Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Acta Pharmacol Sin ; 42(11): 1913-1920, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34561552

RESUMEN

Sepsis is a dysregulated immune response to infection and potentially leads to life-threatening organ dysfunction, which is often seen in serious Covid-19 patients. Disulfiram (DSF), an old drug that has been used to treat alcohol addiction for decades, has recently been identified as a potent inhibitor of the gasdermin D (GSDMD)-induced pore formation that causes pyroptosis and inflammatory cytokine release. Therefore, DSF represents a promising therapeutic for the treatment of inflammatory disorders. Lactoferrin (LF) is a multifunctional glycoprotein with potent antibacterial and anti-inflammatory activities that acts by neutralizing circulating endotoxins and activating cellular responses. In addition, LF has been well exploited as a drug nanocarrier and targeting ligands. In this study, we developed a DSF-LF nanoparticulate system (DSF-LF NP) for combining the immunosuppressive activities of both DSF and LF. DSF-LF NPs could effectively block pyroptosis and inflammatory cytokine release from macrophages. Treatment with DSF-LF NPs showed remarkable therapeutic effects on lipopolysaccharide (LPS)-induced sepsis. In addition, this therapeutic strategy was also applied to treat ulcerative colitis (UC), and substantial treatment efficacy was achieved in a murine colitis model. The underlying mode of action of these DSF-LF-NPs may contribute to efficiently suppressing macrophage-mediated inflammatory responses and ameliorating the complications caused by sepsis and UC. As macrophage pyroptosis plays a pivotal role in inflammation, this safe and effective biomimetic nanomedicine may offer a versatile therapeutic strategy for treating various inflammatory diseases by repurposing DSF.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Colitis Ulcerosa , Disulfiram/farmacocinética , Lactoferrina , Síndrome de Respuesta Inflamatoria Sistémica , Inhibidores del Acetaldehído Deshidrogenasa/farmacología , Animales , Antiinflamatorios/farmacología , Materiales Biomiméticos/farmacología , COVID-19/inmunología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Modelos Animales de Enfermedad , Disulfiram/farmacología , Portadores de Fármacos/farmacología , Humanos , Inmunosupresores/farmacología , Lactoferrina/metabolismo , Lactoferrina/farmacología , Lipopolisacáridos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Nanopartículas/uso terapéutico , Piroptosis/efectos de los fármacos , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Resultado del Tratamiento
2.
Carbohydr Polym ; 261: 117846, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33766342

RESUMEN

In the clinical treatment of cancer, improving the effectiveness and targeting of drugs has always been a bottleneck problem that needs to be solved. In this contribution, inspired by the targeted inhibition on cancer from combination application of disulfiram and divalent copper ion (Cu2+), we optimized the concentration of disulfiram and Cu2+ ion for inhibiting esophageal cancer cells, and loaded them in hyaluronic acid (HA)/polyethyleneimine (PEI) nanoparticles with specific scales, in order to improve the effectiveness and targeting of drugs. The in vitro cell experiments demonstrated that more drug loaded HA/PEI nanoparticles accumulated to the esophageal squamous cell carcinoma (Eca109) and promoted higher apoptosis ratio of Eca109. Both in vitro and in vivo biological assessment verified that the disulfiram/Cu2+ loaded HA/PEI nanoparticles promoted the apoptosis of cancer cells and inhibited the tumor proliferation, but had no toxicity on other normal organs.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Cobre/administración & dosificación , Disulfiram/administración & dosificación , Neoplasias Esofágicas/tratamiento farmacológico , Ácido Hialurónico/química , Nanopartículas/química , Polietileneimina/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Células Cultivadas , Cobre/farmacocinética , Disulfiram/farmacocinética , Portadores de Fármacos/síntesis química , Portadores de Fármacos/química , Portadores de Fármacos/uso terapéutico , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Iones Pesados , Humanos , Ácido Hialurónico/síntesis química , Ácido Hialurónico/uso terapéutico , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/uso terapéutico , Polietileneimina/síntesis química , Polietileneimina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Mol Pharm ; 17(10): 3857-3869, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32833457

RESUMEN

Disulfiram (DSF) is an FDA-approved anti-alcoholic drug that has recently proven to be effective in cancer treatment. However, the short half-life in the bloodstream and the metal ion-dependent antitumor activity significantly limited the further application of DSF in the clinical field. To this end, we constructed a silk fibroin modified disulfiram/zinc oxide nanocomposites (SF/DSF@ZnO) to solubilize and stabilize DSF, and, more importantly, achieve pH triggered Zn2+ release and subsequent synergistic antitumor activity. The prepared SF/DSF@ZnO nanocomposites were spherical and had a high drug loading. Triggered by the lysosomal pH, SF/DSF@ZnO could induce the rapid release of Zn2+ under the acidic conditions and caused nanoparticulate disassembly along with DSF release. In vitro experiments showed that cytotoxicity of DSF could be enhanced by the presence of Zn2+, and further amplified when encapsulated into SF/DSF@ZnO nanocomposites. It was confirmed that the significantly amplified cytotoxicity of SF/DSF@ZnO was resulted from pH-triggered Zn2+ release, inhibited cell migration, and increased ROS production. In vivo study showed that SF/DSF@ZnO nanocomposites significantly increased the tumor accumulation and prolonged the retention time. In vivo antitumor experiments in the xenograft model showed that SF/DSF@ZnO exerted the highest tumor-inhibition rate among all the drug treatments. Therefore, this exquisite study established silk fibroin-modified disulfiram/zinc oxide nanocomposites, SF/DSF@ZnO, where ZnO not only acted as a delivery carrier but also served as a metal ion reservoir to achieve synergistic antitumor efficacy. The established DSF nanoformulation displayed excellent therapeutic potential in future cancer treatment.


Asunto(s)
Antineoplásicos/farmacocinética , Nanocompuestos/administración & dosificación , Neoplasias/tratamiento farmacológico , Zinc/farmacocinética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Bombyx/química , Cationes Bivalentes/farmacocinética , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Disulfiram/administración & dosificación , Disulfiram/química , Disulfiram/farmacocinética , Composición de Medicamentos/métodos , Liberación de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Fibroínas/química , Semivida , Humanos , Concentración de Iones de Hidrógeno , Ratones , Neoplasias/patología , Óxido de Zinc/administración & dosificación , Óxido de Zinc/química , Óxido de Zinc/farmacocinética
4.
Biomater Sci ; 8(12): 3310-3319, 2020 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-32400782

RESUMEN

Herein, we successfully constructed a combination therapeutic nanoplatform with high tumor targeting for cancer treatment by integrating gold nanorods with disulfiram (denoted Au-DSF). The Au-DSF integrates possess a uniform length (70 nm), excellent photothermal conversion ability and a high DSF loading content (23.2%), and the loaded DSFs show glutathione-, acid-, and laser-responsive release properties. The Au-DSF integrates show significantly enhanced cellular uptake efficiency in breast cancer cells due to the ability of DSF to chelate to the intracellular copper (Cu) which is present at high concentrations. Furthermore, the Au-DSF exhibits improved circulation time (mean residence time = 28.4 h) and increased tumor accumulation (12.0%), due to the targeting of DSF to the abundant Cu ions at the tumor site. Moreover, the DSF/Cu complexes potently elevate reactive oxygen species, which effectively induce cancer cell apoptosis. In vivo experiments show that the Au-DSF integrates dramatically decrease tumor size via photothermal therapy and chemotherapy. Hematoxylin-eosin and TUNEL staining show that the Au-DSF integrates induce necrosis and apoptosis in cancer cells. The high therapeutic efficiency of the Au-DSF integrates for breast cancer is further demonstrated by the reduced elasticity seen in ultrasound elastography, and the absence of perfusion of the contrast agent in contrast-enhanced ultrasound imaging in tumors.


Asunto(s)
Antineoplásicos/administración & dosificación , Disulfiram/administración & dosificación , Oro/administración & dosificación , Neoplasias Mamarias Experimentales/terapia , Nanotubos , Animales , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Disulfiram/farmacocinética , Femenino , Oro/química , Oro/farmacocinética , Humanos , Rayos Láser , Células MCF-7 , Neoplasias Mamarias Experimentales/metabolismo , Ratones Desnudos , Nanotubos/química , Terapia Fototérmica
5.
Daru ; 27(2): 853-862, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31758497

RESUMEN

BACKGROUND: Disulfiram (DSF) has a long history of being used as a first-line promising therapy for treatment of alcoholism in human. Besides its prominence in the treatment of alcoholism, extensive investigations have been carried out to explore other biomedical and pharmacological effects of DSF. Amongst other biomedical implications, plenty researches have shown evidence of promising anticancer efficacy of this agent for treatment of wide range of cancers such as breast cancer, liver cancer and lung carcinoma. METHODS: Electronic databases, including Google scholar, PubMed and Web of science were searched with the keywords disulfiram, nanoparticles, cancer, drug delivery systems. RESULT: Despite its excellent anticancer efficacy, the pharmaceutical significance and clinical applicability of DSF are hampered due to poor stability, low solubility, short plasma half-life, rapid metabolism, and early clearance from systemic circulation. Various attempts have been made to eradicate these issues. Nanotechnology based interventions have gained remarkable recognition in improving pharmacokinetic and pharmacodynamic profile of DSF by improving its stability and avoiding its degradation. CONCLUSION: The aim of the present review is to critically analyse all recent developments in designing various nanotechnology-based delivery systems, to ponder their relevance in improving stability, pharmacokinetic and pharmacodynamic profile, and achieving target-specific delivery of this agent to cancer cells to effectively eradicate cancer and abolish its metastasis. Nanotechnology is a novel approach for overcoming such obstacles faced presently, the results obtained so far using different novel drug delivery systems seem to be very promising to increase the stability and half-life of DSF. Graphical abstract Nanocrrier mediated drug delivery systems for disulfiram.


Asunto(s)
Antineoplásicos/administración & dosificación , Disulfiram/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disulfiram/química , Disulfiram/farmacocinética , Portadores de Fármacos , Semivida , Humanos , Nanopartículas , Solubilidad
6.
Int J Pharm ; 557: 304-313, 2019 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-30599232

RESUMEN

Multidrug resistance (MDR) is a common intractable barrier in success of clinical cancer chemotherapy. Codelivery of two drugs using nanocarriers is a commonly used approach to treat the MDR cancer. However, the drug payload in the conventional nanocarriers is low and thus compromises the treatment outcomes. Disulfiram (DSF) is promising to reverse MDR and increases the sensitivity of cancer cells to chemotherapy. While, paclitaxel (PTX) is one of the frequently used anticancer drug. Here, by using a drug-delivering-drug (DDD) strategy based on nanocrystals, hybrid PTX-DSF nanocrystals (PTX-DSF Ns) were developed for codelivery of PTX and DSF to reverse MDR in cancer. The 160-nm PTX-DSF Ns with rod-like morphology had drug-loading up to 43% at mass ratio of 5:1. Interestingly, the nanoparticles entered cells via caveolar endocytosis. By reducing intracellular ATP level and GST activity, PTX-DSF Ns killed the Taxol resistant A549 cells with higher efficiency than PTX alone, exhibiting as 6-fold increase of apoptosis in MDR tumor. The nanoparticles circulated in blood over time, accumulated in tumor efficiently and reduced the tumor volume by 12-fold in MDR tumor-bearing BALB/c nude mice and allowed 12-fold apoptosis in tumor. Additionally, the immunohistochemical examination demonstrated the safety of the nanoparticles. Overall, the DDD strategy-based PTX-DSF Ns have promising potential for the treatment of MDR cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Disulfiram/administración & dosificación , Portadores de Fármacos/administración & dosificación , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Nanopartículas/administración & dosificación , Paclitaxel/administración & dosificación , Células A549 , Animales , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Transporte Biológico , Disulfiram/farmacocinética , Portadores de Fármacos/farmacocinética , Combinación de Medicamentos , Eritrocitos/efectos de los fármacos , Femenino , Hemólisis/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Paclitaxel/farmacocinética , Ratas Sprague-Dawley , Distribución Tisular , Carga Tumoral/efectos de los fármacos
7.
ACS Appl Mater Interfaces ; 10(48): 41118-41128, 2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30444340

RESUMEN

Disulfiram (DSF), an alcohol-aversion drug, has been explored for cancer treatment. Copper diethyldithiocarbamate (Cu(DDC)2) complex formed by DSF and copper ions is a major active ingredient for its anticancer activity. Direct administration of Cu(DDC)2 is a promising strategy to enhance the anticancer efficacy of DSF. However, efficient drug delivery remains a significant challenge for Cu(DDC)2 and hinders its clinical use. In this study, we developed a facile stabilized metal ion ligand complex (SMILE) method to prepare Cu(DDC)2 nanoparticles (NPs). The SMILE method could prepare Cu(DDC)2 NPs with different types of stabilizers including 1,2-distearoyl- sn-glycerol-3-phosphoethanolamine-poly(ethylene glycol) (PEG) 2000, d-α-tocopherol PEG 1000 succinate, methoxy PEG 5000- b-poly(l-lactide) 5000, and other generally recognized as safe excipients approved by the US Food and Drug Administration. The optimized formulations demonstrated excellent drug-loading efficiency (close to 100%), high drug concentrations (increased drug concentration by over 200-fold compared to the traditional micelle formulation), and an optimal particle size in the sub-100 nm range. Cu(DDC)2 NPs exhibited outstanding stability in serum for 72 h and can also be stored at room temperature for at least 1 month. The anticancer effects of Cu(DDC)2 NP formulations were determined by multiple assays including 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, colony-forming assay, calcein-AM/propidium iodide staining, and others. Cu(DDC)2 NPs showed excellent activity against drug-resistant prostate cancer cells and other cancer cells with a half-maximal inhibitory concentration (IC50) of around 100 nM. Our study also demonstrated that Cu(DDC)2 NPs induced cell death in drug-resistant prostate cancer cells (DU145-TXR) through paraptosis, which is a nonapoptotic cell death. To our best knowledge, the SMILE method provides, for the first time, a simple yet efficient process for generating Cu(DDC)2 NPs with high drug concentration, excellent loading efficiency, and desirable physicochemical properties. This method could potentially address drug delivery challenges of DSF/copper-based chemotherapy and facilitate its clinical translation.


Asunto(s)
Complejos de Coordinación , Cobre , Disulfiram , Ditiocarba , Resistencia a Antineoplásicos/efectos de los fármacos , Nanopartículas , Neoplasias de la Próstata , Complejos de Coordinación/química , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/farmacología , Cobre/química , Cobre/farmacocinética , Cobre/farmacología , Disulfiram/química , Disulfiram/farmacocinética , Disulfiram/farmacología , Ditiocarba/química , Ditiocarba/farmacocinética , Ditiocarba/farmacología , Humanos , Células MCF-7 , Masculino , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
8.
Colloids Surf B Biointerfaces ; 160: 305-314, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28950195

RESUMEN

The purpose of the study was to design a novel octa-arginine (R8) modified lipid emulsion (LE) system for the ocular delivery of the lipophilic drug disulfiram (DSF). The influence of the particle size of the lipid emulsions and the presence of R8 on corneal permeation was studied. DSF-loaded lipid emulsions with different particle sizes (DSF-LE1, DSF-LE2, DSF-LE3) and DSF-loaded lipid emulsions modified with R8 (DSF-LE1-R8 and DSF-LE2-R8) were prepared. The Zeta potential of the lipid emulsions was changed from negative to a positive value after modification of R8. The mucoadhesion of different preparations was investigated, and DSF-LE1-R8 was found to produce the strongest mucoadhesion. The in vitro corneal penetration study and in vivo ocular distribution study showed that the R8 modified lipid emulsion (DSF-LE1-R8) with a nano particle size, exhibited the highest permeability and the largest amount of DDC distributed in ocular issues. Coumarin-6 labelled LE1-R8 displayed more homogeneous fluorescence with the deeper penetration into the cornea compared with other preparations at various times. Confocal laser scanning microscopy showed that, in addition to paracellular routes, LE-R8 could also transport across the corneal epithelium by transcellular routes as a result of increased uptake due to the R8 modification. Furthermore, the anti-cataract effect was evaluated and it was found that DSF-LE1-R8 exhibited a marked anti-cataract effect. Therefore, the lipid emulsions with nano-sized particles and modification of R8 were proposed as a potential ocular delivery system to improve the corneal penetration and ocular delivery of DSF.


Asunto(s)
Catarata/tratamiento farmacológico , Córnea/efectos de los fármacos , Disulfiram/farmacocinética , Sistemas de Liberación de Medicamentos , Soluciones Oftálmicas/farmacocinética , Péptidos/química , Animales , Catarata/inducido químicamente , Catarata/metabolismo , Catarata/patología , Córnea/metabolismo , Córnea/patología , Cumarinas/química , Disulfiram/metabolismo , Disulfiram/farmacología , Emulsiones , Colorantes Fluorescentes/química , Masculino , Soluciones Oftálmicas/síntesis química , Soluciones Oftálmicas/farmacología , Tamaño de la Partícula , Permeabilidad , Conejos , Ratas , Ratas Sprague-Dawley , Selenito de Sodio , Electricidad Estática
9.
J Neurooncol ; 128(2): 259-66, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26966095

RESUMEN

Disulfiram, a generic alcohol aversion drug, has promising preclinical activity against glioblastoma (GBM). This phase I study aims to evaluate its safety, maximum tolerated dose (MTD), pharmacodynamic effect, and preliminary efficacy when combined with adjuvant temozolomide in GBM patients after standard chemoradiotherapy. Patients received disulfiram 500-1000 mg once daily, in combination with 150-200 mg/m(2) temozolomide. A modified 3 + 3 dose-escalation design was used to determine the MTD. The pharmacodynamic effect of proteasome inhibition was assessed using fluorometric 20S proteasome assay on peripheral blood cells. The MTD was determined based on the dose-limiting toxicities (DLTs) within the first month of therapy. Twelve patients were enrolled to two dose levels: 500 and 1000 mg. Two DLTs of grade 3 delirium occurred after 15 days of administration at 1000 mg per day. Other possible grade 2-3 DSF-related toxicities included fatigue, ataxia, dizziness, and peripheral neuropathy. The toxicities were self-limiting or resolved after discontinuing DSF. The MTD was determined to be 500 mg per day. Limited proteasome inhibition was observed at week 4 and showed an increased trend with escalated disulfiram. Median progression-free survival with 500 mg of DSF was 5.4 months from the start of disulfiram and 8.1 months from the start of chemoradiotherapy. Disulfiram can be safely combined with temozolomide but can cause reversible neurological toxicities. The MTD of disulfiram with adjuvant temozolomide appears to produce limited proteasome inhibition on peripheral blood cells.


Asunto(s)
Antineoplásicos/uso terapéutico , Dacarbazina/análogos & derivados , Disulfiram/uso terapéutico , Glioblastoma/terapia , Neoplasias Supratentoriales/terapia , Administración Oral , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Quimioradioterapia , Dacarbazina/uso terapéutico , Disulfiram/efectos adversos , Disulfiram/farmacocinética , Relación Dosis-Respuesta a Droga , Reposicionamiento de Medicamentos , Quimioterapia Combinada , Femenino , Glioblastoma/sangre , Humanos , Masculino , Persona de Mediana Edad , Complejo de la Endopetidasa Proteasomal/sangre , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/efectos adversos , Inhibidores de Proteasoma/farmacocinética , Inhibidores de Proteasoma/uso terapéutico , Neoplasias Supratentoriales/sangre , Temozolomida , Resultado del Tratamiento , Adulto Joven
10.
ACS Appl Mater Interfaces ; 7(45): 25147-61, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26501354

RESUMEN

Disulfiram, which exhibits marked tumor inhibition mediated by copper, was encapsulated in lipid nanocapsules modified with TAT peptide (TATp) and pH-triggered sheddable PEG to target cancer cells on the basis of tumor environmental specificity. PEG-shedding lipid nanocapsules (S-LNCs) were fabricated from LNCs by decorating short PEG chains with TATp (HS-PEG(1k)-TATp) to form TATp-LNCs and then covered by pH-sensitive graft copolymers of long PEG chains (PGA-g-PEG(2k)). The DSF-S-LNCs had sizes in the range of 60-90 nm and were stable in the presence of 50% plasma. DSF-S-LNCs exhibited higher intracellular uptake and antitumor activity at pH 6.5 than at pH 7.4. The preincubation of Cu showed that the DSF cytotoxicity was based on the accumulation of Cu in Hep G2 cells. Pharmacokinetic studies showed the markedly improved pharmacokinetic profiles of DSF-S-LNCs (AUC= 3921.391 µg/L·h, t(1/2z) = 1.294 h) compared with free DSF (AUC = 907.724 µg/L·h, t(1/2z) = 0.252 h). The in vivo distribution of S-LNCs was investigated using Cy5.5 as a fluorescent probe. In tumor-bearing mice, the delivery efficiency of S-LNCs was found to be 496.5% higher than that of free Cy5.5 and 74.5% higher than that of LNCs in tumors. In conclusion, DSF-S-LNCs increased both the stability and tumor internalization and further increased the cytotoxicity because of the higher copper content.


Asunto(s)
Cobre/química , Disulfiram/uso terapéutico , Lípidos/química , Nanocápsulas/química , Neoplasias/tratamiento farmacológico , Polietilenglicoles/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Disulfiram/farmacocinética , Disulfiram/farmacología , Liberación de Fármacos , Endocitosis/efectos de los fármacos , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Concentración 50 Inhibidora , Espectrometría de Masas , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Polietilenglicoles/síntesis química , Polisorbatos/química , Espectroscopía de Protones por Resonancia Magnética , Espectroscopía Infrarroja Corta , Electricidad Estática , Distribución Tisular/efectos de los fármacos
11.
Artículo en Inglés | MEDLINE | ID: mdl-24013191

RESUMEN

Disulfiram (DSF) has been used to treat alcoholism for many years and it has been suggested to play a key role in combatting many kinds of tumors. However, disulfiram has complex pharmacokinetics and is rapidly eliminated which limits its use as a tumor treatment. Therefore, a rapid and sensitive analytical method based on ultra performance liquid chromatography coupled to electrospray ionization-tandem mass spectrometry (UPLC-ESI-MS/MS) was developed and validated for the determination of disulfiram in rat plasma. Blood samples were pre-stabilized with a stabilizing agent and then plasma was obtained and subjected to solid phase extraction (SPE), and chromatographed on a Phenomenex Kinetex(®) XB C18 column with gradient elution using a mobile phase consisting of acetonitrile-water (containing 0.1% formic acid and 1mM ammonium acetate) at a flow rate of 0.2mL/min for 3min. Multiple reactions monitoring in positive mode was carried out with disulfiram at 296.95/115.94 and diphenhydramine (internal standard, IS) at 256.14/167.02 over a linear range from 0.6 to 1200ng/mL. The extraction recovery of disulfiram for different concentrations ranged from 75.7% to 78.3%. The intra- and inter-day precision was less than 8.93% and 12.39%, respectively, and the accuracy was within ±7.75%. The validated method was successfully applied to a pharmacokinetic study of disulfiram in rat plasma after oral administration of a dose of 180mg/kg.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Disulfiram/sangre , Espectrometría de Masa por Ionización de Electrospray/métodos , Animales , Disulfiram/química , Disulfiram/farmacocinética , Estabilidad de Medicamentos , Modelos Lineales , Masculino , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Prostate Cancer Prostatic Dis ; 16(4): 357-61, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23958896

RESUMEN

BACKGROUND: Preclinical drug screens identified disulfiram as a potent in vitro inhibitor of prostate cancer (PCa) cell growth. Although many mechanisms for its anticancer activity have been proposed, tumor suppressor gene re-expression through promoter demethylation emerged as one of the more plausible. METHODS: We conducted an open-label, dose escalation trial of disulfiram in men with non-metastatic recurrent PCa after local therapy. Dose escalation occurred if a demethylating 'response' (that is, 10% decrease in peripheral blood mononuclear cell (PBMC) global 5-methyl cytosine (5(me)C) content) was observed in <3 patients in cohort 1. Cohorts 1 and 2 received disulfiram 250 mg and 500 mg daily, respectively. The primary end point was the proportion of subjects with a demethylation response. Secondary end points included the rate of PSA progression at 6 months, changes in PSA doubling time and safety/tolerability. RESULTS: Changes in global 5(me)C content were observed in two of nine patients (22.2%) in cohort 1 and 3 of 10 (30.0%) in cohort 2. Only five subjects were on trial for 6 months, all were in cohort 1 and all had PSA progression by 6 months. No changes in PSA kinetics were observed in either cohort. Disulfiram was poorly tolerated with six patients experiencing grade 3 adverse events (three per cohort). Three of the responders displayed pretreatment instability in their 5(me)C content. CONCLUSIONS: A minority of patients had transient global PBMC demethylation changes. Instability in 5(me)C may limit the reproducibility of these findings, limiting our ability to confirm our hypothesis. Given the toxicities and no clinical benefits, further development of disulfiram should not be pursued in this population.


Asunto(s)
Antineoplásicos/farmacocinética , Disulfiram/farmacocinética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Anciano , Antineoplásicos/efectos adversos , Ceruloplasmina/metabolismo , Metilación de ADN/efectos de los fármacos , Disulfiram/efectos adversos , Epigénesis Genética/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/genética
14.
J Pharm Biomed Anal ; 54(4): 799-806, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21145687

RESUMEN

Liquid chromatography-tandem mass spectrometry methodology is described for the determination of S-(N,N-diethylcarbamoyl)glutathione (carbamathione) in human plasma samples. Sample preparation consisted of a straightforward perchloric acid medicated protein precipitation, with the resulting supernatant containing the carbamathione (recovery ~98%). For optimized chromatography/mass spec detection a carbamathione analog, S-(N,N-di-i-propylcarbamoyl)glutathione, was synthesized and used as the internal standard. Carbamathione was found to be stable over the pH 1-8 region over the timeframe necessary for the various operations of the analytical method. Separation was accomplished via reversed-phase gradient elution chromatography with analyte elution and re-equilibration accomplished within 8 min. Calibration was established and validated over the concentration range of 0.5-50 nM, which is adequate to support clinical investigations. Intra- and inter-day accuracy and precision determined and found to be <4% and <10%, respectively. The methodology was utilized to demonstrate the carbamathione plasma-time profile of a human volunteer dosed with disulfiram (250 mg/d). Interestingly, an unknown but apparently related metabolite was observed with each human plasma sample analyzed.


Asunto(s)
Glutatión/análogos & derivados , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Disuasivos de Alcohol/farmacocinética , Cromatografía Líquida de Alta Presión , Disulfiram/farmacocinética , Femenino , Glutatión/análisis , Glutatión/sangre , Glutatión/química , Humanos , Concentración de Iones de Hidrógeno , Límite de Detección , Fase II de la Desintoxicación Metabólica , Persona de Mediana Edad , Profármacos/farmacocinética , Reproducibilidad de los Resultados , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
15.
Biol Pharm Bull ; 31(7): 1444-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18591790

RESUMEN

We previously prepared 2-hydroxypropyl-beta-cyclodextrin (HPbetaCD) solutions containing disulfiram (DSF) and hydroxypropylmethylcellulose (HPMC, DSF solutions), and found the instillation of this DSF solutions delayed lens opacification in ICR/f rats, a recessive-type hereditary cataractous strain. In this study, we determined the corneal penetration mechanism of DSF solutions using human cornea epithelial cell monolayers based on the immortalized human cornea epithelial cell line (HCE-T) developed by Tropainen et al. [Invest. Ophthalmol. Vis. Sci., 42, 2942-2948 (2001)]. The transepithelial electrical resistance (TER) values of HCE-T cells increases from approximately 275 to 388 Omega.cm(2) by exposure to an air-liquid interface for 2 weeks. The penetration of DSF into the basolateral chamber was prevented by the increase in TER values. The DSF in solution was converted to diethyldithiocarbamate (DDC) during the penetration experiment using HCE-T cell monolayers, and a close relationship between the penetration coefficient of DDC and aldehyde dehydrogenase (ALDH) 3A1 mRNA expression (y=41.202x+18.587, R=0.9413) was observed. In addition, an anti-ALDH3A1 antibody significantly inhibited the DSF-DDC conversion. These results suggest that DSF in DSF solutions is converted to DDC via catalysis by an ALDH3A1 in the cornea, and then transited from the apical side to the basolateral side.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Córnea/metabolismo , Disulfiram/farmacocinética , Células Epiteliales/metabolismo , Aldehído Deshidrogenasa/antagonistas & inhibidores , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Células Cultivadas , Córnea/citología , Ditiocarba/metabolismo , Células Epiteliales/enzimología , Humanos , ARN/biosíntesis , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Absorción Cutánea
16.
Pharmazie ; 62(10): 767-72, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18236782

RESUMEN

Immortalized human corneal epithelial cells (HCECs) and human lens epithelial cells (HLECs) were cultured in vitro. Cells were observed under a phase-contrast microscope and the integrity of cell monolayers was assayed by transepithelial electrical resistance (TEER) determination. The permeability of disulfiram (DSF) through a HCECs monolayer was compared with that of DSF through an excised rabbit cornea. The permeability coefficients of DSF through a HCECs monolayer and excised rabbit cornea were 29.5 +/- 4.8 x 10(-6) cm/s and 34.7 +/- 5.2 x 10(-6) cm/s, respectively. Diethyldithiocarbamate (DDC) had high permeability through HLECs monolayer with a permeability coefficient of 44.6 +/- 7.1 x 10(-6) cm/s. The cytotoxicity of DDC against HLECs was investigated using the trypan blue exclusion test. For a DDC concentration of 5 mmol/l, more than 85% cells were viable. DH3a1 mRNA was expressed in cultured HLECs. The expression of aldehyde dehydrogenase 3a1 (ALDH3a1), which may be be responsible for DSF-DDC conversion, was detected using RT-PCR and agarose gels electrophoresis. These results demonstrate that the permeability of DSF can be detected and intra-ocular drug action may be predicted using the cultured HCEC and HLEC monolayers as model.


Asunto(s)
Catarata/tratamiento farmacológico , Catarata/metabolismo , Aldehído Deshidrogenasa/genética , Animales , Línea Celular , Permeabilidad de la Membrana Celular , Colágeno Tipo IV/metabolismo , Colorantes , Córnea/metabolismo , Disulfiram/farmacocinética , Ditiocarba/farmacocinética , Conductividad Eléctrica , Células Epiteliales/metabolismo , Epitelio Corneal/metabolismo , Humanos , Técnicas In Vitro , Cristalino/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Azul de Tripano
17.
Neurotoxicology ; 25(3): 365-75, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15019299

RESUMEN

3,4-Dihydroxyphenylacetaldehyde (DOPAL) is formed by the oxidative deamination of dopamine (DA) catalyzed by monoamine oxidases (MAO); then, the aldehyde is oxidized to 3,4-dihydroxyphenylacetic acid (DOPAC) by aldehyde dehydrogenases (ALDH) or reduced to 3,4-dihydroxyphenylethanol (DOPET) by aldose/aldehyde reductases. The present work aimed at evaluating the in vitro toxicity of DOPAL on catecholaminergic neuroblastoma SH-SY5Y cells which accumulate DA. DOPAL synthesis was stimulated by incubating cells with DA and blocking DOPAL oxidation by disulfiram, an irreversible inhibitor of ALDH. As evidenced by MTT reduction assays, DA and disulfiram treatments produced cell losses which increased with time. 10(-2)M DA reduced by 40% cell viability after a 1h treatment, when its TC(50) (concentration reducing viability by 50%) value was 7.3 x 10(-5) M after a 24 h treatment. For the same treatment periods, TC(50) values for disulfiram were 8 x 10(-5) and 8.7 x 10 (-7) M, respectively. MTT reduction assay performed after a 24h treatment followed by a 24h incubation in a drug-free medium evidenced that the toxicity of 10(-4)M DA or 10(-6)M disulfiram was potentiated by the second drug. HPLC measurements showed that DOPAL was produced at the early stages of the treatment by DA and disulfiram. This was evidenced by the significant increase in the ((DOPAL + DOPET)/DOPAC ratio observed after a combined 3h treatment by 10(-4)M DA and 10(-6)M disulfiram. Total contents in DA and DOPAL were greatly reduced at the end of a 15 h treatment, and disulfiram did not significantly enhanced the (DOPAL + DOPET)/DOPAC ratio. For both treatment durations, DOPAL and DOPET were detectable only in the extracellular medium. So, these results suggest that an early production of DOPAL could produce delayed toxic effects on SH-SY5Y cells. Production of DOPET and release of DOPAL could be important means for reducing DOPAL concentrations in dopaminergic neurons.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/análogos & derivados , Ácido 3,4-Dihidroxifenilacético/metabolismo , Disulfiram/toxicidad , Dopamina/toxicidad , Neuroblastoma/metabolismo , Neuroblastoma/patología , Catecolaminas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Disulfiram/farmacocinética , Dopamina/farmacocinética , Humanos
18.
Biol Pharm Bull ; 23(5): 616-20, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10823675

RESUMEN

Disulfiram, a dimer of diethyldithiocarbamate (DDC) which is a strong radical scavenger, is known to prevent cataract development. However, disulfiram is hardly absorbed from the cornea and its bioavailability is extremely low. In this study, we attempted to prepare disulfiram solid dispersion for the improvement of ocular bioavailability. Solid dispersions of disulfiram were prepared by either an evaporation method or a spray-drying method, using polyvinylpyrrolidone (PVP) as a carrier. Preparations were analyzed by scanning electron microscopy, powder X-ray diffractometry and differential scanning calorimetry, and confirmed to be a solid dispersion. The particle size of the solid dispersion prepared by the spray-drying method was smaller than the preparation by the evaporation method (spray-drying: 3.3+/-0.04 microm, evaporation: 34.3+/-18.0 microm). An in vivo ocular absorption experiment was conducted by instilling solid dispersions to rabbit eye and measuring the DDC in the aqueous humor. After instillation of disulfiram and PVP physical mixture, DDC was not detected in the aqueous humor. On the other hand, DDC appeared in the aqueous humor after the instillation of a solid dispersion. Maximal concentration and the area under the aqueous humor concentration-time curve were greater in the solid dispersion prepared by the spray-drying method than the preparation by the evaporation method. Disulfiram solid dispersion, especially prepared by the spray-drying method, improved ocular bioavailability.


Asunto(s)
Disulfiram/farmacocinética , Ojo/metabolismo , Absorción , Animales , Catarata/tratamiento farmacológico , Dimerización , Disulfiram/química , Disulfiram/metabolismo , Ditiocarba/química , Ditiocarba/metabolismo , Portadores de Fármacos , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Masculino , Povidona/administración & dosificación , Conejos
19.
Mol Carcinog ; 22(4): 235-46, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9726816

RESUMEN

Prolinedithiocarbamate (PDTC) and diethyldithiocarbamate (DDTC) are cancer chemopreventive agents and can be biotransformed to prolinethiuramdisulfide (PTDS) and tetraethylthiuramdisulfide (disulfiram; DTDS), respectively. We found that the reactive metabolites PTDS and DTDS induced apoptosis after G1/S arrest. Phosphorylation of cyclin E, inhibition of cyclin-dependent kinase 2 activity, and degradation of cyclin E were found in human hepatoma Hep G2 cells during apoptosis. Moreover, PTDS and DTDS decreased the level of bcl-2 but increased the level of p53. In contrast, PDTC, DDTC, and ammonium dithiocarbamate (ADTC) did not induce apoptosis; rather they led to the induction of p53 and p21 followed by G1/S arrest. PDTC, DDTC, and ADTC also arrested cells in G1 phase. We then examined the effects of PTDS and DTDS on the signal transduction mechanisms leading to apoptosis. Although the transcription factors NFkappaB and AP-1 cooperatively decreased their DNA-binding activities to kappaB and 12-O-tetradecanoylphorbol-13-acetate-responsive elements, respectively, and p53 increased DNA-binding activity in the early stage but decreased it in the latter stage after treatment with PTDS, when the human Hep G2 cells were undergoing apoptosis. In summary, our results indicated that (i) PTDS and DTDS induced apoptosis and G1/S arrest mediated by p53, whereas PDTC, DDTC, and ADTC induced p53-dependent p21 expression leading to G1/S arrest; (ii) PDTC, DDTC, and ADTC induced p21/KIP1/CIP1 expression in a p53-dependent pathway leading to G1/S arrest; and (iii) NFkappaB, AP-1, and bcl-2 were downregulated during PTDS- and DTDS-induced apoptosis. These results suggested that PTDS and DTDS induced p53-dependent apoptosis, whereas PDTC, DDTC, and ADTC induced G1/S arrest. Apoptosis is regulated by the modulation of intracellular effectors such as NFkappaB, AP-1, and bcl-2 and activation of p53 in early stages.


Asunto(s)
Disuasivos de Alcohol/farmacocinética , Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular , Ditiocarba/farmacología , FN-kappa B/fisiología , Prolina/análogos & derivados , Proteínas Proto-Oncogénicas c-fos/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Tiocarbamatos/farmacología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor , Disuasivos de Alcohol/farmacología , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Antioxidantes/metabolismo , Antioxidantes/farmacocinética , Apoptosis/fisiología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Disulfiram/farmacocinética , Disulfiram/farmacología , Ditiocarba/metabolismo , Ditiocarba/farmacocinética , Fase G1/efectos de los fármacos , Fase G1/fisiología , Humanos , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/fisiología , FN-kappa B/biosíntesis , Oxidación-Reducción , Prolina/metabolismo , Prolina/farmacocinética , Prolina/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-jun/biosíntesis , Tiocarbamatos/metabolismo , Tiocarbamatos/farmacocinética , Factor de Transcripción AP-1/biosíntesis , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis
20.
J Pharmacol Exp Ther ; 281(2): 611-7, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9152363

RESUMEN

Recent studies on the mechanism by which disulfiram inhibits aldehyde dehydrogenase have provided evidence for the formation of reactive intermediates that are thought to carbamoylate, and thereby inactivate the enzyme. In our study, rats were dosed with either disulfiram (0.25 mmol kg-1 i.p.) or its reduced metabolite diethyldithiocarbamate (DDTC; 0.5 mmol kg-1 i.p.) and urine was collected for the analysis of metabolites derived from putative reactive intermediates. By means of ionspray LC-MS/MS, two novel N-acetylcysteine (NAC) conjugates, i.e., N-acetyl-S-(N, N-diethylcarbamoyl)cysteine and N-acetyl-S-(N, N-diethylthiocarbamoyl)cysteine, were identified in urine specimens. Quantitative analyses indicated that, over the 0- to 24-hr period after drug administration, urinary excretion of N-acetyl-S-(N, N-diethylcarbamoyl)cysteine accounted for 7.5 +/- 4.0 and 6.2 +/- 1.0%, respectively, of the dose of disulfiram and diethyldithiocarbamate, while the corresponding thiocarbamoyl conjugate, N-acetyl-S-(N, N-diethylthiocarbamoyl)cysteine, accounted for a further 0.5 +/- 0.3 and 0.3 +/- 0.1%, respectively, of the dose. These conjugates are believed to derive from reactive sulfoxide and sulfone metabolites of disulfiram, namely S-methyl-N, N-diethylthiocarbamate sulfoxide (DETC-MeSO), S-methyl-N, N-diethylthiocarbamate sulfone (DETC-MeSO2), S-methyl-N, N-diethyldithiocarbamate sulfoxide (DDTC-MeSO) and S-methyl-N, N-diethyldithiocarbamate sulfone (DDTC-MeSO2), which were found to carbamoylate N-acetylcysteine in vitro with the following rank order of reactivity: DDTC-MeSO2 > DETC-MeSO2 > DDTC-MeSO > DETC-MeSO. In vitro experiments with aldehyde dehydrogenase showed that all four S-oxygenated metabolites inhibited the enzyme effectively. Furthermore, inclusion of NAC in incubation media attenuated significantly the inhibition by DDTC-MeSO2, DETC-MeSO2 and DDTC-MeSO, but had little effect on that by DETC-MeSO. Our results are consistent with the hypothesis that disulfiram and diethyldithiocarbamate undergo activation by a sequence of metabolic reactions leading to the formation of electrophilic S-methyl sulfoxides and sulfones that carbamoylate, and thereby inhibit, aldehyde dehydrogenase and possibly other enzymes.


Asunto(s)
Acetilcisteína/orina , Disuasivos de Alcohol/farmacocinética , Aldehído Deshidrogenasa/antagonistas & inhibidores , Disulfiram/farmacocinética , Acetilcisteína/química , Animales , Biotransformación , Cromatografía Liquida/métodos , Masculino , Espectrometría de Masas/métodos , Oxígeno/química , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA