1
|
Tanaka T, Sano K, Kawakami R, Tanaka S, Munekane M, Yamasaki T, Mukai T. Electrostatically self-assembled gold nanorods with sulfated hyaluronic acid for targeted photothermal therapy for CD44-positive tumors. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102781. [PMID: 39163902 DOI: 10.1016/j.nano.2024.102781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/27/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Gold nanorods (GNR) produce heat upon irradiation with near-infrared light, enabling a tumor-targeted photothermal therapy. In this study, we prepared GNR coated with sulfated hyaluronic acid (sHA) with a binding affinity for CD44 via electrostatic interactions to deliver GNR to tumors efficiently and stably, and evaluated their usefulness for photothermal therapy. Cationic GNR modified with trimethylammonium groups electrostatically interacted with native HA or sHA with varying degrees of sulfation to form complexes. While GNR/HA was unstable in saline, GNR/sHA maintained the absorbance peak in the near-infrared region, particularly for GNR/sHA with higher degrees of sulfation. GNR/sHA exhibited an intense photothermal effect upon irradiation with near-infrared light. Furthermore, in vitro and in vivo studies revealed that GNR coated with sHA containing approximately 1.2 sulfated groups per HA unit could accumulate in CD44-positive tumors via an HA-specific pathway. These findings indicate the effectiveness of GNR/sHA as a tumor-targeted photothermal therapeutic agent.
Collapse
Affiliation(s)
- Toshie Tanaka
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Rin Kawakami
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Shiho Tanaka
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan.
| |
Collapse
|
2
|
Gholami A, Mohkam M, Soleimanian S, Sadraeian M, Lauto A. Bacterial nanotechnology as a paradigm in targeted cancer therapeutic delivery and immunotherapy. MICROSYSTEMS & NANOENGINEERING 2024; 10:113. [PMID: 39166136 PMCID: PMC11333603 DOI: 10.1038/s41378-024-00743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/02/2024] [Accepted: 06/23/2024] [Indexed: 08/22/2024]
Abstract
Cancer, a multifaceted and diverse ailment, presents formidable obstacles to traditional treatment modalities. Nanotechnology presents novel prospects for surmounting these challenges through its capacity to facilitate meticulous and regulated administration of therapeutic agents to malignant cells while concurrently modulating the immune system to combat neoplasms. Bacteria and their derivatives have emerged as highly versatile and multifunctional platforms for cancer nanotherapy within the realm of nanomaterials. This comprehensive review delves into the multifaceted and groundbreaking implementations of bacterial nanotechnology within cancer therapy. This review encompasses four primary facets: the utilization of bacteria as living conveyors of medicinal substances, the employment of bacterial components as agents that stimulate the immune system, the deployment of bacterial vectors as tools for delivering genetic material, and the development of bacteria-derived nano-drugs as intelligent nano-medications. Furthermore, we elucidate the merits and modalities of operation pertaining to these bacterial nano-systems, along with their capacity to synergize with other cutting-edge nanotechnologies, such as CRISPR-Cas systems. Additionally, we offer insightful viewpoints regarding the forthcoming trajectories and prospects within this expanding domain. It is our deduction that bacterial nanotechnology embodies a propitious and innovative paradigm in the realm of cancer therapy, which has the potential to provide numerous advantages and synergistic effects in enhancing the outcomes and quality of life for individuals afflicted with cancer.
Collapse
Affiliation(s)
- Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Mohkam
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Sadraeian
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Antonio Lauto
- School of Science, University of Western Sydney, Campbelltown, NSW 2560 Australia
- School of Medicine, University of Western Sydney, Campbelltown, NSW 2560 Australia
| |
Collapse
|
3
|
Bokatyi AN, Dubashynskaya NV, Skorik YA. Chemical modification of hyaluronic acid as a strategy for the development of advanced drug delivery systems. Carbohydr Polym 2024; 337:122145. [PMID: 38710553 DOI: 10.1016/j.carbpol.2024.122145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024]
Abstract
Hyaluronic acid (HA) has emerged as a promising biopolymer for various biomedical applications due to its biocompatibility, biodegradability, and intrinsic ability to interact with cell surface receptors, making it an attractive candidate for drug delivery systems and tissue engineering. Chemical modification of HA has opened up versatile possibilities to tailor its properties, enabling the development of advanced drug delivery systems and biomaterials with enhanced functionalities and targeted applications. This review analyzes the strategies and applications of chemically modified HA in the field of drug delivery and biomaterial development. The first part of the review focuses on the different methods and functional groups used for the chemical modification of HA, highlighting the impact of these modifications on its physicochemical properties, degradation behavior and interactions with drugs. The second part of the review evaluates the use of chemically modified HA in the development of advanced biomedical materials including nano- and microparticles, hydrogels and mucoadhesive materials with tailored drug release profiles, site-specific targeting and stimuli-responsive behavior. Thus, the review consolidates the current advances and future perspectives in the field of chemical modification of HA, underscoring its immense potential to drive the development of advanced drug delivery systems and biomaterials with diverse biomedical applications.
Collapse
Affiliation(s)
- Anton N Bokatyi
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, St. Petersburg 199004, Russian Federation
| | - Natallia V Dubashynskaya
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, St. Petersburg 199004, Russian Federation
| | - Yury A Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, St. Petersburg 199004, Russian Federation.
| |
Collapse
|
4
|
Yetisgin AA, Durak S, Kutlu O, Cetinel S. Hyaluronan-Sphingosine Polymersomes for Treatment of Ocular Neovascularization: Synthesis and Evaluation. Macromol Biosci 2024; 24:e2300531. [PMID: 38318988 DOI: 10.1002/mabi.202300531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/21/2024] [Indexed: 02/07/2024]
Abstract
Ocular neovascularization is a hallmark of several sight-threatening diseases, including diabetic retinopathy and age-related macular degeneration. Currently, available treatments are limited and often associated with side effects. Therefore, a novel approach to ocular neovascularization treatment through utilization of polymersomes from self-assembled sphingosine-grafted hyaluronic acid (HA-Sph) amphiphilic polymers is presented. The polymersomes are generated in spherical morphologies and sizes between 97.95 - 161.9 nm with homogenous size distributions. Experiments reveal that HA-Sph polymersomes, with concentrations ≥150 µg mL-1, significantly inhibit the proliferation of human umbilical vein endothelial cells (HUVECs), while concurrently promoting the proliferation of retinal pigment epithelial cells. The polymersomes demonstrate gradual disintegration in vitro, leading to sustained release of sphingosine, which prolongs the inhibition of HUVEC proliferation (from 87.5% at 24 h to 35.2% viability at 96 h). The efficacy of polymersomes in inhibiting angiogenesis is confirmed through tube formation assay, revealing a substantial reduction in tube length compared to the control group. The findings also validate the ocular penetration capability of polymersomes through ex vivo whole porcine eye ocular penetration study, indicating their suitability for topical administration. Potentially, HA-Sph polymersomes can be harnessed to develop intricate drug delivery systems that protect the retina and effectively treat ocular diseases.
Collapse
Affiliation(s)
- Abuzer Alp Yetisgin
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, 34956, Turkey
- Faculty of Engineering and Natural Sciences, Materials Science and Nano-Engineering Program, Sabanci University, Istanbul, 34956, Turkey
| | - Saliha Durak
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, 34956, Turkey
| | - Ozlem Kutlu
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, 34956, Turkey
| | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, 34956, Turkey
| |
Collapse
|
5
|
Saroj S, Us P, Patil S, Paul D, Saha S, Ali A, Pal S, Lochab B, Rakshit T. Herb Extracellular Vesicle-Chitosan-PEGylated Graphene Oxide Conjugate Delivers Estrogen Receptor α Targeting siRNA to Breast Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:2741-2751. [PMID: 38630629 DOI: 10.1021/acsabm.3c01108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Herb-based extracellular vesicles (EV), inherently replete with bioactive proteins, RNA, lipids, and other medicinal compounds, are noncytotoxic and uniquely capable of cellular delivery to meet the ever-stringent challenges of ongoing clinical applications. EVs are abundant in nature, affordable, and scalable, but they are also incredibly fragile and stuffed with many biomolecules. To address the low drug binding abilities and poor stability of EVs, we demonstrated herb-based EVs (isolated from neem, mint, and curry leaves) conjugated with chitosan (CS) and PEGylated graphene oxide (GP) that led to their transformation into robust and efficient vectors. The designed conjugates successfully delivered estrogen receptor α (ERα1)-targeting siRNA to breast cancer MCF7 cells. Our data revealed that neem-based EV-CS-GP conjugates were most efficient in cellular siRNA delivery, which could be attributed to hyaluronic acid-mediated recognition of neem EVs by MCF7 cells via CD44 receptors. Our approach shows a futuristic direction in designing clinically viable, sustainable, nontoxic EV-based vehicles that can deliver a variety of functional siRNA cargos.
Collapse
Affiliation(s)
- Saroj Saroj
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi-NCR, Uttar Pradesh 201314, India
| | - Poornima Us
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi-NCR, Uttar Pradesh 201314, India
| | - Sachin Patil
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi-NCR, Uttar Pradesh 201314, India
| | - Debashish Paul
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi-NCR, Uttar Pradesh 201314, India
| | - Sunita Saha
- Department of Chemistry, IIT Bhilai, Durg, Chhattisgarh 491001, India
| | - Akbar Ali
- Department of Chemistry, IIT Bhilai, Durg, Chhattisgarh 491001, India
| | - Suchetan Pal
- Department of Chemistry, IIT Bhilai, Durg, Chhattisgarh 491001, India
- Department of Bioscience and Biomedical Engineering, IIT Bhilai, Durg 491001, India
| | - Bimlesh Lochab
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi-NCR, Uttar Pradesh 201314, India
| | - Tatini Rakshit
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi-NCR, Uttar Pradesh 201314, India
| |
Collapse
|
6
|
Mahdieh A, Motasadizadeh H, Maghsoudian S, Sabzevari A, Khalili F, Yeganeh H, Nyström B. Novel polyurethane-based ionene nanoparticles electrostatically stabilized with hyaluronic acid for effective gene therapy. Colloids Surf B Biointerfaces 2024; 236:113802. [PMID: 38382225 DOI: 10.1016/j.colsurfb.2024.113802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
Gene therapy is considered to be a valuable strategy for effective cancer treatment. However, the development of effective delivery systems that can specifically deliver gene materials, such as siRNA to tumor tissues plays a critical role in cancer therapy. In the present study, we have developed a novel complex that is based on an electrostatic interaction between cationic polyurethane ionene (CPUI) nanoparticles and an anti-signal transducer and activator of transcription 3 (STAT3) siRNA. For active targeting, hyaluronic acid (HA) was used to coat the complexes, which significantly reduced the cytotoxicity of the blank nanocarriers while demonstrating high transport efficiency of the siRNA via the CD44-mediated endocytosis pathway in MCF-7 breast cancer cells. The targeted nanocarriers (HA/CPUI/siRNA) showed significantly higher cellular internalization in flow cytometry and confocal microscopy compared with the non-targeted system (CPUI/siRNA). In addition, the incorporation of HA on the surface of the complexes resulted in significantly greater suppression of the STAT3 gene compared to the corresponding non-targeted formulation. Whole-body fluorescence images showed more significant tumor accumulation of the targeted nanocarriers in 4T1 breast tumor-bearing mice. Therefore, HA/CPUI/siRNA nanocarriers are an interesting option for the siRNA-targeted treatment of breast cancer cells.
Collapse
Affiliation(s)
- Athar Mahdieh
- Department of Pharmacy, Section for Pharmaceutics and Social Pharmacy, University of Oslo, Oslo, Norway
| | - Hamidreza Motasadizadeh
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Samane Maghsoudian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Sabzevari
- Polymer Faculty, Biomedical Engineering Department, Meybod University, Meybod, Yazd, Iran; Polymer Engineering Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fereshte Khalili
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Yeganeh
- Iran Polymer and Petrochemical Institute, Tehran, Iran
| | - Bo Nyström
- Department of Chemistry, University of Oslo, Oslo, Norway.
| |
Collapse
|
7
|
Abd-Rabou AA, Abdelaziz AM, Shaker OG, Ayeldeen G. Hyaluronated nanoparticles deliver raloxifene to CD44-expressed colon cancer cells and regulate lncRNAs/miRNAs epigenetic cascade. Cancer Nanotechnol 2023; 14:32. [DOI: 10.1186/s12645-023-00183-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/27/2023] [Indexed: 09/02/2023] Open
Abstract
Abstract
Background
Colorectal malignant cells (CRC) are one of the world’s main causes of cancer mortality and morbidity. Notwithstanding the plenty of anti-CRC therapeutics, its prognosis remains not selective owing to cancer resistance to these therapeutics. Raloxifene (RX), a medication firstly used to treat osteoporosis, was recently licenced for the prevention of CRC. Unfortunately, due to medication resistance, many RX-based therapies are likely to become ineffective. Recently, we identified a novel method of administration to lengthen the half-life of RX by mixing it with chitosan (CS) and hyaluronic acid (HA). Thus, the rationale of the current study was to investigate how colon cancer cells were affected by RX-HA-CS nanoparticles (RX NPs) in terms of targetability, cytotoxicity, and epigenetic cascade alteration.
Results
RX NP had an entrapment efficiency (EE%) of 90.0 ± 8.12%. Compared to HCT 116 cells, Caco-2 cells were more susceptible to the cytotoxic effects of RX and its NP as well as they had a higher binding affinity to CD44 receptors compared to normal WI-38 cells. In comparison to the free RX, the RX NP’s cytotoxic fold changes in HCT 116 and Caco-2 cells were 2.16 and 2.52, respectively. Furthermore, the epigenetic cascade of some noncoding RNAs was examined. Moreover, particular protein concentrations were investigated in all tested cells after application of the proposed therapies. Our results showed that the RX NP recorded higher remarkable cytotoxic impact on CRC cells compared to the free RX. Intriguingly, it was hypothesized that RX nanoparticles attacked colon cancerous cells by up-regulating miR-944 and E-cadherin (ECN) expressions, while down-regulating the expressions of PPARγ, YKL-40, VEGF, H-19, LINC00641, HULC, HOTTIP, miR-92a, miR-200, and miR-21.
Conclusions
We may conclude that the RX NP effectively targets CRC cells in vitro via altering lncRNAs and miRNAs epigenetic cascade as well as cellular uptake through CD44-expressed CRC cells.
Collapse
|
8
|
Myint SS, Laomeephol C, Thamnium S, Chamni S, Luckanagul JA. Hyaluronic Acid Nanogels: A Promising Platform for Therapeutic and Theranostic Applications. Pharmaceutics 2023; 15:2671. [PMID: 38140012 PMCID: PMC10747897 DOI: 10.3390/pharmaceutics15122671] [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: 11/02/2023] [Revised: 11/18/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Hyaluronic acid (HA) nanogels are a versatile class of nanomaterials with specific properties, such as biocompatibility, hygroscopicity, and biodegradability. HA nanogels exhibit excellent colloidal stability and high encapsulation capacity, making them promising tools for a wide range of biomedical applications. HA nanogels can be fabricated using various methods, including polyelectrolyte complexation, self-assembly, and chemical crosslinking. The fabrication parameters can be tailored to control the physicochemical properties of HA nanogels, such as size, shape, surface charge, and porosity, enabling the rational design of HA nanogels for specific applications. Stimulus-responsive nanogels are a type of HA nanogels that can respond to external stimuli, such as pH, temperature, enzyme, and redox potential. This property allows the controlled release of encapsulated therapeutic agents in response to specific physiological conditions. HA nanogels can be engineered to encapsulate a variety of therapeutic agents, such as conventional drugs, genes, and proteins. They can then be delivered to target tissues with high efficiency. HA nanogels are still under development, but they have the potential to become powerful tools for a wide range of theranostic or solely therapeutic applications, including anticancer therapy, gene therapy, drug delivery, and bioimaging.
Collapse
Affiliation(s)
- Su Sundee Myint
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.M.); (S.C.)
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Chavee Laomeephol
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Biomaterial Engineering in Medical and Health, Chulalongkorn University, Bangkok 10330, Thailand
| | - Sirikool Thamnium
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.M.); (S.C.)
- Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok 10330, Thailand
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence in Biomaterial Engineering in Medical and Health, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
9
|
Hogan KJ, Perez MR, Mikos AG. Extracellular matrix component-derived nanoparticles for drug delivery and tissue engineering. J Control Release 2023; 360:888-912. [PMID: 37482344 DOI: 10.1016/j.jconrel.2023.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/16/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The extracellular matrix (ECM) consists of a complex combination of proteins, proteoglycans, and other biomolecules. ECM-based materials have been demonstrated to have high biocompatibility and bioactivity, which may be harnessed for drug delivery and tissue engineering applications. Herein, nanoparticles incorporating ECM-based materials and their applications in drug delivery and tissue engineering are reviewed. Proteins such as gelatin, collagen, and fibrin as well as glycosaminoglycans including hyaluronic acid, chondroitin sulfate, and heparin have been employed for cancer therapeutic delivery, gene delivery, and wound healing and regenerative medicine. Strategies for modifying and functionalizing these materials with synthetic and natural polymers or to enable stimuli-responsive degradation and drug release have increased the efficacy of these materials and nano-systems. The incorporation and modification of ECM-based materials may be used to drive drug targeting and increase tissue-specific cell differentiation more effectively.
Collapse
Affiliation(s)
- Katie J Hogan
- Department of Bioengineering, Rice University, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Marissa R Perez
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
10
|
Quílez-Alburquerque J, Saad MA, Descalzo AB, Orellana G, Hasan T. Hyaluronic acid-poly(lactic-co-glycolic acid) nanoparticles with a ruthenium photosensitizer cargo for photokilling of oral cancer cells. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2022.114349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
11
|
Ramasundaram S, Saravanakumar G, Sobha S, Oh TH. Dextran Sulfate Nanocarriers: Design, Strategies and Biomedical Applications. Int J Mol Sci 2022; 24:ijms24010355. [PMID: 36613798 PMCID: PMC9820219 DOI: 10.3390/ijms24010355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Dextran sulfate (DXS) is a hydrophilic, non-toxic, biodegradable, biocompatible and safe biopolymer. These biomedically relevant characteristics make DXS a promising building block in the development of nanocarrier systems for several biomedical applications, including imaging and drug delivery. DXS polyanion can bind with metal oxide nanomaterials, biological receptors and therapeutic drug molecules. By taking advantage of these intriguing properties, DXS is used to functionalize or construct nanocarriers for specific applications. In particular, the diagnostic or therapeutic active agent-loaded DXS nanoparticles are prepared by simple coating, formation of polyelectrolyte complexes with other positively charged polymers or through self-assembly of amphiphilic DXS derivatives. These nanoparticles show a potential to localize the active agents at the pathological site and minimize undesired side effects. As DXS can recognize and be taken up by macrophage surface receptors, it is also used as a targeting ligand for drug delivery. Besides as a nanocarrier scaffold material, DXS has intrinsic therapeutic potential. DXS binds to thrombin, acts as an anticoagulant and exhibits an inhibitory effect against coagulation, retrovirus, scrapie virus and human immunodeficiency virus (HIV). Herein, biomedical applications involving the use of DXS as nanocarriers for drugs, biomolecules, and imaging agents have been reviewed. A special focus has been made on strategies used for loading and delivering of drugs and biomolecules meant for treating several diseases, including cancer, inflammatory diseases and ocular disease.
Collapse
Affiliation(s)
| | | | | | - Tae Hwan Oh
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38436, Republic of Korea
- Correspondence:
| |
Collapse
|
12
|
Liwinska W, Waleka-Bagiel E, Stojek Z, Karbarz M, Zabost E. Enzyme-triggered- and tumor-targeted delivery with tunable, methacrylated poly(ethylene glycols) and hyaluronic acid hybrid nanogels. Drug Deliv 2022; 29:2561-2578. [PMID: 35938558 PMCID: PMC9477489 DOI: 10.1080/10717544.2022.2105443] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/03/2022] Open
Abstract
Enzyme-responsive polymeric-based nanostructures are potential candidates for serving as key materials in targeted drug delivery carriers. However, the major risk in their prolonged application is fast disassembling of the short-lived polymeric-based structures. Another disadvantage is the limited accessibility of the enzyme to the moieties that are located inside the network. Here, we report on a modified environmentally responsive and enzymatically cleavable nanogel carrier that contains a hybrid network. A properly adjusted volume phase transition (VPT) temperature allowed independent shrinking of a) poly(ethylene glycol) methyl ether methacrylate (OEGMA) with di(ethylene glycol) and b) methyl ether methacrylate (MEO2MA) part of the network, and the exposition of hyaluronic acid methacrylate (MeHa) network based carboxylic groups for its targeted action with the cellular based receptors. This effect was substantial after raising temperature in typical hyperthermia-based treatment therapies. Additionally, novel tunable NGs gained an opportunity to store- and to efficient-enzyme-triggered release relatively low but highly therapeutic doses of doxorubicin (DOX) and mitoxantrone (MTX). The controlled enzymatic degradation of NGs could be enhanced by introducing more hyaluronidase enzyme (HAdase), that is usually overexpressed in cancer environments. MTT assay results revealed effective cytotoxic activity of the NGs against the human MCF-7 breast cancer cells, the A278 ovarian cancer cells and also cytocompatibility against the MCF-10A and HOF healthy cells. The obtained tunable, hybrid network NGs might be used as a useful platform for programmed delivery of other pharmaceuticals and diagnostics in therapeutic applications.
Collapse
Affiliation(s)
- Wioletta Liwinska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, PL, Poland
| | - Ewelina Waleka-Bagiel
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, PL, Poland
- Faculty of Chemistry, Warsaw University of Technology, Warsaw, PL, Poland
| | - Zbigniew Stojek
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, PL, Poland
| | - Marcin Karbarz
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, PL, Poland
| | - Ewelina Zabost
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, PL, Poland
| |
Collapse
|
13
|
Zhu L, Zhao Y, Liu T, Chen M, Qian WP, Jiang B, Barwick BG, Zhang L, Styblo TM, Li X, Yang L. Inhibition of NADPH Oxidase-ROS Signal using Hyaluronic Acid Nanoparticles for Overcoming Radioresistance in Cancer Therapy. ACS NANO 2022; 16:18708-18728. [PMID: 36256454 PMCID: PMC9764083 DOI: 10.1021/acsnano.2c07440] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Upregulation of NADPH oxidases (NOXs) in cancer cells leads to chronic increase in intracellular reactive oxygen species (ROS) and adaptation to a high ROS level for cell survival and, thereby, low sensitivity to radiotherapy. To overcome resistance to radiotherapy, we have developed a bioactive and CD44 targeted hyaluronic acid nanoparticle encapsulated with an NOX inhibitor, GKT831 (HANP/GKT831). We found that HANP/GKT831 had stronger inhibitory effects on ROS generation and cell proliferation than that of GKT831 alone in cancer cells. Systemic delivery of HANP/GKT831 led to the targeted accumulation in breast cancer patient derived xenograft (PDX) tumors in nude mice. Importantly, the combination of systemic delivery of HANP/GKT831 with a low dose of local radiotherapy significantly enhanced tumor growth inhibition in breast cancer PDX models. Our results showed that HANP/GKT831 primed tumor cells to radiation-induced DNA damage and cell death by downregulation of DNA repair function and oncogenic signal pathways.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Yi Zhao
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Tongrui Liu
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Minglong Chen
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Wei Ping Qian
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Binghua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia 19107, Pennsylvania, United States
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Lumeng Zhang
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Toncred M Styblo
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| | - Lily Yang
- Department of Surgery and Winship Cancer Institute, Emory University School of Medicine, Atlanta 30322, Georgia, United States
| |
Collapse
|
14
|
Iannazzo D, Celesti C, Espro C, Ferlazzo A, Giofrè SV, Scuderi M, Scalese S, Gabriele B, Mancuso R, Ziccarelli I, Visalli G, Di Pietro A. Orange-Peel-Derived Nanobiochar for Targeted Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14102249. [PMID: 36297682 PMCID: PMC9607014 DOI: 10.3390/pharmaceutics14102249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer-targeted drug delivery systems (DDS) based on carbon nanostructures have shown great promise in cancer therapy due to their ability to selectively recognize specific receptors overexpressed in cancer cells. In this paper, we have explored a green route to synthesize nanobiochar (NBC) endowed with graphene structure from the hydrothermal carbonization (HTC) of orange peels and evaluated the suitability of this nanomaterial as a nanoplatform for cancer therapy. In order to compare the cancer-targeting ability of different widely used targeting ligands (TL), we have conjugated NBC with biotin, riboflavin, folic acid and hyaluronic acid and have tested, in vitro, their biocompatibility and uptake ability towards a human alveolar cancer cell line (A549 cells). The nanosystems which showed the best biological performances-namely, the biotin- and riboflavin- conjugated systems-have been loaded with the poorly water-soluble drug DHF (5,5-dimethyl-6a-phenyl-3-(trimethylsilyl)-6,6a-dihydrofuro[3,2-b]furan-2(5H)-one) and tested for their anticancer activity. The in vitro biological tests demonstrated the ability of both systems to internalize the drug in A549 cells. In particular, the biotin-functionalized NBC caused cell death percentages to more than double with respect to the drug alone. The reported results also highlight the positive effect of the presence of oxygen-containing functional groups, present on the NBC surface, to improve the water dispersion stability of the DDS and thus make the approach of using this nanomaterial as nanocarrier for poorly water-soluble drugs effective.
Collapse
Affiliation(s)
- Daniela Iannazzo
- Department of Engineering, University of Messina, Contrada Di Dio, 98166 Messina, Italy
- Correspondence: (D.I.); (C.C.)
| | - Consuelo Celesti
- Department of Engineering, University of Messina, Contrada Di Dio, 98166 Messina, Italy
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
- Correspondence: (D.I.); (C.C.)
| | - Claudia Espro
- Department of Engineering, University of Messina, Contrada Di Dio, 98166 Messina, Italy
| | - Angelo Ferlazzo
- Department of Engineering, University of Messina, Contrada Di Dio, 98166 Messina, Italy
| | - Salvatore V. Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres, 98166 Messina, Italy
| | - Mario Scuderi
- Institute for Microelectronics and Microsystems, National Research Council (CNR-IMM), Ottava Strada n.5, 95121 Catania, Italy
| | - Silvia Scalese
- Institute for Microelectronics and Microsystems, National Research Council (CNR-IMM), Ottava Strada n.5, 95121 Catania, Italy
| | - Bartolo Gabriele
- Laboratory of Industrial and Synthetic Organic Chemistry (LISOC), Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 12/C, 87036 Arcavacata di Rende, Italy
| | - Raffaella Mancuso
- Laboratory of Industrial and Synthetic Organic Chemistry (LISOC), Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 12/C, 87036 Arcavacata di Rende, Italy
| | - Ida Ziccarelli
- Laboratory of Industrial and Synthetic Organic Chemistry (LISOC), Department of Chemistry and Chemical Technologies, University of Calabria, Via Pietro Bucci 12/C, 87036 Arcavacata di Rende, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphological and Functional Images, University Hospital of Messina, Via Consolare Valeria, 1, 98100 Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphological and Functional Images, University Hospital of Messina, Via Consolare Valeria, 1, 98100 Messina, Italy
| |
Collapse
|
15
|
Zhang W, Zhang Z, Lou S, Chang Z, Wen B, Zhang T. Hyaluronic Acid–Stabilized Fe3O4 Nanoparticles for Promoting In Vivo Magnetic Resonance Imaging of Tumors. Front Pharmacol 2022; 13:918819. [PMID: 35910362 PMCID: PMC9337838 DOI: 10.3389/fphar.2022.918819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
The use of iron oxide (Fe3O4) nanoparticles as novel contrast agents for magnetic resonance imaging (MRI) has attracted great interest due to their high r2 relaxivity. However, both poor colloidal stability and lack of effective targeting ability have impeded their further expansion in the clinics. Here, we reported the creation of hyaluronic acid (HA)-stabilized Fe3O4 nanoparticles prepared by a hydrothermal co-precipitation method and followed by electrostatic adsorption of HA onto the nanoparticle surface. The water-soluble HA functions not only as a stabilizer but also as a targeting ligand with high affinity for the CD44 receptor overexpressed in many tumors. The resulting HA-stabilized Fe3O4 nanoparticles have an estimated size of sub-20 nm as observed by transmission electron microscopy (TEM) imaging and exhibited long-term colloidal stability in aqueous solution. We found that the nanoparticles are hemocompatible and cytocompatible under certain concentrations. As verified by quantifying the cellular uptake, the Fe3O4@HA nanoparticles were able to target a model cell line (HeLa cells) overexpressing the CD44 receptor through an active pathway. In addition, we showed that the nanoparticles can be used as effective contrast agents for MRI both in vitro in HeLa cells and in vivo in a xenografted HeLa tumor model in rodents. We believe that our findings shed important light on the use of active targeting ligands to improve the contrast of lesion for tumor-specific MRI in the nano-based diagnosis systems.
Collapse
Affiliation(s)
- Weijie Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Weijie Zhang,
| | - Zhongyue Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shitong Lou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Baohong Wen
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Zhang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
16
|
Sivasubramanian M, Chu CH, Cheng SH, Chen NT, Chen CT, Chuang YC, Yu H, Chen YL, Liao LD, Lo LW. Multimodal Magnetic Resonance and Photoacoustic Imaging of Tumor-Specific Enzyme-Responsive Hybrid Nanoparticles for Oxygen Modulation. Front Bioeng Biotechnol 2022; 10:910902. [PMID: 35910012 PMCID: PMC9326367 DOI: 10.3389/fbioe.2022.910902] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Multimodal imaging contrast agents for cancer that can not only perform diagnostic functions but also serve as tumor microenvironment–responsive biomaterials are encouraging. In this study, we report the design and fabrication of a novel enzyme-responsive T1 magnetic resonance imaging (MRI) contrast agent that can modulate oxygen in the tumor microenvironment via the catalytic conversion of H2O2 to O2. The T1 contrast agent is a core–shell nanoparticle that consists of manganese oxide and hyaluronic acid (HA)–conjugated mesoporous silica nanoparticle (HA-MnO@MSN). The salient features of the nanoparticle developed in this study are as follows: 1) HA serves as a targeting ligand for CD44-expressing cancer cells; 2) HA allows controlled access of water molecules to the MnO core via the digestion of enzyme hyaluronidase; 3) the generation of O2 bubbles in the tumor by consuming H2O2; and 4) the capability to increase the oxygen tension in the tumor. The r1 relaxivity of HA-MnO@MSN was measured to be 1.29 mM−1s−1 at a magnetic field strength of 9.4 T. In vitro results demonstrated the ability of continuous oxygen evolution by HA-MnO@MSN. After intratumoral administration of HA-MnO@MSN to an HCT116 xenograft mouse model, T1 weighted MRI contrast was observed after 5 h postinjection and retained up to 48 h. In addition, in vivo photoacoustic imaging of HA-MnO@MSN demonstrated an increase in the tumor oxygen saturation over time after i. t. administration. Thus, the core–shell nanoparticles developed in this study could be helpful in tumor-targeted T1 MR imaging and oxygen modulation.
Collapse
Affiliation(s)
- Maharajan Sivasubramanian
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Hui Chu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Hsun Cheng
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Radiology, The University of Chicago, Chicago, IL, United States
| | - Nai-Tzu Chen
- College of Biopharmaceutical and Food Sciences, Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Chin-Tu Chen
- Department of Radiology, The University of Chicago, Chicago, IL, United States
| | - Yao Chen Chuang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Hsia Yu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Lin Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
- *Correspondence: Lun-De Liao, ; Leu-Wei Lo,
| | - Leu-Wei Lo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
- *Correspondence: Lun-De Liao, ; Leu-Wei Lo,
| |
Collapse
|
17
|
Xie YJ, Huang M, Li D, Hou JC, Liang HH, Nasim AA, Huang JM, Xie C, Leung ELH, Fan XX. Bacteria-based nanodrug for anticancer therapy. Pharmacol Res 2022; 182:106282. [PMID: 35662630 DOI: 10.1016/j.phrs.2022.106282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 12/15/2022]
Abstract
Bacteria-based immunotherapy has become a promising strategy to induce innate and adaptive responses for fighting cancer. The advantages of bacteriolytic tumor therapy mainly lie in stimulation of innate immunity and colonization of some bacteria targeting the tumor microenvironment (TME). These bacteria have cytotoxic proteins and immune modulating factors that can effectively restrain tumor growth. However, cancer is a multifactorial disease and single therapy is typically unable to eradicate tumors. Rapid progress has been made in combining bacteria with nanotechnology. Using the nanomolecular properties of bacterial products for tumor treatment preserves many features from the original bacteria while providing some unique advantages. Nano-bacterial therapy can enhance permeability and retention of drugs, increase the tolerability of the targeted drugs, promote the release of immune cell mediators, and induce immunogenic cell death pathways. In addition, combining nano-bacterial mediated antitumor therapeutic systems with modern therapy is an effective strategy for overcoming existing barriers in antitumor treatment and can achieve satisfactory therapeutic efficacy. Overall, exploring the immune antitumor characteristics of adjuvant clinical treatment with bacteria can provide potential efficacious treatment strategies for combatting cancer.
Collapse
Affiliation(s)
- Ya-Jia Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Min Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Dan Li
- Beijing Wante'er Biological Pharmaceutical Co., Ltd., No. 32 Yard, East 2nd Road, Yanqi Economic Development Zone, Huairou District, Beijing, China
| | - Jin-Cai Hou
- Beijing Wante'er Biological Pharmaceutical Co., Ltd., No. 32 Yard, East 2nd Road, Yanqi Economic Development Zone, Huairou District, Beijing, China
| | - Hai-Hai Liang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ali Adnan Nasim
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Ju-Min Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chun Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Elaine Lai-Han Leung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xing-Xing Fan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
18
|
Aghamiri S, Noofeli M, Saffarian P, Salehi Najafabadi Z, Goudarzi HR. Investigating preparation and characterisation of diphtheria toxoid-loaded on sodium alginate nanoparticles. IET Nanobiotechnol 2022; 16:199-209. [PMID: 35610737 PMCID: PMC9178656 DOI: 10.1049/nbt2.12088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/18/2022] [Accepted: 05/05/2022] [Indexed: 11/30/2022] Open
Abstract
This paper aims to investigate the preparation and characterisation of the alginate nanoparticles (NPs) as antigen delivery system loaded by diphtheria toxoid (DT). For this purpose, both the loading capacity (LC) and Loading efficiency (LE) of the alginate NPs burdened by DT are evaluated. Moreover, the effects of different concentrations of sodium alginate and calcium chloride on the NPs physicochemical characteristics are surveyed in addition to other physical conditions such as homogenization time and rate. To do so, the NPs are characterised using particle size and distribution, zeta potential, scanning electron microscopy, encapsulation efficiency, in vitro release study and FT‐IR spectroscopy. Subsequently, the effects of homogenization time and rate on the NPs are assessed. At the meantime, the NPs LC and efficiency in several DT concentrations are estimated. The average size of the NPs was 400.7 and 276.6 nm for unloaded and DT loaded, respectively. According to the obtained results, the zeta potential of the blank and DT loaded NPs are estimated as −23.7 mV and −21.2 mV, respectively. Whereas, the LC and LE were >80% and >90%, in that order. Furthermore, 95% of the releasing DT loaded NPs occurs at 140 h in the sustained mode without any bursting release. It can be concluded that the features of NPs such as morphology and particle size are strongly depended on the calcium chloride, sodium alginate concentrations and physicochemical conditions in the NPs formation process. In addition, appropriate concentrations of the sodium alginate and calcium ions would lead to obtaining the desirable NPs formation associated with the advantageous LE, LC (over 80%) and sustained in vitro release profile. Ultimately, the proposed NPs can be employed in vaccine formulation for the targeted delivery, controlled and slow antigen release associated with the improved antigen stability.
Collapse
Affiliation(s)
- Samira Aghamiri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mojtaba Noofeli
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agriculture Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Parvaneh Saffarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Salehi Najafabadi
- Department of Human Bacterial Vaccine, Razi Vaccine and Serum Research Institute, Agriculture Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Hamid Reza Goudarzi
- Department of Research and Development, Razi Vaccine and Serum Research Institute, Agriculture Research Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
19
|
Platelet Lysate as a Promising Medium for Nanocarriers in the Management and Treatment of Ocular Diseases. CURRENT OPHTHALMOLOGY REPORTS 2022. [DOI: 10.1007/s40135-022-00285-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Silva-Carvalho R, Leão T, Gama FM, Tomás AM. Covalent Conjugation of Amphotericin B to Hyaluronic Acid: An Injectable Water-Soluble Conjugate with Reduced Toxicity and Anti-Leishmanial Potential. Biomacromolecules 2022; 23:1169-1182. [DOI: 10.1021/acs.biomac.1c01451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ricardo Silva-Carvalho
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Teresa Leão
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Francisco M. Gama
- CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ana M. Tomás
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
21
|
Le TN, Lin CJ, Shen YC, Lin KY, Lee CK, Huang CC, Rao NV. Hyaluronic Acid Derived Hypoxia-Sensitive Nanocarrier for Tumor Targeted Drug Delivery. ACS APPLIED BIO MATERIALS 2021; 4:8325-8332. [PMID: 35005953 DOI: 10.1021/acsabm.1c00847] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hyaluronic acid (HA) is conjugated with BHQ3 moiety with azo bonds to prepare hypoxia-responsive polymer conjugate. Because of the amphiphilic nature, the polymer conjugate self-assembles to HA-BHQ3 nanoparticles (NPs). The anticancer drug doxorubicin (DOX) is loaded into the NPs. In the physiological environment, DOX is released slowly. In contrast, under hypoxic conditions, the azo bond in BHQ3 is cleaved, thus significantly enhancing the DOX release rate. For instance, after 24 h, 25% of DOX is released under normal conditions, while 74% of DOX is released under hypoxic conditions. In vitro cytotoxicity demonstrates higher toxicity in the hypoxic conditions. DOX@HA-BHQ3 NPs exhibit greater toxicity levels against 4T1 cells in hypoxic conditions. The fluorescent microscope images confirm the oxygen-dependent intracellular DOX release from the NPs. The in vivo biodistribution results suggest the tumor targetability of HA-BHQ3 NPs in 4T1 tumor-bearing mice.
Collapse
Affiliation(s)
- Trong-Nghia Le
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Chin-Jung Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Yen Chen Shen
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Kuan-Yu Lin
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Cheng-Kang Lee
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan.,Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan.,School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - N Vijayakameswara Rao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 106335, Taiwan
| |
Collapse
|
22
|
Imiquimod-gemcitabine nanoparticles harness immune cells to suppress breast cancer. Biomaterials 2021; 280:121302. [PMID: 34894584 DOI: 10.1016/j.biomaterials.2021.121302] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 11/21/2021] [Accepted: 11/29/2021] [Indexed: 12/20/2022]
Abstract
Monotherapy with a single chemotherapeutic regimen has met with significant hurdles in terms of clinical efficacy. The complexity of cancer accentuates the need for an alternative approach with a combination of two or more therapeutic regimens to win the battle. However, it is still a challenge to develop a successful combination of drugs with high efficiency and low toxicity to control cancer growth. While gemcitabine monotherapy remains a choice of standard treatment for advanced breast cancer, the approach has not prolonged the median survival time of metastatic breast cancer patients. Here, we report a hyaluronic acid (HA)-based drug combination of gemcitabine (GEM) with imiquimod (IMQ) to stimulate immune cells for anticancer activity. Treatment of the drug combination (IMQ-HA-GEM) showed enhanced anticancer activity against 4T1 breast tumor cells in vitro. Our study with a microfluidics-based 3D, compartmentalized cancer model showed that infiltration of THP-1 monocytes occurred particularly at the site of cancer cells treated with IMQ-HA-GEM. Moreover, IMQ-HA-GEM significantly suppressed the volume of 4T1 breast tumor of mice in vivo. Flow cytometry study displayed a significantly higher activation of CD11b+ immune cells in the blood of mice treated with IMQ-HA-GEM, whereas immunohistochemistry study revealed greater prevalence of CD68+ tumor-associated macrophages in the tumor. Histological examination of isolated tumors of mice treated with IMQ-HA-GEM further confirmed the efficacy of drug combination on cancer cells. This study supports the conclusion that imiquimod potentiates the effect of gemcitabine by activating immune cells to suppress tumors in the form of combination nanoparticles.
Collapse
|
23
|
Design, Synthesis, Characterization, and In Vitro Evaluation of a New Cross-Linked Hyaluronic Acid for Pharmaceutical and Cosmetic Applications. Pharmaceutics 2021; 13:pharmaceutics13101672. [PMID: 34683965 PMCID: PMC8540713 DOI: 10.3390/pharmaceutics13101672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
Hyaluronic acid (HA), an excellent biomaterial with unique bio properties, is currently one of the most interesting polymers for many biomedical and cosmetic applications. However, several of its potential benefits are limited as it is rapidly degraded by hyaluronidase enzymes. To improve the half-life and consequently increase performance, native HA has been modified through cross-linking reactions with a natural and biocompatible amino acid, Ornithine, to overcome the potential toxicity commonly associated with traditional linkers. 2-chloro-dimethoxy-1,3,5-triazine/4-methylmorpholine (CDMT/NMM) was used as an activating agent. The new product (HA–Orn) was extensively characterized to confirm the chemical modification, and rheological analysis showed a gel-like profile. In vitro degradation experiments showed an improved resistance profile against enzymatic digestions. Furthermore, in vitro cytotoxicity studies were performed on lung cell lines (Calu-3 and H441), which showed no cytotoxicity.
Collapse
|
24
|
Chen TY, Tseng CL, Lin CA, Lin HY, Venkatesan P, Lai PS. Effects of Eye Drops Containing Hyaluronic Acid-Nimesulide Conjugates in a Benzalkonium Chloride-Induced Experimental Dry Eye Rabbit Model. Pharmaceutics 2021; 13:1366. [PMID: 34575442 PMCID: PMC8469214 DOI: 10.3390/pharmaceutics13091366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Dry eye syndrome (DES) is a common ocular disease worldwide. Currently, anti-inflammatory agents and immunosuppressive drugs, such as cyclosporine A, have been widely used to treat this chronic condition. However, the multifactorial etiology of DES, poor tolerance, low bioavailability, and prolonged treatment to response time have limited their usage. In this study, nimesulide, a cyclooxygenase (COX)-2 selective inhibitor, was conjugated with hyaluronic acid (HA), and the HA-nimesulide conjugates were expected to increase the solubility and biocompatibility for alleviating the DES in the benzalkonium chloride (BAC)-induced goblet cell-loss dry eye model. The therapeutic efficacy of HA-nimesulide was assessed using fluorescein staining, goblet cell density by conjunctival impression cytology, and histology and immunohistochemistry of corneal tissues. Compared to commercial artificial tears and Restasis®, the HA-nimesulide conjugates could promote goblet cell recovery and enhance the regeneration of the corneal epithelium. Importantly, immunofluorescent staining studies demonstrated that the HA-nimesulide conjugates could decrease the number of infiltrating CD11b-positive cells after two weeks of topical application. In the anti-inflammatory test, the HA-nimesulide conjugates could inhibit the production of pro-inflammatory cytokines and prostaglandin E2 (PGE2) in the lipopolysaccharide (LPS)-stimulated Raw 264.7 cell model. In conclusion, we demonstrated that HA-nimesulide conjugates had anti-inflammatory activity, and promoted goblet cell recovery and corneal epithelium regeneration when used as topical eye drops; accordingly, the HA-nimesulide conjugates could potentially be effective for the treatment of DES.
Collapse
Affiliation(s)
- Tzu-Yang Chen
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; (T.-Y.C.); (P.V.)
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan;
| | - Chih-An Lin
- Ph.D. Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Hua-Yang Lin
- Preclinical Development Research Department, Holy Stone Healthcare Co., Ltd., Taipei 11493, Taiwan;
| | - Parthiban Venkatesan
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; (T.-Y.C.); (P.V.)
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, Taichung 40227, Taiwan; (T.-Y.C.); (P.V.)
- Ph.D. Program of Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan;
| |
Collapse
|
25
|
Hyaluronic Acid-Functionalized Nanomicelles Enhance SAHA Efficacy in 3D Endometrial Cancer Models. Cancers (Basel) 2021; 13:cancers13164032. [PMID: 34439185 PMCID: PMC8394402 DOI: 10.3390/cancers13164032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary One of the major limitations to cancer therapies are the side effects caused by the drug interacting with any tissue in the body. There is often a balance between patient health and effectively treating the disease. To by-pass this balancing act nanoparticles are being used to deliver therapeutics straight to the tumors, acting as “Trojan Horses”. Endometrial cancers are known to have more of the cell surface protein CD44 than healthy tissues. Here, to efficiently target endometrial cancer, hyaluronic acid, which naturally binds to the CD44 protein was attached to the surface of nanoparticles and tested on microtissues or spheroids to better model a tumor and understand drug delivery performance. We show that our hyaluronic acid-nanoparticle formulations improve drug effects and interact with the cancer cells more than without this targeting agent. Abstract Histone Deacetylase (HDAC) enzymes are upregulated in cancer leading to the development of HDAC inhibiting compounds, several of which are currently in clinical trials. Side effects associated with toxicity and non-specific targeting indicate the need for efficient drug delivery approaches and tumor specific targeting to enhance HDAC efficacy in solid tumor cancers. SAHA encapsulation within F127 micelles functionalized with a surface hyaluronic acid moiety, was developed to target endometrial cancer cells expressing elevated levels of CD44. In vitro viability and morphology analyses was conducted in both 2D and 3D models to assess the translational potential of this approach. Encapsulation enhanced SAHA delivery and activity, demonstrating increased cytotoxic efficacy in 2D and 3D endometrial cancer models. High-content imaging showed improved nanoparticle internalization in 2D and CD44 enhanced penetration in 3D models. In addition, the nano-delivery system enhanced spheroid penetration resulting in cell growth suppression, p21 associated cell cycle arrest, as well as overcoming the formation of an EMT associated phenotype observed in free drug treated type II endometrial cancer cells. This study demonstrates that targeted nanoparticle delivery of SAHA could provide the basis for improving its efficacy in endometrial cancer. Using 3D models for endometrial cancer allows the elucidation of nanoparticle performance and CD44 targeting, likely through penetration and retention within the tumor model.
Collapse
|
26
|
Zhang Y, Fowlkes JB. Liposomes-based nanoplatform enlarges ultrasound-related diagnostic and therapeutic precision. Curr Med Chem 2021; 29:1331-1341. [PMID: 34348609 DOI: 10.2174/0929867328666210804092624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 12/07/2022]
Abstract
Ultrasound (US) is notable in the medical field as a safe and effective imaging modality due to its lack of ionizing radiation, non-invasive approach, and real-time monitoring capability. Accompanying recent progress in nanomedicine, US has been providing hope of theranostic capability not only for imaging-based diagnosis but also for US-based therapy by taking advantage of the bioeffects induced by US. Cavitation, sonoporation, thermal effects, and other cascade effects stimulated by acoustic energy conversion have contributed to medical problem-solving in the past decades although to varying degrees of efficacy in comparisons to other methods. Recently, the usage of liposomes-based nanoplatform fuels the development of nanomedicine and provides novel clinical strategies for antitumor, thrombolysis, and controlled drug release. Merging of novel liposome-based nanoplatforms and US-induced reactions has promise for a new blueprint for future medicine. In the present review article, the value of liposome-based nanoplatforms in US-related diagnosis and therapy will be discussed and summarized along with potential future directions for further investigations.
Collapse
Affiliation(s)
- Ying Zhang
- Dept. Radiology, University of Michigan, Ann Arbor, Michigan, 48109. United States
| | - J Brian Fowlkes
- Dept. Radiology, University of Michigan, Ann Arbor, Michigan, 48109. United States
| |
Collapse
|
27
|
Kotla NG, Bonam SR, Rasala S, Wankar J, Bohara RA, Bayry J, Rochev Y, Pandit A. Recent advances and prospects of hyaluronan as a multifunctional therapeutic system. J Control Release 2021; 336:598-620. [PMID: 34237401 DOI: 10.1016/j.jconrel.2021.07.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
Hyaluronan (HA) is a naturally occurring non-sulfated glycosaminoglycan (GAG), cell-surface-associated biopolymer and is the key component of tissue extracellular matrix (ECM). Along with remarkable physicochemical properties, HA also has multifaceted biological effects that include but not limited to ECM organization, immunomodulation, and various cellular processes. Environmental cues such as tissue injury, infection or cancer change downstream signaling functionalities of HA. Unlike native HA, the fragments of HA have diversified effects on inflammation, cancer, fibrosis, angiogenesis and autoimmune response. In this review, we aim to discuss HA as a therapeutic delivery system development process, source, biophysical-chemical properties, and associated biological pathways (especially via cell surface receptors) of native and fragmented HA. We also tried to address an overview of the potential role of HA (native HA vs fragments) in the modulation of inflammation, immune response and various cancer targeting delivery applications. This review will also highlight the HA based therapeutic systems, medical devices and future perspectives of various biomedical applications were discussed in detail.
Collapse
Affiliation(s)
- Niranjan G Kotla
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris F-75006, France
| | - Swetha Rasala
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Jitendra Wankar
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Raghvendra A Bohara
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris F-75006, France; Indian Institute of Technology Palakkad, Palakkad 678 623, Kerala, India
| | - Yury Rochev
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland; Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, Moscow 119992, Russia.
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, H91 W2TY, Ireland.
| |
Collapse
|
28
|
Strategies to load therapeutics into polysaccharide-based nanogels with a focus on microfluidics: A review. Carbohydr Polym 2021; 266:118119. [PMID: 34044935 DOI: 10.1016/j.carbpol.2021.118119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/04/2021] [Accepted: 04/15/2021] [Indexed: 01/05/2023]
Abstract
Nowadays nanoparticles are increasingly investigated for the targeted and controlled delivery of therapeutics, as suggested by the high number of research articles (2400 in 2000 vs 8500 in 2020). Among them, almost 2% investigated nanogels in 2020. Nanogels or nanohydrogels (NGs) are nanoparticles formed by a swollen three-dimensional network of synthetic polymers or natural macromolecules such as polysaccharides. NGs represent a highly versatile nanocarrier, able to deliver a number of therapeutics. Currently, NGs are undergoing clinical trials for the delivery of anti-cancer vaccines. Herein, the strategies to load low molecular weight drugs, (poly)peptides and genetic material into polysaccharide NGs as well as to formulate NGs-based vaccines are summarized, with a focus on the microfluidics approach.
Collapse
|
29
|
Attia YA, Al Nazawi AM, Elsayed H, Sadik MW. Carbon nanotubes catalyzed UV-trigger production of hyaluronic acid from Streptococcus equi. Saudi J Biol Sci 2021; 28:484-491. [PMID: 33424331 PMCID: PMC7783678 DOI: 10.1016/j.sjbs.2020.10.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/05/2020] [Accepted: 10/19/2020] [Indexed: 01/15/2023] Open
Abstract
Hyaluronic acid (HA) has great importance in biomedical applications. In this work, a novel nanoparticle-based method that stimulates the hyaluronic acid (HA) production by the bacteria Streptococcus equi subsp. Zooepidemicus has been reported. CNTs with diameters of 40-50 nm and lengths of about 20 mm were used at four different concentrations (0, 10, 25, 50, and 100 μg) to the bacteria and determined the mass of the produced HA in dependence on the exposure time under UV-irradiation. The results clearly showed that the exposure for one minute with low power UV light (254 nm) and 100 µg (CNTs) treatments steadily increased HA production from the control (0.062 g/L) to the highest value (0.992) g/L of HA. The incubation of the streptococci with CNTs led to an increase of the HA production by a factor of 4.23 after 300S exposure time under UV light, whereas the HA production was no significant enhancement under visible light. It is explained that the CNTs nanoparticle-stimulated increase of the HA production with the internalization of the nanoparticles by the bacteria since they "serve as co-enzymes" under induced mutation by UV-irradiation. Transformation process was carried out and showed that the major protein band of Streptococcus equi was observed in the Streptococcus DH5α. RAPD analysis indicates that the amplified DNA fragments and the percentage of polymorphism was similar between Streptococcus equi and Streptococcus DH50α. The chemical structure and molecular weight of the photoproduced HA from Streptococcus equi was similar to the chemical structure of the standard sample.
Collapse
Affiliation(s)
- Yasser A. Attia
- National Institute of Laser Enhanced Sciences, Cairo University, Giza 12613, Egypt
| | - Ashwaq M. Al Nazawi
- Preventive Medicine Department, Public Health Directorate, Ministry of Health, Jeddah 22246, Saudi Arabia
| | - Hassan Elsayed
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Division, National Research Centre, Dokki, Giza 12622, Egypt
| | - Mahmoud W. Sadik
- Microbiology Department, Faculty of Agriculture, Cairo University, Giza 12613, Egypt
| |
Collapse
|
30
|
Bumrung J, Chanchao C, Intasanta V, Palaga T, Wanichwecharungruang S. Water-dispersible unadulterated α-mangostin particles for biomedical applications. ROYAL SOCIETY OPEN SCIENCE 2020; 7:200543. [PMID: 33391780 PMCID: PMC7735336 DOI: 10.1098/rsos.200543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/02/2020] [Indexed: 06/12/2023]
Abstract
α-Mangostin, the extract from pericarp of Garcinia mangostana L . or mangosteen fruit, has been applied in various biomedical products because of its minimal skin irritation, and prominent anti-inflammatory, antimicrobial and immune-modulating activities. Owing to its low water solubility, the particle formulations are necessary for the applications of α-mangostin in aqueous media. The particle formulations are usually prepared using surfactants and/or polymers, usually at a larger amount of these auxiliaries than the amount of α-mangostin itself. Here, we show the self-assembly of α-mangostin molecules into water-dispersible particles without a need of any polymers/surfactants. Investigations on chemical structure, crystallinity and thermal properties of the obtained α-mangostin particles, in comparison to the conventional α-mangostin crystalline solid, confirm no formation of the new compound during the particle formation and suggest changes in intermolecular interactions among α-mangostin molecules and significantly more hydroxyl functionality positioned at the particles' surface. The ability of the water suspension of the α-mangostin to inhibit the growth of Propionibacterium acnes, the acne-causing bacteria, is similar to that of the solution of the conventional α-mangostin in 5% dimethyl sulfoxide. Moreover, at 12.7 ppm in an aqueous environment of RAW 264.7 cell culture, α-mangostin suspension exhibits five times higher anti-inflammatory activity than the conventional α-mangostin solution, with the same acceptable cytotoxicity of less than 20% cell death.
Collapse
Affiliation(s)
- Jutamad Bumrung
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Advanced Materials and Biointerfaces, Chulalongkorn University, Bangkok, Thailand
| | - Chanpen Chanchao
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Varol Intasanta
- National Nanotechnology Center, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Supason Wanichwecharungruang
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Advanced Materials and Biointerfaces, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
31
|
Jain NK, Dimri S, Prasad R, Ravichandran G, Naidu V, De A, Srivastava R. Characteristics of Molecularly Engineered Anticancer Drug Conjugated Organic Nanomicelles for Site-Selective Cancer Cell Rupture and Growth Inhibition of Tumor Spheroids. ACS APPLIED BIO MATERIALS 2020; 3:7067-7079. [PMID: 35019366 DOI: 10.1021/acsabm.0c00913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Site-selective uptake and specific biodistribution of chemotherapeutic drugs are essential prerequisites for targeted cancer therapy. Especially, antibody and peptide conjugated drugs have been attempted as localized therapeutic agents. However, the characteristics of drug conjugated nanosystems are less explored, which are limited with their toxicity, low therapeutic efficacy, complicated synthesis, and high costs. Herein, we report a biocompatible (about 95%) molecularly engineered anticancer drug conjugated nanomicelles (∼200 nm in size) for site-selective CD44 overexpressed cancer cell rupture and tumor growth inhibition. Microscopic analysis demonstrates the distinct visualization of organic-organic interfaces (∼5 nm), which are corroborated with spectroscopic measurements confirmed the conjugation of niclosamide drug with hyaluronic acid (NIC-HA). Uniformly distributed hemocompatible (about 99%) organic nanomicelles exhibit the cellular membrane and cytoplasmic targeting with significant cellular rupture (IC50 of 4 μM for MDA MB 231 cells) indicating their inherent targeting ability for cancer cells and cancer stem cells. An inclusive in vitro and in vivo analysis for targeted antitumor activity (HT1080 tumor xenograft model) of NIC-HA nanoconjugates (∼24.6% loading) exhibited promising cancer cell death and tumor growth inhibition (60%, p < 0.05) due to STAT-3 signaling pathway inhibition and induction of apoptosis in CD44-positive triple negative breast cancer cells.
Collapse
Affiliation(s)
- Nishant Kumar Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT-B), Powai, Mumbai, India
| | - Shalini Dimri
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT-B), Powai, Mumbai, India
| | - Gayathri Ravichandran
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Vegi Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IIT-B), Powai, Mumbai, India
| |
Collapse
|
32
|
Idrees H, Zaidi SZJ, Sabir A, Khan RU, Zhang X, Hassan SU. A Review of Biodegradable Natural Polymer-Based Nanoparticles for Drug Delivery Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1970. [PMID: 33027891 PMCID: PMC7600772 DOI: 10.3390/nano10101970] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 01/19/2023]
Abstract
Biodegradable natural polymers have been investigated extensively as the best choice for encapsulation and delivery of drugs. The research has attracted remarkable attention in the pharmaceutical industry. The shortcomings of conventional dosage systems, along with modified and targeted drug delivery methods, are addressed by using polymers with improved bioavailability, biocompatibility, and lower toxicity. Therefore, nanomedicines are now considered to be an innovative type of medication. This review critically examines the use of natural biodegradable polymers and their drug delivery systems for local or targeted and controlled/sustained drug release against fatal diseases.
Collapse
Affiliation(s)
- Humaira Idrees
- Department of Polymer Engineering and Technology, University of the Punjab, Lahore 54590, Pakistan; (A.S.); (R.U.K.)
| | - Syed Zohaib Javaid Zaidi
- Institute of Chemical Engineering and Technology, University of the Punjab, Lahore 54000, Punjab, Pakistan
| | - Aneela Sabir
- Department of Polymer Engineering and Technology, University of the Punjab, Lahore 54590, Pakistan; (A.S.); (R.U.K.)
| | - Rafi Ullah Khan
- Department of Polymer Engineering and Technology, University of the Punjab, Lahore 54590, Pakistan; (A.S.); (R.U.K.)
- Institute of Chemical Engineering and Technology, University of the Punjab, Lahore 54000, Punjab, Pakistan
| | - Xunli Zhang
- Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK;
| | - Sammer-ul Hassan
- Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK;
| |
Collapse
|
33
|
Rao N, Rho JG, Um W, EK PK, Nguyen VQ, Oh BH, Kim W, Park JH. Hyaluronic Acid Nanoparticles as Nanomedicine for Treatment of Inflammatory Diseases. Pharmaceutics 2020; 12:E931. [PMID: 33003609 PMCID: PMC7600604 DOI: 10.3390/pharmaceutics12100931] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023] Open
Abstract
Owing to their unique biological functions, hyaluronic acid (HA) and its derivatives have been explored extensively for biomedical applications such as tissue engineering, drug delivery, and molecular imaging. In particular, self-assembled HA nanoparticles (HA-NPs) have been used widely as target-specific and long-acting nanocarriers for the delivery of a wide range of therapeutic or diagnostic agents. Recently, it has been demonstrated that empty HA-NPs without bearing any therapeutic agent can be used therapeutically for the treatment of inflammatory diseases via modulating inflammatory responses. In this review, we aim to provide an overview of the significant achievements in this field and highlight the potential of HA-NPs for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- N.Vijayakameswara Rao
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10617, Taiwan
| | - Jun Gi Rho
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Korea;
| | - Wooram Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Pramod Kumar EK
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Van Quy Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Byeong Hoon Oh
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Wook Kim
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Korea;
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
- Department Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
34
|
Lierova A, Kasparova J, Pejchal J, Kubelkova K, Jelicova M, Palarcik J, Korecka L, Bilkova Z, Sinkorova Z. Attenuation of Radiation-Induced Lung Injury by Hyaluronic Acid Nanoparticles. Front Pharmacol 2020; 11:1199. [PMID: 32903478 PMCID: PMC7435052 DOI: 10.3389/fphar.2020.01199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Therapeutic thorax irradiation as an intervention in lung cancer has its limitations due to toxic effects leading to pneumonitis and/or pulmonary fibrosis. It has already been confirmed that hyaluronic acid (HA), an extracellular matrix glycosaminoglycan, is involved in inflammation disorders and wound healing in lung tissue. We examined the effects after gamma irradiation of hyaluronic acid nanoparticles (HANPs) applied into lung prior to that irradiation in a dose causing radiation-induced pulmonary injuries (RIPI). Materials and Methods Biocompatible HANPs were first used for viability assay conducted on the J774.2 cell line. For in vivo experiments, HANPs were administered intratracheally to C57Bl/6 mice 30 min before thoracic irradiation by 17 Gy. Molecular, cellular, and histopathological parameters were measured in lung and peripheral blood at days 113, 155, and 190, corresponding to periods of significant morphological and/or biochemical alterations of RIPI. Results Modification of linear hyaluronic acid molecule into nanoparticles structure significantly affected the physiological properties and caused long-term stability against ionizing radiation. The HANPs treatments had significant effects on the expression of the cytokines and particularly on the pro-fibrotic signaling pathway in the lung tissue. The radiation fibrosis phase was altered significantly in comparison with a solely irradiated group. Conclusions The present study provides evidence that application of HANPs caused significant changes in molecular and cellular patterns associated with RIPI. These findings suggest that HANPs could diminish detrimental radiation-induced processes in lung tissue, thereby potentially decreasing the extracellular matrix degradation leading to lung fibrosis.
Collapse
Affiliation(s)
- Anna Lierova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | - Jitka Kasparova
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technologies, University of Pardubice, Pardubice, Czechia
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | - Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | - Marcela Jelicova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| | - Jiri Palarcik
- Institute of Environmental and Chemical Engineering, Faculty of Chemical Technology, University of Pardubice, Pardubice, Czechia
| | - Lucie Korecka
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technologies, University of Pardubice, Pardubice, Czechia
| | - Zuzana Bilkova
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technologies, University of Pardubice, Pardubice, Czechia
| | - Zuzana Sinkorova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czechia
| |
Collapse
|
35
|
Janik-Hazuka M, Szafraniec-Szczęsny J, Kamiński K, Odrobińska J, Zapotoczny S. Uptake and in vitro anticancer activity of oleic acid delivered in nanocapsules stabilized by amphiphilic derivatives of hyaluronic acid and chitosan. Int J Biol Macromol 2020; 164:2000-2009. [PMID: 32781133 DOI: 10.1016/j.ijbiomac.2020.07.288] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/24/2020] [Accepted: 07/26/2020] [Indexed: 12/25/2022]
Abstract
The nanoemulsion-based delivery systems have gained particular attention due to effective encapsulation and protection of hydrophobic active compounds. However, several features like limited stability, cellular uptake or release of payloads still need to be addressed. We investigated the uptake of the nanocapsules based on the amphiphilic derivative of hyaluronate with oleic acid cores (oil-in-water nanoemulsion) and their anticancer activity in vitro. The core-shell nanocapsules exhibiting long term stability in dispersion showed an enhanced uptake by cancer cells and effectively killed them only if composed of hyaluronate-based shells and oleic acid cores - the anionic chitosan-based shells and/or corn oil cores were used for control experiments. We concluded that the nanocapsules stabilized by the amphiphilic derivative of hyaluronic acid may serve as very stable and efficient delivery systems for oil-soluble compounds without necessity of application of low molecular weight (co)surfactants. The in vitro studies indicated anticancer activity of such delivered oleic acid and crucial role of hyaluronate shell of the nanocapsules in its efficient delivery and enzyme-triggered disintegration inside cells. Corn oil was shown as a nutrient that can serve as an inert vehicle in the studied nanoemulsion that exhibit application potential in food, dietary supplement industry and medicine.
Collapse
Affiliation(s)
| | - Joanna Szafraniec-Szczęsny
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Kamil Kamiński
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Joanna Odrobińska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Szczepan Zapotoczny
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
36
|
Tang Y, Chen M, Xie Q, Li L, Zhu L, Ma Q, Gao S. Construction and evaluation of hyaluronic acid-based copolymers as a targeted chemotherapy drug carrier for cancer therapy. NANOTECHNOLOGY 2020; 31:305702. [PMID: 32272454 DOI: 10.1088/1361-6528/ab884d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Melanoma (MM) is a highly aggressive skin cancer with limited treatment options. Although chemotherapy has been using for advanced melanoma treatment, the lack of targetability, the poor biocompatibility and the severe side effects still hamper the wide applications of chemotherapy agents in MM management. Herein, a biocompatible and biodegradable polymeric hyaluronic acid nanoparticle (HANP) encapsulated with Paclitaxel (PTX) was developed for MM targeted therapy. Our results showed that PTX at 37 ± 2.1% (w/w) was able to be loaded into HANP with over 5 d of stability under physiological conditions. In vitro, HANP/PTX presented hyaluronidase-dependent drug release. Compared to free PTX, HANP/PTX demonstrated a 6-75 times higher growth inhibition in five different cancer cells, while only presenting minimum toxicity to normal cells. After intravenous administration at a 10 mg kg-1 equivalent dose of PTX, HANP/PTX significantly ablated MM tumor growth in a mouse model. As confirmed by 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) imaging, the tumors started to respond to the HANP/PTX as early as 7 d after the initial treatment, which will significantly benefit for personalized treatment. In conclusion, the HANP/PTX nanocomplex demonstrated great promise as a translational nanomedicine for cancer chemotherapy.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Dacoba TG, Anfray C, Mainini F, Allavena P, Alonso MJ, Torres Andón F, Crecente-Campo J. Arginine-Based Poly(I:C)-Loaded Nanocomplexes for the Polarization of Macrophages Toward M1-Antitumoral Effectors. Front Immunol 2020; 11:1412. [PMID: 32733469 PMCID: PMC7358452 DOI: 10.3389/fimmu.2020.01412] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Tumor-associated macrophages (TAMs), with M2-like immunosuppressive profiles, are key players in the development and dissemination of tumors. Hence, the induction of M1 pro-inflammatory and anti-tumoral states is critical to fight against cancer cells. The activation of the endosomal toll-like receptor 3 by its agonist poly(I:C) has shown to efficiently drive this polarization process. Unfortunately, poly(I:C) presents significant systemic toxicity, and its clinical use is restricted to a local administration. Therefore, the objective of this work has been to facilitate the delivery of poly(I:C) to macrophages through the use of nanotechnology, that will ultimately drive their phenotype toward pro-inflammatory states. Methods: Poly(I:C) was complexed to arginine-rich polypeptides, and then further enveloped with an anionic polymeric layer either by film hydration or incubation. Physicochemical characterization of the nanocomplexes was conducted by dynamic light scattering and transmission electron microscopy, and poly(I:C) association efficiency by gel electrophoresis. Primary human-derived macrophages were used as relevant in vitro cell model. Alamar Blue assay, ELISA, PCR and flow cytometry were used to determine macrophage viability, polarization, chemokine secretion and uptake of nanocomplexes. The cytotoxic activity of pre-treated macrophages against PANC-1 cancer cells was assessed by flow cytometry. Results: The final poly(I:C) nanocomplexes presented sizes lower than 200 nm, with surface charges ranging from +40 to −20 mV, depending on the envelopment. They all presented high poly(I:C) loading values, from 12 to 50%, and great stability in cell culture media. In vitro, poly(I:C) nanocomplexes were highly taken up by macrophages, in comparison to the free molecule. Macrophage treatment with these nanocomplexes did not reduce their viability and efficiently stimulated the secretion of the T-cell recruiter chemokines CXCL10 and CCL5, of great importance for an effective anti-tumor immune response. Finally, poly(I:C) nanocomplexes significantly increased the ability of treated macrophages to directly kill cancer cells. Conclusion: Overall, these enveloped poly(I:C) nanocomplexes might represent a therapeutic option to fight cancer through the induction of cytotoxic M1-polarized macrophages.
Collapse
Affiliation(s)
- Tamara G Dacoba
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), IDIS Research Institute, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Clément Anfray
- Laboratory of Cellular Immunology, Humanitas Clinical and Research Center IRCCS, Milan, Italy
| | - Francesco Mainini
- Laboratory of Cellular Immunology, Humanitas Clinical and Research Center IRCCS, Milan, Italy
| | - Paola Allavena
- Laboratory of Cellular Immunology, Humanitas Clinical and Research Center IRCCS, Milan, Italy
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), IDIS Research Institute, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), IDIS Research Institute, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Laboratory of Cellular Immunology, Humanitas Clinical and Research Center IRCCS, Milan, Italy
| | - José Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), IDIS Research Institute, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
38
|
Castanheira EJ, Correia TR, Rodrigues JMM, Mano JF. Novel Biodegradable Laminarin Microparticles for Biomedical Applications. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020. [DOI: 10.1246/bcsj.20200034] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Edgar J. Castanheira
- CICECO – Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Tiago R. Correia
- CICECO – Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - João M. M. Rodrigues
- CICECO – Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - João F. Mano
- CICECO – Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
39
|
Lee WH, Rho JG, Han HS, Kweon S, Nguyen VQ, Park JH, Kim W. Self-assembled hyaluronic acid nanoparticle suppresses fat accumulation via CD44 in diet-induced obese mice. Carbohydr Polym 2020; 237:116161. [DOI: 10.1016/j.carbpol.2020.116161] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 01/29/2023]
|
40
|
Targeted delivery of hyaluronic acid nanomicelles to hepatic stellate cells in hepatic fibrosis rats. Acta Pharm Sin B 2020; 10:693-710. [PMID: 32322471 PMCID: PMC7161713 DOI: 10.1016/j.apsb.2019.07.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/21/2022] Open
Abstract
Hepatic fibrosis is one kind of liver diseases with a high mortality rate and incidence. The activation and proliferation of hepatic stellate cells (HSCs) is the most fundamental reason of hepatic fibrosis. There are no specific and effective drug delivery carriers for the treatment of hepatic fibrosis at present. We found that when hepatic fibrosis occurs, the expression of CD44 receptors on the surface of HSCs is significantly increased. Based on this finding, we designed silibinin-loaded hyaluronic acid (SLB-HA) micelles to achieve the treatment of hepatic fibrosis. Meanwhile, we constructed liver fibrosis rat model using Sprague–Dawley rats. We demonstrated that HA micelles had specific uptake to HSCs in vitro while avoiding the distribution in normal liver cells and the phagocytosis of macrophages. Importantly, HA micelles showed a significant liver targeting effect in vivo, especially in fibrotic liver which highly expressed CD44 receptors. In addition, SLB-HA micelles could selectively kill activated HSCs, having an excellent anti-hepatic fibrosis effect in vivo and a significant sustained release effect, and also had a good biological safety and biocompatibility. Overall, HA micelles represented a novel nanomicelle system which showed great potentiality in anti-hepatic fibrosis drugs delivery.
Collapse
|
41
|
Zhong W, Pang L, Feng H, Dong H, Wang S, Cong H, Shen Y, Bing Y. Recent advantage of hyaluronic acid for anti-cancer application: a review of "3S" transition approach. Carbohydr Polym 2020; 238:116204. [PMID: 32299556 DOI: 10.1016/j.carbpol.2020.116204] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/06/2020] [Accepted: 03/20/2020] [Indexed: 12/25/2022]
Abstract
In recent years, nano drug delivery system has been widely concerned because of its good therapeutic effect. However, the process from blood circulation to cancer cell release of nanodrugs will be eliminated by the human body's own defense trap, thus reducing the therapeutic effect. In recent years, a "3S" transition concept, including stability transition, surface transition and size transition, was proposed to overcome the barriers in delivery process. Hyaluronic (HA) acid has been widely used in delivery of anticancer drugs due to its excellent biocompatibility, biodegradability and specific targeting to cancer cells. In this paper, the strategies and methods of HA-based nanomaterials using "3S" theory are reviewed. The applications and effects of "3S" modified nanomaterials in various fields are also introduced.
Collapse
Affiliation(s)
- Wei Zhong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Long Pang
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Haohui Feng
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Haonan Dong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Yu Bing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
42
|
Wang G, Song L, Hou X, Kala S, Wong KF, Tang L, Dai Y, Sun L. Surface-modified GVs as nanosized contrast agents for molecular ultrasound imaging of tumor. Biomaterials 2020; 236:119803. [PMID: 32028170 DOI: 10.1016/j.biomaterials.2020.119803] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 12/13/2022]
Abstract
Nanobubbles, as a kind of new ultrasound contrast agent (UCAs), have shown promise to penetrate tumor vasculature to allow for targeted imaging. However, their inherent physical instability is an ongoing concern that could weaken their imaging ability with ultrasound. Gas vesicles (GVs), which are genetically encoded, naturally stable nanostructures, have been developed as the first ultrasonic biomolecular reporters which showed strong contrast enhancement. However, further development of tumor imaging with GVs is limited by the quick clearance of GVs by the reticuloendothelial system (RES). Here, we developed PEGylated HA-GVs (PH-GVs) for in-tumor molecular ultrasound imaging by integrating polyethylene glycol (PEG) and hyaluronic acid (HA) in GV shells. PH-GVs were observed to accumulate around CD44-positive cells (SCC7) but not be internalized by macrophage cell line RAW 264.7. Green fluorescence from PH-GVs was found around cell nuclei in the tumor site after 6 h and the signal was sustained over 48 h following tail injection, demonstrating PH-GVs' ability to escape the clearance from the RES and to penetrate tumor vasculature through enhanced permeability and retention (EPR) effects. Further, PH-GVs produced strong ultrasound contrast in the tumor site in vivo, with no obvious side-effects detected following intravenous injection. Thus, we demonstrate the potential of PH-GVs as novel, nanosized and targeted UCAs for efficient and specific molecular tumor imaging, paving the way for the application of GVs in precise and personalized medicine.
Collapse
Affiliation(s)
- Guohao Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Lin Song
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Xuandi Hou
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Shashwati Kala
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Kin Fung Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Liya Tang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Yunlu Dai
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau
| | - Lei Sun
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region.
| |
Collapse
|
43
|
Abdel-Mottaleb MM, Abd-Allah H, El-Gogary RI, Nasr M. Versatile hyaluronic acid nanoparticles for improved drug delivery. DRUG DELIVERY ASPECTS 2020:1-18. [DOI: 10.1016/b978-0-12-821222-6.00001-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
44
|
Kumari R, Sunil D, Ningthoujam RS. Hypoxia-responsive nanoparticle based drug delivery systems in cancer therapy: An up-to-date review. J Control Release 2019; 319:135-156. [PMID: 31881315 DOI: 10.1016/j.jconrel.2019.12.041] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 02/08/2023]
Abstract
Hypoxia is a salient feature observed in most solid malignancies that holds a pivotal role in angiogenesis, metastasis and resistance to conventional cancer therapeutic approaches, and thus enables cancer progression. However, the typical characteristics of hypoxic cells such as low oxygen levels and highly bio-reductive environment can offer stimuli-responsive drug release to aid in tumor-specific chemo, radio, photodyanamic and sonodynamic therapies. This approach based on targeting the poorly oxygenated tumor habitats offers the prospective to overcome the difficulties that arises due to heterogenic nature of tumor and could be possibly used in the design of diagnostic as well as therapeutic nanocarriers for targeting various types of solid cancers. Consequently, hypoxia triggered nanoparticle based drug delivery systems is a rapidly progressing research area in developing effective strategies to combat drug-resistance in solid tumors. The present review presents the recent advances in the development of hypoxia-responsive nanovehicles for drug delivery to heterogeneous tumors. The initial sections of the article provides insights into the development of hypoxia in growing cancer and its role in disease progression. The current limitations and the future prospective of hypoxia-stimulated nanomachines for cancer treatment are also discussed.
Collapse
Affiliation(s)
- Rashmi Kumari
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576 104, Karnataka, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576 104, Karnataka, India.
| | | |
Collapse
|
45
|
Metal-Phenolic Network-Coated Hyaluronic Acid Nanoparticles for pH-Responsive Drug Delivery. Pharmaceutics 2019; 11:pharmaceutics11120636. [PMID: 31795253 PMCID: PMC6956368 DOI: 10.3390/pharmaceutics11120636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/24/2019] [Accepted: 11/26/2019] [Indexed: 12/02/2022] Open
Abstract
Although self-assembled nanoparticles (SNPs) have been used extensively for targeted drug delivery, their clinical applications have been limited since most of the drugs are released into the blood before they reach their target site. In this study, metal-phenolic network (MPN)-coated SNPs (MPN-SNPs), which consist of an amphiphilic hyaluronic acid derivative, were prepared to be a pH-responsive nanocarrier to facilitate drug release in tumor microenvironments (TME). Due to their amphiphilic nature, SNPs were capable of encapsulating doxorubicin (DOX), chosen as the model anticancer drug. Tannic acid and FeCl3 were added to the surface of the DOX-SNPs, which allowed them to be readily coated with MPNs as the diffusion barrier. The pH-sensitive MPN corona allowed for a rapid release of DOX and effective cellular SNP uptake in the mildly acidic condition (pH 6.5) mimicking TME, to which the hyaluronic acid was exposed to facilitate receptor-mediated endocytosis. The DOX-loaded MPN-SNPs exhibited a higher cytotoxicity for the cancer cells, suggesting their potential use as a drug carrier in targeted cancer therapy.
Collapse
|
46
|
Montanari E, Di Meo C, Coviello T, Gueguen V, Pavon-Djavid G, Matricardi P. Intracellular Delivery of Natural Antioxidants via Hyaluronan Nanohydrogels. Pharmaceutics 2019; 11:pharmaceutics11100532. [PMID: 31615083 PMCID: PMC6835714 DOI: 10.3390/pharmaceutics11100532] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022] Open
Abstract
Natural antioxidants, such as astaxanthin (AX), resveratrol (RV) and curcumin (CU), are bioactive molecules that show a number of therapeutic effects. However, their applications are remarkably limited by their poor water solubility, physico-chemical instability and low bioavailability. In the present work, it is shown that self-assembled hyaluronan (HA)-based nanohydrogels (NHs) are taken up by endothelial cells (Human Umbilical Vein Endothelial Cells, HUVECs), preferentially accumulating in the perinuclear area of oxidatively stressed HUVECs, as evidenced by flow cytometry and confocal microscopy analyses. Furthermore, NHs are able to physically entrap and to significantly enhance the apparent water solubility of AX, RV and CU in aqueous media. AX/NHs, RV/NHs and CU/NHs systems showed good hydrodynamic diameters (287, 214 and 267 nm, respectively), suitable ζ-potential values (-45, -43 and -37 mV, respectively) and the capability to neutralise reactive oxygen species (ROS) in tube. AX/NHs system was also able to neutralise ROS in vitro and did not show any toxicity against HUVECs. This research suggests that HA-based NHs can represent a kind of nano-carrier suitable for the intracellular delivery of antioxidant agents, for the treatment of oxidative stress in endothelial cells.
Collapse
Affiliation(s)
- Elita Montanari
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Chiara Di Meo
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Tommasina Coviello
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| | - Virginie Gueguen
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Paris 13 University, Sorbonne Paris Cite 99, Av. Jean-Baptiste Clément, 93430 Villetaneuse, France.
| | - Graciela Pavon-Djavid
- INSERM U1148, Laboratory for Vascular Translational Science, Cardiovascular Bioengineering, Paris 13 University, Sorbonne Paris Cite 99, Av. Jean-Baptiste Clément, 93430 Villetaneuse, France.
| | - Pietro Matricardi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
47
|
de Souza AB, Chaud MV, Santana MHA. Hyaluronic acid behavior in oral administration and perspectives for nanotechnology-based formulations: A review. Carbohydr Polym 2019; 222:115001. [DOI: 10.1016/j.carbpol.2019.115001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/15/2019] [Accepted: 06/16/2019] [Indexed: 12/17/2022]
|
48
|
Şahin Ş, Bilgiç E, Salimi K, Tuncel A, Karaosmanoğlu B, Taşkıran EZ, Korkusuz P, Korkusuz F. Development, characterization and research of efficacy on in vitro cell culture of glucosamine carrying hyaluronic acid nanoparticles. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
49
|
Choi KY, Han HS, Lee ES, Shin JM, Almquist BD, Lee DS, Park JH. Hyaluronic Acid-Based Activatable Nanomaterials for Stimuli-Responsive Imaging and Therapeutics: Beyond CD44-Mediated Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803549. [PMID: 30773699 DOI: 10.1002/adma.201803549] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 12/27/2018] [Indexed: 05/24/2023]
Abstract
There is a rapidly increasing interest in developing stimuli-responsive nanomaterials for treating a variety of diseases. By enabling the activation of function locally at the sites of interest, it is possible to increase therapeutic efficacy significantly while simultaneously reducing adverse side effects. While there are many sophisticated nanomaterials available, they are often highly complex and not easily transferrable to industrial scales and clinical settings. However, nanomaterials based on hyaluronic acid offer a compelling strategy for reducing their complexity while retaining several desirable benefits such as active targeting and stimuli-responsive degradation. Herein, the basic properties of hyaluronic acid, its binding partners, and natural routes for degradation by hyaluronidases-hyaluronic-acid-degrading enzymes-and oxidative stresses are discussed. Recent advances in designing hyaluronic acid-based, actively targeted, hyaluronidase- or reactive-oxygen-species-responsive nanomaterials for both diagnostic imaging and therapeutic delivery, which go beyond merely the classical targeting of CD44, are summarized.
Collapse
Affiliation(s)
- Ki Young Choi
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Hwa Seung Han
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, Gangneung, 25451, Republic of Korea
| | - Eun Sook Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung Min Shin
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | | | - Doo Sung Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Theranostic Macromolecules Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
50
|
Fragai M, Ravera E, Tedoldi F, Luchinat C, Parigi G. Relaxivity of Gd-Based MRI Contrast Agents in Crosslinked Hyaluronic Acid as a Model for Tissues. Chemphyschem 2019; 20:2204-2209. [PMID: 31298452 DOI: 10.1002/cphc.201900587] [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: 06/11/2019] [Revised: 07/11/2019] [Indexed: 12/12/2022]
Abstract
The efficiency of MRI contrast agents depends on the relaxation rate enhancement that they can induce at imaging fields. It is well known that, at these fields, large relaxation rates are obtained by binding of gadolinium(III) ions to large molecules. By the same token, the interaction of the gadolinium(III) complexes with macromolecules that are found in biological tissues can be responsible for an increase of the relaxation rate with respect to the value observed in liquids. We investigate here the relaxation enhancement of gadoteridol (Gd-HP-DO3A) in crosslinked hyaluronic acid, taken as model tissue, using fast field-cycling relaxometry. The analysis of the relaxation profiles as a function of the magnetic fields indicates that a sizable increase in the relaxation rates is due to a modest interaction of the contrast agent with the hydrogel and to the slower mobility of the water molecules outside the first-coordination sphere of the gadolinium(III) ion.
Collapse
Affiliation(s)
- Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, Italy.,Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Enrico Ravera
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, Italy.,Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Fabio Tedoldi
- Bracco Imaging Spa, Bracco Research Centre, Via Ribes 5, 10010, Colleretto Giacosa (TO), Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, Italy.,Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Giacomo Parigi
- Magnetic Resonance Center (CERM), University of Florence, and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), via Sacconi 6, Sesto Fiorentino, Italy.,Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|