1
|
Wu D, Li Y, Dai Y, Tian H, Chen Y, Shen G, Yang G. Stabilization of chitosan-based nanomedicines in cancer therapy: a review. Int J Biol Macromol 2025; 309:143016. [PMID: 40216118 DOI: 10.1016/j.ijbiomac.2025.143016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Chitosan (CS), a versatile and alkaline polysaccharide, has gained significant attention in nanomedicine due to its biocompatibility and biodegradability. In recent years, its applications in cancer therapy, particularly for the delivery of chemotherapeutic drugs, diagnostic agents, and genes, have advanced considerably. However, many CS-based nanomedicines suffer from poor stability in biological fluids, especially under physiological conditions. The neutral pH and the presence of electrolytes in physiological environments reduce the charge density of CS, which can account for this application limitation of CS-based nanomedicines. To improve the stability and prevent dissociation or aggregation of these nanomedicines before reaching the target sites, this review summarizes common stabilization strategies including hydrophilic or hydrophobic modification of CS, as well as incorporation with metal ions (e.g. Fe3+ or Zn2+), complexation with anionic cross-linkers (e.g. TPP) or anionic polymers. Additionally, the review highlights the application of stabilized CS-based nanocarriers in drug delivery, with a particular focus on cancer therapy. The challenges and future perspectives for accelerating the clinical translation of these nanomedicines are also discussed.
Collapse
Affiliation(s)
- Danjun Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yazhen Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yiwei Dai
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Tian
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yifei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gongmin Shen
- Hangzhou Guoguang Pharmaceutical Co., Ltd., Hangzhou 310018, China.
| | - Gensheng Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
2
|
Quiroga AC, Gimeno-Martos S, Lorenzo PL, Arias Álvarez M, G Rebollar P, García-García RM. Microencapsulated rrBNGF as an alternative ovulation induction method in rabbits. BMC Vet Res 2025; 21:133. [PMID: 40025466 PMCID: PMC11874391 DOI: 10.1186/s12917-025-04547-9] [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: 02/14/2024] [Accepted: 01/30/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Rabbits are an induced-ovulatory species such that exogenous hormone factors are needed to induce ovulation. Traditionally, intramuscular injections of gonadotropin-releasing hormone (GnRH) analogues are given at the time of artificial insemination (AI). To avoid the need for injections, the intravaginal delivery of molecules naturally present in seminal plasma has been explored. Here, we examined the possibility of using nerve growth factor (NGF) microencapsulated with chitosan to induce ovulation. First, the biological activity of these NGF microcapsules was assessed in pheochromocytoma of rat adrenal medulla cell (PC12) cultures, along with their effects on semen. Next, we examined the ability of the intravaginal NGF-chitosan delivery system administered at AI (NGFch-0) or 30 min before AI (NGFch-30) to elicit ovulation. To this end, progesterone concentrations on Day 7 post AI, pregnancy rates and prolificacy (kits born alive and stillbirths per doe) were determined in nulliparous and multiparous rabbit does and then compared amongst treatments: intravaginal NGFch-0 and NGFch-30, intramuscular injection of GnRH analogue, intravaginal empty-catheter (C-e) or intravaginal semen-containing catheter (C-s). RESULTS NGF-chitosan promoted similar PC12 differentiation to free NGF without impairing cell viability. The presence of the NGF-containing microcapsules did not interfere with semen motility, viability or capacitation status. In our in vivo experiments, nulliparous rabbits showed similar rates of ovulating females across treatments (GnRH 90%, NGFch-30 100%, NGFch-0 66.7%, C-e 83.3%), yet higher pregnancy rates were observed in response to GnRH and NGFch-30 (90% and 100%, respectively) than to NGFch-0 (60%). Prolificacy results in these does were similar across treatments. In multiparous does, GnRH treatment gave rise to the highest rate of ovulating female and pregnancy rates (100 and 90%, respectively). In contrast, the NGF-chitosan groups showed the lowest ovulating female and pregnancy rates (NGFch-30 50% and 25%, NGFch-0 41.7% and 21%, respectively). An intermediate ovulatory response was obtained in does stimulated with the catheter (C-e 70%, C-s 57.1%), and a pregnancy rate of 20% was obtained if the catheter contained diluted semen (C-s). CONCLUSIONS Intravaginal NGF-chitosan administered 30 min before AI induced ovulation at a similar rate to GnRH injection in nulliparous, but not multiparous, rabbit females. A better receptivity status of nulliparous females could be a determining factor for this response. However, mechanical stimulation gave rise to a high ovulation rate, so this could be masking or, in some cases, directly replacing the NGF-chitosan effect.
Collapse
Affiliation(s)
- Alejandra C Quiroga
- Department of Physiology, Veterinary Faculty, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Silvia Gimeno-Martos
- Department of Physiology, Veterinary Faculty, Complutense University of Madrid, 28040, Madrid, Spain
- Department of Biochemistry and Molecular and Cellular Biology, Veterinary Faculty Zaragoza University, 50013, Zaragoza, Spain
| | - Pedro L Lorenzo
- Department of Physiology, Veterinary Faculty, Complutense University of Madrid, 28040, Madrid, Spain
| | - María Arias Álvarez
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, 28040, Madrid, Spain
| | - Pilar G Rebollar
- Department of Agrarian Production, ETSIAAB, Universidad Politécnica de Madrid, 28040, Madrid, Spain.
| | - Rosa M García-García
- Department of Physiology, Veterinary Faculty, Complutense University of Madrid, 28040, Madrid, Spain
| |
Collapse
|
3
|
Rochima E, Ilman B, Sektiaji RGB, Lili W, Pratama RI, Utama GL, Damayanti W, Azhary SY, Panatarani C, Joni IM. The influence of nanochitosan-incorporated edible coating on the characteristics of Pangasius (Pangasius sp.) fillet. Food Chem 2025; 464:141623. [PMID: 39442215 DOI: 10.1016/j.foodchem.2024.141623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Nanochitosan is known to have the potential to protect fish fillets, highly perishable products, as it can penetrate deep into the meat tissue and has high antibacterial activity. This study aimed to analyze the effect of nanochitosan incorporation to edible coating in inhibiting the degradation of Pangasius fillet quality during storage. The fillets were soaked with 2 % chitosan and nanochitosan concentrations (1 %, 2 %, and 3 %), stored for 15 days, and its quality was assessed periodically. On the 10th day of storage, the fillet coated with 2 % nanochitosan-incorporated edible coating (NEC) showed the lowest weight loss (1.41 ± 0.01 %) and maintained the pH close to neutral (6.92 ± 0.08), along with low TVB-N (33.55 mg N/100 g). and TPC (4.7 × 105 CFU/g) value, fulfilling the standard set by Indonesian National Standardization Agency. Based on the results, nanochitosan proved to extended the storage duration of Pangasius fillet while maintaining its quality.
Collapse
Affiliation(s)
- Emma Rochima
- Department of Fishery, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Research Collaboration Center for Marine Biomaterials, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Bachrul Ilman
- Department of Fishery, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Radika Gilang Bayu Sektiaji
- Department of Fishery, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Walim Lili
- Department of Fishery, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Rusky Intan Pratama
- Department of Fishery, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Gemilang Lara Utama
- Faculty of Agro-Industrial Technology, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Nanotechnology and Graphene Research Center, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Windi Damayanti
- Biogeochemical Laboratory, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - Sundoro Yoga Azhary
- Department of Fishery, Faculty of Fisheries and Marine Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia
| | - Camellia Panatarani
- Research Collaboration Center for Marine Biomaterials, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| | - I Made Joni
- Research Collaboration Center for Marine Biomaterials, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia; Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor 45363, West Java, Indonesia.
| |
Collapse
|
4
|
Aydemir D, Öztürk K, Arslan FB, Çalis S, Ulusu NN. Gemcitabine-loaded chitosan nanoparticles enhanced apoptotic and ferroptotic response of gemcitabine treatment alone in the pancreatic cancer cells in vitro. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9051-9066. [PMID: 38884675 PMCID: PMC11522156 DOI: 10.1007/s00210-024-03193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024]
Abstract
Gemcitabine (GEM) is a first-line treatment for pancreatic ductal adenocarcinoma (PDAC) patients, causing side effects and poor overall survival. Eighty percent of patients often develop resistance rapidly to GEM. Developing therapeutic approaches and increasing sensitivity to gemcitabine in PDAC has become one of the challenges in cancer research. We synthesized GEM-loaded NPs prepared with a method that combines ultrasonication and ionotropic gelation to overcome GEM-related limitations in PDAC. CFPAC-1 cells were treated with increased concentrations of GEM, empty chitosan, and GEM-loaded NPs (0.66, 1.32, 2.64, 5.32 µg/ml) for up to 48 h. Empty chitosan NPs did not show toxicity on L929 cells. Antioxidant enzyme activities, including glucose 6-phosphate dehydrogenase (G6PD), 6-phosphogluconate dehydrogenase (6-PGD), glutathione reductase (GR), glutathione s-transferase (GST), and glutathione peroxidase (GPx), significantly reduced in GEM-loaded NPs compared to the GEM associated with increased oxidative stress, PPP, and glycolysis. Bcl-xL, NOXA/mcl-1, and Ca2+ levels significantly increased in GEM-loaded NP-administered cells compared to the GEM and control groups. In contrast, JNK, p38, STAT3, Akt, and CREB levels significantly decreased in the GEM-loaded NP group, addressing enhanced apoptotic response compared to the GEM alone. Increased ferroptosis activity in GEM-loaded NP-administered groups has been validated via decreased antioxidant enzyme activities, increased cytosolic Fe, Zn, Mg, and Mn levels, and reduced GPx activity compared to the GEM and control groups. For the first time in the literature, we showed biocompatible GEM-loaded NPs enhanced apoptotic and ferroptotic response in CFPAC-1 cells via downregulation of antioxidant, glycolysis, and PPP metabolism compared to the GEM alone.
Collapse
Affiliation(s)
- Duygu Aydemir
- School of Medicine, Department of Medical Biochemistry, Koc University, Istanbul, Turkey.
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
- Biochemistry Department, Koc University School of Medicine, Rumelifeneri Yolu, Sariyer, Istanbul, 34450, Turkey.
| | - Kıvılcım Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Fatma Betül Arslan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Çalis
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Nuriye Nuray Ulusu
- School of Medicine, Department of Medical Biochemistry, Koc University, Istanbul, Turkey.
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
- Biochemistry Department, Koc University School of Medicine, Rumelifeneri Yolu, Sariyer, Istanbul, 34450, Turkey.
| |
Collapse
|
5
|
Maršík D, Maťátková O, Kolková A, Masák J. Exploring the antimicrobial potential of chitosan nanoparticles: synthesis, characterization and impact on Pseudomonas aeruginosa virulence factors. NANOSCALE ADVANCES 2024; 6:3093-3105. [PMID: 38868829 PMCID: PMC11166115 DOI: 10.1039/d4na00064a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/21/2024] [Indexed: 06/14/2024]
Abstract
The escalating antibiotic resistance observed in bacteria poses a significant threat to society, with the global prevalence of resistant strains of Pseudomonas aeruginosa on the rise. Addressing this challenge necessitates exploring strategies that would complement existing antimicrobial agents, e.g. by substances mitigating bacterial virulence without eliciting selective pressure for resistance emergence. In this respect, free-form chitosan has demonstrated promising efficacy, prompting our investigation into reinforcing its effects through nanoparticle formulations. Our study focuses on the preparation of chitosan nanoparticles under suitable conditions while emphasizing the challenges associated with stability that can affect biological activity. These challenges are mitigated by introducing quaternized chitosan, which ensures colloidal stability in the culture media. Our approach led to the production of trimethylchitosan nanoparticles with a median size of 103 nm, circularity of 0.967, and a charge of 14.9 ± 3.1 mV, stable within a one-month period in a water stock solution, showing promising attributes for further valorization. Furthermore, the study delves into the antimicrobial activity of trimethylchitosan nanoparticles on Pseudomonas aeruginosa and confirms the benefits of both nanoformulation and modification of chitosan, as our prepared nanoparticles inhibit 50% of the bacterial population at concentration ≥160 mg L-1 within tested strains. Additionally, we identified a concentration of 5 mg L-1 that no longer impedes bacterial growth, allowing reliable verification of the effect of the prepared nanoparticles on Pseudomonas aeruginosa virulence factors, including motility, protease activity, hemolytic activity, rhamnolipids, pyocyanin, and biofilm production. Although trimethylchitosan nanoparticles exhibit promise as an effective antibiofilm agent (reducing biofilm development by 50% at concentrations ranging from 80 to 160 mg L-1) their impact on virulence manifestation is likely not directly associated with quorum sensing. Instead, it can probably be attributed to non-specific interactions with the bacterial surface. This exploration provides valuable insights into the potential of quaternized chitosan nanoparticles in addressing Pseudomonas aeruginosa infections and underscores the multifaceted nature of their antimicrobial effects.
Collapse
Affiliation(s)
- Dominik Maršík
- Department of Biotechnology, University of Chemistry and Technology Technická 5, Prague 6 Prague 166 28 Czechia
| | - Olga Maťátková
- Department of Biotechnology, University of Chemistry and Technology Technická 5, Prague 6 Prague 166 28 Czechia
| | - Anna Kolková
- Department of Biotechnology, University of Chemistry and Technology Technická 5, Prague 6 Prague 166 28 Czechia
| | - Jan Masák
- Department of Biotechnology, University of Chemistry and Technology Technická 5, Prague 6 Prague 166 28 Czechia
| |
Collapse
|
6
|
Meher K, Radha G, Lopus M. Induction of autophagy-dependent and caspase- and microtubule-acetylation-independent cell death by phytochemical-stabilized gold nanopolygons in colorectal adenocarcinoma cells. NANOSCALE 2024; 16:7976-7987. [PMID: 38567463 DOI: 10.1039/d4nr00730a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Collective functionalization of the phytochemicals of medicinal herbs on nanoparticles is emerging as a potential cancer therapeutic strategy. This study presents the facile synthesis of surface-functionalized gold nanoparticles using Bacopa monnieri (Brahmi; Bm) phytochemicals and their therapeutically relevant mechanism of action in the colorectal cancer cell line, HT29. The nanoparticles were characterized using UV-visible spectroscopy, TEM-EDAX, zeta potential analysis, TGA, FTIR and 1H NMR spectroscopy, and HR-LC-MS. The particles (Bm-GNPs) were of polygonal shape and were stable against aggregation. They entered the target cells and inhibited the viability and clonogenicity of the cells with eight times more antiproliferative efficacy (25 ± 1.5 μg mL-1) than Bm extract (Bm-EX). In vitro studies revealed that Bm-GNPs bind tubulin (a protein crucial in cell division and a target of anticancer drugs) and disrupt its helical structure without grossly altering its tertiary conformation. Like other antitubulin agents, Bm-GNPs induced G2/M arrest and ultimately killed the cells, as confirmed using flow cytometry analyses. ZVAD-FMK-mediated global pan-caspase inhibition and the apparent absence of cleaved caspase-3 in treated cells indicated that the death did not involve the classic apoptosis pathway. Cellular ultrastructure analyses, western immunoblots, and in situ immunofluorescence visualization of cellular microtubules revealed microtubule-acetylation-independent induction of autophagy as the facilitator of cell death. Together, the data indicate strong antiproliferative efficacy and a possible mechanism of action for these designer nanoparticles. Bm-GNPs, therefore, merit further investigations, including preclinical evaluations, for their therapeutic potential as inducers of non-apoptotic cell death.
Collapse
Affiliation(s)
- Kimaya Meher
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai-400098, India.
| | - Gudapureddy Radha
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai-400098, India.
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai-400098, India.
| |
Collapse
|
7
|
Fasquelle F, Scuotto A, Howsam M, Betbeder D. Maltodextrin-Nanoparticles as a Delivery System for Nasal Vaccines: A Review Article. Pharmaceutics 2024; 16:247. [PMID: 38399301 PMCID: PMC10892173 DOI: 10.3390/pharmaceutics16020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/24/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Nanoparticles are increasingly being studied as antigen delivery systems for immunization with nasal vaccines. The addition of adjuvants is still generally required in many nanoparticle formulations, which can induce potential side effects owing to mucosal reactogenicity. In contrast, maltodextrin nanoparticles do not require additional immunomodulators, and have been shown to be efficient vaccine delivery systems. In this review, the development of maltodextrin nanoparticles is presented, specifically their physico-chemical properties, their ability to load antigens and deliver them into airway mucosal cells, and the extent to which they trigger protective immune responses against bacterial, viral, and parasitic infections. We demonstrate that the addition of lipids to maltodextrin nanoparticles increases their potency as a vaccine delivery system for nasal administration.
Collapse
Affiliation(s)
| | | | - Michael Howsam
- Université de Lille, Inserm, Centre Hospitalier de Lille, Institut Pasteur de Lille, U1167—RID-AGE—Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | | |
Collapse
|
8
|
Souiade L, Domingo-Diez J, Alcaide C, Gámez B, Gámez L, Ramos M, Serrano Olmedo JJ. Improving the Efficacy of Magnetic Nanoparticle-Mediated Hyperthermia Using Trapezoidal Pulsed Electromagnetic Fields as an In Vitro Anticancer Treatment in Melanoma and Glioblastoma Multiforme Cell Lines. Int J Mol Sci 2023; 24:15933. [PMID: 37958913 PMCID: PMC10648011 DOI: 10.3390/ijms242115933] [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: 10/08/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Magnetic hyperthermia (MHT) is an oncological therapy that uses magnetic nanoparticles (MNPs) to generate localized heat under a low-frequency alternating magnetic field (AMF). Recently, trapezoidal pulsed alternating magnetic fields (TPAMFs) have proven their efficacy in enhancing the efficiency of heating in MHT as compared to the sinusoidal one. Our study aims to compare the TPAMF waveform's killing effect against the sinusoidal waveform in B16F10 and CT2A cell lines to determine more efficient waveforms in causing cell death. For that purpose, we used MNPs and different AMF waveforms: trapezoidal (TP), almost-square (TS), triangular (TR), and sinusoidal signal (SN). MNPs at 1 and 4 mg/mL did not affect cell viability during treatment. The exposition of B16F10 and CT2A cells to only AMF showed nonsignificant mortality. Hence, the synergetic effect of the AMF and MNPs causes the observed cell death. Among the explored cases, the nonharmonic signals demonstrated better efficacy than the SN one as an MHT treatment. This study has revealed that the application of TP, TS, or TR waveforms is more efficient and has considerable capability to increase cancer cell death compared to the traditional sinusoidal treatment. Overall, we can conclude that the application of nonharmonic signals enhances MHT treatment efficiency against tumor cells.
Collapse
Affiliation(s)
- Lilia Souiade
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (L.S.); (J.D.-D.); (C.A.); (M.R.)
| | - Javier Domingo-Diez
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (L.S.); (J.D.-D.); (C.A.); (M.R.)
| | - Cesar Alcaide
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (L.S.); (J.D.-D.); (C.A.); (M.R.)
| | - Berta Gámez
- Escula Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, 28006 Madrid, Spain; (B.G.); (L.G.)
| | - Linarejos Gámez
- Escula Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, 28006 Madrid, Spain; (B.G.); (L.G.)
| | - Milagros Ramos
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (L.S.); (J.D.-D.); (C.A.); (M.R.)
- Centro de Investigación Biomédica en Red para Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Javier Serrano Olmedo
- Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain; (L.S.); (J.D.-D.); (C.A.); (M.R.)
- Centro de Investigación Biomédica en Red para Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
9
|
Visan RM, Leonties AR, Anastasescu M, Angelescu DG. Towards understanding the interaction of quercetin with chitosan-phytate complex: An experimental and computational investigation. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
10
|
Bulatao BP, Nalinratana N, Jantaratana P, Vajragupta O, Rojsitthisak P, Rojsitthisak P. Lutein-loaded chitosan/alginate-coated Fe 3O 4 nanoparticles as effective targeted carriers for breast cancer treatment. Int J Biol Macromol 2023; 242:124673. [PMID: 37137353 DOI: 10.1016/j.ijbiomac.2023.124673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/07/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Magnetic drug targeting can be a strategy for effectively delivering phytochemicals in cancer treatment. Here, we demonstrate the benefit of magnetic targeting with superparamagnetic iron oxide nanoparticles for cytotoxicity enhancement of lutein (LUT) against breast cancer cells. Fabrication of LUT-loaded chitosan/alginate iron oxide nanoparticles (LUT-CS/Alg-Fe3O4-NPs) was optimized by a statistical approach using response surface methodology based on the Box-Behnken design. The optimized LUT-CS/Alg-Fe3O4-NPs with a balance among LUT concentration, copolymer coating, and iron ion concentration exhibited controlled size, narrow size distribution, better crystallinity, excellent saturation magnetization, and sustained-release profile. The negligible magnetic coercivity and remanent magnetization confirmed the superparamagnetism of the prepared NPs. The optimized LUT-CS/Alg-Fe3O4-NPs were biocompatible while exhibiting a significantly enhanced cytotoxicity towards breast cancer MCF-7 cells upon exposure to a permanent magnet compared to free LUT with a 4-fold increase, suggesting the potential of LUT-CS/Alg-Fe3O4-NPs as magnetically targeted delivery for breast cancer.
Collapse
Affiliation(s)
- Bryan Paul Bulatao
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand; Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Nonthaneth Nalinratana
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Pongsakorn Jantaratana
- Department of Physics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand.
| | - Opa Vajragupta
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand; Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Pranee Rojsitthisak
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand; Metallurgy and Materials Science Research Institute, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Pornchai Rojsitthisak
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok 10330, Thailand; Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
11
|
Studies on anti-colon cancer potential of nanoformulations of curcumin and succinylated curcumin in mannosylated chitosan. Int J Biol Macromol 2023; 235:123827. [PMID: 36858085 DOI: 10.1016/j.ijbiomac.2023.123827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/05/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023]
Abstract
Colon cancer (CRC) is the second leading cause of death and the third most diagnosed cancer worldwide. Although curcumin (CUR) has demonstrated a potent anticancer activity, it is characterized by its poor solubility, low bioavailability, and instability. This study is a projection from a previous investigation where CUR and succinylated CUR (CUR.SA) were separately encapsulated in mannosylated-chitosan nanoparticles (CM-NPs) to form CUR-NPs and CUR.SA-NPs, respectively. Here, we aim to assess the anti-CRC activity of these two nanoformulations. Cytotoxicity studies using CCK-8 assay indicated that both CUR-NPs and CUR.SA-NPs have a dose and time-dependent toxicity towards CRC human cell-lines (HCT116 and SW480), and more cytotoxic compared to free CUR or CUR-SA in a time-dependent manner. A significant induction of early and late apoptosis in the CUR-NPs and CUR.SA-NPs treated CRC cell lines compared to untreated cells was observed. Western blotting analyses confirmed the induction of apoptosis through activation of Caspase signaling compared to untreated cells. Based on the physicochemical properties of CUR-NPs and CUR.SA-NPs along with the data from the in vitro studies, we may conclude these nanoparticle formulations hold very promising attributes, worthy of further investigations for its role in the management of CRC.
Collapse
|
12
|
Structural characterization, stability, and cytocompatibility study of chitosan BaTiO 3@ZnO:Er heterostructures. Int J Biol Macromol 2023; 235:123796. [PMID: 36822293 DOI: 10.1016/j.ijbiomac.2023.123796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/15/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023]
Abstract
New imaging agents are required in cancer diagnosis to enhance the diagnostic accuracy, classification, and therapeutic management of tumors. Nanomaterials have emerged as a promising alternative to developing new nanostructures with imaging applications. In this study, a heterostructure based on barium titanate (BT), zinc oxide (ZnO), and erbium (Er) was prepared and coated with Chitosan (CS) to investigate their stability and compatibility with biological systems. The structure, particle morphology, luminescence properties, stability, and cytotoxicity of different nanoparticles (NPs) were assessed. The results demonstrated the formation of a [BT@ZnO:Er]-CS heterostructure, which is consistent with the relative intensities and positions of peaks in the X-ray diffraction (XRD) with an average crystallite size of ~76 nm. The electrokinetic measurement results indicate that the coated NPs are the most stable and have an average size close to 200 nm when the pH is between 3 and 5. Finally, we presented a cytotoxicity study of naked and CS-coated NPs. The results indicate that naked NPs exhibit varying cellular toxicity, as indicated by decreased cell viability, morphological changes, and an increase in an apoptotic marker. The CS-coated NPs prevented the cytotoxic effect of the naked NPs, demonstrating the significance of CS as a stabilizing agent.
Collapse
|
13
|
Arslan FB, Öztürk K, Tavukçuoğlu E, Öztürk SC, Esendağlı G, Çalış S. A novel combination for the treatment of small cell lung cancer: Active targeted irinotecan and stattic co-loaded PLGA nanoparticles. Int J Pharm 2023; 632:122573. [PMID: 36592892 DOI: 10.1016/j.ijpharm.2022.122573] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022]
Abstract
Polymeric nanoparticles are widely used drug delivery systems for cancer treatment due to their properties such as ease of passing through biological membranes, opportunity to modify drug release, specifically targeting drugs to diseased areas, and potential of reducing side effects. Here, we formulated irinotecan and Stattic co-loaded PLGA nanoparticles targeted to small cell lung cancer. Nanoparticles were successfully conjugated with CD56 antibody with a conjugation efficiency of 84.39 ± 1.01%, and characterization of formulated nanoparticles was conducted with in-vitro and in-vivo studies. Formulated particles had sizes in the range of 130-180 nm with PDI values smaller than 0.3. Encapsulation and active targeting of irinotecan and Stattic resulted in increased cytotoxicity and anti-cancer efficiency in-vitro. Furthermore, it was shown with ex-vivo biodistribution studies that conjugated nanoparticles were successfully targeted to CD56-expressing SCLC cells and distributed mainly to tumor tissue and lungs. Compliant with our hypothesis and literature, the STAT3 pathway was successfully inhibited with Stattic solution and Stattic loaded nanoparticles. Additionally, intravenous injection of conjugated co-loaded nanoparticles resulted in decreased side effects and better anti-tumor activity than individual solutions of drugs in SCLC tumor-bearing mice. These results may indicate a new treatment option for clinically aggressive small cell lung cancer.
Collapse
Affiliation(s)
- Fatma Betül Arslan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkiye
| | - Kıvılcım Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkiye
| | - Ece Tavukçuoğlu
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkiye
| | - Süleyman Can Öztürk
- Centre for Laboratory Animals Research and Application, Hacettepe University, Ankara, Turkiye
| | - Güneş Esendağlı
- Department of Basic Oncology, Hacettepe University Cancer Institute, 06100 Ankara, Turkiye
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkiye.
| |
Collapse
|
14
|
Mabrouk AA, El-Mezayen NS, Awaad AK, Tadros MI, El-Gazayerly ON, El-Refaie WM. Novel celecoxib-loaded chitosan-fucoidan nanoparticles as potential immunotherapy for oral squamous cell carcinoma: Mechanistic insights. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
15
|
Sarkar A, Roy S, Bhatia P, Jaiswal A. Quaternary ammonium substituted dextrin‐based biocompatible cationic nanoparticles with ultrahigh
pH
stability for drug delivery. J Appl Polym Sci 2023. [DOI: 10.1002/app.53626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Ankita Sarkar
- School of Biosciences and Bioengineering Indian Institute of Technology Mandi Mandi India
| | - Shounak Roy
- School of Biosciences and Bioengineering Indian Institute of Technology Mandi Mandi India
| | - Prachi Bhatia
- School of Biosciences and Bioengineering Indian Institute of Technology Mandi Mandi India
| | - Amit Jaiswal
- School of Biosciences and Bioengineering Indian Institute of Technology Mandi Mandi India
| |
Collapse
|
16
|
Recent developments of nanomedicine delivery systems for the treatment of pancreatic cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
17
|
Esim O, Oztuna A, Sarper M, Hascicek C. Chitosan-coated bovine serum albumin nanocarriers mediate efficient delivery of methotrexate in breast cancer therapeutics. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
18
|
Nunes R, Serra AS, Simaite A, Sousa Â. Modulation of Chitosan-TPP Nanoparticle Properties for Plasmid DNA Vaccines Delivery. Polymers (Basel) 2022; 14:1443. [PMID: 35406316 PMCID: PMC9003200 DOI: 10.3390/polym14071443] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 12/27/2022] Open
Abstract
Nucleic acid vaccines have become a revolutionary technology to give a fast, safe, cost-effective and efficient response against viral infections, such as SARS-CoV-2 or Human papillomavirus (HPV). However, to ensure their effectiveness, the development of adequate methods to protect, carry, and deliver nucleic acids is fundamental. In this work, nanoparticles (NPs) of chitosan (CS)-tripolyphosphate (TPP)-plasmid DNA (pDNA) were thoroughly modulated and characterized, by measuring the charge and size through dynamic light scattering (DLS) and morphology by scanning electron microscopy (SEM). Stability, cytotoxicity and cellular uptake of NPs were also evaluated. Finally, the effect of polyplexes on the expression of HPV E7 antigen in human fibroblast and RAW cells was investigated through polymerase chain reaction (PCR) and real-time PCR. The results showed NPs with a spherical/oval shape, narrow size distribution <180 nm and positive zeta potentials (>20 mV) and good stability after one month of storage at 4 °C in formulation buffer or when incubated in culture medium and trypsin. In vitro studies of NPs cytotoxicity revealed that the elimination of formulation buffers led to an improvement in the rate of cell viability. The E7 antigen transcription was also increased for NPs obtained with high pDNA concentration (60 μg/mL). The analyzed CS-TPP-pDNA polyplexes can offer a promising vehicle for nucleic acid vaccines, not only in the prevention or treatment of viral infections, but also to fight emergent and future pathogens.
Collapse
Affiliation(s)
- Renato Nunes
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (R.N.); (A.S.S.)
- InoCure s.r.o, R&D Laboratory Center, Prumyslová 1960, 250 88 Celákovice, Czech Republic;
| | - Ana Sofia Serra
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (R.N.); (A.S.S.)
| | - Aiva Simaite
- InoCure s.r.o, R&D Laboratory Center, Prumyslová 1960, 250 88 Celákovice, Czech Republic;
| | - Ângela Sousa
- CICS-UBI—Health Sciences Research Center, University of Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (R.N.); (A.S.S.)
| |
Collapse
|
19
|
Visan RM, Leonties AR, Aricov L, Chihaia V, Angelescu DG. Polymorphism of chitosan-based networks stabilized by phytate investigated by molecular dynamics simulations. Phys Chem Chem Phys 2021; 23:22601-22612. [PMID: 34591050 DOI: 10.1039/d1cp02961d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chitosan can associate in the presence of polyphosphates into insoluble hydrogels capable of drug encapsulation and safe and efficient release. On the one hand, chitosan hydrogels were synthesized using the phytate anion as a crosslinking agent and were characterized by employing dynamic light scattering (DLS) and Fourier transform infrared spectroscopy (FTIR). On the other hand, an effective chitosan-phytate model with atomistic details was created to examine the underlying physical crosslinking pattern, and the structure and dynamics of the chitosan-phytate complex were systematically investigated by using molecular dynamics (MD) simulations. To harbor the crosslinker potential for obtaining chitosan-based hydrogels, the impact of the phytate concentration and the functional groups of the chitosan on the reticulation process was addressed. The phytate association was determined by the phosphates' capacity for H-bonding to the amine and hydroxyl groups belonging to two consecutive glucosidic units. The physical crosslinking pattern was determined by the number of chitosan chains bound by one phytate anion and the phytate orientation relative to the glucopyranose neighbors. Cross-linking of two up to six chitosan chains mediated by a phytate anion represented favorable states, and the number distribution of cross-linked chains depended on the phytate concentration. The circular distribution of the cross-linkable phosphates regulated the nearly isotropic orientation of the chitosan chains and phytate at the junction, and the variety of topological crosslinking demonstrated the phytate ion's potential for developing chitosan-based hydrogels with improved structural attributes.
Collapse
Affiliation(s)
- Raluca M Visan
- Romanian Academy, "Ilie Murgulescu" Institute of Physical Chemistry, Splaiul Independentei 202, 060021 Bucharest, Romania.
| | - Anca R Leonties
- Romanian Academy, "Ilie Murgulescu" Institute of Physical Chemistry, Splaiul Independentei 202, 060021 Bucharest, Romania.
| | - Ludmila Aricov
- Romanian Academy, "Ilie Murgulescu" Institute of Physical Chemistry, Splaiul Independentei 202, 060021 Bucharest, Romania.
| | - Viorel Chihaia
- Romanian Academy, "Ilie Murgulescu" Institute of Physical Chemistry, Splaiul Independentei 202, 060021 Bucharest, Romania.
| | - Daniel G Angelescu
- Romanian Academy, "Ilie Murgulescu" Institute of Physical Chemistry, Splaiul Independentei 202, 060021 Bucharest, Romania.
| |
Collapse
|
20
|
Passos Gibson V, Nunes JBB, Falcao DQ, Roullin VG, Leblond Chain J. Lipid Coating of Chitosan Nanogels for Improved Colloidal Stability and In Vitro Biocompatibility. AAPS PharmSciTech 2021; 22:159. [PMID: 34019243 DOI: 10.1208/s12249-021-02027-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/27/2021] [Indexed: 11/30/2022] Open
Abstract
Chitosan-based carriers have coined their position as delivery agents. When assembled with polyanions into nanogels (NG), these vectors have enabled the delivery of drugs, genes, and proteins to a myriad of applications. However, the chemical and colloidal instability of chitosan nanoformulations in physiologically compatible media prejudices in vitro biocompatibility and, thus, scale-up applications. To overcome this issue, we envisaged the coating of chitosan nanogel with phospholipids. In this investigation, we report a two-stage synthesis of hybrid lipid-coated chitosan nanogels, named nanolipogels (NLG), to improve colloidal stability and in vitro biocompatibility over chitosan NG. Practically, we employed a mixing platform to first prepare chitosan NG by ionic gelation, dilute the suspension, and, in a second stage, coat the NG with lipids. We demonstrate that lipid coating increased particle size and reversed the ζ-potential to negative values, suggesting the successful formation of NLG, while maintaining a homogeneous size distribution (PDI < 0.25). Furthermore, multiple light scattering analysis confirmed NLG improved colloidal stability in phosphate buffer saline and cell culture medium, with respect to NG. Finally, lipid coating completely abrogated the cytotoxicity of NG when incubated at 50 μg·mL-1 with HeLa, U87, or b.End3 cell lines and significantly improved the biocompatibility at 100 and 150 μg·mL-1. Future investigations will explore how the lipid coating affects drug loading, release profile, and the ability of NLG to deliver drugs and genes in vitro.
Collapse
|
21
|
Mushtaq A, Li L, A A, Grøndahl L. Chitosan Nanomedicine in Cancer Therapy: Targeted Delivery and Cellular Uptake. Macromol Biosci 2021; 21:e2100005. [PMID: 33738977 DOI: 10.1002/mabi.202100005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/22/2021] [Indexed: 12/11/2022]
Abstract
Nanomedicine has gained much attention for the management and treatment of cancers due to the distinctive physicochemical properties of the drug-loaded particles. Chitosan's cationic nature is attractive for the development of such particles for drug delivery, transfection, and controlled release. The particle properties can be improved by modification of the polymer or the particle themselves. The physicochemical properties of chitosan particles are analyzed in 126 recent studies, which allows to highlight their impact on passive and active targeted drug delivery, cellular uptake, and tumor growth inhibition (TGI). From 2012 to 2019, out of 40 in vivo studies, only 4 studies are found reporting a reduction in tumor size by using chitosan particles while all other studies reported tumor growth inhibition relative to controls. A total of 23 studies are analyzed for cellular uptake including 12 studies reporting cellular uptake mechanisms. Understanding and exploiting the processes involved in targeted delivery, endocytosis, and exocytosis by controlling the physicochemical properties of chitosan particles are important for the development of safe and efficient nanomedicine. It is concluded based on the recent literature available on chitosan particles that combination therapies can play a pivotal role in transformation of chitosan nanomedicine from bench to bedside.
Collapse
Affiliation(s)
- Asim Mushtaq
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 68, Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Corner of College and Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Anitha A
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 68, Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Lisbeth Grøndahl
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 68, Cooper Road, Brisbane, Queensland, 4072, Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Corner of College and Cooper Road, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
22
|
Öztürk-Atar K, Kaplan M, Çalış S. Development and evaluation of polymeric micelle containing tablet formulation for poorly water-soluble drug: tamoxifen citrate. Drug Dev Ind Pharm 2020; 46:1695-1704. [PMID: 32893676 DOI: 10.1080/03639045.2020.1820037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Poor aqueous solubility is one of the key reasons for slow dissolution rate and poor intestinal absorption and finally that causes low therapeutic efficacy of many existing drugs. Tamoxifen citrate (TMX) (BCS Class II drug) with low water solubility has poor oral bioavailability in the range of 20%-30%, therefore, high doses are required for treatment with TMX. Self-assemblage of amphiphilic polymers leads to the formation of polymeric micelles which makes them unique nano-carriers with excellent biocompatibility, low toxicity, enhanced blood circulation time, and solubilization of poorly water-soluble drugs. In this study poly(ethylene glycol)-block-poly(propylene glycol)-block-poly(ethylene glycol) triblock copolymer, which has been approved by FDA for oral application was used to benefit its micellar solubilization effect. Self-assembled micelles were prepared for the delivery of TMX and this way TMX solubility was increased approximately 60 times. TMX-treated cells showed 38.06 ± 1.5% viability at 50 µM concentration for 24 h; 66.71 ± 11.6% viability at 25 µM concentration for 48 h, at the same conditions TMX-loaded micelles exhibited 24.994 ± 0.25% and 43.36 ± 4.37% cell viability, respectively (p < 0.05). These results showed that the encapsulation of TMX into PEG-PPG-PEG micelles facilitated the cellular uptake, which led to an increased cytotoxicity in MCF-7 cancer cells. Tablet formulation containing lyophilized TMX-loaded micelles was showed an improved dissolution than commercial TMX tablet (Tamoxifen® TEVA). It can be reasonably expected that the obtained drug dissolution rate and increased cytotoxicity to tumor cells will result in an increase of TMX bioavailability and tolerability associated with an important dose reduction and decreased side effects.
Collapse
Affiliation(s)
- Kıvılcım Öztürk-Atar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Meryem Kaplan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|