumor efficiency in vivo.
Collapse
Affiliation(s)
- Shuang Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Yuqi Zhao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Wei Ma
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Dun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Hongbing Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Wei Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Dongdong Peng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & Department of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China.
| |
Collapse
|
8
|
Gui J, Chen H, Liu J, Liu Y, Wu C, Zhu X, Wei M, Liu M, Zhang Y, Yao S. Consuming intracellular glucose and regulating the levels of O 2/H 2O 2 via the closed cascade catalysis system of Cu-CeO 2 nanozyme and glucose oxidase. J Colloid Interface Sci 2023; 651:191-199. [PMID: 37542894 DOI: 10.1016/j.jcis.2023.07.190] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/17/2023] [Accepted: 07/29/2023] [Indexed: 08/07/2023]
Abstract
Imbalances in the intracellular environment caused by high levels of glucose, H2O2, and hypoxia can greatly impact cancer development and treatment. However, there is limited research on regulating the levels of these species simultaneously in tumor cells. Here, a pH-responsive nanozyme-enzyme hybrid system was developed to regulate intracellular glucose, H2O2 and O2. The system, named DMSN@Cu-CeO2@GOx, consists of Cu-CeO2 nanoparticles and glucose oxidase (GOx) immobilized in dendritic mesoporous silica (DMSN) spheres. GOx efficiently consumes glucose in tumor cells, causing a drop in pH and producing a significant amount of H2O2. Cu-CeO2 then catalyzes the conversion of H2O2 to O2 due to its high catalase-like (CAT) activity in weakly acidic conditions. The process was monitored by fluorescence probes, and the mechanism was investigated through fluorescence spectroscopy and confocal laser scanning microscopy. The cascade catalytic system with excellent biocompatibility continuously consumes glucose and elevates the level of O2 in cells. This hybrid nanomaterial offers a means to regulate the glucose/H2O2/O2 levels in cells and may provide insights into starvation therapy by modulating reactive species within cells.
Collapse
Affiliation(s)
- Jialing Gui
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Haoyu Chen
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Jing Liu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Yani Liu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Cuiyan Wu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Xiaohua Zhu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Mingjie Wei
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, PR China.
| | - Meiling Liu
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China.
| | - Youyu Zhang
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| | - Shouzhuo Yao
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research (Ministry of Education, China), College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha 410081, PR China
| |
Collapse
|
9
|
Li J, Lou K, Dang Y, Tian H, Luo Q, Huang C, Liu R, Gong X, Wang S, Liu H, Wang P, Liu X. Precise tumor resection under the navigation of Tumor-Microenvironment pH-activated NIR-II fluorescence imaging via calcium Carbonate/Polydopamine Co-packed Nd-doped downshifting nanoprobes. MATERIALS & DESIGN 2023; 227:111703. [DOI: 10.1016/j.matdes.2023.111703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
|
10
|
de Melo Santana B, Pieretti JC, Gomes RN, Cerchiaro G, Seabra AB. Cytotoxicity towards Breast Cancer Cells of Pluronic F-127/Hyaluronic Acid Hydrogel Containing Nitric Oxide Donor and Silica Nanoparticles Loaded with Cisplatin. Pharmaceutics 2022; 14:pharmaceutics14122837. [PMID: 36559330 PMCID: PMC9780945 DOI: 10.3390/pharmaceutics14122837] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The incorporation of both nitric oxide (NO) donor (S-nitrosoglutathione, GSNO) and silica nanoparticles loaded with cisplatin (SiO2@CisPt NPs) into a polymeric matrix represents a suitable approach to creating a drug-delivery system with sustained and localized drug release against tumor cells. Herein, we report the synthesis, characterization, and cytotoxicity evaluation of Pluronic F-127/hyaluronic acid hydrogel containing GSNO and SiO2@CisPt NPs against breast cancer cells. SiO2@CisPt NPs were successfully synthesized, revealing a spherical morphology with an average size of 158 ± 20 nm. Both GSNO and SiO2@CisPt NPs were incorporated into the thermoresponsive Pluronic/hyaluronic hydrogel for sustained and localized release of both NO and cisplatin. The kinetics of NO release from a hydrogel matrix revealed spontaneous and sustained release of NO at the millimolar range for 24 h. The MTT assay showed concentration-dependent cytotoxicity of the hydrogel. The combination of GSNO and SiO2@CisPt incorporated into a polymeric matrix decreased the cell viability 20% more than the hydrogel containing only GSNO or SiO2@CisPt. At 200 µg/mL, this combination led to a critical cell viability of 30%, indicating a synergistic effect between GSNO and SiO2@CisPt NPs in the hydrogel matrix, and, therefore, highlighting the potential application of this drug-delivery system in the field of biomedicine.
Collapse
|
11
|
Shah S, Famta P, Bagasariya D, Charankumar K, Sikder A, Kashikar R, Kotha AK, Chougule MB, Khatri DK, Asthana A, Raghuvanshi RS, Singh SB, Srivastava S. Tuning Mesoporous Silica Nanoparticles in Novel Avenues of Cancer Therapy. Mol Pharm 2022; 19:4428-4452. [PMID: 36109099 DOI: 10.1021/acs.molpharmaceut.2c00374] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The global menace of cancer has led to an increased death toll in recent years. The constant evolution of cancer therapeutics with novel delivery systems has paved the way for translation of innovative therapeutics from bench to bedside. This review explains the significance of mesoporous silica nanoparticles (MSNs) as delivery vehicles with particular emphasis on cancer therapy, including novel opportunities for biomimetic therapeutics and vaccine delivery. Parameters governing MSN synthesis, therapeutic agent loading characteristics, along with tuning of MSN toward cancer cell specificity have been explained. The advent of MSN in nanotheranostics and its potential in forming nanocomposites for imaging purposes have been illustrated. Additionally, various hurdles encountered during the bench to bedside translation have been explained along with potential avenues to circumvent them. This also opens up new horizons in drug delivery, which could be useful to researchers in the years to come.
Collapse
Affiliation(s)
- Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Deepkumar Bagasariya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Kondasingh Charankumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Anupama Sikder
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rama Kashikar
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, Georgia 30341, United States
| | - Arun K Kotha
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, Georgia 30341, United States
| | - Mahavir Bhupal Chougule
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, Georgia 30341, United States
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, Raj Nagar, Ghaziabad 201002, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| |
Collapse
|
12
|
Nassar MY, El-Salhy HI, El-Shiwiny WH, Abdelaziz G, El-Shiekh R. Composite Nanoarchitectonics of Magnetic Silicon Dioxide-Modified Chitosan for Doxorubicin Delivery and In Vitro Cytotoxicity Assay. J Inorg Organomet Polym Mater 2022. [DOI: 10.1007/s10904-022-02498-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2022]
Abstract
AbstractDeveloping drug delivery carriers for highly selective, controlled, and sustained release of the anti-cancer drugs is one of the crucial issues in the cancer strive. We herein report the synthesis of Fe3O4 (M) and SiO2 (S) nanoparticles and their nanocomposites with chitosan (CS) for high loading efficiency and subsequent release potentiality of Doxorubicin (DOX) anticancer drug. The as-synthesized nanostructures were characterized using Fourier transform infrared (FT-IR), X-ray diffraction (XRD), field emission scanning electron microscopy (FE-SEM), high-resolution transmission electron microscopy (HR-TEM), and thermal analysis techniques. The average crystallite sizes of the as-prepared M, S, CS/M, CS/S, and CS/M/S nanostructures were found to be 5, 15, 70, 22, and 29 nm, respectively. The loading and cumulative release of Doxorubicin for the produced nanostructures were examined, and the results exhibited loading efficacy of 71%, 95%, 96%, 79%, 17%, and 42% for M, S, CS, CS/M, CS/S, and CS/M/S nanostructures, respectively. The Doxorubicin releasing results revealed a promising cumulative release percentages at pH 4.2 and pH 5 compared with those at pH 7.4. At pH 4.2, the cumulative release percentages for DOX-M, DOX-S, DOX-CS, DOX/M, and DOX/CS/M/S were 94%, 96%, 92%, 95%, and 98%, respectively. While the corresponding percentages at pH 5 were 97%, 90%, 46%, 43%, and 70%. The percentage for DOX-CS/S was 60% at pH 5, though. The in-vitro cytotoxicity of M-DOX, CS-DOX, and M/CS-DOX was explored against two human cancer cell lines (MCF-7 and Hep-G2) using SRB (Sulforhodamine B) assay. The DOX-loaded M/CS exhibited the highest cytotoxicity and its IC50 values were 2.65 and 2.25 μg/mL against Hep-G2 and MCF-7 cell lines, respectively, compared to the corresponding values of 5.1 and 4.5 μg/mL for free DOX. The results indicated that M/CS nanocomposite is a good candidate as drug delivery nano-carrier for the Doxorubicin anti-cancer drug.
Collapse
|
13
|
Cini N, Calisir F. Layer-by-layer self-assembled emerging systems for nanosized drug delivery. Nanomedicine (Lond) 2022; 17:1961-1980. [PMID: 36645082 DOI: 10.2217/nnm-2022-0254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
New frontiers in the development of stimuli-responsive surfaces that offer switchable properties according to the end-use application have added a new dimension to the design of drug-delivery systems (DDS). In this respect, layer-by-layer (LbL) self-assembled technologies have attracted interest in nano-DDS (NDDS) design due to the advantage of encapsulating different drug types either individually or in multiple formulations as an easy-to-apply and cost-effective strategy. LbL-based microcapsules and core-shell structures in the form of polyelectrolyte multilayers (PEMs) have been proposed as versatile vehicles for NDDS over the last quarter. This review aims to provide a global view of LbL-PEMs used as templates in NDDS for the last 5 years with an emphasis on emerging drug loading and release strategies.
Collapse
Affiliation(s)
- Nejla Cini
- Istanbul Technical University, Science and Letters Faculty, Chemistry Department, Maslak, Istanbul, 34469, Turkiye
| | - Ferah Calisir
- Istanbul Technical University, Science and Letters Faculty, Chemistry Department, Maslak, Istanbul, 34469, Turkiye
| |
Collapse
|
14
|
Stimuli-responsive polyelectrolyte multilayer films and microcapsules. Adv Colloid Interface Sci 2022; 310:102773. [DOI: 10.1016/j.cis.2022.102773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 08/20/2022] [Accepted: 09/05/2022] [Indexed: 12/28/2022]
|
15
|
Zhao L, Zhang Y, Yang Y, Yu C. Silica-based Nanoparticles for Enzyme Immobilization and Delivery. Chem Asian J 2022; 17:e202200573. [PMID: 35796745 DOI: 10.1002/asia.202200573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2022] [Indexed: 11/06/2022]
Abstract
Enzymes play an indispensable role in biosystems, catalyzing a variety of chemical and biochemical reactions with exceptionally high efficiency and selectivity. These features render them uniquely positioned in developing novel catalytic systems and therapeutics. However, their practical application is largely hindered by the vulnerability, low reusability and the inability to overcome the biological barriers of enzymes. Silica-based nanoparticles (SNPs) are a classic family of nanomaterials with tunable physicochemical properties, making them ideal candidates to address the intrinsic shortcomings of natural enzymes. SNPs not only improve the activity and durability of enzymes, but also provide precise spatiotemporal control over their intracellular as well as systemic biodistributions for boosting the catalytic outcome. Herein, the recent progress in SNPs for enzyme immobilization and delivery is summarized. The therapeutic applications, including cancer therapy and bacterial inhibition, are particularly highlighted. Our perspectives in this field, including current challenges and possible future research directions are also provided.
Collapse
Affiliation(s)
- Liang Zhao
- East China Normal University, School of Chemistry and Molecular Engineering, CHINA
| | - Yue Zhang
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, AUSTRALIA
| | - Yannan Yang
- The University of Queensland, Australian Institute for Bioengineering and Nanotechnology, AUSTRALIA
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Building 75,Cnr College Rd & Cooper Rd, 4067, Brisbane, AUSTRALIA
| |
Collapse
|
16
|
Weak Polyelectrolytes as Nanoarchitectonic Design Tools for Functional Materials: A Review of Recent Achievements. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103263. [PMID: 35630741 PMCID: PMC9145934 DOI: 10.3390/molecules27103263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/23/2022]
Abstract
The ionization degree, charge density, and conformation of weak polyelectrolytes can be adjusted through adjusting the pH and ionic strength stimuli. Such polymers thus offer a range of reversible interactions, including electrostatic complexation, H-bonding, and hydrophobic interactions, which position weak polyelectrolytes as key nano-units for the design of dynamic systems with precise structures, compositions, and responses to stimuli. The purpose of this review article is to discuss recent examples of nanoarchitectonic systems and applications that use weak polyelectrolytes as smart components. Surface platforms (electrodeposited films, brushes), multilayers (coatings and capsules), processed polyelectrolyte complexes (gels and membranes), and pharmaceutical vectors from both synthetic or natural-type weak polyelectrolytes are discussed. Finally, the increasing significance of block copolymers with weak polyion blocks is discussed with respect to the design of nanovectors by micellization and film/membrane nanopatterning via phase separation.
Collapse
|
17
|
Fan X, Luo Z, Chen Y, Yeo JCC, Li Z, Wu YL, He C. Oxygen self-supplied enzyme nanogels for tumor targeting with amplified synergistic starvation and photodynamic therapy. Acta Biomater 2022; 142:274-283. [PMID: 35114372 DOI: 10.1016/j.actbio.2022.01.056] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023]
Abstract
Tumor tissues need vast supply of nutrients and energy to sustain the rapid proliferation of cancer cells. Cutting off the glucose supply represents a promising cancer therapy approach. Herein, a tumor tissue-targeted enzyme nanogel (rGCP nanogel) with self-supply oxygen capability was developed. The enzyme nanogel synergistically enhanced starvation therapy and photodynamic therapy (PDT) to mitigate the rapid proliferation of cancer cells. The rGCP nanogel was fabricated by copolymerizing two monomers, porphyrin and cancer cells-targeted, Arg-Gly-Asp (RGD), onto the glucose oxidase (GOX) and catalase (CAT) surfaces. The cascade reaction within the rGCP nanogel could efficiently consume intracellular glucose catalyzed by GOX. Concurrently, CAT safely decomposed the produced H2O2 with systemic toxicity to promote oxygen generation and achieved low toxicity starvation therapy. The produced oxygen subsequently facilitated the glucose oxidation reaction and significantly enhanced the generation of cytotoxic singlet oxygen (1O2) in the presence of 660 nm light irradiation. Combining starvation therapy and PDT, the designed enzyme nanogel system presented an amplified synergic cancer therapy effect. This approach potentially paved a new way to fabricate a combinatorial therapy approach by employing cascaded catalytic nanomedicines with good tumor selectivity and efficient anti-cancer effect. STATEMENT OF SIGNIFICANCE: The performance of starvation and photodynamic therapy (PDT) is usually suppressed by intrinsic tumorous hypoxia. Herein, an oxygen self-supplied and tumor tissue-targeted enzyme nanogel was created by copolymerization of two monomers, porphyrin and cancer cell-targeted Arg-Gly-Asp (RGD), onto the surface of glucose oxidase (GOX) and catalase (CAT), which synergistically enhanced starvation therapy and PDT. Moreover, the enzyme nanogels possessed high stability and could be synthesized straightforwardly. This anti-cancer system provides an approach for constructing a combinatorial therapy approach by employing cascaded catalytic nanomedicine with good tumor selectivity and therapeutic efficacy.
Collapse
Affiliation(s)
- Xiaotong Fan
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Ying Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jayven Chee Chuan Yeo
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore
| | - Zibiao Li
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore.
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.
| | - Chaobin He
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore; Institute of Materials Research and Engineering, Agency for Science, Technology, and Research (A*STAR), 2 Fusionopolis Way, Innovis, Singapore 138634, Singapore.
| |
Collapse
|
18
|
Chen XX, Hou MJ, Wang WX, Tan M, Tan ZK, Mao GJ, Yang B, Li Y, Li CY. ATP-responsive near-infrared fluorescent nanoparticles for synergistic chemotherapy and starvation therapy. NANOSCALE 2022; 14:3808-3817. [PMID: 35191447 DOI: 10.1039/d1nr07233a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cancer is a major public health problem worldwide, and traditional chemotherapy or a single therapeutic strategy often fails to achieve expected results in cancer treatment. Thus, the development of a method to realize controlled drug delivery and synergistic therapy is required. Herein, MOF-based nanoparticles named RhI-DOX-GOD@ZIF-90 are synthesized using RhI (a near-infrared fluorescent dye), DOX (an anti-cancer drug) and GOD (glucose oxidase). RhI and DOX are encapsulated inside the ZIF-90 framework and GOD is loaded on the surface of ZIF-90. Owing to the fact that the ATP level in cancer cells is abnormally higher than that in normal cells, RhI-DOX-GOD@ZIF-90 nanoparticles are destructed only in cancer cells. RhI is released to give outstanding NIR emission and realize controlled drug delivery. DOX is released and cancer cells are killed by chemotherapy. Also, GOD is released to consume glucose and achieve the purpose of starving the cancer cells. By making full use of the advantages of near-infrared emission, RhI-DOX-GOD@ZIF-90 nanoparticles can be used to image ATP in tumor-bearing mice. At the same time, DOX and GOD can be released accurately at tumor sites of mice and excellent anti-tumor efficiency by synergistic chemotherapy and starvation therapy is achieved.
Collapse
Affiliation(s)
- Xi-Xi Chen
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| | - Mei-Jia Hou
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| | - Wen-Xin Wang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| | - Min Tan
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| | - Zhi-Ke Tan
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| | - Guo-Jiang Mao
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, PR China
| | - Bin Yang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| | - Yongfei Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
- College of Chemical Engineering, Xiangtan University, Xiangtan, 411105, P. R. China
| | - Chun-Yan Li
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan, 411105, PR China.
| |
Collapse
|
19
|
Characteristics of glucose oxidase immobilized on carbon-encapsulated iron nanoparticles decorated with polyethyleneimine. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
20
|
Tong Z, Wang T, Cai Y, Sha JQ, Peng T. Oxygen-Powered Flower-like FeMo 6@CeO 2 Self-cascade Nanozyme: Turn-on Enhancement Fluorescence Sensor. J Mater Chem B 2022; 10:6425-6432. [DOI: 10.1039/d2tb01466a] [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
Enzyme cascade reactions in organisms have sparked tremendous interest for their coupled catalysis-facilitated efficient biochemical reactions. However, multi-enzyme cascade nanozymes remain largely unpracticed. In the work, flower-like porous ceria-based integrated...
Collapse
|
21
|
Qin X, Qin L, He J, Wang Q, Li Y, Niu D. A confined crosslinking strategy towards an intelligent organosilica-micellar hybrid drug delivery system. Biomater Sci 2021; 10:524-535. [PMID: 34904973 DOI: 10.1039/d1bm01485d] [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
An ideal drug delivery system must have a high level of stability to ensure effective circulation and passive aggregation, good retention performance, and dynamic delivery and treatment monitoring. Thus, the development of a smart drug delivery carrier with both precise drug release and real-time detection remains a challenge. Herein, we propose a confined crosslink protocol to prepare an intelligent hybrid delivery system for auto-fluorescent monitoring, protonation-induced retention and precise drug release. The construction of this system involves the hydrolysis and condensation of (3-aminopropyl)triethoxysilane (APTES) silanes inside the Pluronic polymer micelles and thereafter a confined Schiff base crosslinking between glutaraldehyde (GA) and residual silane amino groups. The size of the intelligent docetaxel (DTX)-loaded nanosystem changes from ∼25 nm in blood circulation or normal tissues (pH ∼ 7.4) to ∼250 nm in slightly acidic environments (pH ∼ 6.5-7.0) owing to intra-molecular hydrogen bond-induced aggregation and imine cleavage-induced disintegration in the endosome (pH ∼ 5.0-6.2) along with auto-fluorescent monitoring contributing to the high-efficiency chemotherapy. This work provides a new method to construct smart, acid-responsive and fluorescent-guided drug-delivery carrier systems for efficient and safe tumor chemotherapy.
Collapse
Affiliation(s)
- Xing Qin
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, Frontier Science Center of the Materials Biology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Limei Qin
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, Frontier Science Center of the Materials Biology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Jianping He
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, Frontier Science Center of the Materials Biology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Qinghua Wang
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, Frontier Science Center of the Materials Biology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| | - Yongsheng Li
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, Frontier Science Center of the Materials Biology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China. .,Key Laboratory for Green Processing of Chemical Engineering of Xinjiang Bingtuan, School of Chemistry and Chemical Engineering, Shihezi University, Shihezi 832003, China
| | - Dechao Niu
- Low Dimensional Materials Chemistry Laboratory, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, Frontier Science Center of the Materials Biology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, 200237, P. R. China.
| |
Collapse
|
22
|
Liu M, Wu H, Wang S, Hu J, Sun B. Glutathione-triggered nanoplatform for chemodynamic/metal-ion therapy. J Mater Chem B 2021; 9:9413-9422. [PMID: 34746940 DOI: 10.1039/d1tb01330k] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The integration of metal-ion therapy and hydroxyl radical (˙OH)-mediated chemodynamic therapy (CDT) holds great potential for anticancer treatment with high specificity and efficiency. Herein, Ag nanoparticles (Ag NPs) were enveloped with Cu2+-based metal-organic frameworks (MOFs) and further decorated with hyaluronic acid (HA) to construct a glutathione (GSH)-activated nanoplatform (Ag@HKU-HA) for specific chemodynamic/metal-ion therapy. The obtained nanoplatform could avoid the premature leakage of Ag in circulation, but realize the release of Ag at the tumor site owing to the degradation of external MOFs triggered by Cu2+-reduced glutathione. The generated Cu+ could catalyze endogenous H2O2 to the highly toxic ˙OH by a Fenton-like reaction. Meanwhile, Ag NPs were oxidized to toxic Ag ions in the tumor environment. As expect, the effect of CDT combined with metal-ion therapy exhibited an excellent inhibition of tumor cells growth. Therefore, this nanoplatform may provide a promising strategy for on-demand site-specific cancer combination treatment.
Collapse
Affiliation(s)
- Min Liu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Hongshuai Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Senlin Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Jinzhong Hu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| |
Collapse
|
23
|
Song S, Peng J, Wu Y, Li C, Shen D, Yang G, Liu J, Gong P, Liu Z. Biomimetic synthesis of a novel O 2-regeneration nanosystem for enhanced starvation/chemo-therapy. NANOTECHNOLOGY 2021; 33:025102. [PMID: 34544066 DOI: 10.1088/1361-6528/ac2843] [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: 06/17/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Glucose oxidase-mediated starvation therapy that effectively cuts off energy supply holds great promise in cancer treatment. However, high glutathione (GSH) contents and anoxic conditions severely reduce therapy efficiency and cannot fully kill cancer cells. Herein, to resolve the above problem, this study constructed a biomimetic nanosystem based on nanreproo-MnO2with porous craspedia globose-like structure and high specific surface area, and it was further modified with dopamine and folic acid to guarantee good biocompatibility and selectivity toward cancer cells. This nanosystem responsively degraded and reacted with GSH and acid to regenerate O2, which significantly increased intracellular O2levels, accelerated glucose consumption, and improved starvation therapy efficiency. Moreover, anticancer drug of camptothecin was further loaded, and notably enhanced cancer growth inhibition was obtained at very low drug concentrations. Most importantly, this novel therapy could unprecedentedly inhibit cancer cell migration to a very low ratio of 19%, and detailed cell apoptosis analyses revealed late stage apoptosis contributed most to the good therapeutic effect. This work reported a new train of thought to improve starvation therapy in biomedicine, and provided a new strategy to design targeted nanocarrier to delivery mixed drugs to overcome the restriction of starvation therapy and develop new therapy patterns.
Collapse
Affiliation(s)
- Shaohua Song
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Jingyi Peng
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Yuting Wu
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Cheng Li
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Duyi Shen
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Ge Yang
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Jinfeng Liu
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| | - Peiwei Gong
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
- State Key Laboratory of Solidification Processing, Center of Advanced Lubrication and Seal Materials, Northwestern Polytechnical University, Xi'an, 710072, People's Republic of China
| | - Zhe Liu
- College of Life Sciences, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, Shandong, People's Republic of China
| |
Collapse
|
24
|
Hu L, Xiong C, Wei G, Yu Y, Li S, Xiong X, Zou JJ, Tian J. Stimuli-responsive charge-reversal MOF@polymer hybrid nanocomposites for enhanced co-delivery of chemotherapeutics towards combination therapy of multidrug-resistant cancer. J Colloid Interface Sci 2021; 608:1882-1893. [PMID: 34749141 DOI: 10.1016/j.jcis.2021.10.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022]
Abstract
Combination chemotherapy is a promising strategy for cancer treatment in clinics especially when multidrug-resistant cancer is emerging. One significant challenge remains in achieving sufficient multi-drug delivery into tumor cells to maximize the synergetic therapeutic effect, as it is hard to concentrate drugs in drug-resistant cancer. Therefore herein, metal-organic framework (MOF)-based polymer-coated hybrid nanoparticles (NPs) were devised and constructed for the co-delivery of doxorubicin and cisplatin to enhance combination therapy of multidrug-resistant cancer. The MOF@polymer nanocarrier combined the merits of high multi-drug loading capacity, physiological stability, and tumor microenvironment pH-responsiveness, facilitating simultaneous delivery of drugs into cancer cells and making the most of synergistic antitumor effect. Remarkably, this hybrid nanocarrier maintains a negative surface charge during circulation to guarantee a stable and prolonged process in vivo, and then exposes inner positive MOF after degradation of the outer polymer in the acidic tumor microenvironment to promote multi-drug release, cellular internalization, nuclear localization, and tumor penetration. In vitro and in vivo studies with drug-resistant MCF-7/ADR cancer suggested that the nanocarrier could achieve increased accumulation of drugs in solid tumors, remarkable tumor elimination results as well as minimized side effects, indicating an improved efficacy and safety of combination chemotherapy. MOF@polymer hybrid nanocarriers provide new insights into the development of stimuli-responsive co-delivery systems of multiple drugs.
Collapse
Affiliation(s)
- Liefeng Hu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Chuxiao Xiong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Gaohui Wei
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Yunhao Yu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Sihui Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430071, PR China.
| | - Jun-Jie Zou
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China
| | - Jian Tian
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE), Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
25
|
Wu CY, Hsu YH, Chen Y, Yang LC, Tseng SC, Chen WR, Huang CC, Wan D. Robust O 2 Supplementation from a Trimetallic Nanozyme-Based Self-Sufficient Complementary System Synergistically Enhances the Starvation/Photothermal Therapy against Hypoxic Tumors. ACS APPLIED MATERIALS & INTERFACES 2021; 13:38090-38104. [PMID: 34342219 DOI: 10.1021/acsami.1c10656] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Much effort has been focused on novel nanomedicine for cancer therapy. However, tumor hypoxia limits the efficacy of various cancer therapeutics. Herein, we constructed a self-sufficient hybrid enzyme-based silk fibroin hydrogel system, consisting of Pt-decorated hollow Ag-Au trimetallic nanocages (HGN@Pt) and glucose oxidase (GOx), to supply O2 continuously and consume glucose concurrently and, thereby, synergistically enhance the anti-cancer efficacy of a combined starvation and photothermal therapy operating in a hypoxic tumor microenvironment. Thanks to the cooperative effects of the active surface atoms (resulting from the island-like features of the Pt coating), the intrinsically hollow structure, and the strain effect induced by the trimetallic composition, HGN@Pt displayed efficient catalase-like activity. The enhancement in the generation of O2 through the decomposition of H2O2 mediated by the as-designed nanozyme was greater than 400% when compared with that of hollow Ag-Pt bimetallic nanospheres or tiny Pt nanoparticles. Moreover, in the presence of HGN@Pt, significant amounts of O2 could be generated within a few minutes, even in an acidic buffer solution (pH 5.8-6.5) containing a low concentration of H2O2 (100-500 μM). Because HGN@Pt exhibited a strong surface plasmon resonance peak in the near-infrared wavelength range, it could be used as a photothermal agent for hyperthermia therapy. Furthermore, GOx was released gradually from the SF hydrogel into the tumor microenvironment to mediate the depletion of glucose, leading to glucose starvation-induced cancer cell death. Finally, the O2 supplied by HGN@Pt overcame the hypoxia of the microenvironment and, thereby, promoted the starvation therapeutic effect of the GOx-mediated glucose consumption. Meanwhile, the GOx-produced H2O2 from the oxidation of glucose could be used to regenerate O2 and, thereby, construct a complementary circulatory system. Accordingly, this study presents a self-sufficient hybrid enzyme-based system that synergistically alleviates tumor hypoxia and induces an anti-cancer effect when combined with irradiation of light from a near-infrared laser.
Collapse
Affiliation(s)
- Cheng-Yun Wu
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Yu-Hsuan Hsu
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Ling-Chu Yang
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Shao-Chin Tseng
- Experimental Facility Division, National Synchrotron Radiation Research Center, 101 Hsin-Ann Road, Hsinchu Science Park, Hsinchu 30076, Taiwan
| | - Wan-Ru Chen
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| | - Dehui Wan
- Institute of Biomedical Engineering, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan
| |
Collapse
|
26
|
Tehrani Fateh S, Moradi L, Kohan E, Hamblin MR, Shiralizadeh Dezfuli A. Comprehensive review on ultrasound-responsive theranostic nanomaterials: mechanisms, structures and medical applications. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:808-862. [PMID: 34476167 PMCID: PMC8372309 DOI: 10.3762/bjnano.12.64] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/15/2021] [Indexed: 05/03/2023]
Abstract
The field of theranostics has been rapidly growing in recent years and nanotechnology has played a major role in this growth. Nanomaterials can be constructed to respond to a variety of different stimuli which can be internal (enzyme activity, redox potential, pH changes, temperature changes) or external (light, heat, magnetic fields, ultrasound). Theranostic nanomaterials can respond by producing an imaging signal and/or a therapeutic effect, which frequently involves cell death. Since ultrasound (US) is already well established as a clinical imaging modality, it is attractive to combine it with rationally designed nanoparticles for theranostics. The mechanisms of US interactions include cavitation microbubbles (MBs), acoustic droplet vaporization, acoustic radiation force, localized thermal effects, reactive oxygen species generation, sonoluminescence, and sonoporation. These effects can result in the release of encapsulated drugs or genes at the site of interest as well as cell death and considerable image enhancement. The present review discusses US-responsive theranostic nanomaterials under the following categories: MBs, micelles, liposomes (conventional and echogenic), niosomes, nanoemulsions, polymeric nanoparticles, chitosan nanocapsules, dendrimers, hydrogels, nanogels, gold nanoparticles, titania nanostructures, carbon nanostructures, mesoporous silica nanoparticles, fuel-free nano/micromotors.
Collapse
Affiliation(s)
- Sepand Tehrani Fateh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Lida Moradi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Kohan
- Department of Science, University of Kurdistan, Kurdistan, Sanandaj, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | | |
Collapse
|
27
|
Zhao D, Yang N, Xu L, Du J, Yang Y, Wang D. Hollow structures as drug carriers: Recognition, response, and release. NANO RESEARCH 2021; 15:739-757. [PMID: 34254012 PMCID: PMC8262765 DOI: 10.1007/s12274-021-3595-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 05/19/2023]
Abstract
Hollow structures have demonstrated great potential in drug delivery owing to their privileged structure, such as high surface-to-volume ratio, low density, large cavities, and hierarchical pores. In this review, we provide a comprehensive overview of hollow structured materials applied in targeting recognition, smart response, and drug release, and we have addressed the possible chemical factors and reactions in these three processes. The advantages of hollow nanostructures are summarized as follows: hollow cavity contributes to large loading capacity; a tailored structure helps controllable drug release; variable compounds adapt to flexible application; surface modification facilitates smart responsive release. Especially, because the multiple physical barriers and chemical interactions can be induced by multishells, hollow multishelled structure is considered as a promising material with unique loading and releasing properties. Finally, we conclude this review with some perspectives on the future research and development of the hollow structures as drug carriers.
Collapse
Affiliation(s)
- Decai Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Nailiang Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Lekai Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
- Green Catalysis Center, and College of Chemistry, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001 China
| | - Jiang Du
- Green Catalysis Center, and College of Chemistry, School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001 China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433 China
- School of Materials Science and Engineering, Tongji University, Shanghai, 201804 China
| | - Dan Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
28
|
Kashapov R, Ibragimova A, Pavlov R, Gabdrakhmanov D, Kashapova N, Burilova E, Zakharova L, Sinyashin O. Nanocarriers for Biomedicine: From Lipid Formulations to Inorganic and Hybrid Nanoparticles. Int J Mol Sci 2021; 22:7055. [PMID: 34209023 PMCID: PMC8269010 DOI: 10.3390/ijms22137055] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Encapsulation of cargoes in nanocontainers is widely used in different fields to solve the problems of their solubility, homogeneity, stability, protection from unwanted chemical and biological destructive effects, and functional activity improvement. This approach is of special importance in biomedicine, since this makes it possible to reduce the limitations of drug delivery related to the toxicity and side effects of therapeutics, their low bioavailability and biocompatibility. This review highlights current progress in the use of lipid systems to deliver active substances to the human body. Various lipid compositions modified with amphiphilic open-chain and macrocyclic compounds, peptide molecules and alternative target ligands are discussed. Liposome modification also evolves by creating new hybrid structures consisting of organic and inorganic parts. Such nanohybrid platforms include cerasomes, which are considered as alternative nanocarriers allowing to reduce inherent limitations of lipid nanoparticles. Compositions based on mesoporous silica are beginning to acquire no less relevance due to their unique features, such as advanced porous properties, well-proven drug delivery efficiency and their versatility for creating highly efficient nanomaterials. The types of silica nanoparticles, their efficacy in biomedical applications and hybrid inorganic-polymer platforms are the subject of discussion in this review, with current challenges emphasized.
Collapse
Affiliation(s)
- Ruslan Kashapov
- A.E. Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Street 8, 420088 Kazan, Russia; (A.I.); (R.P.); (D.G.); (N.K.); (E.B.); (L.Z.); (O.S.)
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lu R, Zhou L, Liu Q, Wang S, Yang C, Hai L, Guo L, Wu Y. Skillfully collaborating chemosynthesis with GOx-enabled tumor survival microenvironment deteriorating strategy for amplified chemotherapy and enhanced tumor ablation. Biomater Sci 2021; 9:1855-1871. [PMID: 33464244 DOI: 10.1039/d0bm01950j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The satisfactory efficient tumor treatment and complete tumor ablation using a mono-therapeutic approach are limited owing to the tumor complexity, diversity, heterogeneity and the multiple pathways involved in tumor pathogenesis. Herein, novel, intelligent and tumor microenvironment (TME)-responsive biotin/R8 peptide co-modified nanocarriers (BRNC) loading paclitaxel (PTX)/glucose oxidase (GOx) were constructed. GOx could catalyze the oxidation of intracellular glucose to gluconic acid and poisonous H2O2 to cause the deterioration of the tumor survival microenvironment, simultaneously achieving starvation and oxidation therapy. The acidic amplification during the GOx-mediated oxidation progress could in turn accelerate the cleavage of the acid-degradable hydrazone bond, promoting the deep penetration of nanocarriers into tumors. Even better, the aforementioned two aspects further increased the tumors' sensitivity to chemotherapeutic agents. Both in vitro and in vivo investigations indicated that the co-administration of GOx-BRNC and PTX-BRNC can remarkably improve the therapeutic efficacy and reduce side effects through the high-specific tumor targeting multimodal synergistic starvation/oxidation/chemotherapy, which would be a promising strategy for the next generation cancer therapy.
Collapse
Affiliation(s)
- Runxin Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Lin Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Qijun Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Siqi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Chunyan Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Li Hai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Li Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| | - Yong Wu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China.
| |
Collapse
|
30
|
Niu S, Zhang X, Williams GR, Wu J, Gao F, Fu Z, Chen X, Lu S, Zhu LM. Hollow Mesoporous Silica Nanoparticles Gated by Chitosan-Copper Sulfide Composites as Theranostic Agents for the Treatment of Breast Cancer. Acta Biomater 2021; 126:408-420. [PMID: 33731303 DOI: 10.1016/j.actbio.2021.03.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/19/2022]
Abstract
The combination of chemotherapy and photothermal therapy (PTT) into a single formulation has attracted increasing attention as a strategy for enhancing cancer treatment. Here, hollow mesoporous silica nanoparticles (HMSNs) were used as a base carrier material, loaded with the anti-cancer drug doxorubicin (DOX), and surface functionalized with chitosan (CS) and copper sulfide (CuS) nanodots to give HMSNs-CS-DOX@CuS. In this formulation, the CuS dots act as gatekeepers to seal the surface pores of the HMSNs, preventing a burst release of DOX into the systemic circulation. S-S bonds connect the CuS dots to the HMSNs; these are selectively cleaved under the reducing microenvironment of the tumor, permitting targeted drug release. This, coupled with the PTT properties of CuS, results in a potent chemo/PTT platform. The HMSNs-CS-DOX@CuS nanoparticles have a uniform size (150 ± 13 nm), potent photothermal properties (η = 36.4 %), and tumor-targeted and near infrared (NIR) laser irradiation-triggered DOX release. In vitro and in vivo experimental results confirmed that the material has good biocompatibility, but is effectively taken up by cancer cells. Moreover, the CuS nanodots permit simultaneous thermal/photoacoustic dual-modality imaging. Treatment with HMSNs-CS-DOX@CuS and NIR irradiation caused extensive apoptosis in cancer cells both in vitro and in vivo, and could dramatically extend the lifetimes of animals in a murine breast cancer model. The system developed in this work therefore merits further investigation as a potential nanotheranostic platform for cancer treatment. STATEMENT OF SIGNIFICANCE: Conventional cancer chemotherapy is accompanied by unavoidable off-target toxicity. Combination therapies, which can ameliorate these issues, are attracting significant attention. Here, the anticancer drug doxorubicin (DOX) was encapsulated in the central cavity of chitosan (CS)-modified hollow mesoporous silica nanoparticles (HMSNs). The prepared system can target drug release to the tumor microenvironment. When exposed to near infrared laser (NIR) irradiation, CuS nanodots located at the surface pores of the HMSNs generate energy, accelerating drug release. In addition, a systematic in vitro and in vivo evaluation confirmed the HMSNs-CS-DOX@CuS platform to give highly effective synergistic chemotherapeutic-photothermal therapy and have effective thermal/photoacoustic dual-imaging properties. This work may open up a new avenue for NIR-enhanced synergistic therapy with simultaneous thermal/photoacoustic dual imaging.
Collapse
Affiliation(s)
- Shiwei Niu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming 650500, P.R. China
| | - Xuejing Zhang
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | - Jianrong Wu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Feng Gao
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, P.R. China
| | - Zi Fu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Xia Chen
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China
| | - Sheng Lu
- Yunnan Key Laboratory of Digital Orthopaedics, Department of Orthopaedics, the First People's Hospital of Yunnan Province, Kunming 650500, P.R. China.
| | - Li-Min Zhu
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P.R. China.
| |
Collapse
|
31
|
Zhang X, An L, Tian Q, Lin J, Yang S. Tumor microenvironment-activated NIR-II reagents for tumor imaging and therapy. J Mater Chem B 2021; 8:4738-4747. [PMID: 32124909 DOI: 10.1039/d0tb00030b] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Second near-infrared window (NIR-II, 1000-1700 nm) absorption and fluorescent agents have attracted great attention because they can overcome the penetration limitation of the first near-infrared window (NIR-I, 750-1000 nm). However, these always "on" agents face the severe problem of being susceptible to retention and phagocytosis by the reticuloendothelial system after intravenous administration, which results in signal interference during diagnosis and side effects during treatment. Accordingly, tumor microenvironment-responsive smart agents (smart NIR-II agents), whose imaging and therapeutic functions can only be triggered in tumors, can overcome this limitation. Thus, NIR-II smart agents, which exhibit a combined response to the tumor microenvironment and NIR-II, make full use of the advantages of both triggers and improve the precision diagnosis and effective treatment of cancer. This review summarizes the recent advances in tumor microenvironment-activated NIR-II agents for tumor diagnosis and treatment, including smart NIR-II fluorescence imaging, photoacoustic imaging, photothermal therapy and photodynamic therapy. Finally, the challenges and perspectives of NIR-II smart agents for tumor diagnosis and treatment are proposed.
Collapse
Affiliation(s)
- Xue Zhang
- The Key Laboratory of Resource Chemistry of the Ministry of Education, the Shanghai Key Laboratory of Rare Earth Functional Materials, and the Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai, 200234, China.
| | - Lu An
- The Key Laboratory of Resource Chemistry of the Ministry of Education, the Shanghai Key Laboratory of Rare Earth Functional Materials, and the Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai, 200234, China.
| | - Qiwei Tian
- The Key Laboratory of Resource Chemistry of the Ministry of Education, the Shanghai Key Laboratory of Rare Earth Functional Materials, and the Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai, 200234, China.
| | - Jiaomin Lin
- The Key Laboratory of Resource Chemistry of the Ministry of Education, the Shanghai Key Laboratory of Rare Earth Functional Materials, and the Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai, 200234, China.
| | - Shiping Yang
- The Key Laboratory of Resource Chemistry of the Ministry of Education, the Shanghai Key Laboratory of Rare Earth Functional Materials, and the Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai, 200234, China.
| |
Collapse
|
32
|
Chibh S, Katoch V, Kour A, Khanam F, Yadav AS, Singh M, Kundu GC, Prakash B, Panda JJ. Continuous flow fabrication of Fmoc-cysteine based nanobowl infused core-shell like microstructures for pH switchable on-demand anti-cancer drug delivery. Biomater Sci 2021; 9:942-959. [PMID: 33559658 DOI: 10.1039/d0bm01386b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Asymmetric nanostructures such as nanobowls (NBs) can exhibit superior drug delivery performances owing to their concave structure and interior asymmetric cavities. Here, we present a facile one-step method for the fabrication of NB like structures from a mere single amino acid mimetic, N-(9-fluorenylmethoxycarbonyl)-S-triphenylmethyl-l-cysteine following continuous-flow microfluidics enabled supramolecular self-assembly. Following fabrication, NBs were further infused into a vesicular shell consisting of the amino acid N-(tert-butoxycarbonyl)-S-triphenylmethyl-l-cysteine, carrying dual acid labile groups, the triphenylmethyl and the tert-butyloxycarbonyl groups. The NB infused core-shell like microstructures formed after the shell coating will now be addressed as NB-shells. Presence of pH-responsive shells bestowed the core-shell NB like structures with the ability to actively tune their surface pore opening and closing in response to environmental pH switch. To illustrate the potential use of the NB-shells in the field of anticancer drug delivery, the particles were loaded with doxorubicin (Dox) with an encapsulation efficiency of 42% and Dox loaded NB-shells exhibited enhanced efficacy in C6 glioma cells. Additionally, when tested in an animal model of glioblastoma, the nanoformulations demonstrated significantly higher retardation of tumour growth as compared to free Dox. Thus, this work strives to provide a new research area in the development of well turned-out and neatly fabricated pH switchable on/off anti-cancer drug delivery systems with significant translational potential.
Collapse
Affiliation(s)
- Sonika Chibh
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| | - Vibhav Katoch
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| | - Avneet Kour
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| | - Farheen Khanam
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| | - Amit Singh Yadav
- NCCS Complex, University of Pune Campus, University Road, Ganeshkhind, Pune, Maharashtra 411007, India and School of Biotechnology and Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Institute of Eminence, Bhubaneswar, 751024, India
| | - Manish Singh
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| | - Gopal C Kundu
- NCCS Complex, University of Pune Campus, University Road, Ganeshkhind, Pune, Maharashtra 411007, India and School of Biotechnology and Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to be University, Institute of Eminence, Bhubaneswar, 751024, India
| | - Bhanu Prakash
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Phase-10, Sector 64, Mohali, Punjab 160062, India.
| |
Collapse
|
33
|
Wang S, Wu H, Sun K, Hu J, Chen F, Liu W, Chen J, Sun B, Hossain AMS. A novel pH-responsive Fe-MOF system for enhanced cancer treatment mediated by the Fenton reaction. NEW J CHEM 2021. [DOI: 10.1039/d0nj05105e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A novel pH-responsive Fe-MOF system for enhancing cancer treatment mediated by a Fenton reaction.
Collapse
Affiliation(s)
- Senlin Wang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Hongshuai Wu
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Kai Sun
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Jinzhong Hu
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Fanghui Chen
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Wen Liu
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Jian Chen
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- P. R. China
| | | |
Collapse
|
34
|
Bueno V, Ghoshal S. Self-Assembled Surfactant-Templated Synthesis of Porous Hollow Silica Nanoparticles: Mechanism of Formation and Feasibility of Post-Synthesis Nanoencapsulation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:14633-14643. [PMID: 33226821 DOI: 10.1021/acs.langmuir.0c02501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
SiO2 is bioinert and highly functionalizable, thus making it a very attractive material for nanotechnology applications such as drug delivery and nanoencapsulation of pesticides. Herein, we synthesized porous hollow SiO2 nanoparticles (PHSNs) by using cetyltrimethylammonium bromide (CTAB) and Pluronic P123 as the structure-directing agents. The porosity and hollowness of the SiO2 structure allow for the protective and high-density loading of molecules of interest inside the nanoshell. We demonstrate here that loading can be achieved post-synthesis through the pores of the PHSNs. The PHSNs are monodisperse with a mean diameter of 258 nm and a specific surface area of 287 m2 g-1. The mechanism of formation of the PHSNs was investigated using 1-D and 2-D solid-state nuclear magnetic resonance (SS-NMR) and Fourier-transform infrared spectroscopy (FTIR). The data suggest that CTAB and Pluronic P123 interact, forming a hydrophobic spherical hollow cage that serves as a template for the porous hollow structure. After synthesis, the surfactants were removed by calcination at 550 °C and the PHSNs were added to an Fe3+ solution followed by addition of the reductant NaBH4 to the suspension, which led to the formation of Fe(0) NPs both on the PHSNs and inside the hollow shell, as confirmed by transmission electron microscopy imaging. The imaging of the formation of Fe(0) NPs inside the hollow shell provides direct evidence of transport of solute molecules across the shell and their reactions within the PHSNs, making it a versatile nanocarrier and nanoreactor.
Collapse
Affiliation(s)
- Vinicius Bueno
- Department of Civil Engineering, McGill University, Montreal, Quebec H3A 0C3, Canada
| | - Subhasis Ghoshal
- Department of Civil Engineering, McGill University, Montreal, Quebec H3A 0C3, Canada
| |
Collapse
|
35
|
Zhang G, Huang L, Wu J, Liu Y, Zhang Z, Guan Q. Doxorubicin-loaded folate-mediated pH-responsive micelle based on Bletilla striata polysaccharide: Release mechanism, cellular uptake mechanism, distribution, pharmacokinetics, and antitumor effects. Int J Biol Macromol 2020; 164:566-577. [DOI: 10.1016/j.ijbiomac.2020.07.123] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 12/24/2022]
|
36
|
Gao Y, Gao D, Shen J, Wang Q. A Review of Mesoporous Silica Nanoparticle Delivery Systems in Chemo-Based Combination Cancer Therapies. Front Chem 2020; 8:598722. [PMID: 33330389 PMCID: PMC7732422 DOI: 10.3389/fchem.2020.598722] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/20/2020] [Indexed: 01/11/2023] Open
Abstract
Chemotherapy is an important anti-tumor treatment in clinic to date, however, the effectiveness of traditional chemotherapy is limited by its poor selectivity, high systemic toxicity, and multidrug resistance. In recent years, mesoporous silica nanoparticles (MSNs) have become exciting drug delivery systems (DDS) due to their unique advantages, such as easy large-scale production, adjustable uniform pore size, large surface area and pore volumes. While mesoporous silica-based DDS can improve chemotherapy to a certain extent, when used in combination with other cancer therapies MSN based chemotherapy exhibits a synergistic effect, greatly improving therapeutic outcomes. In this review, we discuss the applications of MSN DDS for a diverse range of chemotherapeutic combination anti-tumor therapies, including phototherapy, gene therapy, immunotherapy and other less common modalities. Furthermore, we focus on the characteristics of each nanomaterial and the synergistic advantages of the combination therapies. Lastly, we examine the challenges and future prospects of MSN based chemotherapeutic combination therapies.
Collapse
Affiliation(s)
- Ying Gao
- Department of Pharmacy, School of Medicine, Zhejiang University City College, Hangzhou, China
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Dongruo Gao
- Department of Pharmacy, School of Medicine, Zhejiang University City College, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Jie Shen
- Department of Pharmacy, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Qiwen Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
37
|
Chu Q, Zhu H, Liu B, Cao G, Fang C, Wu Y, Li X, Han G. Delivery of amino acid oxidase via catalytic nanocapsules to enable effective tumor inhibition. J Mater Chem B 2020; 8:8546-8557. [PMID: 32840278 DOI: 10.1039/d0tb01425g] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Amino acids are the fundamental building blocks of proteins in tumor cells. The consumption of amino acid can be an effective approach for destroying the tumor cytoskeleton and malfunctioning of the intracellular metabolic balance. Following this concept, herein, amino acid oxidase (AAO) is delivered by hollow Fe3+/tannic acid nanocapsules (HFe-TA) and incorporated within the cancer cell membrane (M) for the first time for synergistic tumor therapy. In this system (M@AAO@HFe-TA), the intracellularly delivered AAO molecules catalyze the oxidative deamination effectively and consume amino acids significantly. The upregulation of intracellular acid and H2O2 concentration facilitates the HFe-TA mediated Fenton reaction and enhances the induction of cytotoxic ˙OH. With the combined effects, considerable in vitro and in vivo tumor inhibition was achieved by M@AAO@Fe-TA due to the activated Bcl-2/Bax/Cyt C/caspase 3 mitochondrial apoptotic pathway. This study offers an alternative therapeutic platform, functioning as a biomimetic cascade nanozyme, to enable synergistic starvation and chemodynamic tumor therapy with high efficacy.
Collapse
Affiliation(s)
- Qiang Chu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Huimin Zhu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Bin Liu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Guodong Cao
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310000, P. R. China
| | - Chao Fang
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Yulian Wu
- Department of Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310000, P. R. China
| | - Xiang Li
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| | - Gaorong Han
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China.
| |
Collapse
|
38
|
Affiliation(s)
- Chunhui Wang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| | - Jingxian Yang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| | - Chunyan Dong
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| | - Shuo Shi
- Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200092 P. R. China
| |
Collapse
|
39
|
Zhang Y, Li Y, Gao Z, Ding B, An P, Zhang X, Sun B, Sun B. Mesoporous Silica-Coated Silver Nanoframes as Drug-Delivery Vehicles for Chemo/Starvation/Metal Ion Multimodality Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:6345-6351. [PMID: 32388995 DOI: 10.1021/acs.langmuir.0c00191] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cutting off the energy supply by glucose oxidase (GOx) to starve cancer cells has been a feasible and efficient oncotherapy strategy. The employment of GOx can effectively starve tumor cells by aerobic hydrolysis of glucose hopefully strengthening the abnormality (including the decrease in pH, the increase of hypoxia, and toxic hydrogen peroxide) in the tumor microenvironment (TME). On this basis, we designed and fabricated a GOx-conjugated yolk-shell Ag@mSiO2 nanoframe with Ag NPs and GOx-conjugated mesoporous silica as the yolk and the shell, respectively, to make full use of changes the GOx induces in TME. Specifically, lower pH and H2O2 could accelerate the transformation of Ag nanoparticles to poisonous Ag ions. At the same time, the anabatic hypoxia condition in turn activated chemotherapy drug tirapazamine (TPZ) to exert a chemotherapeutic effect, thereby achieving effective chemo/starvation and metal ion multimodality therapy. The drug release experiment in vitro demonstrated that the GOx is the key to the nanocarriers, which can activate the whole system. The excellent cellular uptake performances of nanocarriers were corroborated by a confocal laser scanning microscope (CLSM). In addition, its superb cancer-killing effect has been confirmed by cytotoxicity and apoptosis experiments. These results indicated that the drug-delivery system achieved the cascade cancer-killing process in situ and synergistic chemo/starvation/metal ion therapy, which has a bright prospect for treating cancer.
Collapse
Affiliation(s)
- Yu Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Yaojia Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Zhiguo Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Bowen Ding
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Peijing An
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Xichen Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Bowen Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China
| |
Collapse
|
40
|
Zhang Y, Cao Y, Gao T, Kuang Y, An Z, Mao Z, He Y, Yan J, Lu Z, Pei R. Tumor Microenvironment-Responsive and Catalytic Cascade-Enhanced Nanocomposite for Tumor Thermal Ablation Synergizing with Chemodynamic and Chemotherapy. ACS APPLIED BIO MATERIALS 2020; 3:3880-3893. [DOI: 10.1021/acsabm.0c00042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ye Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yi Cao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tian Gao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Ye Kuang
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou 350004, China
| | - Zhen An
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zheng Mao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yilin He
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jincong Yan
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhongzhong Lu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
41
|
Wibowo FR, Saputra OA, Lestari WW, Koketsu M, Mukti RR, Martien R. pH-Triggered Drug Release Controlled by Poly(Styrene Sulfonate) Growth Hollow Mesoporous Silica Nanoparticles. ACS OMEGA 2020; 5:4261-4269. [PMID: 32149256 PMCID: PMC7057687 DOI: 10.1021/acsomega.9b04167] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/10/2020] [Indexed: 05/03/2023]
Abstract
In the current report, hollow mesoporous silica (HMS) nanoparticles were successfully prepared by means of a hard-templating method and further modified with poly(styrene sulfonate) (PSS) via radical polymerization. Structural analysis, surface spectroscopy, and thermogravimetric characterization confirmed a successful surface modification of HMS nanoparticles. A hairy PSS was clearly visualized by high-resolution transmission electron microscopy measurement, and it is grown on the surface of HMS nanoparticles. The Brunauer-Emmett-Teller surface area and average pore size of HMS nanoparticles were reduced after surface modification because of the pore-blocking effect, which indicated that the PSS lies on the surface of nanoparticles. Nevertheless, the PSS acts as a "nano-gate" to control the release of curcumin which is triggered by pH. The drug-release profile of unmodified HMS nanoparticles showed a stormed release in both pH 7.4 and 5.0 of phosphate buffer saline buffer solution. However, a slow release (9.92% of cumulative release) of curcumin was observed at pH 7.4 when the surface of HMS nanoparticles was modified by PSS. The kinetic release study showed that the curcumin release mechanism from PSS@HMS nanoparticles followed the Ritger-Peppas kinetic model, which is the non-Fickian diffusion. Therefore, the PSS-decorated HMS nanoparticles demonstrate potential for pH-triggered drug release transport.
Collapse
Affiliation(s)
- Fajar R. Wibowo
- Chemistry
Department, Faculty of Mathematics and Natural Sciences, Universitas Sebelas Maret, Jl Ir. Sutami 36A, Surakarta 57126, Indonesia
| | - Ozi A. Saputra
- Master
Program of Chemistry, Graduate School of
Universitas Sebelas Maret, Jl Ir. Sutami 36A, Surakarta 57126, Indonesia
| | - Witri W. Lestari
- Chemistry
Department, Faculty of Mathematics and Natural Sciences, Universitas Sebelas Maret, Jl Ir. Sutami 36A, Surakarta 57126, Indonesia
| | - Mamoru Koketsu
- Department
of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Gifu 501-1193, Japan
| | - Rino R. Mukti
- Division
of Inorganic and Physical Chemistry, Research Center for Nanosciences
and Nanotechnology, Center for Catalysis and Reaction Engineering, Institut Teknologi Bandung, Jl. Ganesha No. 10 Bandung 40132, Indonesia
| | - Ronny Martien
- Department
of Pharmaceutics, Faculty of Pharmacy, Universitas
Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia
| |
Collapse
|
42
|
Sarode A, Annapragada A, Guo J, Mitragotri S. Layered self-assemblies for controlled drug delivery: A translational overview. Biomaterials 2020; 242:119929. [PMID: 32163750 DOI: 10.1016/j.biomaterials.2020.119929] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022]
Abstract
Self-assembly is a prominent phenomenon observed in nature. Inspired by this thermodynamically favorable approach, several natural and synthetic materials have been investigated to develop functional systems for various biomedical applications, including drug delivery. Furthermore, layered self-assembled systems provide added advantages of tunability and multifunctionality which are crucial for controlled and targeted drug release. Layer-by-layer (LbL) deposition has emerged as one of the most popular, well-established techniques for tailoring such layered self-assemblies. This review aims to provide a brief overview of drug delivery applications using LbL deposition, along with a discussion of associated scalability challenges, technological innovations to overcome them, and prospects for commercial translation of this versatile technique. Additionally, alternative self-assembly techniques such as metal-phenolic networks (MPNs) and Liesegang rings are also reviewed in the context of their recent utilization for controlled drug delivery. Blending the sophistication of these self-assembly phenomena with material science and technological advances can provide a powerful tool to develop smart drug carriers in a scalable manner.
Collapse
Affiliation(s)
- Apoorva Sarode
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Akshaya Annapragada
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Junling Guo
- Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Gu D, An P, He X, Wu H, Gao Z, Li Y, Chen F, Cheng K, Zhang Y, You C, Sun B. A novel versatile yolk-shell nanosystem based on NIR-elevated drug release and GSH depletion-enhanced Fenton-like reaction for synergistic cancer therapy. Colloids Surf B Biointerfaces 2020; 189:110810. [PMID: 32014651 DOI: 10.1016/j.colsurfb.2020.110810] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/12/2020] [Accepted: 01/17/2020] [Indexed: 12/22/2022]
Abstract
In this study, a versatile doxorubicin (DOX)-loaded yolk-shell nano-particles (HMCMD) assembled with manganese dioxide (MnO2) as the core and copper sulfide (HMCuS) as the mesoporous (∼ 6.4 nm) shell, was designed and synthesized. The resulting HMCMD possess excellent photothermal conversion efficiency. The DOX release from the yolk-shell nanoparticles could be promoted by laser irradiation, which increased the chemotherapy of DOX. Meanwhile, Mn2+ could be released from the HMCMD through a redox reaction between MnO2 and abundant glutathione (GSH) in tumor cells. The released Mn2+ could promote the decomposition of the intracellular hydrogen peroxide (H2O2) by Fenton-like reaction to generate the highly toxic hydroxyl radicals (·OH), thus exhibiting the effective chemodynamic therapy (CDT). Additionally, the efficiency of Mn2+-mediated CDT could be effectively enhanced by NIR irradiation. Further modification of polyethylene glycol (PEG) would improve the water solubility of the HMCMD to promote the uptake by MCF-7 cells. Hence, the HMCMD with synergistic effects of chemotherapy and chemodynamic/photothermal therapy would provide an alternative strategy in antitumor research.
Collapse
Affiliation(s)
- Dihai Gu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Peijing An
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Xiuli He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Hongshuai Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Zhiguo Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Yaojia Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Fanghui Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Kaiwu Cheng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Yuchen Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Chaoqun You
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 210089, PR China.
| |
Collapse
|
44
|
Li H, Zhang X, Lin X, Zhuang S, Wu Y, Liu Z, Rong J, Zhao J. CaCO 3 nanoparticles pH-sensitively induce blood coagulation as a potential strategy for starving tumor therapy. J Mater Chem B 2020; 8:1223-1234. [PMID: 31950968 DOI: 10.1039/c9tb02684c] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Based on the concept of starving tumor therapy, in this study we put forward a new idea that the pH-sensitive Ca2+ delivery of calcium carbonate nanoparticles (CaCO3 NPs) induced blood coagulation of tumor vessels, and first explored the effect of CaCO3 NPs on the in vitro and in vivo blood coagulation by acid stimulus. CaCO3 NPs with a size of about 100 nm and a porous structure of several nanometers were synthesized in an emulsion system, which showed a high loading capacity (49%) of doxorubicin hydrochloride (DOX) with an encapsulation efficiency of 98% and a pH-sensitive drug delivery. The hemolysis test showed that CaCO3 NPs were blood compatible. The in vitro Ca2+ delivery and blood clotting tests indicated that CaCO3 NPs pH-sensitively released Ca2+, and caused rapid blood coagulation at pH 5.0 but no thrombus at pH 7.4. Confocal laser scanning microscopy showed that after uptake by MCF-7 or MDA-MB-231 breast cancer cells, CaCO3 NPs mainly distributed in endosomes/lysosomes within the initial 2 h and then decomposed by acid stimulus, leading to the intracellular delivery of Ca2+ that subsequently migrated outside the cells. CaCO3 NPs were nontoxic to NIH3T3 mouse fibroblasts, but highly toxic to both MCF-7 and MDA-MB-231 cells after loading DOX. After topical administration into the breast tumors of mice, CaCO3 NPs evoked significant thrombosis and hemorrhage of tumor vasculature by hematoxylin-eosin and Masson's trichrome staining. These results indicated that CaCO3 NPs could induce blood coagulation via acid stimulus, showing potential applications in blocking tumor vessels for starving tumor therapy.
Collapse
Affiliation(s)
- Huiru Li
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Xinyue Zhang
- Guangzhoujinan Biomedicine Research and Development Center, Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Xilin Lin
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Shuqiang Zhuang
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Yan Wu
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Zhong Liu
- Guangzhoujinan Biomedicine Research and Development Center, Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Jianhua Rong
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| | - Jianhao Zhao
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
45
|
Seidl C, Simonato S, Zittel E, Schepers U, Feldmann C. Anti‐Tumor Activity of Doxorubicin‐loaded Boehmite Nanocontainers. Z Anorg Allg Chem 2019. [DOI: 10.1002/zaac.201900211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Carmen Seidl
- Institute of Toxicology and Genetics Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Sara Simonato
- Institute of Inorganic Chemistry Karlsruhe Institute of Technology (KIT) Engesserstraße 15 76131 Karlsruhe Germany
| | - Eva Zittel
- Institute of Toxicology and Genetics Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Ute Schepers
- Institute of Toxicology and Genetics Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Claus Feldmann
- Institute of Inorganic Chemistry Karlsruhe Institute of Technology (KIT) Engesserstraße 15 76131 Karlsruhe Germany
| |
Collapse
|
46
|
Wang M, Wang D, Chen Q, Li C, Li Z, Lin J. Recent Advances in Glucose-Oxidase-Based Nanocomposites for Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903895. [PMID: 31747128 DOI: 10.1002/smll.201903895] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/18/2019] [Indexed: 06/10/2023]
Abstract
Glucose oxidase (GOx) can react with intracellular glucose and oxygen (O2 ) to produce hydrogen peroxide (H2 O2 ) and gluconic acid, which can cut off the nutrition source of cancer cells and consequently inhibit their proliferation. Therefore, GOx is recognised as an ideal endogenous oxido-reductase for cancer starvation therapy. This process can further regulate the tumor microenvironment by increasing the hypoxia and the acidity. Thus, GOx offers new possibilities for the elaborate design of multifunctional nanocomposites for tumor therapy. However, natural GOx is expensive to prepare and purify and exhibits immunogenicity, short in vivo half-life, and systemic toxicity. Furthermore, GOx is highly prone to degrade after exposure to biological conditions. These intrinsic shortcomings will undoubtedly limit its biomedical applications. Accordingly, some nanocarriers can be used to protect GOx from the surrounding environment, thus controlling or preserving the activity. A variety of nanocarriers including hollow mesoporous silica nanoparticles, metal-organic frameworks, organic polymers, and magnetic nanoparticles are summarized for the construction of GOx-based nanocomposites for multimodal synergistic cancer therapy. In addition, current challenges and promising developments in this area are highlighted.
Collapse
Affiliation(s)
- Man Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Qing Chen
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Chunxia Li
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Zhengquan Li
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|