Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Pharmacol Exp Ther ; 351(1): 61-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047517

RESUMEN

Esterase hydrolysis of drugs can accelerate their elimination, thereby limiting their efficacy. Polyethylene glycol (PEG) covalently attached to drugs (pegylation) is known to improve the efficiency of many drugs. Using as a test agent the novel phospho-ibuprofen (PI), we examined whether pegylation of PI could abrogate its hydrolytic degradation by esterases; PI, known to inhibit colon cancer growth, has a carboxylic ester hydrolyzable by carboxylesterases (CES). We covalently attached mPEG-2000 to PI (PI-PEG) and studied its stability by exposing it to cells overexpressing CES and by administering it to mice. We also evaluated PI-PEG's anticancer efficacy in human colon cancer xenografts and in Apc(min/+) mice. PI-PEG was stable in the presence of cells overexpressing CES1 or CES2, whereas PI was extensively hydrolyzed (90.2 ± 0.7%, 14.3 ± 1.1%, mean ± S.E.M.). In mice, PI was nearly completely hydrolyzed. Intravenous administration of PI-PEG resulted in significant levels in blood and in colon cancer xenografts (xenograft values in parentheses): area under the curve for 0-24 hours = 2351 (2621) (nmol/g) × h; Cmax = 1965 (886) nmol/g; Tmax = 0.08 (2) hour. The blood levels of ibuprofen, its main hydrolytic product, were minimal. Compared with controls, PI-PEG inhibited the growth of the xenografts by 74.8% (P < 0.01) and reduced intestinal tumor multiplicity in Apc(min/+) mice by 73.1% (P < 0.01), prolonging their survival (100% versus 55.1% of controls; P = 0.013). Pegylation protects PI from esterase hydrolysis and improves its pharmacokinetics. In preclinical models of colon cancer, PI-PEG is a safe and efficacious agent that merits further evaluation.


Asunto(s)
Antineoplásicos/farmacocinética , Ibuprofeno/análogos & derivados , Organofosfatos/farmacocinética , Polietilenglicoles/química , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Células HEK293 , Humanos , Hidrólisis , Ibuprofeno/efectos adversos , Ibuprofeno/síntesis química , Ibuprofeno/farmacocinética , Ibuprofeno/uso terapéutico , Ratones , Organofosfatos/efectos adversos , Organofosfatos/síntesis química , Organofosfatos/uso terapéutico , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Pharmacol Exp Ther ; 340(2): 422-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22085648

RESUMEN

Phospho-nonsteroidal anti-inflammatory drugs (phospho-NSAIDs) are novel NSAID derivatives with improved anticancer activity and reduced side effects in preclinical models. Here, we studied the metabolism of phospho-NSAIDs by carboxylesterases and assessed the impact of carboxylesterases on the anticancer activity of phospho-NSAIDs in vitro and in vivo. The expression of human liver carboxylesterase (CES1) and intestinal carboxylesterase (CES2) in human embryonic kidney 293 cells resulted in the rapid intracellular hydrolysis of phospho-NSAIDs. Kinetic analysis revealed that CES1 is more active in the hydrolysis of phospho-sulindac, phospho-ibuprofen, phospho-naproxen, phospho-indomethacin, and phospho-tyrosol-indomethacin that possessed a bulky acyl moiety, whereas the phospho-aspirins are preferentially hydrolyzed by CES2. Carboxylesterase expression leads to a significant attenuation of the in vitro cytotoxicity of phospho-NSAIDs, suggesting that the integrity of the drug is critical for anticancer activity. Benzil and bis-p-nitrophenyl phosphate (BNPP), two carboxylesterase inhibitors, abrogated the effect of carboxylesterases and resensitized carboxylesterase-expressing cells to the potent cytotoxic effects of phospho-NSAIDs. In mice, coadministration of phospho-sulindac and BNPP partially protected the former from esterase-mediated hydrolysis, and this combination more effectively inhibited the growth of AGS human gastric xenografts in nude mice (57%) compared with phospho-sulindac alone (28%) (p = 0.037). Our results show that carboxylesterase mediates that metabolic inactivation of phospho-NSAIDs, and the inhibition of carboxylesterases improves the efficacy of phospho-NSAIDs in vitro and in vivo.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Carboxilesterasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Organofosfatos/metabolismo , Organofosfatos/farmacología , Animales , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/uso terapéutico , Aspirina/análogos & derivados , Aspirina/metabolismo , Aspirina/farmacología , Carboxilesterasa/antagonistas & inhibidores , Carboxilesterasa/genética , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Interacciones Farmacológicas/fisiología , Quimioterapia Combinada/métodos , Inhibidores Enzimáticos/farmacología , Femenino , Células HEK293 , Humanos , Hidrólisis , Ibuprofeno/análogos & derivados , Ibuprofeno/metabolismo , Ibuprofeno/farmacología , Indometacina/análogos & derivados , Indometacina/metabolismo , Indometacina/farmacología , Concentración 50 Inhibidora , Cinética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Naproxeno/análogos & derivados , Naproxeno/metabolismo , Naproxeno/farmacología , Nitrofenoles/farmacología , Nitrofenoles/uso terapéutico , Organofosfatos/sangre , Organofosfatos/uso terapéutico , Compuestos Organofosforados/sangre , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/farmacología , Compuestos Organofosforados/uso terapéutico , Fenilglioxal/análogos & derivados , Fenilglioxal/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Sulindac/análogos & derivados , Sulindac/sangre , Sulindac/metabolismo , Sulindac/farmacología , Sulindac/uso terapéutico
3.
Pharm Res ; 29(6): 1435-43, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22072052

RESUMEN

PURPOSE: To incorporate phospho-ibuprofen (P-I), a lipophilic, water insoluble novel anti-cancer agent, into pegylated liposomes and upon formulation optimization to evaluate its antitumor activity in vitro and in vivo. METHODS: P-I loaded liposomes were prepared using the thin-film hydration method, and characterized for size, zeta potential, drug content and drug release. We examined their physical stability by particle size changes; their lyophilization ability in the presence of cryoprotectants; and their antitumor activity in vitro in human cancer cell lines and in vivo in a xenograft murine model. RESULTS: P-I was successfully loaded into liposomes consisting of soy-PC and PEG(2000)-PE. These liposomes were <150 nm in diameter; exhibited prolonged stability in suspension and can be lyophilized using sucrose as cryoprotectant. P-I liposomes inhibited the growth of human cancer cell lines in vitro and in vivo of xenograft in nude mice to a greater extent than free P-I. CONCLUSIONS: High levels of P-I can be incorporated into liposomes which can be lyophilized in the presence of sucrose and showed good stability upon storage. Moreover, these drug-incorporating liposomes were capable of inhibiting the growth of xenografted tumors in mice more effectively than free P-I. These results justify further development of the P-I liposomes.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Ibuprofeno/análogos & derivados , Lípidos/química , Organofosfatos/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Química Farmacéutica , Neoplasias del Colon/patología , Crioprotectores/química , Relación Dosis-Respuesta a Droga , Composición de Medicamentos , Estabilidad de Medicamentos , Femenino , Liofilización , Humanos , Ibuprofeno/administración & dosificación , Ibuprofeno/química , Ibuprofeno/farmacología , Concentración 50 Inhibidora , Inyecciones Intraperitoneales , Cinética , Liposomas , Ratones , Ratones SCID , Organofosfatos/administración & dosificación , Organofosfatos/química , Tamaño de la Partícula , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Solubilidad , Propiedades de Superficie , Tecnología Farmacéutica/métodos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Pharm Res ; 29(12): 3292-301, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22782648

RESUMEN

PURPOSE: To investigate the metabolism of phospho-aspirin (PA, MDC-22), a novel anti-cancer and anti-inflammatory agent. METHODS: The metabolism of PA was studied in the liver and intestinal microsomes from mouse, rat and human. RESULTS: PA is rapidly deacetylated to phospho-salicylic acid (PSA), which undergoes regioselective oxidation to generate 3-OH-PSA and 5-OH-PSA. PSA also can be hydrolyzed to give salicylic acid (SA), which can be further glucuronidated. PA is far more stable in human liver or intestinal microsomes compared to those from mouse or rat due to its slowest deacetylation in human microsomes. Of the five major human cytochrome P450 (CYP) isoforms, CYP2C19 and 2D6 are the most active towards PSA. In contrast to PSA, conventional SA is not appreciably oxidized by the CYPs and liver microsomes, indicating that PSA is a preferred substrate of CYPs. Similarly, PA, in contrast to PSA, cannot be directly oxidized by CYPs and liver microsomes, indicating that the acetyl group of PA abrogates its oxidation by CYPs. CONCLUSIONS: Our findings establish the metabolism of PA, reveal significant inter-species differences in its metabolic transformations, and provide an insight into the role of CYPs in these processes.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Aspirina/análogos & derivados , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Microsomas/metabolismo , Organofosfatos/metabolismo , Animales , Aspirina/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Ratones , Oxidación-Reducción , Ratas
5.
Pharm Res ; 29(11): 3090-101, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22723123

RESUMEN

PURPOSE: To evaluate the antitumor efficacy of solid lipid nanoparticle-encapsulated phospho-sulindac (SLN-PS) in human lung cancer. METHODS: PS was incorporated into SLNs using the emulsion evaporation technique. We determined the antitumor activity of SLN-PS in cultured lung cancer cells. The performance of SLN-PS was further evaluated by pharmacokinetic studies in mice and in a model of human lung cancer xenografts in nude mice. RESULTS: SLN-PS was >4-fold more potent than PS in inhibiting the growth of A549 and H510 cells in vitro. SLN-PS enhanced cellular uptake and facilitated PS accumulation in mitochondria, leading to oxidative stress and apoptosis via the mitochondrial-apoptosis pathway. SLN-PS was highly effective in suppressing the growth of A549 xenografts (78% inhibition compared to control, p < 0.01); while PS had no significant effect. Formulation of PS in SLNs resulted in improved pharmacokinetics in mice and an enhanced (≈ 14-fold) accumulation of PS and its metabolites in A549 xenografts. Finally, SLN-PS enhanced urinary F2-isoprostane uniquely in mice bearing A549 xenografts compared to untreated controls, suggesting that SLN-PS specifically induced oxidative stress in tumors. CONCLUSIONS: Our results show that SLN-PS is efficacious in suppressing the growth of lung cancer and merits further evaluation.


Asunto(s)
Antineoplásicos/farmacología , Lípidos/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Nanopartículas/administración & dosificación , Compuestos Organofosforados/farmacología , Sulindac/análogos & derivados , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Química Farmacéutica , Femenino , Humanos , Lípidos/química , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Nanopartículas/química , Compuestos Organofosforados/administración & dosificación , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacocinética , Sulindac/administración & dosificación , Sulindac/química , Sulindac/farmacocinética , Sulindac/farmacología , Superóxidos/metabolismo , Distribución Tisular , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Lipids Health Dis ; 8: 30, 2009 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-19638223

RESUMEN

The aim of this work was to assess the effect of chronic administration of protonated nanostructured aluminosilicate (NSAS) on the plasma cholesterol levels and development of atherosclerotic lesions in Apolipoprotein (ApoE) deficient mice fed a high cholesterol and high fat diet. Apolipoprotein E (ApoE) deficient mice were divided into the following treatment groups: protonated NSAS 1.4% (w/w), untreated control and 2% (w/w) stigmastanol mixed with high-cholesterol/high-fat diet. Animals were treated for 12 weeks, blood samples were withdrawn every 4 weeks for determination of plasma cholesterol and triglyceride levels. At the end of the study the aortic roots were harvested for assessment of atherosclerotic lesions. NSAS at 1.4% (w/w) and stigmastanol at 2% (w/w) treatment groups showed significant decreases in plasma cholesterol concentrations at all time points relative to the control animals. The lesion sum area in 1.4% (w/w) NSAS and 2% (w/w) stigmastanol groups were significantly less from the control animals. In conclusion, in this study, the effectiveness of chronic administration of protonated NSAS material in the reduction of plasma cholesterol levels and decrease in development of atherosclerotic lesions was demonstrated in Apo-E deficient mice model.


Asunto(s)
Silicatos de Aluminio/uso terapéutico , Anticolesterolemiantes/uso terapéutico , Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Colesterol en la Dieta/administración & dosificación , Colesterol/sangre , Nanopartículas del Metal/uso terapéutico , Animales , Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/genética , Bentonita , Peso Corporal/efectos de los fármacos , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Triglicéridos/sangre
7.
Adv Drug Deliv Rev ; 60(6): 757-67, 2008 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-18096269

RESUMEN

Parenteral formulations, particularly intravascular ones, offer a unique opportunity for direct access to the bloodstream and rapid onset of drug action as well as targeting to specific organ and tissue sites. Triglyceride emulsions, liposomes and micellar solutions have been traditionally used to accomplish these tasks and there are several products on the market using these lipid formulations. The broader application of these lipid systems in parenteral drug delivery, however, particularly with new chemical entities, has been limited due primarily to the following reasons: a) only a small number of parenteral lipid excipients are approved, b) there is increasing number of drugs that are partially or not soluble in conventional oils and other lipid solvents, and c) the ongoing requirement for site-specific targeting and controlled drug release. Thus, there is growing need to expand the array of targetable lipid-based systems to deliver a wide variety of drugs and produce stable formulations which can be easily manufactured in a sterile form, are cost-effective and at least as safe and efficacious as the earlier developed systems. These advanced parenteral lipid-based systems are at various stages of preclinical and clinical development which include nanoemulsions, nanosuspensions and polymeric phospholipid micelles. This review article will showcase these parenteral lipid nanosystems and discuss advances in relation to formulation development, processing and manufacturing, and stability assessment. Factors controlling drug encapsulation and release and in vivo biodistribution will be emphasized along with in vitro/in vivo toxicity and efficacy case studies. Emerging lipid excipients and increasing applications of injectable lipid nanocarriers in cancer chemotherapy and other disease indications will be highlighted and in vitro/in vivo case studies will be presented. As these new parenteral lipid systems advance through the clinic and product launch, their therapeutic utility and value will certainly expand.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Preparaciones Farmacéuticas/administración & dosificación , Animales , Emulsiones/administración & dosificación , Emulsiones/química , Humanos , Inyecciones , Lípidos/administración & dosificación , Lípidos/química , Micelas , Nanopartículas/química , Preparaciones Farmacéuticas/química , Polímeros/administración & dosificación , Polímeros/química , Solubilidad , Suspensiones/administración & dosificación , Suspensiones/química
8.
J Pharm Sci ; 96(2): 235-48, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17051593

RESUMEN

The intestinal efflux pump, P-glycoprotein (P-gp), located in the apical membranes of intestinal absorptive cells, can reduce the bioavailability of a wide range of drugs which are substrates for this membrane transporter. In addition to anticancer and anti-HIV drugs, NCEs for other disease indications are P-gp substrates and there is considerable interest in inhibiting P-gp and thus increasing the bioavailability of these molecules. In this review article, an overview of P-gp and its role in drug transport and absorption will be presented first and then formulation strategies to effectively inhibit P-gp will be discussed and compared. These strategies independently and in combination, are: (a) coadministration of another P-gp substrate/specific inhibitor, and (b) incorporation of a nonspecific lipid and/or polymer excipient in the formulation. The first approach, although very effective in inhibiting P-gp, utilizes a second active compound in the formulation and thus imposes regulatory constraints and long development timelines on such combination products. Excipient inhibitors appear to have minimal nonspecific pharmacological activity and thus potential side effects of specific active compound inhibitors can be avoided. Case studies will be presented where specific active compounds, surfactants, polymers, and formulations incorporating these molecules are shown to significantly improve the intestinal absorption of poorly soluble and absorbed drugs as a result of P-gp inhibition and enhanced drug transport in vitro.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Excipientes/farmacología , Absorción Intestinal/efectos de los fármacos , Preparaciones Farmacéuticas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Disponibilidad Biológica , Química Farmacéutica , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Lípidos/farmacología
9.
Adv Drug Deliv Rev ; 56(9): 1243-55, 2004 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-15109767

RESUMEN

Tocols represent a family of tocopherols, tocotrienols, and their derivatives, and are fundamentally derived from the simplest tocopherol, 6-hydroxy-2-methyl-2-phytylchroman, which is referred to as "tocol". The most common tocol is D-alpha-tocopherol, also known as vitamin E. Tocols can be excellent solvents for water insoluble drugs and are compatible with other cosolvents, oils and surfactants. This review highlights the major developments in the use of tocols in parenteral emulsions for drug delivery, with a focus on drug solubilization, physicochemical properties, and biopharmaceutical applications. Tocol emulsions offer an appealing alternative for the parenteral administration of poorly soluble drugs, including major chemotherapeutics such as paclitaxel. Data will be presented on solubilization of paclitaxel, a key chemotherapeutic agent, and its corresponding formulation development, toxicity, efficacy and pharmacokinetic studies in animal models and humans. The breadth of the utility of tocol-based emulsions will be discussed and examples of specific therapeutic drugs and applications will be provided. As these formulations progress further in the clinic, the therapeutic utility of tocol emulsions is anticipated to expand.


Asunto(s)
Portadores de Fármacos/química , Emulsiones/química , Tocoferoles/química , Tocotrienoles/química , Amiodarona/administración & dosificación , Amiodarona/química , Química Farmacéutica , Ensayos Clínicos como Asunto , Emulsiones Grasas Intravenosas/química , Humanos , Paclitaxel/administración & dosificación , Paclitaxel/química , Solubilidad
10.
Acta Pharm Sin B ; 4(2): 112-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26579372

RESUMEN

The enormous progress biotechnology, bioinformatics and nanotechnology made in recent years provides opportunities and scientific framework for development of biomedicine and constitutes a paradigm shift in pharmaceutical R&D and drug innovation. By analyzing the data and related information at R&D level over the past decades, developmental tendency and R&D patterns were summarized. We found that a growing number of biologics in the pipeline of pharma companies with successful products already in the market though, small molecular entities have primarily dominated drug innovation. Additionally, small/medium size companies will continue to play a key role in the development of small molecule drugs and biologics in a multi-channel integrated process. More importantly, modern and effective R&D strategies in biomedicine development to predict and evaluate efficacy and/or safety of 21st century therapeutics are urgently needed. To face new challenges, developmental strategies were proposed, in terms of molecular targeted medicine, generic drugs, new drug delivery system and protein-based drugs. Under the current circumstances, interdisciplinary cooperation mode and policy related to drug innovation in China were deeply discussed as well.

11.
PLoS One ; 8(5): e61532, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23650499

RESUMEN

New agents are needed to treat pancreatic cancer, one of the most lethal human malignancies. We synthesized phospho-valproic acid, a novel valproic acid derivative, (P-V; MDC-1112) and evaluated its efficacy in the control of pancreatic cancer. P-V inhibited the growth of human pancreatic cancer xenografts in mice by 60%-97%, and 100% when combined with cimetidine. The dominant molecular target of P-V was STAT3. P-V inhibited the phosphorylation of JAK2 and Src, and the Hsp90-STAT3 association, suppressing the activating phosphorylation of STAT3, which in turn reduced the expression of STAT3-dependent proteins Bcl-xL, Mcl-1 and survivin. P-V also reduced STAT3 levels in the mitochondria by preventing its translocation from the cytosol, and enhanced the mitochondrial levels of reactive oxygen species, which triggered apoptosis. Inhibition of mitochondrial STAT3 by P-V was required for its anticancer effect; mitochondrial STAT3 overexpression rescued animals from the tumor growth inhibition by P-V. Our results indicate that P-V is a promising candidate drug against pancreatic cancer and establish mitochondrial STAT3 as its key molecular target.


Asunto(s)
Antineoplásicos/farmacología , Organofosfatos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Factor de Transcripción STAT3/metabolismo , Ácido Valproico/análogos & derivados , Animales , Apoptosis , Línea Celular Tumoral , Cimetidina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/prevención & control , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Ácido Valproico/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Nanomedicine (Lond) ; 7(10): 1577-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23148540

RESUMEN

Nanotechnology is revolutionizing our approach to drug delivery, a key determinant of drug efficacy. Here, we present cancer drug delivery strategies that exploit nanotechnology, providing first an overview of tumor biology aspects that critically affect the design of drug delivery carriers, namely the enhanced permeability and retention effect, the lower tumor extracellular pH and tumor-specific antigens. In general, nanoscience-based approaches have circumvented limitations in the delivery of cancer therapeutics, related to their poor aqueous solubility and toxicity issues with conventional vehicles and resulted in improved pharmacokinetics and biodistribution. Included in the discussion are promising examples and pharmaceutical perspectives on liposomes, nanoemulsions, solid lipid nanoparticles, polymeric nanoparticles, dendrimers, carbon nanotubes and magnetic nanoparticles. As the cardinal features of the ideal multifunctional cancer drug nanocarrier are becoming clear, and drug development challenges are proactively addressed, we anticipate that future advances will enhance therapeutic outcomes by refining the delivery and targeting of complex payloads.


Asunto(s)
Antineoplásicos/uso terapéutico , Nanoestructuras , Antineoplásicos/farmacocinética , Humanos , Concentración de Iones de Hidrógeno , Distribución Tisular
13.
Br J Pharmacol ; 167(1): 222-32, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22489789

RESUMEN

BACKGROUND AND PURPOSE: Phospho-ibuprofen (MDC-917) and phospho-sulindac (OXT-328) are highly effective in cancer and arthritis treatment in preclinical models. Here, we investigated their metabolism by major human cytochrome P450s (CYPs) and flavin monooxygenases (FMOs). EXPERIMENTAL APPROACH: The CYP/FMO-catalysed metabolism of phospho-ibuprofen and phospho-sulindac was studied by using in silico prediction modelling and a direct experimental approach. KEY RESULTS: The CYP isoforms catalyse the oxidation of non-steroidal anti-inflammatory drugs (NSAIDs) and phospho-NSAIDs, with distinct activity and regioselectivity. CYP1A2, 2C19, 2D6 and 3A4 oxidize phospho-ibuprofen, but not ibuprofen; whereas CYP2C9 oxidizes ibuprofen, but not phospho-ibuprofen. All CYPs tested oxidize phospho-sulindac, but not sulindac. Among the five CYPs evaluated, CYP3A4 and 2D6 are the most active in the oxidation of phospho-ibuprofen and phospho-sulindac respectively. FMOs oxidized phospho-sulindac and sulindac, but not phospho-ibuprofen or ibuprofen. FMOs were more active towards phospho-sulindac than sulindac, indicating that phospho-sulindac is a preferred substrate of FMOs. The susceptibility of phospho-NSAIDs to CYP/FMO-mediated metabolism was also reflected in their rapid oxidation by human and mouse liver microsomes, which contain a full complement of CYPs and FMOs. Compared with conventional NSAIDs, the higher activity of CYPs towards phospho-ibuprofen and phospho-sulindac may be due to their greater lipophilicity, a key parameter for CYP binding. CONCLUSIONS AND IMPLICATIONS: CYPs and FMOs play an important role in the metabolism of phospho-NSAIDs, resulting in differential pharmacokinetic profiles between phospho-NSAIDs and NSAIDs in vivo. The consequently more rapid detoxification of phospho-NSAIDs is likely to contribute to their greater safety.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Ibuprofeno/análogos & derivados , Organofosfatos/metabolismo , Oxigenasas/metabolismo , Sulindac/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Ibuprofeno/metabolismo , Microsomas Hepáticos/metabolismo , Modelos Biológicos , Oxidación-Reducción , Proteínas Recombinantes/metabolismo
14.
Int J Oncol ; 41(4): 1199-203, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22842609

RESUMEN

Phospho-sulindac (P-S, OXT-328), a novel sulindac derivative, has shown superior anticancer efficacy and safety compared to sulindac. In this study, we investigated the efficacy of topical P-S hydrogel in the treatment of non-melanoma skin cancer in preclinical models. P-S is a potent inhibitor of A431 epidermoid carcinoma in vitro and achieves this effect by inhibiting cell proliferation and inducing apoptosis. The anticancer efficacy of topical and oral P-S was further evaluated in mice bearing A431 intradermal xenografts. Compared to the controls, topical P-S hydrogel inhibited the A431 xenografts by 70.5% (p<0.01), while oral P-S inhibited it by 43.4% (p<0.05), being significantly less effective than topical P-S (p=0.017). Topical P-S hydrogel generated significant levels (>500 nmol/g tumor tissue) of intact P-S in the tumors, accounting for 92.5% of the total metabolites in the A431 xenografts. This local delivery of high levels of intact P-S to the A431 xenografts is an important contributor to the potent activity of topical P-S and no local or systemic side effects were noted in the treatment group. Thus, topical P-S is a promising treatment modality against non-melanoma skin cancer and merits further evaluation.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Compuestos Organofosforados/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Sulindac/análogos & derivados , Administración Tópica , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Neoplasias Cutáneas/patología , Sulindac/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Pharm Sci ; 98(7): 2390-400, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19090562

RESUMEN

The aim of this work was to assess the ability of aqueous suspensions of surface-modified nanostructured aluminosilicate (NSAS) compounds to reduce the intestinal absorption of cholesterol in a rat model. The rats were divided into 10 treatment groups which included several NSAS compounds at various doses, ezetimibe at 10 mg/kg, stigmastanol at 50 mg/kg, and normal saline. All compounds and controls were independently administered by oral gavage and then a mixture of [(3)H]cholesterol and cold cholesterol in 10% Intralipid(R) was immediately administered orally to the animals. Systemic blood was sampled and the concentration of cholesterol in plasma was determined by means of radioactivity. Protonation of NSAS using an ion-exchange column resulted in significant inhibition of cholesterol absorption relative to the control group (31.5% and 38.6% reduction in absorption of cholesterol for 50 and 100 mg/kg doses, respectively). Other surface-ion modifications of NSAS compounds did not show significant effect on intestinal cholesterol absorption. The inhibition of cholesterol absorption by ezetimibe was superior and by stigmastanol was equal to the effect of protonated NSAS in the doses investigated in this study. In conclusion, protonated NSAS material seems to inhibit significantly the intestinal absorption of dietary cholesterol in a rat model.


Asunto(s)
Silicatos de Aluminio/química , Silicatos de Aluminio/farmacología , Colesterol/metabolismo , Absorción Intestinal/efectos de los fármacos , Animales , Anticolesterolemiantes/farmacología , Azetidinas/farmacología , Colesterol/sangre , Ezetimiba , Masculino , Nanoestructuras/química , Tamaño de la Partícula , Protones , Ratas , Ratas Sprague-Dawley , Sitoesteroles/farmacología , Propiedades de Superficie , Viscosidad
16.
Pharm Res ; 23(2): 243-55, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16421666

RESUMEN

There has been increasing interest in recent years in the drug delivery applications of tocols and their derivatives. Their biocompatibility and potential to deliver both poorly soluble and water-soluble drugs make tocols attractive as drug delivery vehicles. This review article will focus primarily on topical, oral, and parenteral drug administration using tocols, although other routes of delivery such as pulmonary and nasal will also be discussed. After an overview of the tocol structures, physicochemical properties with emphasis on their solvent properties, functions, and metabolism, specific case studies will be discussed where tocols have been successfully used in topical, oral, and parenteral drug formulations and marketed drug products. Case studies will be extended to those where tocol-based formulations were administered pulmonarily and nasally. As more clinical data and marketed drug products emerge, the utility and therapeutic value of tocols will certainly increase.


Asunto(s)
Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Tocoferoles , Animales , Química Farmacéutica , Humanos , Solventes , Tocoferoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA