Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 253(Pt 3): 126846, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37717866

ABSTRACT

The worldwide prevalence of cancer and its significantly rising risks with age have garnered the attention of nanotechnology for prompt detection and effective therapy with minimal or no adverse effects. In the current study, heparin (HP) polymer derived heteroatom (N, S-) co-doped CDs were synthesized using hydrothermal synthesis method to efficiently deliver natural anticancer compound baicalin (BA). Heparin carbon dots (HCDs) were passivated with polyethylenimine (PEI) to improve its fluorescence quantum yield. The surface passivation of CDs by polycationic PEI polymer not only facilitated loading of BA, but also played a crucial role in the pH-responsive drug delivery. The sustained release of BA (up to 80 %) in mildly acidic pH (5.5 and 6.5) conditions endorsed its drug delivery potential for cancer-specific microenvironments. BA-loaded PHCDs exhibited enhanced anticancer activity as compared to BA/PHCDs indicating the effectiveness of the nanoformulation, Furthermore, the flow cytometry analysis confirmed that BA-PHCDs treated cells were arrested in the G2/M phase of cell cycle and had a higher potential for apoptosis. Bioimaging study demonstrated the excellent cell penetration efficiency of PHCDs with complete cytoplasmic localization. All this evidence comprehensively demonstrates the potency of BA-loaded PHCDs as a nanotheranostic agent for cancer.


Subject(s)
Neoplasms , Quantum Dots , Humans , Quantum Dots/chemistry , Polyethyleneimine/chemistry , Precision Medicine , Carbon/chemistry , Heparin/pharmacology , Neoplasms/drug therapy , Tumor Microenvironment
2.
Sci Rep ; 11(1): 8721, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888736

ABSTRACT

Emergence of nanotechnology created a drastic change in the field of cancer therapy due to their unique features in drug delivery and imaging. Polysaccharide based nanoparticles have received extensive attention in recent years as promising nanoparticle mediated drug delivery systems. Polysaccharides are endorsed with versatile merits including high drug encapsulation efficiency, efficient drug protection against chemical or enzymatic degradation, unique ability to create a controlled release and cellular internalization. In the current study, we have fabricated doxorubicin-loaded carboxymethylated PST001 coated iron oxide nanoparticles (DOX@CM-PST-IONPs) for better management of cancer. CM-PST coated iron oxide nanoparticles co-encapsulated with chemotherapeutic drug doxorubicin, can be utilized for targeted drug delivery. Biocompatible and non-toxic nanoconjugates was found to be effective in both 2-D and 3-D cell culture system with efficient cancer cell internalization. The bench-marked potential of CM-PIONPs to produce reactive oxygen species makes it a noticeable drug delivery system to compact neoplasia. These nanoconjugates can lay concrete on a better way for the elimination of cancer spheroids and tumor burden.


Subject(s)
Nanoconjugates/chemistry , Tumor Microenvironment , Animals , Antibiotics, Antineoplastic/administration & dosage , Brain Neoplasms/drug therapy , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Glioblastoma/drug therapy , Humans , Mice , Mice, Inbred BALB C , Polysaccharides/administration & dosage
3.
Int J Biol Macromol ; 168: 130-142, 2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33278441

ABSTRACT

Iron oxide nanoparticles (IONPs) are employed as MRI contrast agents and as effective drug delivery vehicles. However, the limited solubility and biodegradability of these nanoparticles need to be improved for safer biomedical applications. In an attempt to improve the bottlenecks associated with IONPs, the current study focuses on the synthesis of folic acid conjugated, galactoxyloglucan-iron oxide nanoparticles (FAPIONPs), for the loading and controlled release of the encapsulated chemotherapeutic agent doxorubicin (DOX). The as-designed DOX@FAPIONPs induced a dose-dependent increase in cytotoxicity in folate receptor-positive cells through a caspase-mediated programmed cell death pathway while bare DOX demonstrated a non-targeted toxicity profile. Using LC-MS/MS analysis, several major biological processes altered in treated cells, from which, cell cycle, cellular function and maintenance were the most affected. Detailed toxicity studies in healthy mice indicated the absence of any major side effects while bare drugs created substantial organ pathology. Gadolinium-based contrast agents have a risk of adverse effects, including nephrogenic systemic fibrosis overcome by the administration of DOX@FAPIONPs in xenograft mice model. Tumor-targeted biodistribution pattern with a favorable DOX pharmacokinetics will be the driving factor behind the appealing tumor reduction capacity and increased survival benefits demonstrated on solid tumor-bearing mice.


Subject(s)
Drug Delivery Systems/methods , Galactose/chemistry , Glucans/chemistry , Magnetic Iron Oxide Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, Liquid/methods , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Female , Folic Acid/chemistry , Folic Acid/metabolism , Folic Acid/pharmacology , Galactose/pharmacology , Glucans/pharmacology , Humans , Magnetite Nanoparticles/therapeutic use , Mice , Particle Size , Polyethylene Glycols/pharmacology , Tandem Mass Spectrometry/methods , Tissue Distribution/drug effects
4.
Colloids Surf B Biointerfaces ; 193: 111082, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32361551

ABSTRACT

Nanotechnology-based drug delivery research has largely focused on developing well efficient localized delivery therapeutic agents to overcome the limitations of non-specificity and toxicity of conventional chemotherapy. Herein, we constructed a nanoplatform based on a self-assembled polysaccharide-protein conjugate to deliver anti-tumor drug doxorubicin and gold nanoparticles (DOX@PST-BSA AuNPs) for cancer therapy. The self-assembled DOX@PST-BSA AuNPs exhibited higher stability and thermal properties which enable them for drug delivery via passive targeting. The fluorescent property of the drug contributes to the self-monitoring of NPs Biodistribution in vitro and in vivo. Furthermore, the NPs showed negligible cytotoxicity and tissue accumulation in normal cells in vivo. Importantly, the NPs could load the anti-tumor drug with high encapsulation efficiency and competently delivered into the tumor microenvironment thereby inhibit tumor growth significantly through apoptotic induction. Notably, DOX@PST-BSA AuNPs exhibits low systemic toxicity and very few side effects in vivo. Based on the explored features, these NPs could serve as a promising multifunctional drug delivery nanoplatform for cancer therapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Galactose/chemistry , Glucans/chemistry , Neoplasms/drug therapy , Serum Albumin, Bovine/chemistry , A549 Cells , Animals , Cattle , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gold/chemistry , Humans , Metal Nanoparticles/chemistry , Mice , Nanotechnology , Neoplasms/pathology , Particle Size , Surface Properties
5.
Int J Biol Macromol ; 142: 712-723, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31622712

ABSTRACT

The extensive advancement with novel wound dressing materials functionalized with desirable properties, often touted as a panacea for cuts and burns afflicting various pathologies. However, it would indeed be a hard task to isolate any such material which perfectly fits the needs of any biomedical issue at hand. Biocompatibility, biodegradability as well as non-toxicity of natural polysaccharide served as a versatile and tunable platform for designing natural polysaccharide based scaffolds as an attractive tool in tissue engineering with a greater degree of acceptability. In this regard, we aimed to fabricate a semi interpenetrating hydrogel via exploiting the nontoxic and immune-stimulatory nature of galacto-xyloglucan (PST001) which was further doped with silver nanoparticles to formulate SNP@PST. The wound healing potential of SNP@PST was then studied both with in vitro and preclinical mice models. The current study gives a formulation for cost effective preparation of polysaccharide hydrogels using acrylamide crosslinking with improved biocompatibility and degradability. Wound healing studies in mice proved the efficiency of gels for the clinical application wherein the incorporation of nanosilver greatly enhanced the antimicrobial activity.


Subject(s)
Anti-Bacterial Agents/chemistry , Glucans/chemistry , Hydrogels/chemistry , Metal Nanoparticles/chemistry , Plant Extracts/chemistry , Silver/chemistry , 3T3 Cells , Administration, Cutaneous , Animals , Anti-Bacterial Agents/pharmacology , Biocompatible Materials/chemistry , Male , Mice , Mice, Inbred BALB C , Permeability , RAW 264.7 Cells , Seeds/chemistry , Silver/pharmacology , Tamarindus/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Wound Healing/drug effects
6.
Mater Sci Eng C Mater Biol Appl ; 107: 110332, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31761162

ABSTRACT

A variety of naturally derived and synthetic biomaterial scaffolds have been investigated as 3D environments for supporting cell growth and can be used to achieve drug delivery with high loading efficiency. Polysaccharides which enhance the tumour-specific drug release are ideal candidates for scaffold preparation in combination with chemotherapeutic agents for the management of solid tumours by local applications. Galactoxyloglucan (PST001) based porous scaffolds (PS) were prepared by crosslinking and freeze drying with a porosity of 90%. FTIR showed the same functional groups as of PST001 with slight peak shifts and 1200% water absorption was observed. Comparing with PBS, macrophage mediated improved degradation up to 40% in 28 days was observed. The scaffold was relatively non toxic towards normal and cancer cells and there was no epithelial mesenchymal transition (EMT) observed. In vitro drug release profile of doxorubicin (DOX)-loaded scaffold (PSD) showed higher release at acidic pH, apparent in tumour microenvironment, than normal physiological pH. In in vitro assays, cell viability was decreased confirming the drug release potential of the scaffold. DLA tumour was significantly reduced with PSD implantation. The excellent biodegradability of the PS overcome the limitations of non-biodegradable systems which support the sustained release of the drug and degrade after a specific time period. The local tumour reduction potential of the PSD embrace immense application in malignant solid tumour management.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Polysaccharides/chemistry , 3T3-L1 Cells , Absorbable Implants , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Drug Implants , Drug Liberation , Epithelial-Mesenchymal Transition/drug effects , Humans , Male , Materials Testing , Mice , Polysaccharides/pharmacokinetics , Porosity , Rats, Wistar , Xenograft Model Antitumor Assays
7.
Int J Biol Macromol ; 136: 266-274, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31201909

ABSTRACT

Paclitaxel, an effective chemotherapeutic drug, is insoluble in aqueous solvents and is usually administered with excipients which have side effects. The use of this drug is also limited due to multi-drug resistance. In this study polysaccharide nanoparticles are used in the delivery of chemotherapeutic drug while minimizing side-effects, solubility issues and drug resistance. The use of biopolymers like galactoxyloglucan to synthesize nanoparticle makes it more biocompatible. This study involves the synthesis of PST-PTX nanoparticles using tamarind seed polysaccharide and Paclitaxel by epichlorohydrin crosslinking. The particles were further characterized by Dynamic Light Scattering (DLS), High-resolution transmission electron microscopy (HR-TEM) Fourier Transform Infrared Spectroscopy (FTIR) and UV-Visible spectroscopy. The cytotoxicity of PST-PTX nanoparticles in cancer cell lines and resistant cancer cell lines were determined by MTT assay. The quantitative analysis of cell death was determined by Annexin V dead cell assay, Caspase 3/7 assay and expression of pro-apoptotic protein Bax. The ability of the nanoparticle to overcome multi-drug resistance was evaluated by the expression of multidrug-resistant proteins P-glycoprotein (P-gp) and Breast cancer resistant protein (BCRP) in lung adenocarcinoma resistant cells (A549R). The present study provides evidence for the ability of PST-PTX nanoparticle to overcome multi-drug resistance and cause apoptotic cell death. The particle was found to be more effective than Paclitaxel in causing cell death in resistant cancer cells. Moreover, the particles were found to downregulate the expression of multi-drug resistant proteins P-gp and BCRP in resistant cell lines suggesting the ability of PST-PTX nanoparticles to overcome multi-drug resistance.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Galactose/chemistry , Glucans/chemistry , Lung Neoplasms/pathology , Nanoparticles/chemistry , Paclitaxel/chemistry , Paclitaxel/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Apoptosis/drug effects , Biological Transport , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Carriers/chemistry , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Neoplasm Proteins/metabolism , Paclitaxel/metabolism
8.
Int Immunopharmacol ; 73: 280-292, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31125927

ABSTRACT

An unexploited homo-polysaccharide (PSM001) isolated from the seed kernel of Kottukonam variety of Mangifera indica, demonstrated selective cytotoxicity against cancer cells both in vitro and in murine models while maintaining the immunostimulatory potential. Galactoxyloglucan (PST001) isolated from the seeds of Tamarindus indica, was previously established to be an effective anticancer and immunomodulatory agent. Cancer metastasis, with key features including invasion, migration, increased angiogenesis and colony formation is only likely to accentuate in the coming decades, considering the ground realities of the modern lifestyle and environmental factors and hence both the polysaccharides were tested towards the management of malignancy. It was a startling observation with both the biopolymers in inhibiting various processes involved in the metastatic cascade. A quick perusal of the issue at hand would throw up the promising ability of both PSM001 and PST001 to inhibit lung metastatic nodules of C57BL/6 mice wherein the combinatorial treatment of these polysaccharides with vincristine delivered superior therapeutic output. Later, vascular endothelial growth factor and multiple matrix metalloproteinases were found to be the lead players in the polysaccharide mediated metastatic inhibition. Having considered the complexities associated with the chemotherapy in metastatic cancer in terms of palpable immunosuppression, the aftermaths with the co-administration of an immunostimulatory agent which itself possess unique anticancer and anti-metastatic potentials with a potent chemotherapeutic agent will be enormously consequential.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Mangifera , Neoplasms/drug therapy , Polysaccharides/therapeutic use , Tamarindus , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chickens , Chorioallantoic Membrane/drug effects , Female , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Polysaccharides/pharmacology , Wound Healing/drug effects
9.
Int J Biol Macromol ; 118(Pt A): 1174-1182, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30001604

ABSTRACT

Galactomannan isolated from the fruit rind of Punica granatum was previously reported to have excellent antioxidant, immunomodulatory and anticancer properties against both human and murine cancer cells. This polysaccharide was proved to be an anticancer agent either alone or as an adjuvant to chemotherapy. An exploration leads to the fabrication of silver nanoparticles with an average size around 30 nm and a negative surface charge of 35.2 mV using this biopolymer which acted both as reducing and capping agent and displayed good stability and biocompatibility. UV-vis spectrum of the aqueous medium containing silver nanoparticles showed an absorption peak at around 440 nm. The nanoparticles displayed an upgraded and selective cytotoxicity towards human adenocarcinoma, colorectal carcinoma and hepatocellular carcinoma cells. The induction of cancer cell toxicity was proved to be through the induction of programmed cell death pathway mediated with the active involvement of caspases. The significant anti-metastatic properties will further favour the safer in vivo application of these silver nanoparticles against neoplasia. The nontoxic nature of polysaccharide endowed the resultant silver nanoparticles with excellent biocompatibility towards red blood cells and extended the biomedical potential of this candidate. Hence, the surfactant-free green method mediated orchestration of biogenic silver nanoparticles resembled a potential theransonstic nano-construct with synergistic anticancer and immunomodulatory potential in a single platform.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Lythraceae/chemistry , Mannans , Materials Testing , Metal Nanoparticles , Silver , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Erythrocytes/metabolism , Erythrocytes/pathology , Galactose/analogs & derivatives , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mannans/chemistry , Mannans/pharmacology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Silver/chemistry , Silver/pharmacology
10.
Tumour Biol ; 39(3): 1010428317695946, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28345463

ABSTRACT

Imatinib mesylate, a BCR/ABL fusion protein inhibitor, is the first-line treatment against chronic myelogenous leukemia. In spite of its advantageous viewpoints, imatinib still has genuine impediments like undesirable side effects and tumor resistance during chemotherapy. Nanoparticles with sustainable release profile will help in targeted delivery of anticancer drugs while minimizing deleterious side effects and drug resistance. The use of biopolymers like galactoxyloglucan (PST001) for the fabrication of imatinib mesylate nanoparticles could impart its use in overcoming multidrug resistance in chronic myelogenous leukemia patients with minimal side effects. This study involved in the synthesis of PST-Imatinib nanoconjugates with appreciable drug payload and excellent cytotoxicity against drug-resistant chronic myelogenous leukemia cell line (K562) in comparison with free drug. The use of bioinformatics tool revealed better binding affinity for the drug-polysaccharide complex than the drug alone with three proteins: 3QX3 (Topoisomerase), 1M17 (EGFR tyrosine kinase domain), and 3QRJ (ABL1 kinase domain). Assessment of the biochemical, hematological, and histopathological parameters in mice upheld the security and adequacy of the nanoconjugate compared to free drug. Although perspective investigations are warranted, in a condition like drug resistance in leukemia, this nanoconjugate would display a productive approach in cancer therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Delayed-Action Preparations/therapeutic use , Glucans/therapeutic use , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Nanoconjugates/therapeutic use , Cell Line, Tumor , DNA Topoisomerases/genetics , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/genetics , Humans , K562 Cells , Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...