Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 16(34): 44518-44527, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39145481

RESUMEN

Glioblastoma (GBM) is the most common and aggressive malignant brain tumor. Standard therapy includes maximal surgical resection, radiotherapy, and adjuvant temozolomide (TMZ) administration. However, the rapid development of TMZ resistance and the impermeability of the blood-brain barrier (BBB) significantly hinder the therapeutic efficacy. Herein, we developed spatiotemporally controlled microneedle patches (BMNs) loaded with TMZ and niclosamide (NIC) to overcome GBM resistance. We found that hyaluronic acid (HA) increased the viscosity of bovine serum albumin (BSA) and evidenced that concentrations of BSA/HA exert an impact degradation rates exposure to high-temperature treatment, showing that the higher BSA/HA concentrations result in slower drug release. To optimize drug release rates and ensure synergistic antitumor effects, a 15% BSA/HA solution constituting the bottoms of BMNs was chosen to load TMZ, showing sustained drug release for over 28 days, guaranteeing long-term DNA damage in TMZ-resistant cells (U251-TR). Needle tips made from 10% BSA/HA solution loaded with NIC released the drug within 14 days, enhancing TMZ's efficacy by inhibiting the activity of O6-methylguanine-DNA methyltransferase (MGMT). BMNs exhibit superior mechanical properties, bypass the BBB, and gradually release the drug into the tumor periphery, thus significantly inhibiting tumor proliferation and expanding median survival in mice. The on-demand delivery of BMNs patches shows a strong translational potential for clinical applications, particularly in synergistic GBM treatment.


Asunto(s)
Glioblastoma , Ácido Hialurónico , Niclosamida , Albúmina Sérica Bovina , Temozolomida , Temozolomida/química , Temozolomida/farmacología , Temozolomida/farmacocinética , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/metabolismo , Animales , Humanos , Ratones , Niclosamida/farmacología , Niclosamida/química , Niclosamida/farmacocinética , Albúmina Sérica Bovina/química , Ácido Hialurónico/química , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Agujas , Sistemas de Liberación de Medicamentos/instrumentación , Ratones Desnudos , Liberación de Fármacos
2.
Int J Mol Sci ; 23(3)2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35163010

RESUMEN

Repurposing of the anthelminthic drug niclosamide was proposed as an effective treatment for inflammatory airway diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease. Niclosamide may also be effective for the treatment of viral respiratory infections, such as SARS-CoV-2, respiratory syncytial virus, and influenza. While systemic application of niclosamide may lead to unwanted side effects, local administration via aerosol may circumvent these problems, particularly when the drug is encapsulated into small polyethylene glycol (PEG) hydrospheres. In the present study, we examined whether PEG-encapsulated niclosamide inhibits the production of mucus and affects the pro-inflammatory mediator CLCA1 in mouse airways in vivo, while effects on mucociliary clearance were assessed in excised mouse tracheas. The potential of encapsulated niclosamide to inhibit TMEM16A whole-cell Cl- currents and intracellular Ca2+ signalling was assessed in airway epithelial cells in vitro. We achieved encapsulation of niclosamide in PEG-microspheres and PEG-nanospheres (Niclo-spheres). When applied to asthmatic mice via intratracheal instillation, Niclo-spheres strongly attenuated overproduction of mucus, inhibited secretion of the major proinflammatory mediator CLCA1, and improved mucociliary clearance in tracheas ex vivo. These effects were comparable for niclosamide encapsulated in PEG-nanospheres and PEG-microspheres. Niclo-spheres inhibited the Ca2+ activated Cl- channel TMEM16A and attenuated mucus production in CFBE and Calu-3 human airway epithelial cells. Both inhibitory effects were explained by a pronounced inhibition of intracellular Ca2+ signals. The data indicate that poorly dissolvable compounds such as niclosamide can be encapsulated in PEG-microspheres/nanospheres and deposited locally on the airway epithelium as encapsulated drugs, which may be advantageous over systemic application.


Asunto(s)
Niclosamida/administración & dosificación , Neumonía/tratamiento farmacológico , Sistema Respiratorio/efectos de los fármacos , Animales , Asma/tratamiento farmacológico , Asma/metabolismo , Asma/patología , COVID-19/complicaciones , Células Cultivadas , Modelos Animales de Enfermedad , Portadores de Fármacos/química , Composición de Medicamentos , Humanos , Hidrogeles/química , Instilación de Medicamentos , Ratones , Microesferas , Moco/efectos de los fármacos , Moco/metabolismo , Nanosferas/administración & dosificación , Nanosferas/química , Niclosamida/química , Niclosamida/farmacocinética , Neumonía/patología , Polietilenglicoles/química , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Sistema Respiratorio/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Tráquea , Tratamiento Farmacológico de COVID-19
3.
Mol Cancer Ther ; 19(7): 1448-1461, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32371588

RESUMEN

Therapeutic advances for osteosarcoma have stagnated over the past several decades, leading to an unmet clinical need for patients. The purpose of this study was to develop a novel therapy for osteosarcoma by reformulating and validating niclosamide, an established anthelminthic agent, as a niclosamide stearate prodrug therapeutic (NSPT). We sought to improve the low and inefficient clinical bioavailability of oral dosing, especially for the relatively hydrophobic classes of anticancer drugs. Nanoparticles were fabricated by rapid solvent shifting and verified using dynamic light scattering and UV-vis spectrophotometry. NSPT efficacy was then studied in vitro for cell viability, cell proliferation, and intracellular signaling by Western blot analysis; ex vivo pulmonary metastatic assay model; and in vivo pharmacokinetic and lung mouse metastatic model of osteosarcoma. NSPT formulation stabilizes niclosamide stearate against hydrolysis and delays enzymolysis; increases circulation in vivo with t 1/2 approximately 5 hours; reduces cell viability and cell proliferation in human and canine osteosarcoma cells in vitro at 0.2-2 µmol/L IC50; inhibits recognized growth pathways and induces apoptosis at 20 µmol/L; eliminates metastatic lesions in the ex vivo lung metastatic model; and when injected intravenously at 50 mg/kg weekly, it prevents metastatic spread in the lungs in a mouse model of osteosarcoma over 30 days. In conclusion, niclosamide was optimized for preclinical drug delivery as a unique prodrug nanoparticle injected intravenously at 50 mg/kg (1.9 mmol/L). This increased bioavailability of niclosamide in the blood stream prevented metastatic disease in the mouse. This chemotherapeutic strategy is now ready for canine trials, and if successful, will be targeted for human trials in patients with osteosarcoma.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Niclosamida/farmacología , Osteosarcoma/tratamiento farmacológico , Profármacos/farmacología , Estearatos/farmacología , Animales , Antinematodos/química , Antinematodos/farmacocinética , Antinematodos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Proliferación Celular , Perros , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Humanos , Ratones , Ratones Endogámicos C57BL , Niclosamida/química , Niclosamida/farmacocinética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Profármacos/química , Profármacos/farmacocinética , Estearatos/química , Estearatos/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Anticancer Agents Med Chem ; 19(13): 1618-1626, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31284876

RESUMEN

BACKGROUND: Niclosamide is an FDA-approved and old anti-helminthic drug used to treat parasitic infections. Recent studies have shown that niclosamide has broad anti-tumor effects relevant to the treatment of cancer. However, this drug has a low aqueous solubility hindering its systemic use. Herein, we report the preparation and characterization of niclosamide nanoliposomes and their in vivo anti-tumor effects. METHODS: Nanoliposomes were prepared using thin-film method and the drug was encapsulated with a remote loading method. The nanoliposomes were investigated by the observation of morphology, analysis of particle size and zeta potential. Additionally, qualitative and quantitative analyses were performed using HPLC. We assessed the in vitro cytotoxicity of the nanoliposomal niclosamide on B16F10 melanoma cells. Inhibition of tumor growth was investigated in C57BL/6 mice bearing B16F0 melanoma cancer. RESULTS: Analytical results indicated that the nanoliposomal system is a homogeneous and stable colloidal dispersion of niclosamide particles. Atomic force microscopy images and particle size analysis revealed that all niclosamide particles had a spherical shape with a diameter of approximately 108nm. According to in vitro and in vivo studies, nanoliposomal niclosamide exhibited a better anti-tumor activity against B16F10 melanoma tumor compared with free niclosamide. CONCLUSION: Nanoliposomal encapsulation enhanced the aqueous solubility of niclosamide and improved its anti-tumor properties.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos , Liposomas , Melanoma Experimental/tratamiento farmacológico , Nanoestructuras , Niclosamida/administración & dosificación , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Composición de Medicamentos , Masculino , Ratones , Ratones Endogámicos C57BL , Niclosamida/farmacocinética , Microambiente Tumoral
5.
Drug Metab Dispos ; 47(7): 756-763, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31040114

RESUMEN

Niclosamide, an antiparasitic, has been repositioned as a potential therapeutic drug for systemic diseases based on its antiviral, anticancer, and anti-infection properties. However, low bioavailability limits its in vivo efficacy. Our aim was to determine whether metabolic disposition by microsomal P450 enzymes in liver and intestine influences niclosamide's bioavailability in vivo, by comparing niclosamide metabolism in wild-type, liver-Cpr-null (LCN), and intestinal epithelium-Cpr-null (IECN) mice. In vitro stability of niclosamide in microsomal incubations was greater in the intestine than in liver in the presence of NADPH, but it was much greater in liver than in intestine in the presence of UDPGA. NADPH-dependent niclosamide metabolism and hydroxy-niclosamide formation were inhibited in hepatic microsomes of LCN mice, but not IECN mice, compared with wild-type mice. In intestinal microsomal reactions, hydroxy-niclosamide formation was not detected, but rates of niclosamide-glucuronide formation were ∼10-fold greater than in liver, in wild-type, LCN, and IECN mice. Apparent Km and V max values for microsomal niclosamide-glucuronide formation showed large differences between the two tissues, with the intestine having higher Km (0.47 µM) and higher V max (15.8) than the liver (0.09 µM and 0.75, respectively). In vivo studies in LCN mice confirmed the essential role of hepatic P450 in hydroxy-niclosamide formation; however, pharmacokinetic profiles of oral niclosamide were only minimally changed in LCN mice, compared with wild-type mice, and the changes seem to reflect the compensatory increase in hepatic UDP-glucuronosyltransferase activity. SIGNIFICANCE STATEMENT: These results suggest that efforts to increase the bioavailability of niclosamide by blocking its metabolism by P450 enzymes will unlikely be fruitful. In contrast, inhibition of niclosamide glucuronidation in both liver and intestine may prove effective for increasing niclosamide's bioavailability, thereby making it practical to repurpose this drug for treating systemic diseases.


Asunto(s)
Antiparasitarios/farmacocinética , Reposicionamiento de Medicamentos , Mucosa Intestinal/metabolismo , Microsomas Hepáticos/metabolismo , Niclosamida/farmacocinética , Animales , Disponibilidad Biológica , Humanos , Ratones
6.
Drug Dev Ind Pharm ; 45(2): 304-313, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30348020

RESUMEN

In the present study, solid lipid nanoparticles (SLNs) have been formulated as a carrier system for effective intracellular delivery of STAT3 inhibitor, niclosamide (Niclo) to triple negative breast cancer (TNBC) cells. Emulsification-solvent evaporation method was employed in formulation of Niclo-loaded SLNs (Niclo-SLNs). The formula of Niclo-SLN was optimized by Box-Behnken design and characterized for their shape, size, and surface charge. The in vitro anti-cancer efficacy of Niclo-SLNs was studied in TNBC cells. The prepared Niclo-SLNs were found to be spherical with the particle size of 112.18 ± 1.73 nm and zetapotential of 23.8 ± 2.7 mV. In the in vitro anticancer study the Niclo SLNs show a better cytotoxicity than the naïve Niclo, which is attributed to improved cell uptake of SLN formulation. In conclusion, the results of the present study demonstrate that the formulation of Niclo as SLNs will improve the anticancer efficacy against TNBC.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Niclosamida/administración & dosificación , Niclosamida/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Portadores de Fármacos , Composición de Medicamentos , Emulsiones , Femenino , Humanos , Lípidos/química , Nanopartículas , Niclosamida/farmacocinética , Tamaño de la Partícula , Neoplasias de la Mama Triple Negativas/metabolismo
7.
PLoS One ; 13(6): e0198389, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856824

RESUMEN

BACKGROUND: Niclosamide, an FDA-approved anti-helminthic drug, has activity in preclinical models of castration-resistant prostate cancer (CRPC). Potential mechanisms of action include degrading constitutively active androgen receptor splice variants (AR-Vs) or inhibiting other drug-resistance pathways (e.g., Wnt-signaling). Published pharmacokinetics data suggests that niclosamide has poor oral bioavailability, potentially limiting its use as a cancer drug. Therefore, we launched a Phase I study testing oral niclosamide in combination with enzalutamide, for longer and at higher doses than those used to treat helminthic infections. METHODS: We conducted a Phase I dose-escalation study testing oral niclosamide plus standard-dose enzalutamide in men with metastatic CRPC previously treated with abiraterone. Niclosamide was given three-times-daily (TID) at the following dose-levels: 500, 1000 or 1500mg. The primary objective was to assess safety. Secondary objectives, included measuring AR-V expression from circulating tumor cells (CTCs) using the AdnaTest assay, evaluating PSA changes and determining niclosamide's pharmacokinetic profile. RESULTS: 20 patients screened and 5 enrolled after passing all screening procedures. 13(65%) patients had detectable CTCs, but only one was AR-V+. There were no dose-limiting toxicities (DLTs) in 3 patients on the 500mg TID cohort; however, both (N = 2) subjects on the 1000mg TID cohort experienced DLTs (prolonged grade 3 nausea, vomiting, diarrhea; and colitis). The maximum plasma concentration ranged from 35.7 to 182 ng/mL and was not consistently above the minimum effective concentration in preclinical studies. There were no PSA declines in any enrolled subject. Because plasma concentrations at the maximum tolerated dose (500mg TID) were not consistently above the expected therapeutic threshold, the Data Safety Monitoring Board closed the study for futility. CONCLUSIONS: Oral niclosamide could not be escalated above 500mg TID, and plasma concentrations were not consistently above the threshold shown to inhibit growth in CRPC models. Oral niclosamide is not a viable compound for repurposing as a CRPC treatment. CLINICAL TRIAL REGISTRY: Clinicaltrials.gov: NCT02532114.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niclosamida/administración & dosificación , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Benzamidas , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Metástasis de la Neoplasia , Niclosamida/efectos adversos , Niclosamida/farmacocinética , Nitrilos , Feniltiohidantoína/administración & dosificación , Feniltiohidantoína/efectos adversos , Feniltiohidantoína/farmacocinética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología
8.
Cell Signal ; 41: 89-96, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28389414

RESUMEN

Niclosamide is an oral antihelminthic drug used to treat parasitic infections in millions of people worldwide. However recent studies have indicated that niclosamide may have broad clinical applications for the treatment of diseases other than those caused by parasites. These diseases and symptoms may include cancer, bacterial and viral infection, metabolic diseases such as Type II diabetes, NASH and NAFLD, artery constriction, endometriosis, neuropathic pain, rheumatoid arthritis, sclerodermatous graft-versus-host disease, and systemic sclerosis. Among the underlying mechanisms associated with the drug actions of niclosamide are uncoupling of oxidative phosphorylation, and modulation of Wnt/ß-catenin, mTORC1, STAT3, NF-κB and Notch signaling pathways. Here we provide a brief overview of the biological activities of niclosamide, its potential clinical applications, and its challenges for use as a new therapy for systemic diseases.


Asunto(s)
Antihelmínticos/uso terapéutico , Niclosamida/uso terapéutico , Animales , Antihelmínticos/farmacocinética , Antihelmínticos/farmacología , Artritis Reumatoide/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Modelos Animales de Enfermedad , Endometriosis/tratamiento farmacológico , Femenino , Humanos , Infecciones/tratamiento farmacológico , Ratones , Neoplasias/tratamiento farmacológico , Niclosamida/farmacocinética , Niclosamida/farmacología , Ratas , Esclerodermia Sistémica/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Oncotarget ; 7(8): 8993-9006, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26848771

RESUMEN

Ovarian cancer treatment remains a challenge and targeting cancer stem cells presents a promising strategy. Niclosamide is an "old" antihelminthic drug that uncouples mitochondria of intestinal parasites. Although recent studies demonstrated that niclosamide could be a potential anticancer agent, its poor water solubility needs to be overcome before further preclinical and clinical investigations can be conducted. Therefore, we evaluated a novel nanosuspension of niclosamide (nano-NI) for its effect against ovarian cancer. Nano-NI effectively inhibited the growth of ovarian cancer cells in which it induced a metabolic shift to glycolysis at a concentration of less than 3 µM in vitro and suppressed tumor growth without obvious toxicity at an oral dose of 100 mg/kg in vivo. In a pharmacokinetic study after oral administration, nano-NI showed rapid absorption (reaching the maximum plasma concentration within 5 min) and improved the bioavailability (the estimated bioavailability for oral nano-NI was 25%). In conclusion, nano-NI has the potential to be a new treatment modality for ovarian cancer and, therefore, further clinical trials are warranted.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Niclosamida/farmacología , Niclosamida/farmacocinética , Neoplasias Ováricas/tratamiento farmacológico , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Femenino , Glucólisis/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Consumo de Oxígeno/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Suspensiones/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Microencapsul ; 32(5): 496-502, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26079596

RESUMEN

Niclosamide (NL) has demonstrated its great potential in fighting against leukaemia recently. However, either oral or systemic delivery of NL is challenged by its insoluble nature. Here, we developed two different NL-loaded submicron lipid emulsions (NL-SLEs) and compared their suitability in bioavailability enhancement. Conventional and PEGylated NL-SLEs (NL-CSLEs and NL-PSLEs) were prepared by melt dispersion/high pressure homogenisation technique. They were about 307.8 and 162.2 nm in particle size, respectively, and both of them possessed satisfactory stability and drug load (>9.0%). After oral administration, significantly enhanced bioavailability was achieved through NL-CSLEs and NL-PSLEs (441.11 and 463.55% relative to the reference). Apart from global size, NL-CSLEs and NL-PSLEs exhibited similar attributes in release, lipolysis, mucin binding, etc. Taken together, SLEs with or without PEG-lipid have shown to be promising for oral delivery of NL. PEG-lipid could significantly reduce the particle size of SLEs. But, macromolecular PEG-lipid was required to effectively stealth the lipid carriers.


Asunto(s)
Portadores de Fármacos , Lípidos , Niclosamida , Polietilenglicoles , Administración Oral , Animales , Disponibilidad Biológica , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Emulsiones , Lípidos/química , Lípidos/farmacocinética , Lípidos/farmacología , Niclosamida/química , Niclosamida/farmacocinética , Niclosamida/farmacología , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Ratas , Ratas Sprague-Dawley
11.
Colloids Surf B Biointerfaces ; 131: 170-81, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25988281

RESUMEN

Since the turn of the 21st century, nanofiber based drug delivery systems have evolved drastically to attain controlled and sustained delivery of various bioactive molecules. In spite of such efforts, the tangible interface existing between the target cells and the drug molecules could not be narrowed down. This drawback has been overcome in this work by realizing nanofiber based scaffold for delivery of polymer-drug complexes rather than just the drug. In course with this, in the present study a differentially cross-linkable bPEI-PEO (branched-polyethylenimine-poly(ethylene oxide)) based nanofiber is fabricated for tunable delivery of bPEI-niclosamide complexes. Hydrophilic bPEI-niclosamide complexes are pre-synthesized and stabilized by crosslinking agent, which were then incorporated into bPEI-PEO nanofibers by electrospinning. The niclosamide loaded nanofibers by virtue of bPEI moieties presence were then cross-linked to different degrees which in turn altered bPEI-niclosamide release profile. The release kinetics of bPEI-niclosamide complexes from nanofibers was elucidated further by Korsmeyer-Peppas model. Apart from this, the versatile nature of bPEI-PEO nanofibers was also validated for different drug loading concentration and extent of crosslinking. The fibers antitumor efficacy was then assessed against A549 (Non-small cell lung cancer cells) and U-87 MG (glioblastoma cells) at two different time points (at 48h and 96h) in order to realize the importance of release profile in manifestation of different therapeutic outcomes. Thus, this work endows niclosamide a new life for anticancer application which has remained elusive till date due to its hydrophobic nature.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanofibras/química , Niclosamida/administración & dosificación , Polietilenglicoles/química , Polietileneimina/química , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Cinética , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Nanofibras/ultraestructura , Niclosamida/farmacocinética , Niclosamida/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectroscopía Infrarroja por Transformada de Fourier , Factores de Tiempo
12.
Chin J Cancer ; 31(4): 178-84, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22237038

RESUMEN

Niclosamide, an oral antihelminthic drug, has been used to treat tapeworm infection for about 50 years. Niclosamide is also used as a molluscicide for water treatment in schistosomiasis control programs. Recently, several groups have independently discovered that niclosamide is also active against cancer cells, but its precise mechanism of antitumor action is not fully understood. Evidence supports that niclosamide targets multiple signaling pathways (NF-κB, Wnt/ß-catenin, Notch, ROS, mTORC1, and Stat3), most of which are closely involved with cancer stem cells. The exciting advances in elucidating the antitumor activity and the molecular targets of this drug will be discussed. A method for synthesizing a phosphate pro-drug of niclosamide is provided. Given its potential antitumor activity, clinical trials for niclosamide and its derivatives are warranted for cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Niclosamida/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Niclosamida/farmacocinética , Especies Reactivas de Oxígeno/metabolismo , Receptores Notch/metabolismo , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
13.
Cancer Res ; 71(12): 4172-82, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21531761

RESUMEN

Wnt/ß-catenin pathway activation caused by adenomatous polyposis coli (APC) mutations occurs in approximately 80% of sporadic colorectal cancers (CRC). The antihelminth compound niclosamide downregulates components of the Wnt pathway, specifically Dishevelled-2 (Dvl2) expression, resulting in diminished downstream ß-catenin signaling. In this study, we determined whether niclosamide could inhibit the Wnt/ß-catenin pathway in human CRCs and whether its inhibition might elicit antitumor effects in the presence of APC mutations. We found that niclosamide inhibited Wnt/ß-catenin pathway activation, downregulated Dvl2, decreased downstream ß-catenin signaling, and exerted antiproliferative effects in human colon cancer cell lines and CRC cells isolated by surgical resection of metastatic disease, regardless of mutations in APC. In contrast, inhibition of NF-κB or mTOR did not exert similar antiproliferative effects in these CRC model systems. In mice implanted with human CRC xenografts, orally administered niclosamide was well tolerated, achieved plasma and tumor levels associated with biologic activity, and led to tumor control. Our findings support clinical explorations to reposition niclosamide for the treatment of CRC.


Asunto(s)
Antihelmínticos/farmacología , Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Genes APC , Mutación , Niclosamida/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/análisis , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Proteínas Dishevelled , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Ratones SCID , FN-kappa B/antagonistas & inhibidores , Niclosamida/farmacocinética , Compuestos Organoplatinos/farmacología , Oxaliplatino , Fosfoproteínas/análisis , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , beta Catenina/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA