Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Biomacromolecules ; 14(12): 4226-35, 2013 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-24219381

RESUMEN

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.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Dendrímeros/farmacología , Portadores de Fármacos/farmacología , Uniones Estrechas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Amidinas/química , Animales , Células CACO-2 , Dendrímeros/síntesis química , Dendrímeros/metabolismo , Perros , Portadores de Fármacos/síntesis química , Portadores de Fármacos/metabolismo , Humanos , Células de Riñón Canino Madin Darby , Tamaño de la Partícula , Permeabilidad , Polietilenglicoles/química , Uniones Estrechas/efectos de los fármacos
2.
Cytotherapy ; 14(9): 1064-79, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22775077

RESUMEN

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.


Asunto(s)
Técnicas de Cultivo de Célula , Citosol/metabolismo , Sangre Fetal/citología , Leucocitos Mononucleares/citología , Células Madre Mesenquimatosas/citología , Animales , Muerte Celular/genética , Fusión Celular , Proliferación Celular , Supervivencia Celular , Transdiferenciación Celular , Técnicas de Cocultivo , Proteínas Fluorescentes Verdes/análisis , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Células 3T3 NIH
3.
Nanomedicine ; 7(6): 834-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21371568

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Liposomas/química , Quercetina/administración & dosificación , Vincristina/administración & dosificación , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mama/efectos de los fármacos , Mama/patología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones SCID , Quercetina/farmacocinética , Quercetina/uso terapéutico , Trastuzumab , Vincristina/farmacocinética , Vincristina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Anticancer Drugs ; 21(4): 401-10, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20110806

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Quercetina/administración & dosificación , Vincristina/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Femenino , Humanos , Liposomas , Quercetina/uso terapéutico , Receptores de Estrógenos/análisis , Vincristina/uso terapéutico
5.
Int J Pharm ; 349(1-2): 38-46, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-17728083

RESUMEN

A conventional, cholesterol-containing liposome formulation of cisplatin has demonstrated insignificant activity in clinical trials, due in part, to insufficient release of encapsulated content following localization within solid tumors. For this reason, the development of a triggered release liposome formulation is desirable. In this report, cisplatin was encapsulated into lysolipid-containing thermosensitive liposomes (LTSL) using a novel technique, which relies on the equilibration of cisplatin across the liposomal membrane at temperatures above the gel-to-liquid crystalline phase transition temperature (TC) of the bulk phospholipid. Mild heating and drug loading into LTSL did not induce morphological changes of the liposomes. In vitro data demonstrated that >95% of encapsulated cisplatin was released from LTSL within 5 min following mild heating at 42 degrees C, while <5% was released at 37 degrees C. Under similar conditions, lysolipid-free thermosensitive liposomes exhibited 70% release of cisplatin at 42 degrees C, and cholesterol-containing liposomes exhibited negligible drug release at 42 degrees C. The pharmacokinetic profiles of LTSL- and TSL-cisplatin indicated that these formulations were rapidly eliminated from circulation (terminal t(1/2) of 1.09 and 2.83 h, respectively). The therapeutic utility of LTSL-cisplatin formulation will be based on strategies where hyperthermia is applied prior to the administration of the liposomal drug-a strategy similar to that used in the clinical assessment of LTSL-doxorubicin formulation.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Química Farmacéutica , Cisplatino/farmacocinética , Microscopía por Crioelectrón , Portadores de Fármacos , Composición de Medicamentos , Calor , Liposomas , Membranas Artificiales , Ratones , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Permeabilidad , Solubilidad , Temperatura
6.
Mol Cancer Ther ; 6(3): 844-55, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17339368

RESUMEN

Various methods have been explored to enhance antibody-based cancer therapy. The use of multivalent antibodies or fragments against tumor antigens has generated a great deal of interest, as various cellular signals, including induction of apoptosis, inhibition of cell growth/survival, or internalization of the surface molecules, can be triggered or enhanced on extensive cross-linking of the target/antibody complex by the multivalent form of the antibody. The goal of the studies reported here was to develop multivalent antibody constructs via grafting of antibody molecules onto liposome membranes to enhance antibody activity. Using trastuzumab and rituximab as examples, up to a 25-fold increase in the antibody potency in cell viability assay was observed when the antibodies were presented in the multivalent liposome formulation. Key cell survival signaling molecules, such as phosphorylated Akt and phosphorylated p65 nuclear factor-kappaB, were down-regulated on treatment with multivalent liposomal trastuzumab and liposomal rituximab, respectively. Potent in vivo antitumor activity was shown for liposomal trastuzumab. The data presented here showed the potential of liposome technology to enhance the therapeutic effect of antibodies via a mechanism that modulates cell survival through clustering of the target/antibody complex.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias de la Mama/terapia , Animales , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Anticuerpos Antineoplásicos , Antígenos CD20/inmunología , Antígenos de Neoplasias/inmunología , Western Blotting , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Supervivencia Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación hacia Abajo , Femenino , Citometría de Flujo , Genes erbB-2/genética , Genes erbB-2/inmunología , Humanos , Liposomas , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/inmunología , Rituximab , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Trastuzumab
7.
Biochim Biophys Acta ; 1611(1-2): 63-9, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12659946

RESUMEN

Although small, 100-nm liposomes are known to selectively accumulate in solid tumors, the individual contributions of liposome influx and egress rates are not well understood. The aim of this work was to determine influx and efflux kinetics for 100-nm, 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC)/cholesterol (Chol) liposomes by inducing aggregate formation of biotinylated liposomes upon administering avidin. Injecting 50 microg of neutravidin intravenously to mice that had previously been administered 100 mg/kg DPSC/Chol liposomes containing 0.5 mol% biotin-conjugated lipid resulted in >90% elimination of the liposomes from plasma within 1 h. This rapid removal by the reticuloendothelial system (RES) permitted the determination of the tumor efflux kinetics due to negligible tumor influx after neutravidin injection. The tumor efflux rate constant (k(-1)) was determined to be 0.041 h(-1) when neutravidin was injected 4 h after liposome injection. This allowed the determination of the tumor influx rate constant (k(1)), which under these conditions was 0.022 h(-1). Therefore, DSPC/Chol liposomal accumulation, in LS180 solid tumors, is dictated primarily by plasma liposome concentrations and liposome egress is comparable or slightly faster than influx into the tumors. This method is applicable for a wide range of lipid doses, and can be used to characterize influx and efflux parameters at different time points after accumulation. The application, therefore, has the potential to be used to fully characterize the impact of different liposome parameters such as lipid composition, steric stabilization, size and dose on tumor accumulation kinetics.


Asunto(s)
Neoplasias del Colon/metabolismo , Liposomas/farmacocinética , Animales , Avidina/farmacología , Biotina , Neoplasias del Colon/sangre , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/química , Modelos Animales de Enfermedad , Femenino , Liposomas/sangre , Liposomas/química , Ratones , Fosfatidilcolinas/química , Factores de Tiempo , Distribución Tisular
8.
Biochim Biophys Acta ; 1613(1-2): 115-21, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12832092

RESUMEN

Phosphatidylserine (PS) membrane exposure plays an important role in blood coagulation, and the development of a liposome formulation containing PS may be of potential therapeutic utility if they can be designed to achieve tumor selective thrombosis. The objective of this study was to develop proof-of-principle data for a thrombogenic PS liposome targeted to vascular cell adhesion molecule 1 (VCAM-1) via the attachment of an anti-VCAM-1 monoclonal antibody (Ab). We have evaluated binding of the anti-VCAM-1 Ab-conjugated PS liposomes to VCAM-1 using two in vitro models, as well as assessing the ability of these liposomes to catalyze blood coagulation reactions. Binding of the Ab-conjugated PS liposomes containing 2 or 14 mol% 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[poly(ethylene glycol) 2000] (DSPE-PEG(2000)) to interleukin 1alpha stimulated human umbilical vein endothelial cells was 8- and 16-fold higher than those without conjugated Ab, respectively, based on the percentage relative increase in cell associated lipid for these liposomes. Binding to VCAM-1-coated ELISA plates produced similar results. The VCAM-1-bound Ab-conjugated PS liposomes were capable of catalyzing blood coagulation reactions upon the exposure of the thrombogenic PS membrane surface. This control of PS surface exposure was achieved using exchangeable PEG-derivatized phosphatidylethanolamines (PE-PEG), with 97% of clotting activity recovered after PE-PEG exchanged out. Our results demonstrate the potential for considering further development of procoagulant liposomes that selectively target thrombogenesis in tumor vasculature.


Asunto(s)
Endotelio Vascular/fisiología , Liposomas , Fosfatidilserinas/farmacología , Molécula 1 de Adhesión Celular Vascular/química , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Sitios de Unión , Coagulación Sanguínea/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Fibrinolíticos/uso terapéutico , Humanos , Neoplasias/irrigación sanguínea , Neovascularización Patológica/prevención & control , Trastuzumab , Venas Umbilicales , Molécula 1 de Adhesión Celular Vascular/inmunología
9.
Biochim Biophys Acta ; 1560(1-2): 37-50, 2002 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-11958774

RESUMEN

Liposomes containing the acidic phospholipid phosphatidylserine (PS) have been shown to avidly interact with proteins involved in blood coagulation and complement activation. Membranes with PS were therefore used to assess the shielding properties of poly(ethylene glycol 2000)-derivatized phosphatidylethanolamine (PE-PEG(2000)) with various acyl chain lengths on membranes containing reactive lipids. The desorption of PE-PEG(2000) from PS containing liposomes was studied using an in vitro assay which involved the transfer of PE-PEG(2000) into multilamellar vesicles, and the reactivity of PS containing liposomes was monitored by quantifying interactions with blood coagulation proteins. The percent inhibition of clotting activity of PS liposomes was dependent on the PE-PEG(2000) content. 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine (DSPE)-PEG(2000) which transferred out slowly from PS liposomes was able to abolish >80% of clotting activity of PS liposomes at 15 mol%. This level of DSPE-PEG(2000) was also able to extend the mean residence time of PS liposomes from 0.2 h to 14 h. However, PE-PEG(2000) with shorter acyl chains such as 1,2-dimyristyl-sn-glycero-3-phosphoethanolamine-PEG(2000) were rapidly transferred out from PS liposomes, which resulted in a 73% decrease in clotting activity inhibition and 45% of administered intravenously liposomes were removed from the blood within 15 min after injection. Thus, PS facilitates the desorption of PE-PEG(2000) from PS containing liposomes, thereby providing additional control of PEG release rates from membrane surfaces. These results suggest that membrane reactivity can be selectively regulated by surface grafted PEGs coupled to phosphatidylethanolamine of an appropriate acyl chain length.


Asunto(s)
Membrana Celular/efectos de los fármacos , Liposomas/química , Fosfatidilserinas/farmacología , Polietilenglicoles/química , Animales , Sistemas de Liberación de Medicamentos , Lipoproteínas/sangre , Liposomas/sangre , Ratones , Micelas , Neoplasias/irrigación sanguínea , Fosfatidiletanolaminas/sangre , Fosfatidiletanolaminas/química , Fosfatidilserinas/administración & dosificación , Fosfatidilserinas/sangre , Propiedades de Superficie , Factores de Tiempo
10.
J Control Release ; 104(2): 271-88, 2005 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-15907579

RESUMEN

In the present study, doxorubicin was encapsulated into two thermosensitive liposome formulations which were composed of DPPC/MSPC/DSPE-PEG(2000) (90/10/4 mole ratio) or DPPC/DSPE-PEG(2000) (95/5 mole ratio). Doxorubicin loading was achieved through the use of a pH gradient or a novel procedure that involved doxorubicin complexation with manganese. Regardless of the initial drug-to-lipid ratios (D:L), the final D:L reached a maximum of 0.05 (w/w) when doxorubicin was encapsulated via a pH gradient for both thermosensitive liposome formulations. In contrast, the final maximum D:L achieved through manganese complexation was 0.2 (w/w), and this loading method did not affect temperature-induced drug release, with 85% of drug released from MSPC-containing liposomes within 10 min at 42 degrees C but <5% released over 60 min at 37 degrees C. When the thermosensitive liposomes prepared via the two different loading methods were injected into mice, similar plasma elimination profiles were observed. Cryo-transmission electron microscopy analysis indicated the presence of doxorubicin fiber bundles in liposomes loaded via pH gradient, compared to a stippled and diffuse morphology in those loaded via manganese complexation. To investigate the effect of intraliposomal pH on drug precipitate morphology, the A23187 ionophore (mediates Mn(2+)/H(+) exchange) was added to liposomes loaded with doxorubicin-manganese complex, and the stippled and diffuse appearance could be converted to one exhibiting fiber bundles after acidification of the liposome core. This suggests that the formation of doxorubicin-manganese complex is favored when the intraliposomal pH is >6.5. During the conversion to the fiber bundle morphology, no doxorubicin release was observed when A23187 was added to liposomes exhibiting a 0.05 (w/w), whereas a significant release was noted when the initial D:L was 0.2 (w/w). Following acidification of the liposomal interior and establishment of an apparent new D:L equilibrium, the measured D:L ratio was 0.05 (w/w). In conclusion, the manganese complexation loading method increased the encapsulation efficiency of doxorubicin in thermosensitive liposomes with no major impact on temperature-triggered drug release or pharmacokinetics.


Asunto(s)
Doxorrubicina/administración & dosificación , Manganeso/administración & dosificación , Calcimicina/farmacología , Microscopía por Crioelectrón , Portadores de Fármacos , Concentración de Iones de Hidrógeno , Liposomas
11.
J Control Release ; 220(Pt A): 438-446, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26551345

RESUMEN

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.


Asunto(s)
1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , Antineoplásicos Fitogénicos/administración & dosificación , Dendrímeros/química , Portadores de Fármacos , Nanopartículas , Nylons/química , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/administración & dosificación , 1,2-Dipalmitoilfosfatidilcolina/química , 1,2-Dipalmitoilfosfatidilcolina/metabolismo , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dendrímeros/metabolismo , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Femenino , Humanos , Ratones SCID , Nanotecnología , Nylons/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Paclitaxel/química , Paclitaxel/metabolismo , Solubilidad , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Pharm Pharmacol ; 67(11): 1492-502, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26303576

RESUMEN

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.


Asunto(s)
Células Sanguíneas/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Dendrímeros/toxicidad , Polietilenglicoles/química , Animales , Dendrímeros/química , Hemoglobinas/metabolismo , Humanos , Tiempo de Tromboplastina Parcial , Unión Proteica , Tiempo de Protrombina , Ratas , Albúmina Sérica/metabolismo , Compuestos de Sulfhidrilo/química , Células U937
13.
Curr Pharm Des ; 21(19): 2629-42, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25876918

RESUMEN

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.


Asunto(s)
Dendrímeros/administración & dosificación , Dendrímeros/efectos adversos , Sistemas de Liberación de Medicamentos , Preparaciones Farmacéuticas/administración & dosificación , Administración Oral , Disponibilidad Biológica , Dendrímeros/farmacocinética , Absorción Gastrointestinal/efectos de los fármacos , Humanos , Preparaciones Farmacéuticas/química , Propiedades de Superficie
14.
J Control Release ; 84(3): 161-70, 2002 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-12468219

RESUMEN

The therapeutic activity of numerous drugs can be dramatically improved by liposomal encapsulation. However, this requires that liposomes retain their encapsulated drugs following systemic administration. Often, in vitro drug release assays do not accurately predict the liposomal drug retention properties observed in vivo. We postulate that this discrepancy is due to the large membrane pool present in blood cells and tissues, into which drugs can distribute after in vivo administration. Herein we describe an in vitro drug release assay that more accurately predicts in vivo drug release from liposomes following systemic administration. Drug-encapsulated large unilamellar vesicles (LUVs) approximately 100 nm in diameter were incubated with a 100-fold excess of multilamellar vesicles (MLVs) containing 300 mM sucrose, which served as 'acceptors' for drug release and transfer from 'donor' LUVs. Following incubation at 37 degrees C, the donor and acceptor populations were separated with greater than 90% efficiency by centrifugation at 1600xg for 10 min. The amount of drug in the MLV pellet reflects the degree of drug leakage from the donor liposomes. Drug release profiles using this in vitro assay were compared to those obtained using dialysis-based assays and in vivo results following systemic administration to mice. Our results indicate that this release assay is a better predictor of in vivo drug transfer than dialysis-based systems. We also demonstrate its utility in measuring exchange of lipophilic components.


Asunto(s)
Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Ensayo de Materiales/métodos , Animales , Doxorrubicina/química , Doxorrubicina/farmacocinética , Portadores de Fármacos , Femenino , Inyecciones Intravenosas , Cinética , Liposomas , Ratones , Microesferas , Tamaño de la Partícula , Valor Predictivo de las Pruebas , Distribución Tisular , Verapamilo/química , Verapamilo/farmacocinética
15.
Nanomedicine (Lond) ; 9(11): 1665-79, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24294981

RESUMEN

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.


Asunto(s)
Antineoplásicos/química , Sistemas de Liberación de Medicamentos , Leucemia Mieloide Aguda/tratamiento farmacológico , Liposomas/química , Animales , Proliferación Celular , Supervivencia Celular , Ceramidas/química , Colesterol/química , Cromatografía Liquida , Femenino , Humanos , Concentración 50 Inhibidora , Leucemia Mieloide Aguda/metabolismo , Lípidos/química , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Fosfatidilcolinas/química , Esfingosina/análogos & derivados , Esfingosina/química , Espectrometría de Masas en Tándem , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Int J Oncol ; 42(5): 1605-12, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23467622

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Diterpenos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fenantrenos/farmacología , Neoplasias de la Mama/metabolismo , Butadienos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos Clínicos como Asunto , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Compuestos Epoxi/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Nitrilos/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo
17.
J Control Release ; 172(3): 852-61, 2013 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-24459693

RESUMEN

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.


Asunto(s)
Antineoplásicos/administración & dosificación , Camptotecina/análogos & derivados , Doxorrubicina/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Camptotecina/administración & dosificación , Camptotecina/farmacología , Camptotecina/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sinergismo Farmacológico , Femenino , Humanos , Irinotecán , Liposomas/química , Manganeso/química , Ratones , Ratones SCID , Neoplasias Ováricas/patología , Ovario/efectos de los fármacos , Ovario/patología
18.
Eur J Med Chem ; 70: 400-10, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24177367

RESUMEN

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.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Pirazinas/farmacología , Timidina Fosforilasa/antagonistas & inhibidores , Triazinas/farmacología , Uracilo/análogos & derivados , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Pirazinas/síntesis química , Pirazinas/química , Relación Estructura-Actividad , Timidina Fosforilasa/metabolismo , Triazinas/síntesis química , Triazinas/química , Uracilo/química
19.
Int J Nanomedicine ; 7: 739-51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359453

RESUMEN

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.


Asunto(s)
Carboplatino/administración & dosificación , Transportadores de Ácido Fólico/metabolismo , Liposomas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Animales , Carboplatino/química , Carboplatino/farmacocinética , Línea Celular Tumoral , Femenino , Histocitoquímica , Humanos , Estimación de Kaplan-Meier , Liposomas/química , Liposomas/farmacocinética , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones SCID , Terapia Molecular Dirigida , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Oncol ; 40(3): 703-10, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22086152

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Carboplatino/administración & dosificación , Carboplatino/farmacocinética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Folato Anclados a GPI/metabolismo , Liposomas/administración & dosificación , Antineoplásicos/química , Carboplatino/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática , Humanos , Células KB , Liposomas/química , Liposomas/farmacocinética , Fosforilación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA