1
|
Rafiudeen RA, Anwardeen NH, Lavanya V, Jamal S, Ahmed N. Coral Reef Metabolites for Cancer Treatment. CURRENT PHARMACOLOGY REPORTS 2024; 11:3. [DOI: 10.1007/s40495-024-00386-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 01/06/2025]
|
2
|
Kameni SL, Dlamini NH, Feugang JM. Exploring the full potential of sperm function with nanotechnology tools. Anim Reprod 2024; 21:e20240033. [PMID: 39176004 PMCID: PMC11340799 DOI: 10.1590/1984-3143-ar2024-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/20/2024] [Indexed: 08/24/2024] Open
Abstract
Sperm quality is essential to guarantee the success of assisted reproduction. However, selecting high-quality sperm and maintaining it during (cryo)preservation for high efficiency remains challenging in livestock reproduction. A comprehensive understanding of sperm biology allows for better assessment of sperm quality, which could replace conventional sperm analyses used today to predict fertility with low accuracy. Omics approaches have revealed numerous biomarkers associated with various sperm phenotypic traits such as quality, survival during storage, freezability, and fertility. At the same time, nanotechnology is emerging as a new biotechnology with high potential for use in preparing sperm intended to improve reproduction in livestock. The unique physicochemical properties of nanoparticles make them exciting tools for targeting (e.g., sperm damage and sexing) and non-targeting bioapplications. Recent advances in sperm biology have led to the discovery of numerous biomarkers, making it possible to target specific subpopulations of spermatozoa within the ejaculate. In this review, we explore potential biomarkers associated with sperm phenotypes and highlight the benefits of combining these biomarkers with nanoparticles to further improve sperm preparation and technology.
Collapse
Affiliation(s)
- Serge Leugoué Kameni
- Mississippi State University, Department of Animal and Dairy Sciences, Mississippi State, MS, USA
| | - Notsile Hleliwe Dlamini
- Mississippi State University, Department of Animal and Dairy Sciences, Mississippi State, MS, USA
| | - Jean Magloire Feugang
- Mississippi State University, Department of Animal and Dairy Sciences, Mississippi State, MS, USA
| |
Collapse
|
3
|
Britton D, Legocki J, Paul D, Katsara O, Aristizabal O, Pandya N, Mishkit O, Xiao Y, Aristizabal M, Rahman N, Schneider R, Wadghiri YZ, Montclare JK. Coiled-Coil Protein Hydrogels Engineered with Minimized Fiber Diameters for Sustained Release of Doxorubicin in Triple-Negative Breast Cancer. ACS Biomater Sci Eng 2024; 10:3425-3437. [PMID: 38622760 PMCID: PMC11094684 DOI: 10.1021/acsbiomaterials.4c00349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024]
Abstract
Triple-negative breast cancer (TNBC) lacks expressed protein targets, making therapy development challenging. Hydrogels offer a promising new route in this regard by improving the chemotherapeutic efficacy through increased solubility and sustained release. Moreover, subcutaneous hydrogel administration reduces patient burden by requiring less therapy and shorter treatment times. We recently established the design principles for the supramolecular assembly of single-domain coiled-coils into hydrogels. Using a modified computational design algorithm, we designed Q8, a hydrogel with rapid assembly for faster therapeutic hydrogel preparation. Q8 encapsulates and releases doxorubicin (Dox), enabling localized sustained release via subcutaneous injection. Remarkably, a single subcutaneous injection of Dox-laden Q8 (Q8•Dox) significantly suppresses tumors within just 1 week. This work showcases the bottom-up engineering of a fully protein-based drug delivery vehicle for improved TBNC treatment via noninvasive localized therapy.
Collapse
Affiliation(s)
- Dustin Britton
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Jakub Legocki
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Deven Paul
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Olga Katsara
- Department
of Microbiology, New York University Grossman
School of Medicine, New York, New York 10016, United States
| | - Orlando Aristizabal
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Neelam Pandya
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Orin Mishkit
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Yingxin Xiao
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
| | - Matias Aristizabal
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Neha Rahman
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Robert Schneider
- Department
of Microbiology, New York University Grossman
School of Medicine, New York, New York 10016, United States
- Department
of Radiation Oncology, New York University
Grossman School of Medicine, New
York, New York 10016, United States
| | - Youssef Z. Wadghiri
- Center
for Advanced Imaging Innovation and Research (CAI2R), New York University Grossman School of Medicine, New York, New York 10016, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
| | - Jin Kim Montclare
- Department
of Chemical and Biomolecular Engineering, New York University Tandon School of Engineering, Brooklyn, New York 11201, United States
- Bernard
and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York 10016, United States
- Department
of Biomedical Engineering, New York University
Tandon School of Engineering, Brooklyn ,New York11201, United States
- Department
of Chemistry, New York University, New York, New York 10012, United States
- Department
of Biomaterials, New York University College
of Dentistry, New York, New York 10010, United States
| |
Collapse
|
4
|
Yu M, Hu S, Tang B, Yang H, Sun D. Engineering Escherichia coli Nissle 1917 as a microbial chassis for therapeutic and industrial applications. Biotechnol Adv 2023; 67:108202. [PMID: 37343690 DOI: 10.1016/j.biotechadv.2023.108202] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/19/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Genetically engineered microbes, especially Escherichia coli, have been widely used in the biosynthesis of proteins and metabolites for medical and industrial applications. As a traditional probiotic with a well-established safety record, E. coli Nissle 1917 (EcN) has recently emerged as a microbial chassis for generating living therapeutics, drug delivery vehicles, and microbial platforms for industrial production. Despite the availability of genetic tools for engineering laboratory E. coli K-12 and B strains, new genetic engineering systems are still greatly needed to expand the application range of EcN. In this review, we have summarized the latest progress in the development of genetic engineering systems in EcN, as well as their applications in the biosynthesis and delivery of valuable small molecules and biomacromolecules of medical and/or industrial interest, followed by a glimpse of how this rapidly growing field will evolve in the future.
Collapse
Affiliation(s)
- Mingjing Yu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Shilong Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Biao Tang
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Hua Yang
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, China
| | - Dongchang Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
5
|
Kim R, Kim JH. Engineered Extracellular Vesicles with Compound-Induced Cargo Delivery to Solid Tumors. Int J Mol Sci 2023; 24:ijms24119368. [PMID: 37298320 DOI: 10.3390/ijms24119368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Efficient delivery of functional factors into target cells remains challenging. Although extracellular vesicles (EVs) are considered to be potential therapeutic delivery vehicles, a variety of efficient therapeutic delivery tools are still needed for cancer cells. Herein, we demonstrated a promising method to deliver EVs to refractory cancer cells via a small molecule-induced trafficking system. We generated an inducible interaction system between the FKBP12-rapamycin-binding protein (FRB) domain and FK506 binding protein (FKBP) to deliver specific cargo to EVs. CD9, an abundant protein in EVs, was fused to the FRB domain, and the specific cargo to be delivered was linked to FKBP. Rapamycin recruited validated cargo to EVs through protein-protein interactions (PPIs), such as the FKBP-FRB interaction system. The released EVs were functionally delivered to refractory cancer cells, triple negative breast cancer cells, non-small cell lung cancer cells, and pancreatic cancer cells. Therefore, the functional delivery system driven by reversible PPIs may provide new possibilities for a therapeutic cure against refractory cancers.
Collapse
Affiliation(s)
- Raeyeong Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| | - Jong Hyun Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic University, Daegu 42472, Republic of Korea
| |
Collapse
|
6
|
Dzhuzha A, Gandalipov E, Korzhikov-Vlakh V, Katernyuk E, Zakharova N, Silonov S, Tennikova T, Korzhikova-Vlakh E. Amphiphilic Polypeptides Obtained by Post-Polymerization Modification of Poly-l-Lysine as Systems for Combined Delivery of Paclitaxel and siRNA. Pharmaceutics 2023; 15:pharmaceutics15041308. [PMID: 37111793 PMCID: PMC10143851 DOI: 10.3390/pharmaceutics15041308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
The development of effective anti-cancer therapeutics remains one of the current pharmaceutical challenges. The joint delivery of chemotherapeutic agents and biopharmaceuticals is a cutting-edge approach to creating therapeutic agents of enhanced efficacy. In this study, amphiphilic polypeptide delivery systems capable of loading both hydrophobic drug and small interfering RNA (siRNA) were developed. The synthesis of amphiphilic polypeptides included two steps: (i) synthesis of poly-αl-lysine by ring-opening polymerization and (ii) its post-polymerization modification with hydrophobic l-amino acid and l-arginine/l-histidine. The obtained polymers were used for the preparation of single and dual delivery systems of PTX and short double-stranded nucleic acid. The obtained double component systems were quite compact and had a hydrodynamic diameter in the range of 90-200 nm depending on the polypeptide. The release of PTX from the formulations was studied, and the release profiles were approximated using a number of mathematical dissolution models to establish the most probable release mechanism. A determination of the cytotoxicity in normal (HEK 293T) and cancer (HeLa and A549) cells revealed the higher toxicity of the polypeptide particles to cancer cells. The separate evaluation of the biological activity of PTX and anti-GFP siRNA formulations testified the inhibitory efficiency of PTX formulations based on all polypeptides (IC50 4.5-6.2 ng/mL), while gene silencing was effective only for the Tyr-Arg-containing polypeptide (56-70% GFP knockdown).
Collapse
Affiliation(s)
- Apollinariia Dzhuzha
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| | - Erik Gandalipov
- International Institute of Solution Chemistry and Advanced Materials Technologies, ITMO University, Lomonosov Street 9, St. Petersburg 191002, Russia
| | - Viktor Korzhikov-Vlakh
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
| | - Elena Katernyuk
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| | - Natalia Zakharova
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| | - Sergey Silonov
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Cytology, Russian Academy of Sciences, Tihkorezky Pr. 4, St. Petersburg 194064, Russia
| | - Tatiana Tennikova
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Chemistry, Saint-Petersburg State University, Universitetsky Pr. 26, St. Petersburg 198504, Russia
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy Pr. 31, St. Petersburg 199004, Russia
| |
Collapse
|
7
|
Aventaggiato M, Preziosi A, Cheraghi Bidsorkhi H, Schifano E, Vespa S, Mardente S, Zicari A, Uccelletti D, Mancini P, Lotti LV, Sarto MS, Tafani M. ZnO Nanorods Create a Hypoxic State with Induction of HIF-1 and EPAS1, Autophagy, and Mitophagy in Cancer and Non-Cancer Cells. Int J Mol Sci 2023; 24:ijms24086971. [PMID: 37108134 PMCID: PMC10138614 DOI: 10.3390/ijms24086971] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Nanomaterials are gaining increasing attention as innovative materials in medicine. Among nanomaterials, zinc oxide (ZnO) nanostructures are particularly appealing because of their opto-electrical, antimicrobial, and photochemical properties. Although ZnO is recognized as a safe material and the Zn ion (Zn2+) concentration is strictly regulated at a cellular and systemic level, different studies have demonstrated cellular toxicity of ZnO nanoparticles (ZnO-NPs) and ZnO nanorods (ZnO-NRs). Recently, ZnO-NP toxicity has been shown to depend on the intracellular accumulation of ROS, activation of autophagy and mitophagy, as well as stabilization and accumulation of hypoxia-inducible factor-1α (HIF-1α) protein. However, if the same pathway is also activated by ZnO-NRs and how non-cancer cells respond to ZnO-NR treatment, are still unknown. To answer to these questions, we treated epithelial HaCaT and breast cancer MCF-7 cells with different ZnO-NR concentrations. Our results showed that ZnO-NR treatments increased cell death through ROS accumulation, HIF-1α and endothelial PAS domain protein 1 (EPAS1) activation, and induction of autophagy and mitophagy in both cell lines. These results, while on one side, confirmed that ZnO-NRs can be used to reduce cancer growth, on the other side, raised some concerns on the activation of a hypoxic response in normal cells that, in the long run, could induce cellular transformation.
Collapse
Affiliation(s)
- Michele Aventaggiato
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Adele Preziosi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, P.le A. Moro,5, 00185 Rome, Italy
| | - Hossein Cheraghi Bidsorkhi
- Department of Aerospace, Electrical and Energy Engineering, Sapienza University, Via Eudossiana 18, 00184 Rome, Italy
- Research Center for Nanotechnology Applied to Engineering, Sapienza University, Via Eudossiana 18, 00184 Rome, Italy
| | - Emily Schifano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, P.le A. Moro,5, 00185 Rome, Italy
| | - Simone Vespa
- Center for Advanced Studies and Technology, University "G. D'Annunzio" of Chieti-Pescara, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Stefania Mardente
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Alessandra Zicari
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Daniela Uccelletti
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University, P.le A. Moro,5, 00185 Rome, Italy
- Research Center for Nanotechnology Applied to Engineering, Sapienza University, Via Eudossiana 18, 00184 Rome, Italy
| | - Patrizia Mancini
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Lavinia Vittoria Lotti
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| | - Maria Sabrina Sarto
- Department of Aerospace, Electrical and Energy Engineering, Sapienza University, Via Eudossiana 18, 00184 Rome, Italy
- Research Center for Nanotechnology Applied to Engineering, Sapienza University, Via Eudossiana 18, 00184 Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
8
|
pH-Responsive Drug Delivery Nanoplatforms as Smart Carriers of Unsymmetrical Bisacridines for Targeted Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15010201. [PMID: 36678830 PMCID: PMC9861370 DOI: 10.3390/pharmaceutics15010201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Selective therapy and controlled drug release at an intracellular level remain key challenges for effective cancer treatment. Here, we employed folic acid (FA) as a self-navigating molecule in nanoconjugates containing quantum dots (QDs) and β-cyclodextrin (β-CD) for the delivery of antitumor unsymmetrical bisacridine compound (C-2028) to lung and prostate cancers as well as normal cells. The bisacridine derivative can form the inclusion complex with β-cyclodextrin molecule, due to the presence of a planar fragment in its structure. The stability of such a complex is pH-dependent. The drug release profile at different pH values and the mechanism of C-2028 release from QDs-β-CD-FA nanoconjugates were investigated. Next, the intracellular fate of compounds and their influence on lysosomal content in the cells were also studied. Confocal Laser Scanning Microscopy studies proved that all investigated compounds were delivered to acidic organelles, the pH of which promoted an increased release of C-2028 from its nanoconjugates. Since the pH in normal cells is higher than in cancer cells, the release of C-2028 from its nanoconjugates is decreased in these cells. Additionally, we obtained the concentration profiles of C-2028 in the selected cells treated with unbound C-2028 or nanoconjugate by the HPLC analysis.
Collapse
|
9
|
Lopes LB, Apolinário AC, Salata GC, Malagó ID, Passos JS. Lipid Nanocarriers for Breast Cancer Treatment. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
10
|
Dornjak L, Kovačić M, Ostojić K, Angaits A, Szpunar J, Urlić I, Rogina A. Chitosan-Boric Acid Scaffolds for Doxorubicin Delivery in the Osteosarcoma Treatment. Polymers (Basel) 2022; 14:4753. [PMID: 36365746 PMCID: PMC9656110 DOI: 10.3390/polym14214753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 08/20/2023] Open
Abstract
Biologically compatible chitosan-based scaffolds have been considered a promising platform for tissue regeneration, tumor treatment, and targeted drug delivery. Chitosan-based scaffolds can be utilized as pH-sensitive drug carriers with targeted drug delivery resulting in less invasive tumor treatments. Further improvement with bioactive ions, such as borate ions, can result in the dual functionality of chitosan carriers provided by simultaneous antitumor efficacy and tissue regeneration. Here, boric acid-containing crosslinked chitosan scaffolds were prepared as delivery systems of doxorubicin, a chemotherapy drug used in the treatment of osteosarcoma. The encapsulation of boric acid was indicated by FTIR spectroscopy, while the ICP-MS analysis indicated the rapid release of boron in phosphate buffer (pH 6.0) and phosphate-buffered saline solution (pH 7.4). The obtained chitosan-boric acid scaffolds exhibit a highly porous and interconnected structure responsible for high swelling capacity, while enzymatic degradation indicated good scaffolds stability during four weeks of incubation at pH 6.0 and 7.4. Furthermore, the release of doxorubicin investigated in phosphate buffers indicated lower doxorubicin concentrations at pH 7.4 with respect to pH 6.0. Finally, the cytotoxicity of prepared doxorubicin-encapsulated scaffolds was evaluated on human sarcoma cells indicating the scaffolds' potential as cytostatic agents.
Collapse
Affiliation(s)
- Luka Dornjak
- Faculty of Chemical Engineering and Technology, University of Zagreb, Trg Marka Marulića 19, 10000 Zagreb, Croatia
| | - Marin Kovačić
- Faculty of Chemical Engineering and Technology, University of Zagreb, Trg Marka Marulića 19, 10000 Zagreb, Croatia
| | - Karla Ostojić
- Faculty of Science, University of Zagreb, Horvatovac 102a, 10000 Zagreb, Croatia
| | - Ange Angaits
- Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM), UMR 5254, CNRS-University of Pau, Hélioparc, 2, Av. Pr. Angot, 64053 Pau, France
| | - Joanna Szpunar
- Institute of Analytical and Physical Chemistry for the Environment and Materials (IPREM), UMR 5254, CNRS-University of Pau, Hélioparc, 2, Av. Pr. Angot, 64053 Pau, France
| | - Inga Urlić
- Faculty of Science, University of Zagreb, Horvatovac 102a, 10000 Zagreb, Croatia
| | - Anamarija Rogina
- Faculty of Chemical Engineering and Technology, University of Zagreb, Trg Marka Marulića 19, 10000 Zagreb, Croatia
| |
Collapse
|
11
|
Cheng Z, Li Y, Zhao D, Zhao W, Wu M, Zhang W, Cui Y, Zhang P, Zhang Z. Nanocarriers for intracellular co-delivery of proteins and small-molecule drugs for cancer therapy. Front Bioeng Biotechnol 2022; 10:994655. [PMID: 36147526 PMCID: PMC9485877 DOI: 10.3389/fbioe.2022.994655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In the past few decades, the combination of proteins and small-molecule drugs has made tremendous progress in cancer treatment, but it is still not satisfactory. Because there are great differences in molecular weight, water solubility, stability, pharmacokinetics, biodistribution, and the ways of release and action between macromolecular proteins and small-molecule drugs. To improve the efficacy and safety of tumor treatment, people are committed to developing protein and drug co-delivery systems. Currently, intracellular co-delivery systems have been developed that integrate proteins and small-molecule drugs into one nanocarrier via various loading strategies. These systems significantly improve the blood stability, half-life, and biodistribution of proteins and small-molecule drugs, thus increasing their concentration in tumors. Furthermore, proteins and small-molecule drugs within these systems can be specifically targeted to tumor cells, and are released to perform functions after entering tumor cells simultaneously, resulting in improved effectiveness and safety of tumor treatment. This review summarizes the latest progress in protein and small-molecule drug intracellular co-delivery systems, with emphasis on the composition of nanocarriers, as well as on the loading methods of proteins and small-molecule drugs that play a role in cells into the systems, which have not been summarized by others so far.
Collapse
Affiliation(s)
- Zhihong Cheng
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yongshuang Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Meng Wu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Weilin Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Congo Red as a Supramolecular Carrier System for Doxorubicin: An Approach to Understanding the Mechanism of Action. Int J Mol Sci 2022; 23:ijms23168935. [PMID: 36012200 PMCID: PMC9408855 DOI: 10.3390/ijms23168935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
The uptake and distribution of doxorubicin in the MCF7 line of breast-cancer cells were monitored by Raman measurements. It was demonstrated that bioavailability of doxorubicin can be significantly enhanced by applying Congo red. To understand the mechanism of doxorubicin delivery by Congo red supramolecular carriers, additional monolayer measurements and molecular dynamics simulations on model membranes were undertaken. Acting as molecular scissors, Congo red particles cut doxorubicin aggregates and incorporated them into small-sized Congo red clusters. The mixed doxorubicin/Congo red clusters were adsorbed to the hydrophilic part of the model membrane. Such behavior promoted transfer through the membrane.
Collapse
|
13
|
Zhang CW, Zhang JG, Yang X, Du WL, Yu ZL, Lv ZY, Mou XZ. Carbohydrates based stimulus responsive nanocarriers for cancer-targeted chemotherapy: A review of current practices. Expert Opin Drug Deliv 2022; 19:623-640. [PMID: 35611662 DOI: 10.1080/17425247.2022.2081320] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Many nanocarriers have been developed to react physicochemically to exterior stimuli like ultrasonic, light, heat, and magnetic fields, along with various internal stimuli including pH, hypoxia, enzyme, and redox potential. Nanocarriers are capable to respond various stimuli within the cancer cells to enable on-demand drug delivery, activation of bioactive compounds, controlled drug release, and targeting ligands, as well as size, charge, and conformation conversion, enabling sensing and signaling, overcoming multidrug resistance, accurate diagnosis, and precision therapy. AREAS COVERED Carbohydrates are ubiquitous biomolecules with a high proclivity for supramolecular network formation. Numerous carbohydrate-based nanomaterials have been used in biological solicitations and stimuli-based responses. Particular emphasis has been placed on the utilization of carbohydrate-based NPs and nanogels in various fields including imaging, drug administration, and tissue engineering. Because the assembly process is irreversible, carbohydrate-based systems are excellent ingredients for the development of stimulus-responsive nanocarriers for cancer-targeted chemotherapy. This review aims to summarise current research on carbohydrate-based nanomaterials, with an emphasis on stimuli-sensitive nanocarriers for cancer-targeted chemotherapy. EXPERT OPINION Carbohydrates-based stimulus-responsive nanomaterials have been proved highly efficient for targeted delivery of anticancer drugs, thus leading to effective chemotherapy with minimum off-target effects.
Collapse
Affiliation(s)
- Cheng-Wu Zhang
- General Surgery, Cancer Center, Department of hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Jun-Gang Zhang
- General Surgery, Cancer Center, Department of hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Xue Yang
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Wen-Lin Du
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Zi-Lin Yu
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Zhen-Ye Lv
- General Surgery, Cancer Center, Department of Breast Surgery, Zhejiang Provincial People's Hospital (Affiliated Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China.,Department of General Surgery, Zhoushan Dinghai Central Hospital, Zhoushan, Zhejiang, China
| | - Xiao-Zhou Mou
- General Surgery, Cancer Center, Department of hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China.,Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (Affiliated Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Drobota M, Vlad S, Gradinaru LM, Bargan A, Radu I, Butnaru M, Rîmbu CM, Ciobanu RC, Aflori M. Composite Materials Based on Gelatin and Iron Oxide Nanoparticles for MRI Accuracy. MATERIALS (BASEL, SWITZERLAND) 2022; 15:3479. [PMID: 35629506 PMCID: PMC9147670 DOI: 10.3390/ma15103479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023]
Abstract
The majority of recent studies have focused on obtaining MRI materials for internal use. However, this study focuses on a straightforward method for preparing gelatin-based materials with iron oxide nanoparticles (G-Fe2O3 and G-Fe3O4) for external use. The newly obtained materials must be precisely tuned to match the requirements and usage situation because they will be in close touch with human/animal skin. The biocompatible structures formed by gelatin, tannic acid, and iron oxide nanoparticles were investigated by using FTIR spectroscopy, SEM-EDAX analysis, and contact angle methods. The physico-chemical properties were obtained by using mechanical investigations, dynamic vapor sorption analysis, and bulk magnetic determination. The size and shape of iron oxide nanoparticles dictates the magnetic behavior of the gelatin-based samples. The magnetization curves revealed a typical S-shaped superparamagnetic behavior which is evidence of improved MRI image accuracy. In addition, the MTT assay was used to demonstrate the non-toxicity of the samples, and the antibacterial test confirmed satisfactory findings for all G-based materials.
Collapse
Affiliation(s)
- Mioara Drobota
- “Petru Poni” Institute of Macromolecular Chemistry, Aleea Gr. GhicaVoda, 41A, 700487 Iasi, Romania; (S.V.); (L.M.G.); (A.B.); (M.B.)
| | - Stelian Vlad
- “Petru Poni” Institute of Macromolecular Chemistry, Aleea Gr. GhicaVoda, 41A, 700487 Iasi, Romania; (S.V.); (L.M.G.); (A.B.); (M.B.)
| | - Luiza Madalina Gradinaru
- “Petru Poni” Institute of Macromolecular Chemistry, Aleea Gr. GhicaVoda, 41A, 700487 Iasi, Romania; (S.V.); (L.M.G.); (A.B.); (M.B.)
| | - Alexandra Bargan
- “Petru Poni” Institute of Macromolecular Chemistry, Aleea Gr. GhicaVoda, 41A, 700487 Iasi, Romania; (S.V.); (L.M.G.); (A.B.); (M.B.)
| | - Iulian Radu
- Department of Surgery, Regional Institute of Oncology, I-st Surgical Oncology, “Grigore T. Popa” University of Medicine and Pharmacy, 700483 Iasi, Romania;
| | - Maria Butnaru
- “Petru Poni” Institute of Macromolecular Chemistry, Aleea Gr. GhicaVoda, 41A, 700487 Iasi, Romania; (S.V.); (L.M.G.); (A.B.); (M.B.)
- Department of Biomedical Sciences, “Grigore T. Popa” University of Medicine and Pharmacy, Kogalniceanu Street, 9-13, 700115 Iasi, Romania
| | - Cristina Mihaela Rîmbu
- Department of Public Health, Faculty of Veterinary Medicine, “Ion Ionescu de la Brad” University of Life Sciences, Mihail Sadoveanu Alley no. 8, 700490 Iasi, Romania;
| | - Romeo Cristian Ciobanu
- SC All Green SRL, I. Bacalu Street, 5, 700029 Iasi, Romania;
- Electrical Engineering Faculty, “Gheorghe Asachi” Technical University of Iasi, Dimitrie Mangeron Bd., 67, 700050 Iasi, Romania
| | - Magdalena Aflori
- “Petru Poni” Institute of Macromolecular Chemistry, Aleea Gr. GhicaVoda, 41A, 700487 Iasi, Romania; (S.V.); (L.M.G.); (A.B.); (M.B.)
| |
Collapse
|
15
|
Prashanth M, Chaitra K, Krishnaiah P, Prashanth K, Yogesh kumar K, Kumar S, Alharthi FA, Parashuram L, Raghu M. Multifunctional carbon dot anchored halloysite nanotube: Nanovehicle for cisplatin drug release, cytotoxicity on breast cancer cells and DNA binding studies. CHEMICAL DATA COLLECTIONS 2022; 38:100827. [DOI: 10.1016/j.cdc.2021.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Doxorubicin Release from Bovine Serum Albumin Microparticles. Pharm Chem J 2022. [DOI: 10.1007/s11094-022-02552-y] [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]
|
17
|
Mehraj T, Elkanayati RM, Farooq I, Mir TM. A review of Nigella sativa and its active principles as anticancer agents. BLACK SEEDS (NIGELLA SATIVA) 2022:91-118. [DOI: 10.1016/b978-0-12-824462-3.00012-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Rani V, Verma Y, Rana SVS. Zinc Oxide Nanoparticles Ameliorate Dimethylnitrosamine-Induced Renal Toxicity in Rat. Appl Biochem Biotechnol 2021; 194:1699-1715. [PMID: 34855113 DOI: 10.1007/s12010-021-03689-4] [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] [Received: 05/19/2021] [Accepted: 09/08/2021] [Indexed: 11/29/2022]
Abstract
Dimethylnitrosamine (DMN) is an established carcinogen. It is toxic to several organs, viz., the liver, kidney, and lungs, and immune system. Several drugs have been used in the past to modulate its toxicity using experimental animal models. The present study was designed to investigate the effect of zinc oxide nanoparticles (ZnONPs) on renal toxicity caused by DMN in laboratory rat. Since oxidative mechanisms are mainly involved in its toxicity, the proposed study focuses on the amelioration of oxidative stress response by ZnONPs, if any. The present results show that administration of ZnONPs (50 mg/kg body weight/rat) to DMN (2 μl/100 g body weight/rat)-treated rats diminuted the concentration of malonaldehyde, H2O2, and NO in the kidney. However, reduced glutathione (GSH) concentration increased after ZnONP treatment. Results on glutathione S-transferase and glutathione peroxidase favored its antioxidative effects. These results are supported by the recovery of oxidative DNA damage and less pronounced histopathological changes in the kidney. It is hypothesized that ZnONPs might be toxic to renal tissue; however, its strong therapeutic/antioxidative potential helps in ameliorating DMN-induced renal toxicity in rat.
Collapse
Affiliation(s)
- Varsha Rani
- Department of Toxicology, Chaudhary Charan Singh University, Meerut, 250004, India
| | - Yeshvandra Verma
- Department of Toxicology, Chaudhary Charan Singh University, Meerut, 250004, India
| | - S V S Rana
- Department of Toxicology, Chaudhary Charan Singh University, Meerut, 250004, India.
| |
Collapse
|
19
|
Singh S, Moran JL. Autonomously Propelled Colloids for Penetration and Payload Delivery in Complex Extracellular Matrices. MICROMACHINES 2021; 12:mi12101216. [PMID: 34683267 PMCID: PMC8541468 DOI: 10.3390/mi12101216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/01/2021] [Accepted: 10/03/2021] [Indexed: 11/30/2022]
Abstract
For effective treatment of diseases such as cancer or fibrosis, it is essential to deliver therapeutic agents such as drugs to the diseased tissue, but these diseased sites are surrounded by a dense network of fibers, cells, and proteins known as the extracellular matrix (ECM). The ECM forms a barrier between the diseased cells and blood circulation, the main route of administration of most drug delivery nanoparticles. Hence, a stiff ECM impedes drug delivery by limiting the transport of drugs to the diseased tissue. The use of self-propelled particles (SPPs) that can move in a directional manner with the application of physical or chemical forces can help in increasing the drug delivery efficiency. Here, we provide a comprehensive look at the current ECM models in use to mimic the in vivo diseased states, the different types of SPPs that have been experimentally tested in these models, and suggest directions for future research toward clinical translation of SPPs in diverse biomedical settings.
Collapse
Affiliation(s)
- Shrishti Singh
- Department of Bioengineering, George Mason University, Fairfax, VA 22030, USA;
| | - Jeffrey L. Moran
- Department of Bioengineering, George Mason University, Fairfax, VA 22030, USA;
- Department of Mechanical Engineering, George Mason University, Fairfax, VA 22030, USA
- Correspondence:
| |
Collapse
|
20
|
Sharifianjazi F, Irani M, Esmaeilkhanian A, Bazli L, Asl MS, Jang HW, Kim SY, Ramakrishna S, Shokouhimehr M, Varma RS. Polymer incorporated magnetic nanoparticles: Applications for magnetoresponsive targeted drug delivery. MATERIALS SCIENCE AND ENGINEERING: B 2021; 272:115358. [DOI: 10.1016/j.mseb.2021.115358] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
21
|
Iravani S, Varma RS. Plant Pollen Grains: A Move Towards Green Drug and Vaccine Delivery Systems. NANO-MICRO LETTERS 2021; 13:128. [PMID: 34138347 PMCID: PMC8124031 DOI: 10.1007/s40820-021-00654-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 04/25/2021] [Indexed: 05/29/2023]
Abstract
Pollen grains and plant spores have emerged as innovative biomaterials for various applications such as drug/vaccine delivery, catalyst support, and the removal of heavy metals. The natural microcapsules comprising spore shells and pollen grain are designed for protecting the genetic materials of plants from exterior impairments. Two layers make up the shell, the outer layer (exine) that comprised largely of sporopollenin, and the inner layer (intine) that built chiefly of cellulose. These microcapsule shells, namely hollow sporopollenin exine capsules have some salient features such as homogeneity in size, non-toxic nature, resilience to both alkalis and acids, and the potential to withstand at elevated temperatures; they have displayed promising potential for the microencapsulation and the controlled drug delivery/release. The important attribute of mucoadhesion to intestinal tissues can prolong the interaction of sporopollenin with the intestinal mucosa directing to an augmented effectiveness of nutraceutical or drug delivery. Here, current trends and prospects related to the application of plant pollen grains for the delivery of vaccines and drugs and vaccine are discussed.
Collapse
Affiliation(s)
- Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacký University in Olomouc , Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
22
|
Carrion CC, Nasrollahzadeh M, Sajjadi M, Jaleh B, Soufi GJ, Iravani S. Lignin, lipid, protein, hyaluronic acid, starch, cellulose, gum, pectin, alginate and chitosan-based nanomaterials for cancer nanotherapy: Challenges and opportunities. Int J Biol Macromol 2021; 178:193-228. [PMID: 33631269 DOI: 10.1016/j.ijbiomac.2021.02.123] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
Although nanotechnology-driven drug delivery systems are relatively new, they are rapidly evolving since the nanomaterials are deployed as effective means of diagnosis and delivery of assorted therapeutic agents to targeted intracellular sites in a controlled release manner. Nanomedicine and nanoparticulate drug delivery systems are rapidly developing as they play crucial roles in the development of therapeutic strategies for various types of cancer and malignancy. Nevertheless, high costs, associated toxicity and production of complexities are some of the critical barriers for their applications. Green nanomedicines have continually been improved as one of the viable approaches towards tumor drug delivery, thus making a notable impact on which considerably affect cancer treatment. In this regard, the utilization of natural and renewable feedstocks as a starting point for the fabrication of nanosystems can considerably contribute to the development of green nanomedicines. Nanostructures and biopolymers derived from natural and biorenewable resources such as proteins, lipids, lignin, hyaluronic acid, starch, cellulose, gum, pectin, alginate, and chitosan play vital roles in the development of cancer nanotherapy, imaging and management. This review uncovers recent investigations on diverse nanoarchitectures fabricated from natural and renewable feedstocks for the controlled/sustained and targeted drug/gene delivery systems against cancers including an outlook on some of the scientific challenges and opportunities in this field. Various important natural biopolymers and nanomaterials for cancer nanotherapy are covered and the scientific challenges and opportunities in this field are reviewed.
Collapse
Affiliation(s)
- Carolina Carrillo Carrion
- Department of Organic Chemistry, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, Ctra Nnal IV-A Km. 396, E-14014 Cordoba, Spain
| | | | - Mohaddeseh Sajjadi
- Department of Chemistry, Faculty of Science, University of Qom, Qom 37185-359, Iran
| | - Babak Jaleh
- Department of Physics, Bu-Ali Sina University, 65174 Hamedan, Iran
| | | | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
23
|
Al Subeh ZY, Chu NQ, Korunes-Miller JT, Tsai LL, Graf TN, Hung YP, Pearce CJ, Grinstaff MW, Colby AH, Colson YL, Oberlies NH. Delivery of eupenifeldin via polymer-coated surgical buttresses prevents local lung cancer recurrence. J Control Release 2021; 331:260-269. [PMID: 33484778 PMCID: PMC7946725 DOI: 10.1016/j.jconrel.2021.01.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/25/2022]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Unfortunately, high recurrence rates and poor survival remain despite surgical resection and conventional chemotherapy. Local drug delivery systems are a promising intervention for lung cancer treatment with the potential for improved efficacy with reduced systemic toxicity. Here, we describe the development of a chemotherapy-loaded polymer buttress, to be implanted along the surgical margin at the time of tumor resection, for achieving local and prolonged release of a new anticancer agent, eupenifeldin. We prepared five different formulations of buttresses with varying amounts of eupenifeldin, and additional external empty polymer coating layers (or thicknesses) to modulate drug release. The in vitro eupenifeldin release profile depends on the number of external coating layers with the formulation of the greatest thickness demonstrating a prolonged release approaching 90 days. Similarly, the long-term cytotoxicity of eupenifeldin-loaded buttress formulations against murine Lewis lung carcinoma (LLC) and human lung carcinoma (A549) cell lines mirrors the eupenifeldin release profiles and shows a prolonged cytotoxic effect. Eupenifeldin-loaded buttresses significantly decrease local tumor recurrence in vivo and increase disease-free survival in a lung cancer resection model.
Collapse
Affiliation(s)
- Zeinab Y Al Subeh
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, United States
| | - Ngoc-Quynh Chu
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA 02114, United States
| | | | - Lillian L Tsai
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Tyler N Graf
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, United States
| | - Yin P Hung
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA 02114, United States
| | | | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States; Department of Chemistry, Boston University, Boston, MA 02215, United States
| | - Aaron H Colby
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States; Ionic Pharmaceuticals, LLC, Brookline, MA 02445, United States.
| | - Yolonda L Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA 02114, United States.
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, United States.
| |
Collapse
|
24
|
Jahan S, Karim ME, Chowdhury EH. Nanoparticles Targeting Receptors on Breast Cancer for Efficient Delivery of Chemotherapeutics. Biomedicines 2021; 9:114. [PMID: 33530291 PMCID: PMC7910939 DOI: 10.3390/biomedicines9020114] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/25/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
The journey of chemotherapeutic drugs from the site of administration to the site of action is confronted by several factors including low bioavailability, uneven distribution in major organs, limited accessibility of drug molecules to the distant tumor tissues, and lower therapeutic indexes. These unavoidable features of classical chemotherapeutics necessitate an additional high, repetitive dose of drugs to obtain maximum therapeutic responses with the result of unintended adverse side effects. An erratic tumor microenvironment, notable drawbacks of conventional chemotherapy, and multidrug-resistant mechanisms of breast cancer cells warrant precisely designed therapeutics for the treatment of cancers. In recent decades, nanoparticles have been deployed for the delivery of standard anticancer drugs to maximize the therapeutic potency while minimizing the adverse effects to increase the quality and span of life. Several organic and inorganic nanoplatforms that have been designed exploiting the distinctive features of the tumor microenvironment and tumor cells offer favorable physicochemical properties and pharmacokinetic profiles of a parent drug, with delivery of higher amounts of the drug to the pathological site and its controlled release, thereby improving the balance between its efficacy and toxicity. Advances to this front have included design and construction of targeted nanoparticles by conjugating homing devices like peptide, ligand, and Fab on the surface of nanomaterials to navigate nanoparticledrug complexes towards the target tumor cell with minimal destruction of healthy cells. Furthermore, actively targeting nanoparticles can facilitate the delivery and cellular uptake of nanoparticle-loaded drug constructs via binding with specific receptors expressed aberrantly on the surface of a tumor cell. Herein, we present an overview of the principle of targeted delivery approaches, exploiting drug-nanoparticle conjugates with multiple targeting moieties to target specific receptors of breast cancer cells and highlighting therapeutic evaluation in preclinical studies. We conclude that an understanding of the translational gap and challenges would show the possible future directions to foster the development of novel targeted nanotherapeutics.
Collapse
Affiliation(s)
| | | | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Petaling Jaya 47500, Malaysia; (S.J.); (M.E.K.)
| |
Collapse
|
25
|
Verma N, Thapa K, Dua K. Material and strategies used in oncology drug delivery. ADVANCED DRUG DELIVERY SYSTEMS IN THE MANAGEMENT OF CANCER 2021:47-62. [DOI: 10.1016/b978-0-323-85503-7.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Karan S, Debnath S, Kuotsu K, Chatterjee TK. In-vitro and in-vivo evaluation of polymeric microsphere formulation for colon targeted delivery of 5-fluorouracil using biocompatible natural gum katira. Int J Biol Macromol 2020; 158:922-936. [PMID: 32335117 DOI: 10.1016/j.ijbiomac.2020.04.129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 12/20/2022]
Abstract
The aim was to develop oral site-specific rate-controlled anticancer drug delivery to pacify systemic side-effects and offer effective and safe therapy for colon cancer with compressed dose and duration of treatment. The double emulsion solvent evaporation method was employed. To check functionality, DAPI-staining and in-vivo anticancer study of Ehrlich Ascites Carcinoma bearing mice was tested. Histopathology of liver and kidney and Cell morphology of EAC cell was also performed. Formulated and optimized polymeric microsphere of 5-FU showed excellent physicochemical features. In-vitro, DAPI results pointed drug-treated groups displayed the prominent feature of apoptosis. The percentage of apoptotic of entrapped drug played in a dose-dependent manner. Significant decreases in EAC liquid tumors and increased life span of treated mice were observed. Rate of variation of cell morphology was more in 5-FU loaded microsphere than 5-FU injection. Hematological and biochemical parameter's and Histopathology of liver and kidney resulted that due to control released formulation have slow release rate, that gives less trace on liver and kidney function. Finally, we foresee that polymeric microsphere of 5-FU applying natural gum katira could be an assuring micro-carrier for active colon targeting delivery tool with augmented chemotherapeutic efficacy and lowering side effect against colon cancer.
Collapse
Affiliation(s)
- Saumen Karan
- Division of Pharmacology, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Souvik Debnath
- Division of Pharmacology, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India; Department of Basic Medical Sciences, Purdue University, USA
| | - Ketousetuo Kuotsu
- Division of Pharmaceutics, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Tapan Kumar Chatterjee
- Division of Pharmacology, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India; Department of Pharmaceutical Science and Technology, JIS University, Kolkata, India.
| |
Collapse
|
27
|
Abasalizadeh F, Moghaddam SV, Alizadeh E, akbari E, Kashani E, Fazljou SMB, Torbati M, Akbarzadeh A. Alginate-based hydrogels as drug delivery vehicles in cancer treatment and their applications in wound dressing and 3D bioprinting. J Biol Eng 2020; 14:8. [PMID: 32190110 PMCID: PMC7069202 DOI: 10.1186/s13036-020-0227-7] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/05/2020] [Indexed: 12/31/2022] Open
Abstract
Hydrogels are a three-dimensional and crosslinked network of hydrophilic polymers. They can absorb a large amount of water or biological fluids, which leads to their swelling while maintaining their 3D structure without dissolving (Zhu and Marchant, Expert Rev Med Devices 8:607-626, 2011). Among the numerous polymers which have been utilized for the preparation of the hydrogels, polysaccharides have gained more attention in the area of pharmaceutics; Sodium alginate is a non-toxic, biocompatible, and biodegradable polysaccharide with several unique physicochemical properties for which has used as delivery vehicles for drugs (Kumar Giri et al., Curr Drug Deliv 9:539-555, 2012). Owing to their high-water content and resembling the natural soft tissue, hydrogels were studied a lot as a scaffold. The formation of hydrogels can occur by interactions of the anionic alginates with multivalent inorganic cations through a typical ionotropic gelation method. However, those applications require the control of some properties such as mechanical stiffness, swelling, degradation, cell attachment, and binding or release of bioactive molecules by using the chemical or physical modifications of the alginate hydrogel. In the current review, an overview of alginate hydrogels and their properties will be presented as well as the methods of producing alginate hydrogels. In the next section of the present review paper, the application of the alginate hydrogels will be defined as drug delivery vehicles for chemotherapeutic agents. The recent advances in the application of the alginate-based hydrogels will be describe later as a wound dressing and bioink in 3D bioprinting.
Collapse
Affiliation(s)
- Farhad Abasalizadeh
- Department of Traditional Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elahe akbari
- Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Elmira Kashani
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Mohammad Bagher Fazljou
- Department of Traditional Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadali Torbati
- Department of Food Science and Technology, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Tuberculosis and Lung Disease Research Center of Tabriz, Tabriz University of Medical Sciences, Tabriz, 5154853431 Iran
- Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| |
Collapse
|
28
|
Lei M, Ma G, Sha S, Wang X, Feng H, Zhu Y, Du X. Dual-functionalized liposome by co-delivery of paclitaxel with sorafenib for synergistic antitumor efficacy and reversion of multidrug resistance. Drug Deliv 2019; 26:262-272. [PMID: 30856352 PMCID: PMC6419656 DOI: 10.1080/10717544.2019.1580797] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 01/19/2023] Open
Abstract
Multidrug resistance (MDR) remains one of the major reasons for inefficiency of many chemotherapeutic agents in cancer therapy. In this study, a D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) and polylysine-deoxycholic acid copolymer (PLL-DA) co-modified cationic liposome coating with hyaluronic acid (HA) was constructed for co-delivery of paclitaxel (PTX) and chemosensitizing agent, sorafenib (SOR) to treat the MDR cancer. The multifunctional liposome (HA-TPD-CL-PTX/SOR) presented good stability against rat plasma and was capable of reversing surface zeta potential under acidic conditions in the presence of HAase. Additionally, experimental result confirmed that the PLL-DA copolymer would facilitate the endo-lysosomal escape of the liposome. In vitro study demonstrated that HA-TPD-CL-PTX/SOR could significantly enhance drug accumulation in resistant MCF-7/MDR cells by inhibiting the P-gp efflux, and effectively inhibited growth of tumor cells. Furthermore, the liposome showed an enhanced anticancer activity in vivo, with a tumor growth inhibition rate of 78.52%. In summary, HA-TPD-CL-PTX/SOR exhibited a great potential for effective therapy of resistant cancers by combining with chemotherapeutic agents and could be a promising nano-carrier for reversing MDR and improving the effectiveness of chemotherapy.
Collapse
Affiliation(s)
- Meng Lei
- College of Science, Nanjing Forestry University, Nanjing, PR China
| | - Guanglan Ma
- College of Life Science, Nanjing Normal University, Nanjing, PR China
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, PR China
| | - Sijia Sha
- College of Science, Nanjing Forestry University, Nanjing, PR China
| | - Xueyuan Wang
- College of Life Science, Nanjing Normal University, Nanjing, PR China
| | - Haiting Feng
- College of Life Science, Nanjing Normal University, Nanjing, PR China
| | - Yongqiang Zhu
- College of Life Science, Nanjing Normal University, Nanjing, PR China
| | - Xiao Du
- College of Life Science, Nanjing Normal University, Nanjing, PR China
| |
Collapse
|
29
|
Wang P, Liu W, Liu S, Yang R, Pu Y, Zhang W, Wang X, Liu X, Ren Y, Chi B. pH-responsive nanomicelles of poly(ethylene glycol)-poly(ε-caprolactone)-poly(L-histidine) for targeted drug delivery. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2019; 31:277-292. [PMID: 31665964 DOI: 10.1080/09205063.2019.1687132] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Here, a novel pH-responsive block copolymer, poly (ethylene glycol)-poly(ε-caprolactone)-poly(L-histidine) (PEG-PCL-PHis), was synthesized and designed for anti-cancer drug delivery with excellent biocompatible, biodegradable, and strong drug loading efficiency. 1H-NMR, IF-IR, and GPC were used to characterize the structure of the PEG-PCL-PHis copolymer. In addition, the morphology, particle size, Zeta potential, and critical micelle concentration (CMC) of different degree of polymerization were determined by transmission electron microscopy (TEM), dynamic light scattering granulometer (DLS), and fluorescence spectrometer, respectively. The strong affinity between the core of micelles and hydrophobic drug was manifested with 15.09% drug loading content and 84.65% entrapment efficiency. In vitro release of DOX from the block copolymer micelle demonstrated, the PEG-PCL-PHis copolymer micelle has stable and durable drug releasing ability accompanied with pH-sensitivity. From the mechanism of cellular uptake the micelles, the pathway of drug release was captured by confocal laser scanning microscope. These experiments demonstrated the safe delivery for anticancer medicine through this novel copolymer. In conclusion, the PEG-PCL-PHis copolymer micelle has great potential to become a safe drug carrier for cancer chemotherapy.
Collapse
Affiliation(s)
- Penghui Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Wei Liu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Shuai Liu
- School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Rong Yang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Yajie Pu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Wenjie Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Xiaoxue Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Xin Liu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Yanhan Ren
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Bo Chi
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China.,Jiangsu National Synergetic Innovation Center for Advanced Materials, Nanjing Tech University, Nanjing, China
| |
Collapse
|
30
|
Ngo N, Choucair K, Creeden JF, Qaqish H, Bhavsar K, Murphy C, Lian K, Albrethsen MT, Stanbery L, Phinney RC, Brunicardi FC, Dworkin L, Nemunaitis J. Bifidobacterium spp: the promising Trojan Horse in the era of precision oncology. Future Oncol 2019; 15:3861-3876. [PMID: 31668087 DOI: 10.2217/fon-2019-0374] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Selective delivery of therapeutic agents into solid tumors has been a major challenge impeding the achievement of long-term disease remission and cure. The need to develop alternative drug delivery routes to achieve higher drug concentration in tumor tissue, reduce unwanted off-target side effects and thus achieve greater therapeutic efficacy, has resulted in an explosive body of research. Bifidobacterium spp. are anaerobic, nonpathogenic, Gram-positive bacteria, commensal to the human gut that are a possible anticancer drug-delivery vehicle. In this review, we describe Bifidobacterium's microbiology, current clinical applications, overview of the preclinical work investigating Bifidobacterium's potential to deliver anticancer therapy, and review the different strategies used up to date. Finally, we discuss both current challenges and future prospects.
Collapse
Affiliation(s)
- Nealie Ngo
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Khalil Choucair
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Justin F Creeden
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Hanan Qaqish
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Krupa Bhavsar
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Chantal Murphy
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Kendra Lian
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Mary T Albrethsen
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Laura Stanbery
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | | | - F Charles Brunicardi
- Department of Surgery, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
- ProMedica Health System, Toledo, OH 43606, USA
| |
Collapse
|
31
|
Zhang R, Zhang Y, Zhang Y, Wang X, Gao X, Liu Y, Zhang X, He Z, Wang D, Wang Y. Ratiometric delivery of doxorubicin and berberine by liposome enables superior therapeutic index than Doxil Ⓡ. Asian J Pharm Sci 2019; 15:385-396. [PMID: 32636956 PMCID: PMC7327765 DOI: 10.1016/j.ajps.2019.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/20/2019] [Accepted: 04/25/2019] [Indexed: 01/01/2023] Open
Abstract
Although the appearance of Doxil alleviated the cardiotoxicity of DOX, the progression-free survival of patients was not prolonged compared with traditional medication regimens, and side effects such as hand-foot syndrome has occurred. In order to solve this dilemma, we have designed a novel co-delivery strategy to construct a co-loaded liposome of berberine (BER) and doxorubicin (DOX), which was called LipoBeDo. The optimal synergistic ratio of the two drugs was screened by cell cytotoxicity experiments in vitro, and the optimal attenuation ratio was further determined by in vivo cardiac H&E staining pathological sections. The optimal combination treatment caused a robust increase in apoptotic cells of 4T1, as compared to drug alone treatment. The prepared co-loaded liposome, LipoBeDo, had high encapsulation efficiency and good stability. The nanoliposome carrier controlled the biological fate of the drugs and maintained a pre-defined optimal ratio in vivo. The LipoBeDo significantly inhibited tumor growth in 4T1 murine mammary carcinoma model compared with Doxil (P < 0.05), and completely overcame the myocardial rupture toxicity caused by Doxil in mice. Our co-loaded liposome delivery platform technology provided a new direction for the clinical treatment of triple-negative breast cancer and the safe application of DOX.
Collapse
Affiliation(s)
- Ruoshi Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingxi Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yue Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xuanming Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuyan Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xuanbo Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dun Wang
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
32
|
Luchini A, Vitiello G. Understanding the Nano-bio Interfaces: Lipid-Coatings for Inorganic Nanoparticles as Promising Strategy for Biomedical Applications. Front Chem 2019; 7:343. [PMID: 31165058 PMCID: PMC6534186 DOI: 10.3389/fchem.2019.00343] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/25/2019] [Indexed: 12/26/2022] Open
Abstract
Inorganic nanoparticles (NPs) exhibit relevant physical properties for application in biomedicine and specifically for both the diagnosis and therapy (i.e. theranostic) of severe pathologies, such as cancer. The inorganic NP core is often not stable in aqueous suspension and can induce cytotoxic effects. For this reason, over the years, several coating strategies were suggested to improve the NP stability in aqueous solutions as well as the NP biocompatibility. Among the various components which can be used for NP coatings, lipids, and in particular phospholipids emerged as versatile molecular building blocks for the production of NP coatings suitable for biomedical application. The recent synthetic efforts in NP lipid coatings allows today to introduce on the NP surface a large variety of lipid molecules eventually in mixture with amphiphilic or hydrophobic drugs or active molecules for cell targeting. In this review, the most relevant examples of NP lipid-coatings are presented and grouped in two main categories: supported lipid bilayers (SLB) and hybrid lipid bilayers (HLB). The discussed scientific cases take into account the most commonly used inorganic NP for biomedical applications in cancer therapy and diagnosis.
Collapse
Affiliation(s)
| | - Giuseppe Vitiello
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- CSGI, Center for Colloids and Surface Science, Sesto Fiorentino, Italy
| |
Collapse
|
33
|
Senapati S, Mahanta AK, Kumar S, Maiti P. Controlled drug delivery vehicles for cancer treatment and their performance. Signal Transduct Target Ther 2018; 3:7. [PMID: 29560283 PMCID: PMC5854578 DOI: 10.1038/s41392-017-0004-3] [Citation(s) in RCA: 1132] [Impact Index Per Article: 161.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/16/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Although conventional chemotherapy has been successful to some extent, the main drawbacks of chemotherapy are its poor bioavailability, high-dose requirements, adverse side effects, low therapeutic indices, development of multiple drug resistance, and non-specific targeting. The main aim in the development of drug delivery vehicles is to successfully address these delivery-related problems and carry drugs to the desired sites of therapeutic action while reducing adverse side effects. In this review, we will discuss the different types of materials used as delivery vehicles for chemotherapeutic agents and their structural characteristics that improve the therapeutic efficacy of their drugs and will describe recent scientific advances in the area of chemotherapy, emphasizing challenges in cancer treatments. Improving the delivery of cancer therapies to tumor sites is crucial to reduce unwanted side effects and patient mortality rates. Pralay Maiti and colleagues at the Indian Institute of Technology in Varanasi, India, review the latest developments in drug delivery vehicles and treatment approaches designed to enhance the effectiveness of current cancer therapies. New nanoparticle-based carriers, hydrogels and hybrid materials that offer controlled and sustained drug release are showing great promise in animal models. Furthermore, materials that respond to stimuli such as heat, light, magnetic or electric fields are also being tested to aid target-specific drug delivery and, thus, avoid damage to healthy tissues. Although there are some challenges in translating these findings to the clinic, there is no doubt that technological advances are shaping better and safer treatment options.
Collapse
Affiliation(s)
- Sudipta Senapati
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Arun Kumar Mahanta
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sunil Kumar
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Pralay Maiti
- School of Materials Science and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
34
|
Liu Y, Tamam H, Yeo Y. Mixed Liposome Approach for Ratiometric and Sequential Delivery of Paclitaxel and Gemcitabine. AAPS PharmSciTech 2018; 19:693-699. [PMID: 28971370 DOI: 10.1208/s12249-017-0877-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/08/2017] [Indexed: 12/29/2022] Open
Abstract
Paclitaxel (PTX) and gemcitabine (GEM) are often used in combination due to the synergistic anticancer effects. PTX and GEM combination showed a synergistic effect to SKOV-3 cells at a molar ratio of 1 to 1 and in PTX ➔ GEM sequence. Liposomes were explored as a carrier of PTX and GEM combination. We optimized the drug loading in liposomes varying the preparation method and co-encapsulated PTX and GEM in a single liposome preparation maintaining the maximum loading efficiency of each drug. However, drug release kinetics from the co-loaded liposomes (LpPG) was suboptimal because of the detrimental effect of PTX on GEM-release control. Instead, a mixture of LpP and LpG, which were separately optimized according to the desired release kinetics, achieved a greater cytotoxic effect than LpPG, due to the attenuation of GEM release relative to PTX. This study illustrates that co-encapsulation in a single carrier is not always desirable for the delivery of drug combinations, when the activity depends on the dosing sequence. These combinations may benefit from the mixed liposome approach, which offers greater flexibility in controlling the ratio and release kinetics of component drugs.
Collapse
|
35
|
Zheng CX, Zhao Y, Liu Y. Recent Advances in Self-assembled Nano-therapeutics. CHINESE JOURNAL OF POLYMER SCIENCE 2017. [DOI: 10.1007/s10118-018-2078-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Chen LJ, Yang CX, Yan XP. Liposome-Coated Persistent Luminescence Nanoparticles as Luminescence Trackable Drug Carrier for Chemotherapy. Anal Chem 2017; 89:6936-6939. [PMID: 28605896 DOI: 10.1021/acs.analchem.7b01397] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Near-infrared persistent luminescence nanoparticles (NIR-PLNPs) are promising imaging agents due to deep tissue penetration, high signal-to-noise ratio, and repeatedly charging ability. Here, we report liposome-coated NIR-PLNPs (Lipo-PLNPs) as a novel persistent luminescence imaging guided drug carrier for chemotherapy. The Lipo-PLNP nanocomposite shows the advantages of superior persistent luminescence and high drug loading efficiency and enables autofluorescence-free and long-term tracking of drug delivery carriers with remarkable therapeutic effect.
Collapse
Affiliation(s)
- Li-Jian Chen
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Nankai University , 94 Weijin Road, Tianjin 300071, China
| | - Cheng-Xiong Yang
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Nankai University , 94 Weijin Road, Tianjin 300071, China
| | - Xiu-Ping Yan
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Recognition and Biosensing, Nankai University , 94 Weijin Road, Tianjin 300071, China.,Collaborative Innovation Center of Chemical Science and Engineering (Tianjin) , 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
37
|
Abstract
Nanoparticle drug formulations have been extensively investigated, developed, and in some cases, approved by the Food and Drug Administration (FDA). Synergistic combinations of drugs having distinct tumor-inhibiting mechanisms and non-overlapping toxicity can circumvent the issue of treatment resistance and may be essential for effective anti-cancer therapy. At the same time, co-delivery of a combined regimen by a single nanocarrier presents a challenge due to differences in solubility, molecular weight, functional groups and encapsulation conditions between the two drugs. This review discusses cellular and microenvironment mechanisms behind treatment resistance and nanotechnology-based solutions for effective anti-cancer therapy. Co-loading or cascade delivery of multiple drugs using of polymeric nanoparticles, polymer-drug conjugates and lipid nanoparticles will be discussed along with lipid-coated drug nanoparticles developed by our lab and perspectives on combination therapy.
Collapse
Affiliation(s)
- Lei Miao
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shutao Guo
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - C Michael Lin
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
38
|
Gehrke T, Scherzad A, Ickrath P, Schendzielorz P, Hagen R, Kleinsasser N, Hackenberg S. Zinc oxide nanoparticles antagonize the effect of Cetuximab on head and neck squamous cell carcinoma in vitro. Cancer Biol Ther 2017; 18:513-518. [PMID: 28494171 DOI: 10.1080/15384047.2017.1323598] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Zinc oxide nanoparticles (ZnO-NPs) are being used in many cosmetic products and have been shown to induce tumor-selective cell death in human head and neck squamous cell carcinoma (HNSCC) in vitro. Cetuximab is a monoclonal antibody directed against the epidermal growth factor receptor (EGFR), whose effectiveness for HNSCC, alone or in combination with cytostatic drugs, has been demonstrated intensively in the last decades. Nanoparticles are known to interact with protein structures and thus may influence their functionality. The aim of the current study was to evaluate the effect of ZnO-NPs on the antitumor properties of Cetuximab in HNSCC in vitro. Two HNSCC cell lines (FaDu and HLaC-78) were treated with 0.1, 1 or 10 μM Cetuximab as well as 0, 0.1 or 1 μg/ml ZnO-NP. Qualitative assessment of ZnO-NP was conducted via transmission electron microscopy (TEM) and immunofluorescence staining. Evaluation was done via the MTT-assay after 24, 48 and 72 hours of incubation with Cetuximab and ZnO-NPs. ZnO-NPs were shown to antagonize the anti-tumor effects of Cetuximab in a time-dependent as well as dose-dependent way. These findings suggest an inhibitory interaction of ZnO-NPs with Cetuximab, which warrants further investigation.
Collapse
Affiliation(s)
- Thomas Gehrke
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| | - Agmal Scherzad
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| | - Pascal Ickrath
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| | - Philipp Schendzielorz
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| | - Rudolf Hagen
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| | - Norbert Kleinsasser
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| | - Stephan Hackenberg
- a Department of Otorhinolaryngology , Head and Neck Surgery, University Hospital Wuerzburg , Germany
| |
Collapse
|
39
|
Qu D, Liu M, Huang M, Wang L, Chen Y, Liu C, Liu Y. Octanoyl galactose ester-modified microemulsion system self-assembled by coix seed components to enhance tumor targeting and hepatoma therapy. Int J Nanomedicine 2017; 12:2045-2059. [PMID: 28352174 PMCID: PMC5358984 DOI: 10.2147/ijn.s125293] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A nanosized drug delivery platform with a combination of rational components and tumor targeting is significant for enhancement of anticancer therapy and reduction of side effects. In this study, we developed a octanoyl galactose ester-modified microemulsion system self-assembled by coix seed components (Gal(oct)-C-MEs), which improved the tumor accumulation through asialoglycoprotein receptor-mediated endocytosis and promoted the antitumor efficacy through multicomponent-mediated synergistic effect. Octanoyl galactose ester (Gal(oct)) with a yield of 82.3% was synthesized through a green enzymatic reaction and multidimensional characterization. Gal(oct)-C-MEs with a spherical shape had a small and uniform particle size (58.49±1.03 nm), narrow polydispersity index (0.09±0.01) and neutral surface charge (-5.82±0.57 mV). In the cellular uptake studies, the internalized Gal(oct)-C-ME was 2.28-fold higher relative to that of coix seed component-based microemulsions (C-MEs). The half-maximal inhibitory concentration of Gal(oct)-C-MEs against HepG2 cells was 46.5±2.4 μg/mL, which was notably higher than that of C-MEs. Importantly, the intratumor fluorescence of HepG2 xenograft-bearing nude mice treated with Cy5/Gal(oct)-C-MEs was 1.9-fold higher relative to treatment with Cy5/C-MEs. In the study of antitumor efficacy in vivo, HepG2 xenograft-bearing nude mice intragastrically administered Gal(oct)-C-MEs for 14 days exhibited the strongest inhibition of tumor growth and the lowest toxicity against liver and kidney among all the treatments. In summary, Gal(oct)-C-ME, as a highly effective and safe anticancer drug delivery system, showed promising potential for hepatoma therapy.
Collapse
Affiliation(s)
- Ding Qu
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Research Center for Multicomponent Traditional Medicine and Microecology, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Mingjian Liu
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine
| | - Mengmeng Huang
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Research Center for Multicomponent Traditional Medicine and Microecology, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Lixiang Wang
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine
| | - Yan Chen
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Research Center for Multicomponent Traditional Medicine and Microecology, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Congyan Liu
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Research Center for Multicomponent Traditional Medicine and Microecology, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Yuping Liu
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Research Center for Multicomponent Traditional Medicine and Microecology, Jiangsu Provincial Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
40
|
Bardania H, Tarvirdipour S, Dorkoosh F. Liposome-targeted delivery for highly potent drugs. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 45:1478-1489. [DOI: 10.1080/21691401.2017.1290647] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hassan Bardania
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Shabnam Tarvirdipour
- Biomedical Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Farid Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Zununi Vahed S, Salehi R, Davaran S, Sharifi S. Liposome-based drug co-delivery systems in cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:1327-1341. [DOI: 10.1016/j.msec.2016.11.073] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
|
42
|
Hassan HFH, Mansour AM, Abo-Youssef AMH, Elsadek BEM, Messiha BAS. Zinc oxide nanoparticles as a novel anticancer approach; in vitro and in vivo evidence. Clin Exp Pharmacol Physiol 2017; 44:235-243. [DOI: 10.1111/1440-1681.12681] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/23/2016] [Accepted: 10/04/2016] [Indexed: 12/23/2022]
Affiliation(s)
| | - Ahmed Mohamed Mansour
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Al-Azhar University; Cairo Egypt
| | | | - Bakheet E M Elsadek
- Department of Biochemistry; Faculty of Pharmacy; Al-Azhar University; Assuit Egypt
| | | |
Collapse
|
43
|
Shen S, Liu M, Li T, Lin S, Mo R. Recent progress in nanomedicine-based combination cancer therapy using a site-specific co-delivery strategy. Biomater Sci 2017; 5:1367-1381. [DOI: 10.1039/c7bm00297a] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review article highlights the recent progresses in nanomedicine-based combination cancer therapy via site-specific co-delivery strategies.
Collapse
Affiliation(s)
- Shiyang Shen
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Meng Liu
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Teng Li
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Shiqi Lin
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Ran Mo
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| |
Collapse
|
44
|
Chang Q, Geng R, Wang S, Qu D, Kong X. DOPA-based paclitaxel-loaded liposomes with modifications of transferrin and alendronate for bone and myeloma targeting. Drug Deliv 2016; 23:3629-3638. [PMID: 27749106 DOI: 10.1080/10717544.2016.1214989] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Treatment for multiple myeloma (MM) with a combined strategy of bone and tumor targeting remains a crucial technical challenge due to the incorporation of various functional components into one single system. Here, we developed dioleoyl phosphatidic acid (DOPA)-based paclitaxel (PTX)-loaded liposomes with modifications of alendronate and transferrin (Ald-/Tf-modified PTX-L), which were capable of bone affinity mediated by phosphate groups in DOPA and alendronate, and tumor targeting offered by transferrin. Ald-/Tf-modified PTX-L had clear and well-defined spherical shape with an intermediated size of 118.8 ± 4.8 nm, a highly negative surface charge of -46.9 ± 6.8 mV and a drug entrapment efficiency (DEE) of approximately 80%. When the pH was changed from pH 7.4 to pH 6.5, the accumulative release of PTX from Ald-/Tf-modified PTX-L significantly increased from 26.7 ± 3.7% to 41.7 ± 4.9%. Importantly, liposomes based on DOPA displayed an obviously stronger affinity with hydroxyapatite (HAp) than 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)-based liposomes. Compared to PTX-L, Ald-/Tf-modified PTX-L exhibited obvious improvement of cytotoxicity (IC50 = 1.25 ± 0.09 μg/mL), significant enhancement on PTX intracellular accumulation (16.58 ± 0.62 μg/mg) and notable promotion to apoptosis induction (45.21 ± 3.10%) toward myeloma (MM1s) cells. In this study of antitumor efficacy, Ald-/Tf-modified PTX-L with bone-specific targeting showed a significant effect on extending the median survival time (48 days) and terminal survival time (> 58 days) against the MM1S-injected nude mice among all formulations. The results suggested that Ald-/Tf-modified PTX-L had potential as an efficient anticancer drug delivery system for MM therapy.
Collapse
Affiliation(s)
- Qing Chang
- a Department of Orthopaedics , School of Medicine, Zhongda Hospital, Southeast University , Nanjing , P.R. China
| | - Rui Geng
- a Department of Orthopaedics , School of Medicine, Zhongda Hospital, Southeast University , Nanjing , P.R. China
| | - Shanzheng Wang
- a Department of Orthopaedics , School of Medicine, Zhongda Hospital, Southeast University , Nanjing , P.R. China
| | - Ding Qu
- b Jiangsu Province Academy of Traditional Chinese Medicine , Nanjing , P.R. China , and
| | - Xiangfei Kong
- a Department of Orthopaedics , School of Medicine, Zhongda Hospital, Southeast University , Nanjing , P.R. China.,c Surgical Research Center, School of Medicine, Southeast University , Nanjing , P.R. China
| |
Collapse
|
45
|
Hu Q, Sun W, Wang C, Gu Z. Recent advances of cocktail chemotherapy by combination drug delivery systems. Adv Drug Deliv Rev 2016; 98:19-34. [PMID: 26546751 PMCID: PMC4998845 DOI: 10.1016/j.addr.2015.10.022] [Citation(s) in RCA: 450] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/05/2015] [Accepted: 10/27/2015] [Indexed: 12/22/2022]
Abstract
Combination chemotherapy is widely exploited for enhanced cancer treatment in the clinic. However, the traditional cocktail administration of combination regimens often suffers from varying pharmacokinetics among different drugs. The emergence of nanotechnology offers an unparalleled opportunity for developing advanced combination drug delivery strategies with the ability to encapsulate various drugs simultaneously and unify the pharmacokinetics of each drug. This review surveys the most recent advances in combination delivery of multiple small molecule chemotherapeutics using nanocarriers. The mechanisms underlying combination chemotherapy, including the synergistic, additive and potentiation effects, are also discussed with typical examples. We further highlight the sequential and site-specific co-delivery strategies, which provide new guidelines for development of programmable combination drug delivery systems. Clinical outlook and challenges are also discussed in the end.
Collapse
Affiliation(s)
- Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wujin Sun
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA; Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
46
|
Co-delivery of chemotherapeutics and proteins for synergistic therapy. Adv Drug Deliv Rev 2016; 98:64-76. [PMID: 26546464 DOI: 10.1016/j.addr.2015.10.021] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 01/15/2023]
Abstract
Combination therapy with chemotherapeutics and protein therapeutics, typically cytokines and antibodies, has been a type of crucial approaches for synergistic cancer treatment. However, conventional approaches by simultaneous administration of free chemotherapeutic drugs and proteins lead to limitations for further optimizing the synergistic effects, due to the distinct in vivo pharmacokinetics and distribution of small drugs and proteins, insufficient tumor selectivity and tumor accumulation, unpredictable drug/protein ratios at tumor sites, short half-lives, and serious systemic adverse effects. Consequently, to obtain optimal synergistic anti-tumor efficacy, considerable efforts have been devoted to develop the co-delivery systems for co-incorporating chemotherapeutics and proteins into a single carrier system and subsequently releasing the dual or multiple payloads at desired target sites in a more controllable manner. The co-delivery systems result in markedly enhanced blood stability and in vivo half-lives of the small drugs and proteins, elevated tumor accumulation, as well as the capability of delivering the multiple agents to the same target sites with rational drug/protein ratios, which may facilitate maximizing the synergistic effects and therefore lead to optimal antitumor efficacy. This review emphasizes the recent advances in the co-delivery systems for chemotherapeutics and proteins, typically cytokines and antibodies, for systemic or localized synergistic cancer treatment. Moreover, the proposed mechanisms responsible for the synergy of chemotherapeutic drugs and proteins are discussed.
Collapse
|
47
|
Teo PY, Cheng W, Hedrick JL, Yang YY. Co-delivery of drugs and plasmid DNA for cancer therapy. Adv Drug Deliv Rev 2016; 98:41-63. [PMID: 26529199 DOI: 10.1016/j.addr.2015.10.014] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/21/2015] [Accepted: 10/23/2015] [Indexed: 12/12/2022]
Abstract
Cancer is an extremely complex disease involving multiple signaling pathways that enable tumor cells to evade programmed cell death, thus making cancer treatment extremely challenging. The use of combination therapy involving both gene therapy and chemotherapy has resulted in enhanced anti-cancer effects and has become an increasingly important strategy in medicine. This review will cover important design parameters that are incorporated into delivery systems for the co-administration of drug and plasmid-based nucleic acids (pDNA and shRNA), with particular emphasis on polymers as delivery materials. The unique challenges faced by co-delivery systems and the strategies to overcome such barriers will be discussed. In addition, the advantages and disadvantages of combination therapy using separate carrier systems versus the use of a single carrier will be evaluated. Finally, future perspectives in the design of novel platforms for the combined delivery of drugs and genes will be presented.
Collapse
|
48
|
Song Y, Xie Y, Yang J, Li R, Jin X, Yang J. A poly(ascorbyl acrylate)-containing nanoplatform with anticancer activity and the sequential combination therapy with its loaded paclitaxel. J Mater Chem B 2016; 4:6588-6596. [DOI: 10.1039/c6tb01818a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The complex nanocarriers combined with the loaded therapeutic agents to achieve synergistic tumor inhibition.
Collapse
Affiliation(s)
- Yufeng Song
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Yanqi Xie
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Junjiao Yang
- College of Science
- Beijing University of Chemical Technology
- Beijing 100029
- China
| | - Ruiqiong Li
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| | - Xu Jin
- Department of Anesthesiology and Pain Therapy
- Beijing Tiantan Hospital Affiliated to Capital Medical University
- Beijing 100050
- China
| | - Jing Yang
- State Key Laboratory of Chemical Resource
- Beijing Key Laboratory of Bioprocess
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
| |
Collapse
|
49
|
He Q, Guo S, Qian Z, Chen X. Development of individualized anti-metastasis strategies by engineering nanomedicines. Chem Soc Rev 2015; 44:6258-6286. [PMID: 26056688 PMCID: PMC4540626 DOI: 10.1039/c4cs00511b] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Metastasis is deadly and also tough to treat as it is much more complicated than the primary tumour. Anti-metastasis approaches available so far are far from being optimal. A variety of nanomedicine formulae provide a plethora of opportunities for developing new strategies and means for tackling metastasis. It should be noted that individualized anti-metastatic nanomedicines are different from common anti-cancer nanomedicines as they specifically target different populations of malignant cells. This review briefly introduces the features of the metastatic cascade, and proposes a series of nanomedicine-based anti-metastasis strategies aiming to block each metastatic step. Moreover, we also concisely introduce the advantages of several promising nanoparticle platforms and their potential for constructing state-of-the-art individualized anti-metastatic nanomedicines.
Collapse
Affiliation(s)
- Qianjun He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
50
|
Assanhou AG, Li W, Zhang L, Xue L, Kong L, Sun H, Mo R, Zhang C. Reversal of multidrug resistance by co-delivery of paclitaxel and lonidamine using a TPGS and hyaluronic acid dual-functionalized liposome for cancer treatment. Biomaterials 2015; 73:284-95. [PMID: 26426537 DOI: 10.1016/j.biomaterials.2015.09.022] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 09/11/2015] [Accepted: 09/14/2015] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) remains the primary issue in cancer therapy, which is characterized by the overexpressed P-glycoprotein (P-gp)-included efflux pump or the upregulated anti-apoptotic proteins. In this study, a D-alpha-tocopheryl poly (ethylene glycol 1000) succinate (TPGS) and hyaluronic acid (HA) dual-functionalized cationic liposome containing a synthetic cationic lipid, 1,5-dioctadecyl-N-histidyl-L-glutamate (HG2C18) was developed for co-delivery of a small-molecule chemotherapeutic drug, paclitaxel (PTX) with a chemosensitizing agent, lonidamine (LND) to treat the MDR cancer. It was demonstrated that the HG2C18 lipid contributes to the endo-lysosomal escape of the liposome following internalization for efficient intracellular delivery. The TPGS component was confirmed able to elevate the intracellular accumulation of PTX by inhibiting the P-gp efflux, and to facilitate the mitochondrial-targeting of the liposome. The intracellularly released LND suppressed the intracellular ATP production by interfering with the mitochondrial function for enhanced P-gp inhibition, and additionally, sensitized the MDR breast cancer (MCF-7/MDR) cells to PTX for promoted induction of apoptosis through a synergistic effect. Functionalized with the outer HA shell, the liposome preferentially accumulated at the tumor site and showed a superior antitumor efficacy in the xenograft MCF-7/MDR tumor mice models. These findings suggest that this dual-functional liposome for co-delivery of a cytotoxic drug and an MDR modulator provides a promising strategy for reversal of MDR in cancer treatment.
Collapse
Affiliation(s)
- Assogba G Assanhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China; UFR Pharmacie, Faculté des Sciences de la Santé, Université d'Abomey-Calavi, 01 BP 188 Cotonou, Benin; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Wenyuan Li
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Lei Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Hongbin Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|