Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 14: 1142374, 2023.
Article in English | MEDLINE | ID: mdl-37063284

ABSTRACT

Introduction: Triple-negative breast cancer (TNBC) usually has a poor prognosis. Although the immunotherapy of TNBC has achieved great progress, only a few patients can benefit from the current treatment. CD47 is widely expressed on the surface of TNBC cells and may become an immune checkpoint for TNBC treatment. Nevertheless, increasingly more attention has been paid to systemic side effects since the ubiquitous expression of CD47 on normal cells. The toll-like receptor (TLR) agonist resiquimod (R848) can activate dendritic cells (DCs) and promote the maturation of immune cells in the tumor microenvironment, which further enhances the tumor inhibition ability of the immune system and synergizes with CD47 small interfering RNA (siRNA) for TNBC therapy. However, ideal delivery platforms such as nanocarriers are still needed because its weakness of hydrophobicity. Methods: In order to improve efficacy and reduce toxicity, R848 and siCD47 were entrapped in amphiphilic PEG-PLGA nanoparticles by double emulsification and stable nanoparticles NP/R848/siCD47 were generated to investigate their anti-tumor effects in a TNBC tumor-bearing mouse model. Results: Here, we show that PEG-PLGA nanoparticles are effective nanocarriers that can safely and effectively deliver siCD47 and R848 to tumor tissue, as demonstrated by retarded tumor growth. Mechanistically, downregulation of CD47 expression and activation of DCs took part in promoting the immune response of cytotoxic T cells (CTLs). Meanwhile, a decrease of myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) indicated the modulating of the tumor immune microenvironment. Discussion: To our best knowledge, our study pioneered co-delivery system for hydrophilic siCD47 and hydrophobic R848. It can maximize break tumor immune escape caused by CD47 and simultaneously enhance antigen presentation by activating DCs for effector T cell killing while regulating the tumor microenvironment as expected. Not only does it conform to the reports of previous basic research, but also it can break the bottleneck of their clinical application hopefully. Collectively, our findings could lay the foundation for future therapeutic strategies of TNBC.

2.
Exp Parasitol ; 246: 108460, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36642299

ABSTRACT

Chronic toxoplasmosis which is positively correlated with many neuropsychiatric problems has no curative treatment till now; due to the resistant tissue cysts especially in the brain. In search of an effective treatment, guanabenz-loaded polyethylene glycol poly lactic-co-glycolic acid (PEG-PLGA) nanoparticles was evaluated against chronic experimental toxoplasmosis. For this purpose, each mouse was infected with 10 cysts of Toxoplasma gondii (ME 49 strain). Treated mice received either guanabenz alone (5 mg/kg/day) in subgroup IIa or guanabenz-loaded nanoparticles by full dose in subgroup IIb or guanabenz-loaded nanoparticles by the half dose (2.5 mg/kg/day) in subgroup IIc. Subgroup Ie was treated by pyrimethamine and sulfadiazine. The treatment started on day 25 post-infection for 19 successive days. Then Parasitological, histopathological, immunohistochemical, immunological and ultrastructural morphological studies were performed. The results showed that: subgroup IIb showed the highest statistically significant reduction in the neuroinflammation and brain tissue cysts (77%) with a significant higher efficacy in comparison with pyrimethamine and sulfadiazine and showed the highest level of IFN-γ, while the lowest level was in subgroup IIa. All group II mice showed similar changes of depression and compression of the wall of the cyst. This is marked in subgroup IIb with release of crescent shaped bradyzoite outside the cyst. PEG-PLGA nanoparticles had no toxic effect on the liver or the kidney of the mice. It could be concluded that guanabenz-loaded PEG-PLGA nanoparticles could be promising and safe for treatment of chronic toxoplasmosis.


Subject(s)
Guanabenz , Nanoparticles , Toxoplasma , Toxoplasmosis , Animals , Mice , Guanabenz/pharmacology , Guanabenz/therapeutic use , Nanoparticles/therapeutic use , Pyrimethamine/therapeutic use , Pyrimethamine/pharmacology , Sulfadiazine/therapeutic use , Sulfadiazine/pharmacology , Toxoplasmosis/drug therapy
3.
Polymers (Basel) ; 15(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36679166

ABSTRACT

Polyethylene glycol (PEG) coated Poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) for cancer treatment are biocompatible, nonimmunogenic and accumulate in tumour sites due to the enhanced permeability and retention (EPR). Doxorubicin (DOX) is a potent but cardiotoxic anticancer agent. Hyaluronic acid (HA) occurs naturally in the extra-cellar matrix and binds to CD44 receptors which are overexpressed in cancer metastasis, proven to be characteristic of cancer stem cells and responsible for multidrug resistance. In this study, an athymic mice model of breast cancer metastasis was developed using red fluorescent protein (RFP)-labelled triple negative cancer cells. The animals were divided into four treatment groups (Control, HA-PEG-PLGA nanoparticles, PEG-PLGA nanoparticles, and Free DOX). The tumour size growth was assessed until day 25 when animals were sacrificed. Mice treated with HA-PEG-PLGA NPs inhibited tumour growth. The tumour growth at day 25 (118% ± 13.0) was significantly (p < 0.05) less than PEG-PLGA NPs (376% ± 590 and control (826% ± 970). Fluorescent microscopy revealed that HA-PEG-PLGA NPs had significantly (p < 0.05) less metastasis in liver, spleen, colon, and lungs as compared to control and to Free DOX groups. The efficacy of HA-PEG-PLGA NPs was proven in vivo. Further pharmacokinetic and toxicity studies are required for this formulation to be ready for clinical research.

4.
Drug Deliv ; 28(1): 1363-1375, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34180761

ABSTRACT

Targeted treatment of cerebral ischemia/reperfusion injury (CIRI) remains a problem due to the difficulty in drug delivery across the blood-brain barrier (BBB). In this study, we developed Bo-TSA-NP, a novel tanshinone IIA (TSA) loaded nanoparticles modified by borneol, which has long been proved with the ability to enhance other drugs' transport across the BBB. The Bo-TSA-NP, with a particle size of about 160 nm, drug loading of 3.6%, showed sustained release and P-glycoprotein (P-gp) inhibition property. It demonstrated a significantly higher uptake by 16HBE cells in vitro through the clathrin/caveolae-mediated endocytosis and micropinocytosis. Following intranasal (IN) administration, Bo-TSA-NP significantly improved the preventive effect on a rat model of CIRI with improved neurological scores, decreased cerebral infarction areas and a reduced content of malondialdehyde (MDA) and increased activity of superoxide dismutase (SOD) in rat brain. In conclusion, these results indicate that Bo-TSA-NP is a promising nose-to-brain delivery system that can enhance the prevention effect of TSA on CIRI.


Subject(s)
Abietanes/pharmacology , Brain Ischemia/drug therapy , Camphanes/chemistry , Nanoparticles/chemistry , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adjuvants, Pharmaceutic , Administration, Intranasal , Animals , Brain/drug effects , Chemistry, Pharmaceutical , Delayed-Action Preparations , Disease Models, Animal , Drug Carriers , Malondialdehyde/antagonists & inhibitors , Particle Size , Polyethylene Glycols/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats , Succinimides/chemistry , Superoxide Dismutase/biosynthesis
7.
Int J Pharm ; 564: 340-349, 2019 Jun 10.
Article in English | MEDLINE | ID: mdl-31002934

ABSTRACT

Nanoparticles (NPs) conjugated with aptamers have been extensively in recent years, which can efficiently target cancer cells that improve the therapeutic effect. Aptamers (Apt) are small oligonucleotide molecule ligands have specific high-affinity. In this work, we developed a PEG-PLGA nanoparticles (NPs) encapsulated with doxorubicin. The NPs were modified with C2NP, a ssDNA aptamer specifically binding to CD30 protein which was over expressed in anaplastic large cell lymphoma (ALCL) cells. PEG-PLGA nanoparticles (NPs) were formed by nanoprecipitation and loaded with doxorubicin, further conjugated C2NP aptamer via an EDC/NHS technique. Obtained results demonstrated that the targeted agent was successfully conjugated confirming by Urea PAGE and XPS. The physicochemical properties of Apt-DOX-NPs like particle size at 168.07 ±â€¯2.72 nm and zeta potential at -30.76 ±â€¯0.153 mV. The time of the release drugs was efficiently increased in targeted formulations and showed higher accumulation in ALCL cells than non-targeted system. Findings from this work demonstrated the potential efficacy of C2NP-functionalized nanoparticles for a therapy in ALCL.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/administration & dosage , Doxorubicin/administration & dosage , Ki-1 Antigen , Lymphoma, Large-Cell, Anaplastic/drug therapy , Nanoparticles/administration & dosage , Polyesters/administration & dosage , Polyethylene Glycols/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems , Humans
8.
Acta Pharmacol Sin ; 40(1): 143-150, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29950614

ABSTRACT

Mesangial proliferative glomerulonephritis (MsPGN), one of the most common glomerulonephritis pathological types, often leads to end-stage renal disease over a prolonged period. But the current treatment of MsPGN is non-specific and causes serious side effects, thus novel therapeutics and targeting strategies are urgently demanded. By combining the advantages of PEG-PLGA nanoparticles and the size selection mechanism of renal glomerulus, we designed and developed a novel PEG-PLGA nanoparticle delivery system capable of delivering dexamethasone acetate (A-DEX) into glomerular mesangium. We determined that 90 nm was the optimum size to encapsulate A-DEX for glomerular mesangium targeting based on the size-selection mechanism of glomerulus. After intravenous administration in rats, 90 nm DiD-loaded NPs were found to accumulate to a greater extent in the kidney and kidney cortex compared with the free DiD solution. The 90 nm A-DEX NPs are also more stable at room temperature and showed a sustained release pattern. In rat glomerular mesangial cells (HBZY-1) in vitro, we found that the uptake of 90 nm A-DEX NPs was both temperature-dependent and energe-dependent, and they were mostly engulfed via clathrin-dependent endocytosis pathways. In summary, we have successfully developed a glomerular mesangium-targeted PEG-PLGA NPs, which is potential for the treatment of MsPGN.


Subject(s)
Dexamethasone/analogs & derivatives , Drug Carriers/chemistry , Glomerular Mesangium/metabolism , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Animals , Dexamethasone/metabolism , Drug Design , Drug Liberation , Particle Size , Rats, Sprague-Dawley , Tissue Distribution
9.
Biosci Rep ; 38(1)2018 02 28.
Article in English | MEDLINE | ID: mdl-29229675

ABSTRACT

PEG-PLGA nanoparticles (NPs) modified with anti-CD133 and tumor-targeting single-chain antibody fragment (scFV-NPs) for systemic delivery of methioninase (METase) and pemetrexed for gastric carcinoma were successfully formulated. The structure characterization and biological functions of METase-pemetrexed-loaded scFV-PEG-PLGA NPs (scFV-METase/pemetrexed-NPs) in vitro were investigated. Functional scFV-PEG-PLGA NPs or PEG-PLGA NPs present low cell cytoxicity in CD133+ SGC7901 cells. scFV-METase/pemetrexed-NPs (scFv-M/P-NP) was more effective in inhibiting tumor growth (including cell growth and migration ability) in CD133 positive expressed gastric cancer cells than METase/pemetrexed-NPs (M/P-NP). Moreover, METase enhanced the inhibitory effect of pemetrexed on thymidylate synthase (TS) synthesis and cell apoptosis. We have demonstrated the application of scFV-targeted PEG-PLGA NPs as a new potential strategy to enhance treatment benefits for gastric carcinoma.


Subject(s)
Lactic Acid/administration & dosage , Nanoparticles/administration & dosage , Polyglycolic Acid/administration & dosage , Single-Chain Antibodies/administration & dosage , Stomach Neoplasms/drug therapy , AC133 Antigen/administration & dosage , AC133 Antigen/chemistry , Carbon-Sulfur Lyases/administration & dosage , Carbon-Sulfur Lyases/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Humans , Lactic Acid/chemistry , Nanoparticles/chemistry , Pemetrexed/chemistry , Polyesters/administration & dosage , Polyesters/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Single-Chain Antibodies/chemistry , Stomach Neoplasms/pathology
10.
Int J Nanomedicine ; 11: 6547-6559, 2016.
Article in English | MEDLINE | ID: mdl-27994458

ABSTRACT

Sustainable and safe delivery of brain-targeted drugs is highly important for successful therapy in Parkinson's disease (PD). This study was designed to formulate biodegradable poly(ethylene glycol)-poly(lactic-co-glycolic acid) (PEG-PLGA) nanoparticles (NPs), which were surface-modified with lactoferrin (Lf), for efficient intranasal delivery of rotigotine to the brain for the treatment of PD. Rotigotine NPs were prepared by nanoprecipitation, and the effect of various independent process variables on the resulting properties of NPs was investigated by a Box-Behnken experimental design. The physicochemical and pharmaceutical properties of the NPs and Lf-NPs were characterized, and the release kinetics suggested that both NPs and Lf-NPs provided continuous, slow release of rotigotine for 48 h. Neither rotigotine NPs nor Lf-NPs reduced the viability of 16HBE and SH-SY5Y cells; in contrast, free rotigotine was cytotoxic. Qualitative and quantitative cellular uptake studies demonstrated that accumulation of Lf-NPs was greater than that of NPs in 16HBE and SH-SY5Y cells. Following intranasal administration, brain delivery of rotigotine was much more effective with Lf-NPs than with NPs. The brain distribution of rotigotine was heterogeneous, with a higher concentration in the striatum, the primary region affected in PD. This strongly suggested that Lf-NPs enable the targeted delivery of rotigotine for the treatment of PD. Taken together, these results demonstrated that Lf-NPs have potential as a carrier for nose-to-brain delivery of rotigotine for the treatment of PD.


Subject(s)
Brain/drug effects , Drug Delivery Systems , Lactoferrin/administration & dosage , Nanoparticles/administration & dosage , Parkinson Disease/drug therapy , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tetrahydronaphthalenes/administration & dosage , Thiophenes/administration & dosage , Administration, Intranasal , Animals , Blood-Brain Barrier , Bronchi/cytology , Bronchi/drug effects , Cells, Cultured , Drug Carriers/chemistry , Humans , Lactoferrin/chemistry , Male , Mice , Nanoparticles/chemistry , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Particle Size , Tissue Distribution
11.
Oncotarget ; 6(25): 21379-94, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26041888

ABSTRACT

The Mouse Double Minute 2 (MDM2) oncogene plays a critical role in cancer development and progression through p53-dependent and p53-independent mechanisms. Both natural and synthetic MDM2 inhibitors have been shown anticancer activity against several human cancers. We have recently identified a novel ginsenoside, 25-OCH3-PPD (GS25), one of the most active anticancer ginsenosides discovered thus far, and have demonstrated its MDM2 inhibition and anticancer activity in various human cancer models, including prostate cancer. However, the oral bioavailability of GS25 is limited, which hampers its further development as an oral anticancer agent. The present study was designed to develop a novel nanoparticle formulation for oral delivery of GS25. After GS25 was successfully encapsulated into PEG-PLGA nanoparticles (GS25NP) and its physicochemical properties were characterized, the efficiency of MDM2 targeting, anticancer efficacy, pharmacokinetics, and safety were evaluated in in vitro and in vivo models of human prostate cancer. Our results indicated that, compared with the unencapsulated GS25, GS25NP demonstrated better MDM2 inhibition, improved oral bioavailability and enhanced in vitro and in vivo activities. In conclusion, the validated nano-formulation for GS25 oral delivery improves its molecular targeting, oral bioavailability and anticancer efficacy, providing a basis for further development of GS25 as a novel agent for cancer therapy and prevention.


Subject(s)
Antineoplastic Agents/administration & dosage , Ginsenosides/administration & dosage , Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Caco-2 Cells , Cell Line, Tumor , Drug Carriers , Drug Delivery Systems , Ginsenosides/pharmacology , Humans , Male , Mice , Nanomedicine , Neoplasm Transplantation , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Distribution , Tumor Suppressor Protein p53/metabolism
12.
Eur J Pharm Biopharm ; 87(1): 30-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24560967

ABSTRACT

Lectin-functionalized, polyethylene glycol-block-poly-(D,L)-lactic-co-glycolic acid nanoparticles loaded with haloperidol were prepared with narrow size distributions and sizes <135nm. The nanoparticles exhibited high Solanum tuberosum lectin (STL) conjugation efficiencies, encapsulation efficiencies, and drug loading capacities. The in vitro release of haloperidol was 6-8% of the loaded amount in endo-lysosomal conditions over 96h, demonstrating minimal drug leakage and the potential for the efficient drug transport to the targeted brain tissue. The haloperidol released upon erosion was successful in displacing [(3)H] N-propylnorapomorphine and binding to bovine striatal dopamine D2 receptors. Both haloperidol-loaded nanoparticle formulations were found to be highly effective at inducing catalepsy. Intranasal administration of STL-functionalized nanoparticles increased the brain tissue haloperidol concentrations by 1.5-3-fold compared to non-STL-functionalized particles and other routes of administration. This formulation demonstrates promise in the reduction of the drug dose necessary to produce a therapeutic effect with antipsychotic drugs for the treatment of schizophrenia.


Subject(s)
Drug Carriers/chemistry , Haloperidol/administration & dosage , Lectins/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Schizophrenia/drug therapy , Administration, Intranasal , Animals , Catalepsy/chemically induced , Catalepsy/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drug Compounding , Drug Liberation , Haloperidol/pharmacokinetics , Haloperidol/therapeutic use , Male , Microscopy, Electron, Transmission , Particle Size , Rats, Sprague-Dawley , Schizophrenia/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...