Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Toxics ; 11(6)2023 Jun 20.
Article in English | MEDLINE | ID: mdl-37368647

ABSTRACT

Antrocin is a novel compound isolated from Antrodia cinnamomea, and is classified as a sesquiterpene lactone. The therapeutic efficacy of antrocin has been studied, and it has shown an antiproliferative effect on various cancers. The aim of this study was to evaluate the anti-oxidant activity, potential genotoxicity, and oral toxicity of antrocin. Ames tests with five different strains of Salmonella typhimurium, chromosomal aberration tests in CHO-K1 cells, and micronucleus tests in ICR mice were conducted. The results of anti-oxidant capacity assays showed that antrocin has great anti-oxidant activity and is a moderately strong antimutagenic agent. In the results of the genotoxicity assays, antrocin did not show any mutagenic potential. In the 28-day oral toxicity test, Sprague Dawley rats were gavaged with 7.5 or 37.5 mg/kg of antrocin for 28 consecutive days. In addition, 7.5 mg/kg sorafenib, an anti-cancer drug, was used as a positive control for toxicity comparison. At the end of the study, antrocin did not produce any toxic effects according to hematology, serum chemistry, urine analysis, or histopathological examinations. According to the results of the genotoxicity and 28-day oral toxicity study, antrocin, at a dose of 37.5 mg/kg, did not cause adverse effects and can be a reference dose for therapeutic agents in humans.

2.
Sci Rep ; 7(1): 3487, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615716

ABSTRACT

Surgery and radiotherapy cannot fully remove brain glioma; thus, chemotherapy continues to play an important role in treatment of this illness. However, because of the restriction of the blood-brain barrier (BBB) and the regeneration of glioma stem cells, post-chemotherapy relapse usually occurs. Here, we report a potential solution to these issues that involves a type of novel multifunctional vinblastine liposomes equipped with transferrin receptor binding peptide TfR-T12 and octa-arginine conjugate stearyl-R8. Studies were performed on brain glioma and glioma stem cells in vitro and were verified in brain glioma-bearing mice. The liposomes were transported across the BBB, killing brain glioma and glioma stem cells via the induction of necrosis, apoptosis and autophagy. Furthermore, we reveal the molecular mechanisms for treating brain glioma and glioma stem cells via functionalized drug lipid vesicles.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Brain Neoplasms/drug therapy , Glioma/drug therapy , Liposomes/administration & dosage , Neoplastic Stem Cells/drug effects , Vinblastine/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Drug Delivery Systems , Humans , Liposomes/chemistry , Mice , Oligopeptides/chemistry , Receptors, Transferrin/chemistry
3.
Int J Nanomedicine ; 12: 4163-4176, 2017.
Article in English | MEDLINE | ID: mdl-28615943

ABSTRACT

Currently, chemotherapy is less efficient in controlling the continued development of breast cancer because it cannot eliminate extrinsic and intrinsic refractory cancers. In this study, mitochondria were modified by functional epirubicin liposomes to eliminate refractory cancers through initiation of an apoptosis cascade. The efficacy and mechanism of epirubicin liposomes were investigated on human breast cancer cells in vitro and in vivo using flow cytometry, confocal microscopy, high-content screening system, in vivo imaging system, and tumor inhibition in mice. Mechanistic studies revealed that the liposomes could target the mitochondria, activate the apoptotic enzymes caspase 8, 9, and 3, upregulate the proapoptotic protein Bax while downregulating the antiapoptotic protein Mcl-1, and induce the generation of reactive oxygen species (ROS) through an apoptosis cascade. In xenografted mice bearing breast cancer, the epirubicin liposomes demonstrated prolonged blood circulation, significantly increased accumulation in tumor tissue, and robust anticancer efficacy. This study demonstrated that functional epirubicin liposomes could significantly induce programmed death of refractory breast cancer by activating caspases and ROS-related apoptotic signaling pathways, in addition to the direct killing effect of the anticancer drug itself. Thus, we present a simple nanomedicine strategy to treat refractory breast cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Epirubicin/pharmacology , Liposomes/chemistry , Liposomes/pharmacology , Animals , Antibiotics, Antineoplastic/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/pathology , Caspases/metabolism , Epirubicin/administration & dosage , Female , Humans , Liposomes/administration & dosage , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Reactive Oxygen Species/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
4.
Oncotarget ; 8(25): 40906-40921, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28402948

ABSTRACT

Chemotherapy of brain glioma faces a major obstacle owing to the inability of drug transport across the blood-brain barrier (BBB). Besides, neovasculatures in brain glioma site result in a rapid infiltration, making complete surgical removal virtually impossible. Herein, we reported a novel kind of C-type natriuretic peptide (CNP) modified vinorelbine lipid vesicles for transferring drug across the BBB, and for treating brain glioma along with disrupting neovasculatures. The studies were performed on brain glioma U87-MG cells in vitro and on glioma-bearing nude mice in vivo. The results showed that the CNP-modified vinorelbine lipid vesicles could transport vinorelbine across the BBB, kill the brain glioma, and destroy neovasculatures effectively. The above mechanisms could be associated with the following aspects, namely, long circulation in the blood; drug transport across the BBB via natriuretic peptide receptor B (NPRB)-mediated transcytosis; elimination of brain glioma cells and disruption of neovasculatures by targeting uptake and cytotoxic injury. Besides, CNP-modified vinorelbine lipid vesicles could induce apoptosis of the glioma cells. The mechanisms could be related to the activations of caspase 8, caspase 3, p53, and reactive oxygen species (ROS), and inhibition of survivin. Hence, CNP-modified lipid vesicles could be used as a carrier material for treating brain glioma and disabling glioma neovasculatures.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Glioma/drug therapy , Lipids/administration & dosage , Natriuretic Peptide, C-Type/administration & dosage , Animals , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems/methods , Glioma/pathology , Humans , Lipids/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Natriuretic Peptide, C-Type/chemistry , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinblastine/chemistry , Vinorelbine
5.
Int J Nanomedicine ; 11: 1131-46, 2016.
Article in English | MEDLINE | ID: mdl-27042063

ABSTRACT

The efficacy of chemotherapy for brain glioma is restricted by the blood-brain barrier (BBB), and surgery or radiotherapy cannot eliminate the glioma cells because of their unique location. Residual brain glioma cells can form vasculogenic mimicry (VM) channels that can cause a recurrence of brain glioma. In the present study, targeting liposomes incorporating epirubicin and celecoxib were prepared and used for the treatment of brain glioma, along with the destruction of their VM channels. Evaluations were performed on the human brain glioma U87MG cells in vitro and on intracranial brain glioma-bearing nude mice. Targeting epirubicin plus celecoxib liposomes in the circulatory blood system were able to be transported across the BBB, and accumulated in the brain glioma region. Then, the liposomes were internalized by brain glioma cells and killed glioma cells by direct cytotoxic injury and the induction of apoptosis. The induction of apoptosis was related to the activation of caspase-8- and -3-signaling pathways, the activation of the proapoptotic protein Bax, and the suppression of the antiapoptotic protein Mcl-1. The destruction of brain glioma VM channels was related to the downregulation of VM channel-forming indictors, which consisted of MMP-2, MMP-9, FAK, VE-Cad, and VEGF. The results demonstrated that the targeting epirubicin plus celecoxib liposomes were able to effectively destroy the glioma VM channels and exhibited significant efficacy in the treatment of intracranial glioma-bearing nude mice. Therefore, targeting epirubicin plus celecoxib liposomes could be a potential nanostructured formulation to treat gliomas and destroy their VM channels.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/drug therapy , Glioma/drug therapy , Liposomes/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Caspase 8/metabolism , Celecoxib/administration & dosage , Cell Line, Tumor , Epirubicin/administration & dosage , Glioma/pathology , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Male , Mice, Nude , Neoplasm Recurrence, Local
6.
Oncotarget ; 6(34): 36625-42, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26429872

ABSTRACT

Standard chemotherapy cannot eradicate triple-negative breast cancer (TNBC) while the residual cancer cells readily form the vasculogenic mimicry (VM) channels, which lead to the relapse of cancer after treatment. In this study, the functional vincristine plus dasatinib liposomes, modified by a targeting molecule DSPE-PEG2000-c(RGDyK), were fabricated to address this issue. The investigations were performed on TNBC MDA-MB-231 cells and MDA-MB-231 xenografts in nude mice. The liposomes exhibited the superior performances in the following aspects: the enhancement of cellular uptake via targeted action; the induction of apoptosis via activation of caspase 8, 9, and 3, increased expression of Bax, decreased expression of Mcl-1, and generation of reactive oxygen species (ROS); and the deletion of VM channels via inhibitions on the VM channel indicators, which consisted of vascular endothelial-cadherin (VE-Cad), focal adhesion kinase (FAK), phosphatidylinositide 3-kinase (PI3K), and matrix metallopeptidases (MMP-2, and MMP-9). Furthermore, the liposomes displayed the prolonged circulation time in the blood, the increased accumulation in tumor tissue, and the improved therapeutic efficacy along with deletion of VM channels in the TNBC-bearing mice. In conclusion, the nanostructured functional drug-loaded liposomes may provide a promising strategy for the treatment of invasive TNBC along with deletion of VM channels.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Dasatinib/administration & dosage , Disease Models, Animal , Female , Humans , Liposomes/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Nude , Random Allocation , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/pathology , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...