Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 60(25): 14013-14021, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33768682

RESUMEN

The presence of bacteria in the tumor can cause cancer resistance to chemotherapeutics. To fight against bacterium-induced drug resistance, herein we design self-traceable nanoreservoirs that are simultaneously loaded with gemcitabine (an anticancer drug) and ciprofloxacin (an antibiotic) and are decorated with hyaluronic acid for active tumor targeting. The nanoreservoirs have a pH-sensitive gate and an enzyme-responsive gate that can be opened in the acidic and hyaluronidase-abundant tumor microenvironment to control drug release rates. Moreover, the nanoreservoirs can specifically target the tumor regions without eliciting evident toxicity to normal tissues, kill the intratumoral bacteria, and inhibit the tumor growth even in the presence of the bacteria. Unexpectedly, the nanoreservoirs can activate T cell-mediated immune responses through promoting antigen-presenting dendritic cell maturation and depleting immunosuppressive myeloid-derived suppressor cells in bacterium-infected tumors.


Asunto(s)
Antibacterianos/farmacología , Antimetabolitos Antineoplásicos/farmacología , Ciprofloxacina/farmacología , Neoplasias del Colon/terapia , Desoxicitidina/análogos & derivados , Escherichia coli/efectos de los fármacos , Animales , Antibacterianos/química , Antimetabolitos Antineoplásicos/química , Línea Celular , Ciprofloxacina/química , Neoplasias del Colon/microbiología , Desoxicitidina/química , Desoxicitidina/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Humanos , Ratones , Gemcitabina
2.
J Mater Chem B ; 8(26): 5667-5681, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32500886

RESUMEN

Carrier-free nanotheranostics directly assembled by using clinically used photosensitizers and chemotherapeutic drugs are a promising alternative to tumor theranostics. However, the weak interaction-driven assembly still suffers from low structural stability against disintegration, lack of targeting specificity, and poor stimulus-responsive property. Moreover, almost all exogenous ligands possess no therapeutic effect. Enlightened by the concept of metal-organic frameworks, we developed a novel self-recognizing metal-coordinated nanotheranostic agent by the coordination-driven co-assembly of photosensitizer indocyanine green (ICG) and chemo-drug methotrexate (MTX, also served as a specific "targeting ligand" towards folate receptors), in which ferric (FeIII) ions acted as a bridge to tightly associate ICG with MTX. Such carrier-free metal-coordinated nanotheranostics with high dual-drug payload (∼94 wt%) not only possessed excellent structural and physiological stability, but also exhibited prolonged blood circulation. In addition, the nanotheranostics could achieve the targeted on-demand drug release by both stimuli of internal lysosomal acidity and external near-infrared laser. More importantly, the nanotheranostics could self-recognize the cancer cells and selectively target the tumors, and therefore they decreased toxicity to normal tissues and organs. Consequently, the nanotheranostics showed strongly synergistic potency for tumor photo-chemotherapy under the precise guidance of magnetic resonance/photoacoustic/fluorescence imaging, thereby achieving highly effective tumor curing efficiency. Considering that ICG and bi-functional MTX are approved by the Food and Drug Administration, and FeIII ions have high biosafety, the self-recognizing and stimulus-responsive carrier-free metal-coordinated nanotheranostics may hold potential applications in tumor theranostics.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Verde de Indocianina/farmacología , Metotrexato/farmacología , Fármacos Fotosensibilizantes/farmacología , Fototerapia , Nanomedicina Teranóstica , Animales , Antimetabolitos Antineoplásicos/síntesis química , Antimetabolitos Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Verde de Indocianina/síntesis química , Verde de Indocianina/química , Imagen por Resonancia Magnética , Metotrexato/síntesis química , Metotrexato/química , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Imagen Óptica , Tamaño de la Partícula , Técnicas Fotoacústicas , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Propiedades de Superficie
3.
Sci Rep ; 10(1): 1138, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980685

RESUMEN

Halogen-modified nucleic acid molecules, such as trifluorothymidine (FTD) and 5-fluorouracil, are widely used in medical science and clinical site. These compounds have a very similar nucleobase structure. It is reported that both of these compounds could be incorporated into DNA. The incorporation of FTD produces highly anti-tumor effect. However, it is not known whether to occur a significant effect by the incorporation of 5-fluorouracil. Nobody knows why such a difference will occur. To understand the reason why there is large differences between trifluorothymidine and 5-fluorouracil, we have performed the molecular dynamics simulations and molecular orbital calculations. Although the active interaction energy between Halogen-modified nucleic acids or and complementary adenine was increased, in only FTD incorporated DNA, more strongly dispersion force interactions with an adjacent base were detected in many thermodynamic DNA conformations. As the results, the conformational changes occur even if it is in internal body temperature. Then the break of hydrogen bonding between FTD and complementary adenine base occur more frequently. The double helix structural destabilization of DNA with FTD is resulted from autoagglutination caused by the bonding via halogen orbitals such as halogen bonding and the general van der Waals interactions such as CH-[Formula: see text], lone pair (LP)-[Formula: see text], and [Formula: see text]-[Formula: see text] interactions. Therefore, it is strongly speculated that such structural changes caused by trifluoromethyl group is important for the anti-tumor effect of FTD alone.


Asunto(s)
Adenina/química , Antimetabolitos Antineoplásicos/química , ADN/efectos de los fármacos , Fluorouracilo/química , Trifluridina/química , Emparejamiento Base , ADN/química , Daño del ADN , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Estructura Molecular , Conformación de Ácido Nucleico , Teoría Cuántica , Termodinámica
4.
AAPS PharmSciTech ; 20(8): 309, 2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31520191

RESUMEN

A synthetic, dispersible magnesium aminoclay (MgAC) was synthesized in the present study. Besides, structural and spectroscopic detections were conducted to investigate the MgAC nanoclay. With a poor aqueous solubility, methotrexate (MTX) has been applied as a valid antitumor agent in recent years. In our research, an unobtrusive sol-gel process was carried out to manufacture the MgAC-MTX nanohybrids through entrapment of MTX over MgAC in situ. The final product was capable of desquamating and thus dispersed in water, equably. In comparison with rough MTX, the MgAC-MTX nanocomposite with a preferable treatment efficacy against MCF-7 cells was mainly attributed to the preeminent enhanced aqueous solubility, controlled release and the increased cellular uptake capacity. Moreover, with excellent anticancer function and hypotoxicity as vindicated in vivo, the MgAC-MTX nanohybrid was supposed to own the potency in the application of malignant tumors cure as a valid nanomedicine. It turned out that, by virtue of its high bioavailability, the MgAC-MTX nanohybrids with high bioavailability is deserving of further study for the treatment of cancers.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Arcilla/química , Compuestos de Magnesio/química , Metotrexato/administración & dosificación , Vehículos Farmacéuticos/química , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/uso terapéutico , Disponibilidad Biológica , Femenino , Geles , Humanos , Células MCF-7 , Metotrexato/química , Metotrexato/uso terapéutico , Ratones , Nanoestructuras , Neoplasias Experimentales/tratamiento farmacológico , Tamaño de la Partícula
5.
Biol Trace Elem Res ; 187(1): 80-91, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29748931

RESUMEN

Drug resistance is a major challenge of breast and colon cancer therapies leading to treatment failure. The main objective of the current study is to investigate whether selenium nanoparticles (nano-Se) can induce the chemo-sensitivity of 5-fluorouracil (FU)-encapsulated poly (D, L-lactide-co-glycolide) nanoparticles (nano-FU) in breast and colon cancer cell lines. Nano-Se and nano-FU were synthesized and characterized, then applied individually or in combination upon MCF7, MDA-MB-231, HCT 116, and Caco-2 cancerous cell lines. Cytotoxicity, cellular glucose uptake, and apoptosis, as well as malondialdehyde (MDA), nitric oxide (NO), and zinc (Zn) levels, were investigated upon the different treatments. We have resulted that nano-FU induced cell death in MCF7 and Caco-2 more effectively than MDA-MB-231 and HCT 116 cell lines. Moreover, nano-FU plus nano-Se potentiate MCF7 and Caco-2 chemo-sensitivity were higher than MDA-MB-231 and HCT 116 cancerous cell lines. It is relevant to note that Se and FU nano-formulations inhibited cancer cell bioenergetics via glucose uptake slight blockage. Furthermore, nano-FU increased the levels of NO and MDA in media over cancer cells, while their combinations with nano-Se rebalance the redox status with Zn increment. We noticed that MCF7 cell line is sensitive, while MDA-MB-231 cell line is resistant to Se and nano-Se. This novel approach could be of great potential to enhance the chemo-sensitivity in breast and colon cancer cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Fluorouracilo/farmacología , Nanopartículas/química , Selenio/química , Antimetabolitos Antineoplásicos/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Fluorouracilo/química , Humanos , Tamaño de la Partícula , Propiedades de Superficie , Células Tumorales Cultivadas
6.
Int J Nanomedicine ; 13: 3069-3080, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872294

RESUMEN

PURPOSE: Gemcitabine's clinical application is limited due to its short plasma half-life and poor uptake by cells. To address this problem, a drug delivery three-component composite, multiwalled carbon nanotubes (MWNTs)/gemcitabine (Ge)/lentinan (Le; MWNTs-Ge-Le), was fabricated in our study. Moreover, the combination of chemotherapy and photothermal therapy was employed to enhance antitumor efficacy. METHODS: In this study, we conjugated gemcitabine and lentinan with MWNTs via a covalent and noncovalent way to functionalize with MWNTs, and the chemical structure of MWNTs-Ge-Le was characterized by Fourier transform infrared spectroscopy, Raman spectroscopy, thermogravimetric analysis and transmission electron microscopy. Using the composite and an 808 nm laser, we treated tumors, both in vitro and in vivo, and investigated the photothermal responses and the anticancer efficacy. RESULTS: The MWNTs-Ge-Le composite could efficiently cross cell membrane, having a higher antitumor activity than MWNTs, gemcitabine and MWNTs-Ge in vitro and in vivo. Our study on the MWNTs-Ge-Le composite with an 808 nm laser radiation showed the combination of drug therapy and near-infrared photothermal therapy possesses great synergistic antitumor efficacy. CONCLUSION: The MWNTs-Ge-Le three-component anticancer composite can serve as a promising candidate for cancer therapy in the combination of chemotherapy and photothermal therapy.


Asunto(s)
Desoxicitidina/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Lentinano/química , Nanotubos de Carbono/química , Neoplasias Experimentales/terapia , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Desoxicitidina/administración & dosificación , Desoxicitidina/química , Femenino , Humanos , Terapia por Láser/métodos , Rayos Láser , Células MCF-7 , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Fototerapia/métodos , Espectroscopía Infrarroja por Transformada de Fourier , Espectrometría Raman , Termogravimetría , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
7.
Biol Chem ; 399(3): 293-303, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29016350

RESUMEN

Nanoparticles (NPs) are recognized as an attractive vehicles for cancer treatment due to their targeted drug release. Gastric cancer is an important killer disease, and its therapy methods still need improvement. The NPs were prepared using a precipitation method, and were evaluated using transmission electron microscopy (TEM). MTT and Transwell assays were used to determine cell viability and apoptosis. In vivo experiments were performed to validate the effects of NPs on tumor growth. Methioninase (METase)/5-Fu co-encaspulated NPs showed highest ζ size and lowest ζ potential than other NPs. The migration and tumorsphere formation ability of CD44(+) was stronger than CD44(-). The effects of METase/5-Fu co-encaspulated NPs on inhibition cell growth was stronger than that of 5-Fu encaspulated NPs, while HA coated NPs showed significant target ability than that NPs without HA. METase supplementation promoted the inhibition effect of 5-Fu on thymidylate synthetase (TS), as well as cell apoptosis. The in vivo experiments demonstrated that HA coated NPs significantly inhibited tumor growth. It was concluded that HA-coated NPs enhance the target ability, while METase/5-Fu co-encaspulated NPs promote the inhibition effects on tumor growth in gastric cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Liasas de Carbono-Azufre/química , Diseño de Fármacos , Fluorouracilo/farmacología , Ácido Hialurónico/química , Nanopartículas/química , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Liasas de Carbono-Azufre/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/química , Ensayos de Selección de Medicamentos Antitumorales , Fluorouracilo/química , Fluorouracilo/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Plásmidos/metabolismo , Neoplasias Gástricas/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
8.
Molecules ; 22(9)2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28880240

RESUMEN

S-1 (TS-1®) is an oral fluoropyrimidine anticancer agent containing tegafur, oteracil, and gimeracil. Sipjeondaebo-tang (SDT) is a traditional oriental herbal medicine that has potential to alleviate chemotherapy-related adverse effects. The aim of the present study was to evaluate the effect of SDT on the pharmacokinetics of S-1. Sprague-Dawley rats were pretreated with a single dose or repeated doses of SDT for seven consecutive days (1200 mg/kg/day). After the completion of pretreatment with SDT, S-1 was orally administered and plasma concentrations of tegafur, its active metabolite 5-FU, and gimeracil were determined by liquid chromatography-tandem mass spectrometry (LC/MS/MS). A population pharmacokinetic model was developed to evaluate the effect of SDT on pharmacokinetics of tegafur and 5-FU. Although a single dose of SDT did not have any significant effect, the absorption rate of tegafur decreased, and the plasma levels of 5-FU reduced significantly in rats pretreated with SDT for seven days in parallel to the decreased gimeracil concentrations. Population pharmacokinetic modeling also showed the enhanced elimination of 5-FU in the SDT-pretreated group. Repeated doses of SDT may inhibit the absorption of gimeracil, an inhibitor of 5-FU metabolism, resulting in enhanced elimination of 5-FU and decrease its plasma level.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacocinética , Medicamentos Herbarios Chinos/farmacología , Ácido Oxónico/farmacocinética , Piridinas/farmacocinética , Tegafur/farmacocinética , Administración Oral , Animales , Antimetabolitos Antineoplásicos/química , Combinación de Medicamentos , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Fluorouracilo/metabolismo , Interacciones de Hierba-Droga , Humanos , Masculino , Modelos Biológicos , Ácido Oxónico/química , Piridinas/química , Ratas Sprague-Dawley , Tegafur/química
9.
J Control Release ; 262: 192-200, 2017 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-28764995

RESUMEN

Magnetically responsive microbubbles (MagMBs), consisting of an oxygen gas core and a phospholipid coating functionalised with Rose Bengal (RB) and/or 5-fluorouracil (5-FU), were assessed as a delivery vehicle for the targeted treatment of pancreatic cancer using combined antimetabolite and sonodynamic therapy (SDT). MagMBs delivering the combined 5-FU/SDT treatment produced a reduction in cell viability of over 50% when tested against a panel of four pancreatic cancer cell lines in vitro. Intravenous administration of the MagMBs to mice bearing orthotopic human xenograft BxPC-3 tumours yielded a 48.3% reduction in tumour volume relative to an untreated control group (p<0.05) when the tumour was exposed to both external magnetic and ultrasound fields during administration of the MagMBs. In contrast, application of an external ultrasound field alone resulted in a 27% reduction in tumour volume. In addition, activated caspase and BAX protein levels were both observed to be significantly elevated in tumours harvested from animals treated with the MagMBs in the presence of magnetic and ultrasonic fields when compared to expression of those proteins in tumours from either the control or ultrasound field only groups (p<0.05). These results suggest MagMBs have considerable potential as a platform to enable the targeted delivery of combined sonodynamic/antimetabolite therapy in pancreatic cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Fluorouracilo/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Microburbujas , Sonicación , Animales , Antimetabolitos Antineoplásicos/química , Avidina/administración & dosificación , Avidina/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Compuestos Férricos/administración & dosificación , Compuestos Férricos/química , Fluorouracilo/química , Humanos , Fenómenos Magnéticos , Nanopartículas del Metal/química , Ratones SCID , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Rosa Bengala/administración & dosificación , Rosa Bengala/química , Carga Tumoral/efectos de los fármacos
10.
Int J Nanomedicine ; 12: 2793-2811, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28435259

RESUMEN

Heat-based approaches have been considered as promising tools due to their ability to directly eradicate tumor cells and/or increase the sensitivity of tumors to radiation- or chemotherapy. In particular, the heating of magnetic nanoparticles (MNPs) via an alternating magnetic field can provide a handy alternative for a localized tumor treatment. To amplify the efficacy of magnetically induced thermal treatments, we elucidated the superior tumor-destructive effect of methotrexate-coupled MNPs (MTX/MNPs) in combination with magnetic heating (nanochemothermia) over the thermal treatment alone. Our studies in a murine bladder xenograft model revealed the enormous potential of nanochemothermia for a localized and relapse-free destruction of tumors which was superior to the thermal treatment alone. Nanochemothermia remarkably fostered the reduction of tumor volume. It impaired proapoptotic signaling (eg, p-p53), cell survival (eg, p-ERK1/2), and cell cycle (cyclins) pathways. Additionally, heat shock proteins (eg, HSP70) were remarkably affected. Moreover, nanochemothermia impaired the induction of angiogenic signaling by decreasing, for example, the levels of VEGF-R1 and MMP9, although an increasing tumor hypoxia was indicated by elevated Hif-1α levels. In contrast, tumor cells were able to recover after the thermal treatments alone. In conclusion, nanochemothermia on the basis of MTX/MNPs was superior to the thermal treatment due to a modification of cellular pathways, particularly those associated with the cellular survival and tumor vasculature. This allowed very efficient and relapse-free destruction of tumors.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Metotrexato/farmacología , Nanopartículas/química , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Hipertermia Inducida/métodos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Magnetismo , Masculino , Metotrexato/administración & dosificación , Metotrexato/química , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Basic Clin Pharmacol Toxicol ; 119(6): 540-547, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27194111

RESUMEN

The compound 5-fluorouracil (5-FU) is used in cancer chemotherapy and is known to cause diarrhoea. We recently reported that chemokine (C-X-C motif) ligand 1 (CXCL1) and neutrophils in the colonic mucosa were markedly increased by the administration of 5-FU in mice. Curcumin has anti-inflammatory, antitumour and antioxidant properties. Therefore, we examined the effect of curcumin on 5-FU-induced diarrhoea development and CXCL1 and CXCL2 up-regulation in the colon. Mice were given 5-FU (50 mg/kg, i.p.) daily for 4 days. Curcumin (100 or 300 mg/kg, p.o.) was administered on the day before the first administration of 5-FU and administered 30 min. before the administration of 5-FU. Gene expression levels of CXCL1 and CXCL2 in the colon were examined by real-time RT-PCR. Curcumin reduced the 5-FU-induced diarrhoea development. Under this condition, the CXCL1 and CXCL2 gene up-regulated by 5-FU administration was inhibited by curcumin. The gene expression of CXCL1 and CXCL2 was also enhanced by 5-FU application in vitro. The 5-FU-induced up-regulated CXCL1 and CXCL2 gene expressions were inhibited by curcumin, Bay-117082 and bortezomib, nuclear factor kappa B (NF-κB) inhibitors, C646, a p300/cyclic adenosine monophosphate response element-binding protein-histone acetyltransferase (HAT) inhibitor. In conclusion, these findings suggested that curcumin prevented the development of diarrhoea by inhibiting NF-κB and HAT activation.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Antioxidantes/uso terapéutico , Colon Descendente/efectos de los fármacos , Curcumina/uso terapéutico , Diarrea/prevención & control , Suplementos Dietéticos , Fluorouracilo/efectos adversos , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Quimiocina CXCL1/agonistas , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/agonistas , Quimiocina CXCL2/antagonistas & inhibidores , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Colon Descendente/inmunología , Colon Descendente/metabolismo , Colon Descendente/fisiopatología , Curcumina/administración & dosificación , Curcumina/farmacología , Diarrea/inducido químicamente , Diarrea/metabolismo , Diarrea/fisiopatología , Proteína p300 Asociada a E1A/agonistas , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Proteína p300 Asociada a E1A/metabolismo , Inhibidores Enzimáticos/farmacología , Fluorouracilo/antagonistas & inhibidores , Fluorouracilo/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/agonistas , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Índice de Severidad de la Enfermedad , Técnicas de Cultivo de Tejidos
12.
Eur J Pharm Sci ; 92: 276-86, 2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27154260

RESUMEN

Polysaccharide-based nanocomplexes, intended for simultaneous encapsulation and controlled release of 5-Fluorouracil (5-FU) and Temozolomide (TMZ) were developed via the complexation method using chitosan, alginic and polygalacturonic acid. Investigation focused on the influence of polysaccharides on the properties of the system and amelioration of the stability of the drugs, in particular TMZ. The dimensions of particles and their ζ-potential were found to range between 100 and 200nm and -25 to +40mV, respectively. Encapsulation efficiency varied from 16% to over 70%, depending on the given system. The influence of pH on the release and co-release of TMZ and 5-FU was evaluated under different pH conditions. The stability of the loaded drug, in particular TMZ, after release was evaluated and confirmed by LC-MS analysis. Results suggested that the amount of loaded drug(s) and the release rate is connected with the weight ratio of polysaccharides and the pH of the media. One-way ANOVA analysis on the obtained data revealed no interference between the drugs during the encapsulation and release process, and in particular no hydrolysis of TMZ occurred suggesting that CS-ALG and CS-PGA would represent interesting carriers for multi-drug controlled release and drugs protection.


Asunto(s)
Antimetabolitos Antineoplásicos/química , Antineoplásicos Alquilantes/química , Dacarbazina/análogos & derivados , Fluorouracilo/química , Nanopartículas/química , Profármacos/química , Alginatos/química , Quitosano/química , Dacarbazina/química , Preparaciones de Acción Retardada/química , Portadores de Fármacos/química , Combinación de Medicamentos , Composición de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Pectinas/química , Temozolomida
14.
Int J Pharm ; 505(1-2): 96-106, 2016 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26997424

RESUMEN

A novel platform making up of methotrexate intercalated layered double hydroxide (MTX/LDH) hybrid doped with gold nanoparticles (NPs) may have great potential both in chemo-photothermal therapy and the simultaneous drug delivery. In this paper, a promising platform of Au@PDDA-MTX/LDH was developed for anti-tumor drug delivery and synergistic therapy. Firstly, Au NPs were coated using Layer-by-Layer (LbL) technology by alternate deposition of poly (diallyldimethylammonium chloride) (PDDA) and MTX molecules, and then the resulting core-shell structures (named as Au@PDDA-MTX) were directly conjugated onto the surface of MTX/LDH hybrid by electrostatic attraction to afford Au@PDDA-MTX/LDH NPs. Here MTX was used as both the agent for surface modification and the anti-tumor drug for chemotherapy. The platform of Au@PDDA-MTX/LDH NPs not only had a high drug-loading capacity, but also showed excellent colloidal stability and interesting pH-responsive release profile. In vitro drug release studies demonstrated that MTX released from Au@PDDA-MTX/LDH was relatively slow under normal physiological pH, but it was enhanced significantly at a weak acidic pH value. Furthermore, the combined treatment of cancer cells by using Au@PDDA-MTX/LDH for synergistic hyperthermia ablation and chemotherapy was demonstrated to exhibit higher therapeutic efficacy than either single treatment alone, underscoring the great potential of the platform for cancer therapy.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Hipertermia Inducida , Nanopartículas del Metal , Metotrexato/administración & dosificación , Adenocarcinoma/terapia , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Terapia Combinada , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Oro/química , Humanos , Concentración de Iones de Hidrógeno , Neoplasias Pulmonares/terapia , Metotrexato/química , Metotrexato/farmacología , Polietilenos/química , Compuestos de Amonio Cuaternario/química
15.
Int J Nanomedicine ; 11: 485-500, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26893557

RESUMEN

Today, the therapeutic efficacy of cancer is restricted by the heterogeneity of the response of tumor cells to chemotherapeutic drugs. Since those therapies are also associated with severe side effects in nontarget organs, the application of drugs in combination with nanocarriers for targeted therapy has been suggested. Here, we sought to assess whether the coupling of methotrexate (MTX) to magnetic nanoparticles (MNP) could serve as a valuable tool to circumvent the heterogeneity of tumor cell response to MTX by the combined treatment with hyperthermia. To this end, we investigated five breast cancer cell lines of different origin and with different mutational statuses, as well as a bladder cancer cell line in terms of their response to exposure to MTX as a free drug or after its coupling to MNP as well as in presence/absence of hyperthermia. We also assessed whether the effects could be connected to the cell line-specific expression of proteins related to the uptake and efflux of MTX and MNP. Our results revealed a very heterogeneous and cell line-dependent response to an exposure with MTX-coupled MNP (MTX-MNP), which was almost comparable to the efficacy of free MTX in the same cell line. Moreover, a cell line-specific and preferential uptake of MTX-MNP compared with MNP alone was found (probably by receptor-mediated endocytosis), agreeing with the observed cytotoxic effects. Opposed to this, the expression pattern of several cell membrane transport proteins noted for MTX uptake and efflux was only by tendency in agreement with the cellular toxicity of MTX-MNP in different cell lines. Higher cytotoxic effects were achieved by exposing cells to a combination of MTX-MNP and hyperthermal treatment, compared with MTX or thermo-therapy alone. However, the heterogeneity in the response of the tumor cell lines to MTX could not be completely abolished - even after its combination with MNP and/or hyperthermia - and the application of higher thermal dosages might be necessary.


Asunto(s)
Hipertermia Inducida/métodos , Nanopartículas de Magnetita/química , Metotrexato/administración & dosificación , Metotrexato/farmacología , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral/efectos de los fármacos , Terapia Combinada/métodos , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Nanopartículas de Magnetita/administración & dosificación , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Metotrexato/química
16.
J Biomed Nanotechnol ; 11(10): 1808-18, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26502643

RESUMEN

Glioblastoma multiforme is a devastating cerebral tumor with an exceedingly poor prognosis. Methotrexate (MTX) is a folic acid analogue that inhibits DNA synthesis by binding to dihydrofolate reductase. Biodegradable nanoparticles are emerging as a promising system for drug delivery to specific tissues. The aims of the current study were pharmacological improvement and preclinical evaluation of MTX-loaded lipid-core nanocapsules (MTX-LNCs) in a glioblastoma model. Cell viability was assessed using the MTT assay, and the cell cycle was characterized by flow cytometry analysis of propidium iodide staining. Apoptosis was measured using an AnnexinV kit and by examining active caspase-3 immunocontent. In vivo glioma implantation was performed in rats, followed by measurement of the tumor size and tumoral apoptosis, BCL-2 immunohistochemistry and analyses of toxicological parameters. MTX-LNCs with increased encapsulation efficiency were successfully prepared. Our in vitro results showed a decrease in glioma cell viability after MTX-LNC treatment that was preceded by cell cycle arrest, leading the cells to undergo apoptotic death, as indicated by AnnexinV staining and increased active caspase-3 protein levels. In the in vivo glioma model, we observed a decrease in the tumor size and an increase in apoptosis in the tumor microenvironment (based on the AnnexinV assay and BCL-2 measurement). MTX-LNC treatment decreased the leukocyte number but altered neither toxicological tissue marker expression nor metabolic parameters. The present results reveal that MTX-LNCs represented an efficient formulation in a preclinical model of glioma and are a potential candidate for clinical trials.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Liposomas/química , Metotrexato/administración & dosificación , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Difusión , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Glioblastoma/patología , Masculino , Ensayo de Materiales , Metotrexato/química , Nanocápsulas/ultraestructura , Tamaño de la Partícula , Ratas , Ratas Wistar , Resultado del Tratamiento
17.
Int J Pharm ; 492(1-2): 80-91, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26165611

RESUMEN

Gemcitabine is one of the most potent anticancer agents acting on a wide range of solid tumors, however, its use is limited by short half life and high dose leading to serious side effects. The present investigation describes the development and characterization of folate functionalized gemcitabine loaded bovine serum albumin nanoparticles (Fa-Gem-BSANPs). The nanoparticles were prepared by desolvation cross-linking technique and characterized for various parameters including morphology, particle size, zeta potential, drug loading and release profile. The particle size of Gem-BSANPs and Fa-Gem-BSANPs was found to be 159.1±5.29 and 208.7±1.80 nm, respectively. DSC and XRD analysis indicated amorphous nature of the drug within the particles. The encapsulated gemcitabine exhibited less hemolytic properties as compared to native drug. The anticancer activity of Fa-Gem-BSANPs was evaluated in folate receptor over expressing cell lines (Ovcar-5 and MCF-7) and folate receptor deficient cell line (MIAPaCa-2). The Fa-Gem-BSANPs showed superior anticancer activity as compared to Gem-BSANPs in Ovcar-5 and MCF-7 cells while no significant difference in cytotoxicity was found with MIAPaCa-2 cells. Confocal microscopy indicated facilitated intracellular uptake of Fa-Gem-BSANPs in MCF-7, which in turn result in a higher potential for apoptosis. Further, Fa-Gem-BSANPs exhibited improved anti-tumor activity in Ehrlich solid tumor model in mice. In conclusion, our study indicates that folate functionalized nanoparticles confer enhance cellular uptake and cytotoxicity for gemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos , Desoxicitidina/análogos & derivados , Sistemas de Liberación de Medicamentos , Ácido Fólico , Nanopartículas , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Ehrlich/tratamiento farmacológico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/química , Desoxicitidina/farmacología , Liberación de Fármacos , Ácido Fólico/administración & dosificación , Ácido Fólico/química , Ácido Fólico/farmacología , Hemólisis/efectos de los fármacos , Humanos , Masculino , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química , Albúmina Sérica Bovina/administración & dosificación , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/farmacología , Gemcitabina
18.
J Control Release ; 210: 134-46, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26003042

RESUMEN

Delivery and penetration of chemotherapeutic drugs into neoplasm through the tumor vasculature are essential mechanisms to enhance the efficiency of chemotherapy. "Vascular targeting" strategy focuses on promoting the infiltration of chemotherapeutic drugs into neoplastic tissues. In this study, we achieved a targeted therapy by coupling tumor necrosis factor α (TNFα) with TCP-1, a novel vascular-targeting peptide, in an orthotopic colorectal cancer model in mice. High dose of TCP-1-conjugated TNFα (TCP-1/TNFα: 5µg/mouse) displayed potent antitumor activity by inducing apoptosis and reducing microvessel number in tumors than unconjugated TNFα, with no evidence of increased toxicity. In the combined therapy, the antitumor action of 5-fluorouracil (5-FU) was potentiated when the mice were pretreated with a low dose of TNFα (1ng/mouse) and to a greater extent by the same concentration of TCP-1/TNFα. In this regard, TCP-1/TNFα combined with 5-FU synergistically inhibited the tumor growth, induced apoptosis and reduced cell proliferation. More importantly, TCP-1/TNFα normalized the tumor vasculature and facilitated the infiltration of immune cells to neoplasm as well as attenuated the immunosuppressing effects of TNFα in bone marrow and spleen. At the same time, TCP-1/TNFα significantly improved 5-FU absorption into the tumor mass. Taken together, these findings underscore the therapeutic potential of TCP-1 as a drug carrier in cancer therapy. TCP-1 is a novel vascular-targeting peptide and appears to be a promising agent for drug delivery. TCP-1 fused with TNFα holds great promise for colorectal cancer therapy.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Péptidos de Penetración Celular/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Fluorouracilo/administración & dosificación , Neovascularización Patológica/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/uso terapéutico , Médula Ósea/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/uso terapéutico , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Fluorouracilo/química , Fluorouracilo/uso terapéutico , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/química , Humanos , Masculino , Ratones Endogámicos BALB C , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Bazo/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/uso terapéutico
19.
ACS Appl Mater Interfaces ; 7(19): 10132-45, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25840122

RESUMEN

Herein, we prepare nanohybrids by incorporating iron oxide nanocubes (cubic-IONPs) within a thermoresponsive polymer shell that can act as drug carriers for doxorubicin(doxo). The cubic-shaped nanoparticles employed are at the interface between superparamagnetic and ferromagnetic behavior and have an exceptionally high specific absorption rate (SAR), but their functionalization is extremely challenging compared to bare superparamagnetic iron oxide nanoparticles as they strongly interact with each other. By conducting the polymer grafting reaction using reversible addition-fragmentation chain transfer (RAFT) polymerization in a viscous solvent medium, we have here developed a facile approach to decorate the nanocubes with stimuli-responsive polymers. When the thermoresponsive shell is composed of poly(N-isopropylacrylamide-co-polyethylene glycolmethyl ether acrylate), nanohybrids have a phase transition temperature, the lower critical solution temperature (LCST), above 37 °C in physiological conditions. Doxo loaded nanohybrids exhibited a negligible drug release below 37 °C but showed a consistent release of their cargo on demand by exploiting the capability of the nanocubes to generate heat under an alternating magnetic field (AMF). Moreover, the drug free nanocarrier does not exhibit cytotoxicity even when administered at high concentration of nanocubes (1g/L of iron) and internalized at high extent (260 pg of iron per cell). We have also implemented the synthesis protocol to decorate the surface of nanocubes with poly(vinylpyridine) polymer and thus prepare pH-responsive shell coated nanocubes.


Asunto(s)
Preparaciones de Acción Retardada/química , Doxorrubicina/administración & dosificación , Hipertermia Inducida/métodos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico , Neoplasias Experimentales/terapia , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Materiales Biocompatibles Revestidos/síntesis química , Terapia Combinada/métodos , Preparaciones de Acción Retardada/administración & dosificación , Difusión , Doxorrubicina/química , Células HeLa , Calor , Humanos , Nanopartículas de Magnetita/ultraestructura , Ensayo de Materiales , Neoplasias Experimentales/patología , Tamaño de la Partícula , Polímeros/química
20.
Colloids Surf B Biointerfaces ; 128: 498-505, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25794443

RESUMEN

We described a facile synthesis of pH and near-infrared (NIR) light dual-sensitive core/shell hybrid nanospheres, consisting of gold nanorods (GNR) as the core and poly(N-isopropylacrylamide-co-methacrylic acid) as the shell, p(NIPAM-MAA). The resultant GNR/p(NIPAM-MAA) nanospheres showed a core/shell structure, with an average diameter of ∼110nm and a strong longitudinal surface plasmon band at NIR region. Due to the photothermal effect of GNR and pH/thermal-sensitive volume transition of p(NIPAM-MAA) hydrogels, the nanospheres with loading of 5-fluorouracil (5-FU) by electrostatic interactions were developed as a smart carrier for pH- and photothermal-induced release of 5-FU. Experimental results testified that the cumulative release of 5-FU from nanospheres was markedly increased in a mild acidic medium. Moreover, a NIR light (808nm) irradiation triggered a greater and faster release of 5-FU, which was further testified by relevant results from in vitro cytotoxicity assay, in vivo tumor growth inhibition and histological images of ex vivo tumor sections. These results revealed significant applications of GNR/p(NIPAM-MAA) nanospheres in controlled release of anticancer agents and photothermal ablation therapy of tumor tissues, accompanied by synergistic effect of chem-photothermal therapy.


Asunto(s)
Acrilamidas/química , Antimetabolitos Antineoplásicos/farmacología , Portadores de Fármacos , Fluorouracilo/farmacología , Nanosferas/química , Nanotubos/química , Neoplasias/tratamiento farmacológico , Ácidos Polimetacrílicos/química , Animales , Antimetabolitos Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos , Femenino , Fibroblastos , Fluorouracilo/química , Oro/química , Humanos , Concentración de Iones de Hidrógeno , Hipertermia Inducida/métodos , Rayos Infrarrojos , Células MCF-7 , Ratones , Ratones Desnudos , Nanosferas/ultraestructura , Nanotubos/ultraestructura , Neoplasias/metabolismo , Neoplasias/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA