Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Control Release ; 220(Pt A): 438-446, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26551345

ABSTRACT

Combining lipids and dendrimers into one formulation is an emerging platform in the drug delivery field. This study aims to (i) develop and characterize a lipid-dendrimer hybrid (LDH) nanosystem for the hydrophobic anticancer drug paclitaxel, and (ii) evaluate its in vitro and in vivo anti-cancer activity in ovarian cancer models. The LDH nanosystems were prepared from 1,2-dipalmitoyl-sn-glycero-3-phosphocholine and poly (amidoamine) (PAMAM) G4.0. The size and zeta potential of the LDH nanosystem were 37.6 ± 6.1n m and +2.9 ± 0.1 mV, respectively, with vesicular morphology observed under cryo-TEM. The encapsulation efficiency of paclitaxel in the LDH system was 78.0 ± 2.1%. The potency of paclitaxel could be significantly improved by 37-fold when presented in the LDH nanosystem as compared to free drug, whereby paclitaxel and PAMAM G4.0 acted synergistically in killing the ovarian cancer cells. As shown by fluorescence confocal microscopy, majority of the lipids in the LDH nanosystem were located in the plasma membrane, while the dendrimers were distributed intracellularly upon uptake. Despite the use of a 10-fold lower paclitaxel dose, the survival of IGROV-1 ovarian tumor-bearing animals could be significantly prolonged by the paclitaxel-loaded LDH nanosystem, as reflected by a 50% increase in the median survival time. Such hybrid nanosystem emerged from combining two established drug delivery platforms could pave way for the development of multifunctional delivery systems for potential theranostic applications.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/analogs & derivatives , Antineoplastic Agents, Phytogenic/administration & dosage , Dendrimers/chemistry , Drug Carriers , Nanoparticles , Nylons/chemistry , Ovarian Neoplasms/drug therapy , Paclitaxel/administration & dosage , 1,2-Dipalmitoylphosphatidylcholine/chemistry , 1,2-Dipalmitoylphosphatidylcholine/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dendrimers/metabolism , Dose-Response Relationship, Drug , Drug Compounding , Female , Humans , Mice, SCID , Nanotechnology , Nylons/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/chemistry , Paclitaxel/metabolism , Solubility , Time Factors , Xenograft Model Antitumor Assays
2.
J Pharm Pharmacol ; 67(11): 1492-502, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26303576

ABSTRACT

OBJECTIVES: This study aims to examine the blood compatibility of dual-functionalized poly(amidoamine) (PAMAM) dendrimers. METHODS: The cationic PAMAM dendrimer of generation 4.0 (PM4.0) were functionalized by poly(ethylene glycol) (PEG) conjugation or by thiolation or the combination of both methods. Various in-vitro assays including immune cell cytotoxicity, haemoglobin release, serum albumin binding, complement activation and coagulation times were used to characterize the compatibility with blood components. KEY FINDINGS: Although thiolation of polymers has been reported as a strategy to reduce platelet activation or aggregation, thiolation of PM4.0 alone did not offer any protective effect against the dendrimer toxicity on blood components or functions. PEGylation was able to reduce the toxic effect and interactions of the unmodified and thiolated PM4.0 on various blood components and functions; yet, PEGylated PM4.0 displayed prolonged prothrombin times and activated partial thromboplastin times. Among various PM4.0 derivatives, dual-functionalized PM4.0 with PEG and thiol groups displayed the least toxicity to various blood components and functions. CONCLUSIONS: Our findings suggested that comprehensive studies of dendrimer biocompatibility should be performed so as to establish the safe dose window for systemic administration.


Subject(s)
Blood Cells/drug effects , Blood Coagulation/drug effects , Dendrimers/toxicity , Polyethylene Glycols/chemistry , Animals , Dendrimers/chemistry , Hemoglobins/metabolism , Humans , Partial Thromboplastin Time , Protein Binding , Prothrombin Time , Rats , Serum Albumin/metabolism , Sulfhydryl Compounds/chemistry , U937 Cells
3.
Curr Pharm Des ; 21(19): 2629-42, 2015.
Article in English | MEDLINE | ID: mdl-25876918

ABSTRACT

Dendrimers are emerging as potential novel nano-scaled material in drug delivery applications. An interesting area of application is oral drug delivery. In oral drug delivery, many drugs suffer from low bioavailability due to the presence of various biological barriers. Dendrimers have been shown to modulate tight junctions and the integrity of cellular membranes. This effect gives hope for dendrimer to be applied in oral drug delivery. Based on such properties, dendrimers are further surface-modified so that the system will be more suitable for oral delivery applications. Cationic dendrimers are commonly conjugated with neutral or negatively charged ligands, such as polyethylene glycol (PEG), to reduce potential toxicity in gastrointestinal (G.I.) tract. Dendrimers are also surfacemodified to inhibit the efflux effect of P-glycoprotein, which is one of the major drug efflux pumps in G.I. tract. Another interesting strategy is to directly conjugate or mix dendrimer with drugs either to form a dendrimer-drug conjugation or complex to deliver the drug. In this review, application of dendrimers in oral drug delivery will be discussed. The main focus is on the various surface modification strategies to design a more desirable dendrimer-based delivery system that fits the need in oral drug delivery.


Subject(s)
Dendrimers/administration & dosage , Dendrimers/adverse effects , Drug Delivery Systems , Pharmaceutical Preparations/administration & dosage , Administration, Oral , Biological Availability , Dendrimers/pharmacokinetics , Gastrointestinal Absorption/drug effects , Humans , Pharmaceutical Preparations/chemistry , Surface Properties
4.
Nanomedicine (Lond) ; 9(11): 1665-79, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24294981

ABSTRACT

AIM: The aim of this work was to develop a liposomal formulation to facilitate delivery of a synergistic safingol/C2-ceramide combination in the treatment of acute myeloid leukemia (AML). MATERIALS & METHODS: Liposomes were prepared using the extrusion method and the bioactive lipids were encapsulated passively. Drug concentrations were determined by liquid chromatography tandem mass spectrometry. Antileukemic activity was evaluated using human leukemic cell lines, patient samples and U937 leukemic xenograft models. RESULTS: A stable liposome formulation was developed to coencapsulate safingol and C2-ceramide at 1:1 molar ratio with >90% encapsulation efficiency. The liposomal safingol/C2-ceramide was effective in AML cell lines, patient samples and murine xenograft models of AML, compared with liposomal safingol or liposomal C2-ceramide alone despite a dose reduction of 33%. CONCLUSION: Our study provided proof-of-concept evidence to deliver synergistic combination of bioactive lipid to achieve complete remission in AML.


Subject(s)
Antineoplastic Agents/chemistry , Drug Delivery Systems , Leukemia, Myeloid, Acute/drug therapy , Liposomes/chemistry , Animals , Cell Proliferation , Cell Survival , Ceramides/chemistry , Cholesterol/chemistry , Chromatography, Liquid , Female , Humans , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/metabolism , Lipids/chemistry , Mice , Mice, Inbred BALB C , Mice, SCID , Phosphatidylcholines/chemistry , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Tandem Mass Spectrometry , U937 Cells , Xenograft Model Antitumor Assays
5.
Biomacromolecules ; 14(12): 4226-35, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24219381

ABSTRACT

This study aims to surface modify poly(amido amine) or PAMAM dendrimers by sequentially grafting poly(ethylene glycol) or PEG and 4-thiobutylamidine (TBA) so as to reduce PAMAM cytotoxicity while improving the ability of PAMAM to modulate P-glycoprotein (P-gp) efflux and tight junction integrity. Conjugation of functional groups was determined by NMR spectroscopy, FT-IR, thiol group quantification and molecular weight estimation. The yield of the dual-functionalized dendrimers was >80%. The dual-functionalized dendrimer could significantly reduce PAMAM cytotoxicity to <15% as reflected by LDH release in Caco-2 and MDCK/MDR1 cells after 72 h of exposure. Thiolated dendrimers could increase cellular accumulation and permeation of the P-gp substrate R-123, and such effect could be affected by the extent of PEGylation of the dendrimer. Surface-modified PAMAM dendrimers, either by single or dual functionalization, could better modulate tight junction integrity in comparison with unmodified PAMAM, as demonstrated through immunostaining of the tight junction marker ZO-1, permeation of the model compound Lucifer Yellow (LY) and transepithelial electrical resistance (TEER). Of importance, reversible tight junction modulating effect was only observed in the dual-functionalized dendrimers. Collectively, dual functionalization with PEG and TBA represented a promising approach in altering PAMAM dendrimer surface for potential application in oral drug delivery.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Dendrimers/pharmacology , Drug Carriers/pharmacology , Tight Junctions/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amidines/chemistry , Animals , Caco-2 Cells , Dendrimers/chemical synthesis , Dendrimers/metabolism , Dogs , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Humans , Madin Darby Canine Kidney Cells , Particle Size , Permeability , Polyethylene Glycols/chemistry , Tight Junctions/drug effects
6.
Eur J Med Chem ; 70: 400-10, 2013.
Article in English | MEDLINE | ID: mdl-24177367

ABSTRACT

5-Chlorouracil-linked-pyrazolo[1,5-a][1,3,5]triazines were designed as new thymidine phosphorylase inhibitors based on the fragment based drug design approach. Multiple-step convergent synthetic schemes were devised to generate the target compounds. The intermediate 5-chloro-6-chloromethyluracil was synthesized by a 4-step reaction. A series of the second bicyclic intermediates, namely pyrazolo[1,5-a][1,3,5]triazin-2-thioxo-4-one, was obtained from various substituted 3-aminopyrazoles. These two intermediates were coupled finally in the presence of sodium ethoxide and methanol to yield the desirable target compounds. The methylthio coupling spacer was found to be suitable in enabling the interaction of the two fragments at the active site and allosteric site of the enzyme. The best coupled compound (9q) inhibited the thymidine phosphorylase with an IC50 value as low as 0.36 ± 0.1 µM. In addition, 9q demonstrated a mixed-type of enzyme inhibition kinetics, thus suggesting that it might indeed potentially bind at two different sites on the enzyme.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Pyrazines/pharmacology , Thymidine Phosphorylase/antagonists & inhibitors , Triazines/pharmacology , Uracil/analogs & derivatives , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Pyrazines/chemical synthesis , Pyrazines/chemistry , Structure-Activity Relationship , Thymidine Phosphorylase/metabolism , Triazines/chemical synthesis , Triazines/chemistry , Uracil/chemistry
7.
Int J Oncol ; 42(5): 1605-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23467622

ABSTRACT

Since its isolation from Tripterygium wilfordii in 1972, triptolide has been shown to possess potent anticancer activity against a variety of cancers, and has entered phase I clinical trial. It is a diterpenoid triepoxide that acts through multiple molecular targets and signaling pathways. The mitogen-activated protein kinases are well known for their modulation of cell survival and proliferation. In particular, the ERK pathway has a dual role in cell proliferation and cell death. Thus far, data on the effect of triptolide on ERK signaling remain limited. In our current study, we have shown for the first time that ERK activation rather than inhibition occurred in a dose- and time-dependent manner following triptolide treatment in MDA-MB-231 breast cancer cells. ERK activation was crucial in mediating triptolide-induced caspase-dependent apoptosis. Tritpolide-induced ERK activation modulated the expression of the Bcl-2 protein family member Bax but was not involved in the downregulation of Bcl-xL expression. Signals acted upstream of ERK activation included generation of reactive oxygen species (ROS) and endoplasmic reticulum stress predominantly via the PERK­eIF2α pathway, as the MEK inhibitor U0126 did not inhibit the phosphorylation of PERK and eIF2α or the generation of ROS.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/genetics , Diterpenes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Phenanthrenes/pharmacology , Breast Neoplasms/metabolism , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Clinical Trials as Topic , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Epoxy Compounds/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Nitriles/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Reactive Oxygen Species , Signal Transduction/drug effects , eIF-2 Kinase/metabolism
8.
J Control Release ; 172(3): 852-61, 2013 Dec 28.
Article in English | MEDLINE | ID: mdl-24459693

ABSTRACT

Liposome co-encapsulation of synergistic anti-cancer drug combination is an emerging area that has demonstrated therapeutic benefit in clinical trials. Remote loading of two or more drugs into a single liposome constitutes a new challenge that calls for a re-examination of drug loading strategies to allow the loading of the drug combination efficiently and with high drug content. In this study, the Mn(2+) gradient coupled with A23187 ionophore was applied in the sequential co-encapsulation of doxorubicin and irinotecan, as this drug loading method is capable of remotely loading drugs by apparently two different mechanisms, namely, coordination complexation and pH gradient. Doxorubicin and irinotecan could be co-encapsulated into liposomes in a wide range of drug-to-drug ratios, with encapsulation efficiencies of > 80%. The total encapsulated drug content was non-linearly correlated with increases in the intraliposomal Mn(2+) concentration, with a maximum total drug-to-lipid molar ratio of 0.8:1 achieved with 600 mM Mn(2+). This high encapsulated drug content did not affect the stability of the co-encapsulated liposomes upon storage for six months. Regardless of the encapsulated drug amount, the liposomes did not exhibit the fiber bundle precipitate morphology but rather an undefined structural organization in the aqueous core. The co-encapsulated liposome formulation was further tested in an intraperitoneally grown, human ovarian tumor xenograft model, and was shown to significantly improve the survival of the tumor-bearing animals. The improvement in therapeutic efficacy was possibly due to the increase in systemic drug exposure, with the maintenance of the synergistic molar drug ratio of 1:1 in circulation.


Subject(s)
Antineoplastic Agents/administration & dosage , Camptothecin/analogs & derivatives , Doxorubicin/administration & dosage , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Camptothecin/administration & dosage , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Synergism , Female , Humans , Irinotecan , Liposomes/chemistry , Manganese/chemistry , Mice , Mice, SCID , Ovarian Neoplasms/pathology , Ovary/drug effects , Ovary/pathology
9.
Cytotherapy ; 14(9): 1064-79, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22775077

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) have been observed to participate in tissue repair and to have growth-promoting effects on ex vivo co-culture with other stem cells. METHODS: In order to evaluate the mechanism of MSC support on ex vivo cultures, we performed co-culture of MSC with umbilical cord blood (UCB) mononuclear cells (MNC) (UCB-MNC). RESULTS: Significant enhancement in cell growth correlating with cell viability was noted with MSC co-culture (defined by double-negative staining for Annexin-V and 7-AAD; P < 0.01). This was associated with significant enhancement of mitochondrial membrane potential (P < 0.01). We postulated that intercellular transfer of cytosolic substances between MSC and UCB-MNC could be one mechanism mediating the support. Using MSC endogenously expressing green fluorescent protein (GFP) or labeled with quantum dots (QD), we performed co-culture of UCB-MNC with these MSC. Transfer of these GFP and QD was observed from MSC to UCB-MNC as early as 24 h post co-culture. Transwell experiments revealed that direct contact between MSC and UCB-MNC was necessary for both transfer and viability support. UCB-MNC tightly adherent to the MSC layer exhibited the most optimal transfer and rescue of cell viability. DNA analysis of the viable, GFP transfer-positive UCB-MNC ruled out MSC transdifferentiation or MSC-UCB fusion. In addition, there was statistical correlation between higher levels of cytosolic transfer and enhanced UCB-MNC viability (P < 0.0001). CONCLUSIONS: Collectively, the data suggest that intercellular transfer of cytosolic materials could be one novel mechanism for preventing UCB cell death in MSC co-culture.


Subject(s)
Cell Culture Techniques , Cytosol/metabolism , Fetal Blood/cytology , Leukocytes, Mononuclear/cytology , Mesenchymal Stem Cells/cytology , Animals , Cell Death/genetics , Cell Fusion , Cell Proliferation , Cell Survival , Cell Transdifferentiation , Coculture Techniques , Green Fluorescent Proteins/analysis , Humans , Membrane Potential, Mitochondrial , Mice , NIH 3T3 Cells
10.
Int J Pharm ; 430(1-2): 167-75, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22537806

ABSTRACT

The obstacles in translating liposome formulations into marketable products could be attributed to their physical instabilities upon long-term storage as aqueous dispersions. Lyophilization is the most commonly used technique to improve physical stability of liposomes. The development of stable, lyophilized liposomes is focused primarily on the cholesterol-containing liposomes or pure phosphatidylcholine-based liposomes, with minimal studies on cholesterol-free, pegylated (CF-PEG) liposomes which have emerged as an important class of liposome drug carriers. Hence, it is our interest to investigate the effect of lyophilization on CF-PEG liposomes, and specifically, on drug loading via the passive equilibration method. Three different sugar cryoprotectants were used at two different sugar-to-lipid molar ratios (S/L). Our results demonstrated that CF-PEG liposomes lyophilized with sucrose at S/L=5:1 yielded the best cryoprotective effect, as characterized by size, polydispersity indices, and microscopic examination upon liposome reconstitution. The lyophilized liposomes had low water content of 2.59 ± 0.18%. Of note, lyophilized CF-PEG liposomes exhibited two-fold increase in drug content when carboplatin was loaded via the passive equilibration method, and the in vitro drug release profile of these liposomes were not different from that of the non-lyophilized counterparts. Taken together, we envisioned that a stable, lyophilized empty CF-PEG liposome system could be coupled to hydrophilic drug loading via the passive equilibration method to produce a liposomal drug kit product.


Subject(s)
Antineoplastic Agents/chemistry , Carboplatin/chemistry , Freeze Drying , Lipids/chemistry , Polyethylene Glycols/chemistry , Technology, Pharmaceutical/methods , 1,2-Dipalmitoylphosphatidylcholine/analogs & derivatives , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Chemistry, Pharmaceutical , Cryoprotective Agents/chemistry , Drug Compounding , Drug Stability , Glucose/chemistry , Hydrophobic and Hydrophilic Interactions , Kinetics , Liposomes , Particle Size , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Solubility , Sucrose/chemistry , Trehalose/chemistry , Water/chemistry
11.
Int J Nanomedicine ; 7: 739-51, 2012.
Article in English | MEDLINE | ID: mdl-22359453

ABSTRACT

Intraperitoneal (IP) therapy with platinum (Pt)-based drugs has shown promising results clinically; however, high locoregional concentration of the drug could lead to adverse side effects. In this study, IP administration was coupled with a folate receptor-targeted (FRT) liposomal system, in an attempt to achieve intracellular delivery of the Pt-based drug carboplatin in order to increase therapeutic efficacy and to minimize toxicity. In vitro and in vivo activity of FRT carboplatin liposomes was compared with the activity of free drug and nontargeted (NT) carboplatin liposomes using FR-overexpressing IGROV-1 ovarian cancer cells as the model. Significant reduction in cell viability was observed with FRT liposomes, which, compared with the free drug, provided an approximately twofold increase in carboplatin potency. The increase in drug potency was correlated with significantly higher cellular accumulation of Pt resulting from FRT liposomal delivery. Further evaluation was conducted in mice bearing intraperitoneally inoculated IGROV-1 ovarian tumor xenografts. A superior survival rate (five out of six animals) was achieved in animals treated with FRT carboplatin liposomes, injected intraperitoneally with a dose of 15 mg/kg and following a schedule of twice-weekly administration for 3 weeks. In contrast, no survivors were observed in the free drug or NT carboplatin liposome groups. The presence of cancer cells in lung and liver tissues was observed in the saline, free carboplatin, and NT carboplatin liposome groups. However, there was no sign of cancer cells or drug-related toxicity detected in tissues from the animals treated with FRT carboplatin liposomes. The results of this study have demonstrated for the first time that the approach of coupling IP administration with FRT liposomal delivery could provide significantly improved therapeutic efficacy of carboplatin in the treatment of metastatic ovarian cancer.


Subject(s)
Carboplatin/administration & dosage , Folic Acid Transporters/metabolism , Liposomes/administration & dosage , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Animals , Carboplatin/chemistry , Carboplatin/pharmacokinetics , Cell Line, Tumor , Female , Histocytochemistry , Humans , Kaplan-Meier Estimate , Liposomes/chemistry , Liposomes/pharmacokinetics , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Mice , Mice, SCID , Molecular Targeted Therapy , Xenograft Model Antitumor Assays
12.
Int J Nanomedicine ; 7: 651-61, 2012.
Article in English | MEDLINE | ID: mdl-22334787

ABSTRACT

BACKGROUND: Realizing the therapeutic benefits of quercetin is mostly hampered by its low water solubility and poor absorption. In light of the advantages of nanovehicles in the delivery of flavanoids, we aimed to deliver quercetin perorally with nanomicelles made from the diblock copolymer, polyethylene glycol (PEG)-derivatized phosphatidylethanolamine (PE). METHODS: Quercetin-loaded nanomicelles were prepared by using the film casting method, and were evaluated in terms of drug incorporation efficiency, micelle size, interaction with Caco-2 cells, and anticancer activity in the A549 lung cancer cell line and murine xenograft model. RESULTS: The incorporation efficiency into the nanomicelles was ≥88.9% when the content of quercetin was up to 4% w/w, with sizes of 15.4-18.5 nm and polydispersity indices of <0.250. Solubilization of quercetin by the nanomicelles increased its aqueous concentration by 110-fold. The quercetin nanomicelles were stable when tested in simulated gastric (pH 1.2) and intestinal (pH 7.4) fluids, and were non-toxic to the Caco-2 cells as reflected by reversible reduction in transepithelial electrical resistance and ≤25% lactose dehydrogenase release. The anticancer activity of quercetin could be significantly improved over the free drug through the nanomicellar formulation when tested using the A549 cancer cell line and murine xenograft model. The nanomicellar quercetin formulation was well tolerated by the tumor-bearing animals, with no significant weight loss observed at the end of the 10-week study period. CONCLUSION: A stable PEG-PE nanomicellar formulation of quercetin was developed with enhanced peroral anticancer activity and no apparent toxicity to the intestinal epithelium.


Subject(s)
Antineoplastic Agents/administration & dosage , Lung Neoplasms/drug therapy , Micelles , Nanoparticles/chemistry , Quercetin/administration & dosage , Quercetin/chemistry , Administration, Oral , Analysis of Variance , Animals , Antineoplastic Agents/chemistry , Antioxidants/administration & dosage , Antioxidants/chemistry , Caco-2 Cells , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Stability , Female , Humans , Hydrogen-Ion Concentration , Lung Neoplasms/pathology , Mice , Nanoparticles/administration & dosage , Particle Size , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Xenograft Model Antitumor Assays
13.
Int J Oncol ; 40(3): 703-10, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22086152

ABSTRACT

Folate receptor-targeted (FRT) liposomes for carboplatin were developed and evaluated in FR-positive and FR-negative cell lines, KB and A549, respectively, for their cytotoxic effects. Significant enhancement in carboplatin potency and intracellular drug accumulation was observed in KB cells when treated with FRT liposomes, compared to free drug and non-targeted liposomes. No enhancement was observed in the FR-negative A549 cells. The increase in carboplatin potency was hypothesized to be associated with an increase in the formation of DNA-platinum adducts resulted from an increase in cellular accumulation of the drug. Surprisingly, FRT carboplatin liposomes showed significantly lower levels of DNA-platinum adducts in comparison to free drug. To elucidate this discrepancy, activation of extracellular signal-regulated protein kinase (ERK) was probed, which has been suggested as an alternative mechanism of carboplatin action. FRT liposomes loaded with carboplatin exhibited the highest level of ERK phosphorylation, and the cytotoxic effect of FRT carboplatin liposomes could be reversed by the MEK/ERK inhibitors, U0126 and PD98059. Importantly, empty FRT liposomes could significantly increase ERK phosphorylation in a concentration-dependent manner without causing toxicity to cells. For the first time, increased potency of carboplatin delivered by FRT liposomes was found to be associated with other molecular targets in addition to DNA-platinum adduct formation. Collectively, the current study suggests a novel mechanism by which FRT liposomes could sensitize cancer cells to drug treatment via modulation of ERK-related cell survival signals.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Carboplatin/administration & dosage , Carboplatin/pharmacokinetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Folate Receptors, GPI-Anchored/metabolism , Liposomes/administration & dosage , Antineoplastic Agents/chemistry , Carboplatin/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation , Humans , KB Cells , Liposomes/chemistry , Liposomes/pharmacokinetics , Phosphorylation/drug effects
14.
J Control Release ; 160(2): 290-8, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22100388

ABSTRACT

Prognosis of patients with acute myeloid leukemia (AML) remains poor despite the use of first-line induction chemotherapy. Therefore, it is imperative to find effective treatment for AML patients. Safingol is a bioactive sphingolipid which has demonstrated promising in vitro anti-leukemic properties; however, translation into clinical use is hampered by its low water solubility and dose-limiting hemolysis. The present study is the first to describe a rationally designed liposome formulation of safingol and demonstrate the anti-leukemic potential using a panel of human AML cell lines and patient samples as well as a human xenograft model in SCID mice. Encapsulation efficiency of safingol into liposomes was approximately 100%, and the release of drug followed square-root-of-time release model. The presence of a transmembrane pH gradient completely abolished the biological activity of liposomal safingol. A positive zeta potential, which influenced cellular accumulation of liposomal safingol, was crucial to the anti-leukemic activity. Liposomal safingol was effective against a wide range of AML subtypes with minimal hemolytic toxicity, and was able to extend the median survival time of the U937-inoculated mice to 31 days as compared to 23 days by free drug. The increase in therapeutic efficacy could be related to the increase in systemic drug exposure as a result of liposome encapsulation.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Experimental/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Sphingosine/analogs & derivatives , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Cell Culture Techniques , Cell Survival/drug effects , Drug Compounding , Female , Hemolysis/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Solubility , Sphingosine/administration & dosage , Sphingosine/adverse effects , Sphingosine/blood , Sphingosine/therapeutic use , Survival Analysis , U937 Cells , Xenograft Model Antitumor Assays
15.
Int J Nanomedicine ; 6: 2253-63, 2011.
Article in English | MEDLINE | ID: mdl-22125408

ABSTRACT

Although carbon nanomaterials (CNMs) have been increasingly studied for their biomedical applications, there is limited research on these novel materials for oral drug delivery. As such, this study aimed to explore the potential of CNMs in oral drug delivery, and the objectives were to evaluate CNM cytotoxicity and their abilities to modulate paracellular transport and the P-glycoprotein (P-gp) efflux pump. Three types of functionalized CNMs were studied, including polyhydroxy small-gap fullerenes (OH-fullerenes), carboxylic acid functionalized single-walled carbon nanotubes (f SWCNT-COOH) and poly(ethylene glycol) functionalized single-walled carbon nanotubes (f SWCNT-PEG), using the well-established Caco-2 cell monolayer to represent the intestinal epithelium. All three CNMs had minimum cytotoxicity on Caco-2 cells, as demonstrated through lactose dehydrogenase release and 3-(4,5-dimethyliazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. Of the three CNMs, f SWCNT-COOH significantly reduced transepithelial electrical resistance and enhanced transport of Lucifer Yellow across the Caco-2 monolayer. Confocal fluorescence microscopy showed that f SWCNT-COOH treated cells had the highest perturbation in the distribution of ZO-1, a protein marker of tight junction, suggesting that f SWCNT-COOH could enhance paracellular permeability via disruption of tight junctions. This modulating effect of f SWCNT-COOH can be reversed over time. Furthermore, cellular accumulation of the P-gp substrate, rhodamine-123, was significantly increased in cells treated with f SWCNT-COOH, suggestive of P-gp inhibition. Of note, f SWCNT-PEG could increase rhodamine-123 accumulation without modifying the tight junction. Collectively, these results suggest that the functionalized CNMs could be useful as modulators for oral drug delivery, and the differential effects on the intestinal epithelium imparted by different types of CNMs would create unique opportunities for drug-specific oral delivery applications.


Subject(s)
Drug Delivery Systems/methods , Fullerenes/administration & dosage , Fullerenes/chemistry , Nanotubes, Carbon/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Administration, Oral , Analysis of Variance , Biological Transport , Caco-2 Cells , Cell Membrane Permeability , Cell Survival/drug effects , Electric Impedance , Fullerenes/pharmacokinetics , Humans , Intestinal Mucosa , Isoquinolines/pharmacokinetics , Rhodamines/administration & dosage , Rhodamines/pharmacokinetics , Tight Junctions/drug effects , Tight Junctions/metabolism
16.
Oncol Rep ; 26(5): 1315-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21805042

ABSTRACT

Triptolide, a diterpene from Tripterygium wilfordii, has been shown to have potent anticancer activity, exerting its effects through multiple molecular targets and signaling pathways. Yet, its effect on focal adhesion kinase (FAK), a non-receptor tyrosine kinase overexpressed in breast cancer that regulates cellular adhesion and survival, has not been reported. The current study is the first to report on the effect of triptolide on FAK expression, cell adhesion and survival using MCF-7 breast cancer cells. Triptolide significantly reduced MCF-7 anchorage-independent growth in a concentration-dependent manner. Cell rounding and detachment from culture plates were observed as early as 8 h, with significant cell detachment observed after 24 h of triptolide treatment. The adhesion potential of triptolide-treated MCF-7 cells to Matrigel was also compromised. Triptolide induced concentration- and time-dependent cleavage of FAK and PARP, which was dependent on caspase activation. The pan-caspase inhibitor, zVAD-fmk, was the only inhibitor that could significantly reduce FAK and PARP cleavage and cell detachment. However, the presence of zVAD-fmk failed to significantly reverse triptolide-induced cell death. Finally, triptolide-induced FAK cleavage was specific to MCF-7 cells, as no cleaved FAK was observed in MDA-MB-231 cells. In conclusion, our data present the first evidence of triptolide-mediated induction of FAK cleavage that correlates with cell detachment and loss of adhesion potential to the extracellular matrix.


Subject(s)
Breast Neoplasms/drug therapy , Diterpenes/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Phenanthrenes/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Epoxy Compounds/pharmacology , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Humans
17.
Nanomedicine ; 7(6): 834-40, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21371568

ABSTRACT

Hormone- and trastuzumab-insensitive breast cancer has limited and ineffective clinical treatment options. This study sought to develop a liposome formulation containing a synergistic combination of vincristine and quercetin, with prolonged drug circulation times and coordinated drug release in vivo, to develop effective treatments against this subtype of breast cancer. The 2:1 molar ratio of vincristine/quercetin showed strong synergism in the hormone- and trastuzumab-insensitive JIMT-1 cells. Liposome co-encapsulation prolonged plasma circulation of the two drugs and maintained the synergistic drug ratio in vivo. Furthermore, the co-encapsulated liposome formulation demonstrated the most effective tumor growth inhibition in the JIMT-1 human breast tumor xenograft in comparison with vehicle control, free quercetin, free vincristine and free vincristine/quercetin combinations. Specifically, only the co-encapsulated liposome formulation exhibited significant antitumor activity at two-thirds of the maximum tolerated dose of vincristine, without significant body weight loss in the animals. FROM THE CLINICAL EDITOR: In this study, a novel liposome formulation containing a synergistic combination of vincristine and quercetin was utilized in the treatment of breast cancer. Prolonged drug circulation times and coordinated drug release characterize this effective treatment, which may find its way to clinical applications in the near future.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Liposomes/chemistry , Quercetin/administration & dosage , Vincristine/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Synergism , Female , Humans , Mice , Mice, SCID , Quercetin/pharmacokinetics , Quercetin/therapeutic use , Trastuzumab , Vincristine/pharmacokinetics , Vincristine/therapeutic use , Xenograft Model Antitumor Assays
18.
Oncol Lett ; 2(5): 905-910, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-22866148

ABSTRACT

Exploiting the sensitivity of cancer cells to reactive oxygen species (ROS) has been suggested as a strategy for the selective elimination of cancer cells. In this study, the ROS-generating sphingolipid safingol was combined with various conventional chemotherapeutics, and the potential synergism of the safingol-based combination regimen was assessed using a panel of cancer cell lines. The IC(50) values of safingol using as a single agent were 1.4-6.3 µM, which are concentrations that are clinically achievable. While synergism was dependent on the drug molar ratios, a 4:1 molar ratio of safingol to conventional chemotherapeutics exhibited a moderate to strong synergism in MDA-MB-231, JIMT-1, SKOV-3, U937 and KB cells, with combination indices ranging from 0.07 to 0.77. Furthermore, the addition of safingol may reduce the concentrations of conventional chemotherapeutics required to achieve 90% cell-kill by 1 to >3 log-folds. A significant reduction in the cytotoxicity of safingol-based drug combinations was observed in the presence of N-acetyl-L-cysteine, suggesting that ROS is an important factor in mediating the observed synergism. Taken together, our results suggest that the use of safingol-based drug combinations is promising as an effective strategy for cancer therapy and should be investigated.

19.
Int J Oncol ; 37(1): 211-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20514413

ABSTRACT

In the management of metastatic breast cancer, fewer recognized therapeutic standards are available as compared to the early stages of the disease. Thus, it is pertinent to search for new, effective therapy to improve survival, tolerability and quality of life of patients. In this study, a liposomal formulation was developed for a novel dihydrofolate reductase (DHFR) inhibitor, M-V-05. Drug encapsulation into liposomes was achieved by the citrate-based, pH gradient loading technique, with a final drug-to-lipid weight ratio of 0.1:1. The liposome formulation exhibited a sustained release profile of the encapsulated drug that followed first order release kinetics. Liposomal M-V-05 was found to be more effective than the standard DHFR inhibitor, methotrexate, and its activity was comparable to liposomal doxorubicin, with IC50 values of 37 and 59 microM achieved in MDA-MB-231 and JIMT-1 cells, respectively. Similar cytotoxicity was observed in primary patient samples of invasive ductal carcinoma of the breast. The combination of liposomal M-V-05 and liposomal doxorubicin in fixed molar ratio of 3:1 was additive in cytotoxicity, allowing the concentrations of liposomal doxorubicin and liposomal M-V-05 to be reduced by 62 and 46%, respectively. Taken together, liposomal M-V-05 represents a promising agent and offers a potential new adjuvant therapy for breast cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Folic Acid Antagonists/administration & dosage , Liposomes/chemical synthesis , Liposomes/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/metabolism , Carcinoma/pathology , Cell Survival/drug effects , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Drug Compounding/methods , Drug Evaluation, Preclinical , Female , Folic Acid Antagonists/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Liposomes/metabolism , Models, Biological , Spiro Compounds/administration & dosage , Spiro Compounds/chemical synthesis , Spiro Compounds/therapeutic use , Tetrahydrofolate Dehydrogenase/metabolism , Time Factors , Triazines/administration & dosage , Triazines/chemical synthesis , Triazines/therapeutic use , Tumor Cells, Cultured
20.
Anticancer Drugs ; 21(4): 401-10, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20110806

ABSTRACT

Breast cancers are either estrogen receptor-positive (ER) or negative (ER). ER breast cancers are clinically more aggressive and have fewer effective treatment options. Quercetin and vincristine are both active against ER breast cancers and exhibit synergism in vitro. However, the clinical use of quercetin is hampered by its low water solubility. In addition, optimal synergism can only be achieved at a particular ratio of the drugs. Therefore, the objectives of this study are to develop a liposomal formulation to solubilize quercetin, and to co-encapsulate and coordinate the release of quercetin and vincristine in their synergistic ratios to maximize anticancer activity. The optimal synergistic molar ratio of quercetin/vincristine was found to be 1 : 2 by in-vitro MTT assay. Quercetin liposomes were prepared by the film hydration method followed by extrusion, and vincristine was subsequently loaded into the core of the liposomes by remote loading with manganese sulfate and the ionophore A23187. The optimal liposome formulation co-encapsulating quercetin and vincristine comprised egg sphingomyelin/cholesterol/PEG2000 ceramide/quercetin (72.5 : 17.5 : 5 : 5 mol ratio). This formulation was physically stable, enhanced quercetin solubility 8.6 times, co-encapsulated quercetin and vincristine with efficiencies of 78.3 and 78.5%, respectively, and displayed coordinated release of both drugs to maintain the synergistic molar ratio. In-vitro MTT assays of this liposomal formulation showed significant synergism, with a combination index of 0.113 and a dose-reduction index value of 115 at ED50 for vincristine. Therefore, liposomal delivery represents a strategy to solubilize poorly soluble drugs and coordinate the release of two drugs in their synergistic ratio for optimal anticancer effect.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Quercetin/administration & dosage , Vincristine/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Compounding , Drug Evaluation, Preclinical , Drug Stability , Female , Humans , Liposomes , Quercetin/therapeutic use , Receptors, Estrogen/analysis , Vincristine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...