Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 18(12): 4531-4542, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34739255

RESUMEN

Having benefited from the combination of different therapeutic modalities, functionalized nanoplatforms with synergistic strategies have aroused great interest in anticancer treatment. Herein, an engineered, a biodegradable hollow mesoporous organosilica nanoparticle (HMON)-based nanoplatform was fabricated for photothermal-enhanced chemotherapy of tumor. For the first time, we demonstrated that HMONs could serve as nanocarriers for co-delivering of both the paclitaxel and photothermal agent new indocyanine green (IR820), denoted as Paclitaxel/IR820@ HMONs-PEG. The as-prepared nanosystem exhibited a high paclitaxel-loading capacity of 28.4%, much higher than most paclitaxel-loaded nanoformulations. Furthermore, incorporating thioether bonds (S-S) into the HMONs' framework endowed them with GSH-responsive biodegradation behavior, leading to the controllable release of drugs under a tumor reducing microenvironment, and hindered the premature release of paclitaxel. Upon being irradiated with an NIR laser, the obtained co-delivery nanosystem exhibited great photothermal properties generated from IR820. The fabricated nanocomposites could significantly suppress tumor growth under NIR laser irradiation, as validated by in vitro and in vivo assessments. Combined with outstanding biocompatibility, the constructed nanosystem holds great potential in combinational antitumor therapy.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas/química , Neoplasias Experimentales/tratamiento farmacológico , Compuestos de Organosilicio/química , Paclitaxel/química , Fototerapia/métodos , Animales , Liberación de Fármacos , Femenino , Glutatión/metabolismo , Hipertermia Inducida , Ratones , Ratones Endogámicos BALB C , Paclitaxel/farmacocinética , Paclitaxel/uso terapéutico , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nat Nanotechnol ; 16(7): 830-839, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33958764

RESUMEN

Nanoparticulate albumin bound paclitaxel (nab-paclitaxel, nab-PTX) is among the most widely prescribed nanomedicines in clinical use, yet it remains unclear how nanoformulation affects nab-PTX behaviour in the tumour microenvironment. Here, we quantified the biodistribution of the albumin carrier and its chemotherapeutic payload in optically cleared tumours of genetically engineered mouse models, and compared the behaviour of nab-PTX with other clinically relevant nanoparticles. We found that nab-PTX uptake is profoundly and distinctly affected by cancer-cell autonomous RAS signalling, and RAS/RAF/MEK/ERK inhibition blocked its selective delivery and efficacy. In contrast, a targeted screen revealed that IGF1R kinase inhibitors enhance uptake and efficacy of nab-PTX by mimicking glucose deprivation and promoting macropinocytosis via AMPK, a nutrient sensor in cells. This study thus shows how nanoparticulate albumin bound drug efficacy can be therapeutically improved by reprogramming nutrient signalling and enhancing macropinocytosis in cancer cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mutación , Nanopartículas , Neoplasias Experimentales/tratamiento farmacológico , Paclitaxel , Proteínas Proto-Oncogénicas p21(ras)/genética , Albúmina Sérica Humana , Animales , Línea Celular Tumoral , Glucosa/deficiencia , Glucosa/metabolismo , Humanos , Ratones , Ratones Transgénicos , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células RAW 264.7 , Albúmina Sérica Humana/química , Albúmina Sérica Humana/farmacología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
3.
Nanotechnology ; 32(32)2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-33946061

RESUMEN

Pterostilbene (PTE) is known as resveratrol of the next generation and it has attracted extensive attention in recent years. PTE can inhibit the growth of a variety of tumor cells. To overcome the problem of insolubility, PTE was loaded into nanoparticles (NPs) by anti-solvent precipitation technique using soybean lecithin (SPC) and D-α-tocopheryl polyethylene glycol succinate (TPGS) as stabilizers. The obtained PTE-NPs had an average particle size of 71.0 nm, a polydispersity index (PDI) value of 0.258, and a high zeta potential of -40.8 mV. PTE-NPs can maintain particle size stability in various physiological media. The entrapment efficiency of PTE-NPs was 98.24%. And the apparently water solubility of PTE-NPs was about 53 times higher than the solubility of PTE (54.41µg ml-1v-1s-1. 2.89 mg ml-1). M-1T-1T-1assay showed that the antitumor activity of PTE-NPs on 4T1 breast cancer cells, MCF-7 breast cancer cells and Hela cervical cancer cells was significantly increased by 4, 6 and 8 times than that of free PTE, respectively.In vivostudies have shown that PTE-NPs has a certain dose dependence. When injected intraperitoneally, PTE-NPs showed a similar therapeutic effect as paclitaxel injection (TIR was 57.53% versus 57.23%) against 4T1 tumor-bearing mice. This should be due to the improved bioavailability of the drug caused by nano-drug delivery system (nano-DDS). These results indicate that PTE-NPs may be a clinically promising anti-tumor drug for breast cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Nanopartículas/química , Paclitaxel/farmacología , Estilbenos/farmacología , Animales , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Neoplasias de la Mama/patología , Composición de Medicamentos/métodos , Femenino , Células HeLa , Humanos , Lecitinas/química , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Paclitaxel/farmacocinética , Tamaño de la Partícula , Solubilidad , Estilbenos/farmacocinética , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Vitamina E/química , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Int J Pharm ; 584: 119412, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32418898

RESUMEN

Nanofibers based transdermal drug delivery is a promising platform, and it effectively delivers the drug to tumor sites. The objective of the study was to fabricate stimuli-responsive polymeric nanofibers encapsulated with an active targeting micellar system for in situ drug delivery. Stimuli-responsive core-shell nanofibers release thedrug at target sites with minimum side effects to the other organs, decrease the drug administration concentration. Initially, we prepared CA conjugated PCPP polymeric micelles loaded with PTX. Then, core-shell nanofibers were prepared using PHM with coaxial electrospinning and distinct core-shell nanofibers formation confirm by SEM and TEM. Nanofibers showed a homogenous distribution of micelles inside the fiber mesh, diffusion, and erosion processes lead to a controlled release of PTX.In vitro drug release and swelling, revealed the pH based sustained release of the drug for 180 h from the nanofibers mat. Functional and stimuli-responsive nanofibers highly absorb H+ ions and repulsion of cations promoting maximum swelling to release more drugs in acidic pH. An increased transportation rate of 70% drug release through epidermis for 120 h. Nanofibers effectively internalize to the skin, and it confirmed by confocal microscopy. MCF-7 cells grown and spread over the nanofibers, which show the biocompatibility of nanofibers. Compared to PTX, drug-loaded nanofibers exhibited higher cytotoxicity for 8 days which was confirmed by the flow cytometry. These promising results confirm, the novel stimuli-responsive core-shell nanofibers actively target breast cancer cells and lead the way to safe cancer therapy.


Asunto(s)
Portadores de Fármacos/farmacocinética , Epidermis/metabolismo , Micelas , Nanofibras/química , Paclitaxel/farmacocinética , Animales , Neoplasias de la Mama/tratamiento farmacológico , Supervivencia Celular , Ácido Cólico/química , Preparaciones de Acción Retardada , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Liberación de Fármacos , Fibroblastos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Ratones , Microscopía Electroquímica de Rastreo , Paclitaxel/administración & dosificación , Polímeros/química , Psyllium/química , Absorción Subcutánea , Porcinos
5.
Int J Nanomedicine ; 15: 2337-2349, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32308385

RESUMEN

PURPOSE: The aim of this study was to develop an "all-in-one" nanoplatform that integrates at the second near-infrared (NIR-II) region dye IR1061 and anticancer drug paclitaxel (PTX) into an apoferritin (AFN) nanocage (IR-AFN@PTX). Simultaneously, folic acid (FA), tumor target molecule,  was conjugated onto IR-AFN@PTX to be IR-AFN@PTX-FA for tumor-targeted and pH/NIR-II-triggered synergistic photothermal-chemotherapy. METHODS: IR1061 was firstly reacted with PEG and then conjugated with AFN to be IR-AFN. Then, FA was conjugated onto the surface of IR-AFN to be IR-AFN-FA. At last, PTX was incorporated into IR-AFN-FA to fabricate a nanoplatform IR-AFN@PTX-FA. The NIR-II photothermal properties and pH/NIR-II triggered drug release were evaluated. The ability of IR-AFN@PTX-FA to target tumors was estimated using optical bioluminescence. In vitro and in vivo synergistic therapeutic effects of pH/NIR-II-triggered and tumor-targeted photothermal-chemotherapy were investigated in 4T1 tumor model. RESULTS: IR-AFN@PTX-FA showed excellent water solubility and physiological stability, which significantly enhanced the solubility of both IR1061 and PTX. After 5 min of laser irradiation at 1064 nm, IR-AFN@PTX-FA exhibited an effective photothermal effect compared with laser irradiation at 808 nm, even when blocked with 0.6 cm thick chicken breast. Cellular uptake experiments showed IR-AFN@PTX-FA utilized clathrin-mediated and caveolae-mediated endocytosis pathways to enter 4T1 cells, and was then delivered by the endosome to the lysosome. NIR-II laser irradiation and pH could synergistically trigger PTX release, inducing significant tumor inhibition in vitro and in vivo. CONCLUSION: As a novel "all-in-one" nanoplatform, IR-AFN@PTX-FA was found to selectively target tumors and showed very efficient NIR-II photothermal effects and pH/NIR-II triggered drug release effects, showing a remarkable, synergistic photothermal-chemotherapy effect.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Boratos/química , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/administración & dosificación , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Piranos/química , Animales , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/farmacología , Apoferritinas/química , Línea Celular Tumoral , Colorantes/química , Liberación de Fármacos , Ácido Fólico/química , Ácido Fólico/farmacología , Humanos , Concentración de Iones de Hidrógeno , Rayos Láser , Ratones Endogámicos BALB C , Nanopartículas/química , Paclitaxel/farmacocinética , Fototerapia/métodos , Solubilidad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mater Sci Eng C Mater Biol Appl ; 109: 110576, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32228957

RESUMEN

Paclitaxel loaded lipid-polymer nanoparticles (NPs) were successfully synthesized using poly lactide-co-glycolide (PLGA) as polymer and stearyl amine, soya lecithin as lipids via single step nanoprecipitation method. The study was aimed to combine the advantage of structural integrity of hybrid NPs containing PLGA core and lipid in the shell. Surfactants such as polyvinyl alcohol (PVA), tocopheryl polyethylene glycol succinate (TPGS), pluronic 68 (F68) and human serum albumin (HSA) were used as stabilizers. NPs were characterized w.r.t. morphology, particle size, zeta potential, encapsulation efficiency, in vitro drug release, protein binding capability and blood compatibility. NPs were in size range of 150-400 nm and the particle size was greatly influenced by type and concentration of surfactants and lipids. TEM analysis confirmed the spherical shape and coating of the lipid on the NPs surface. Highest percentage entrapment efficiency was observed in NPs prepared with HSA as surfactant. The release rate of paclitaxel from modified NPs was much slower as compared to unmodified NPs. The percent protein binding of P-PVA, P-TPGS, P-F68 and P-HSA (unmodified NPs) was found to be 15.11%, 16.27%, 27.90% and 33.72%, respectively demonstrating effect of surface properties of NPs on protein binding. The hemolytic activity of the NPs was found to be dependent on type of surfactant and not on the lipid employed. PVA, TPGS, F68, HSA surfactants showed ~16%, ~10%, ~13%, ~7% hemolysis rate, respectively. The surface nature of NPs had a significant effect on the circulation profile of formulations. The HSA based NPs showed prolonged blood circulation time when compared to NPs without lipid coating. Thus, the synthesized dual lipid coated PLGA NPs with HSA could act as a potential nano-system for controlled delivery of paclitaxel.


Asunto(s)
Aminas , Portadores de Fármacos , Lecitinas , Nanopartículas , Paclitaxel , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Aminas/química , Aminas/farmacocinética , Aminas/farmacología , Animales , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Lecitinas/química , Lecitinas/farmacocinética , Lecitinas/farmacología , Masculino , Nanopartículas/química , Nanopartículas/uso terapéutico , Paclitaxel/química , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacocinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología , Ratas , Ratas Wistar , Albúmina Sérica Humana/química , Tensoactivos/química
7.
Theranostics ; 10(6): 2621-2630, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194824

RESUMEN

The development of improved or targeted drugs that discriminate between normal and tumor tissues is the key therapeutic issue in cancer research. However, the development of an analytical method with a high accuracy and sensitivity to achieve quantitative assessment of the tumor targeting of anticancer drugs and even intratumor heterogeneous distribution of these drugs at the early stages of drug research and development is a major challenge. Mass spectrometry imaging is a label-free molecular imaging technique that provides spatial-temporal information on the distribution of drugs and metabolites in organisms, and its application in the field of pharmaceutical development is rapidly increasing. Methods: The study presented here accurately quantified the distribution of paclitaxel (PTX) and its prodrug (PTX-R) in whole-body animal sections based on the virtual calibration quantitative mass spectrometry imaging (VC-QMSI) method, which is label-free and does not require internal standards, and then applied this technique to evaluate the tumor targeting efficiency in three treatment groups-the PTX-injection treatment group, PTX-liposome treatment group and PTX-R treatment group-in nude mice bearing subcutaneous A549 xenograft tumors. Results: These results indicated that PTX was widely distributed in multiple organs throughout the dosed body in the PTX-injection group and the PTX-liposome group. Notably, in the PTX-R group, both the prodrug and metabolized PTX were mainly distributed in the tumor tissue, and this group showed a significant difference compared with the PTX-liposome group, the relative targeting efficiency of PTX-R group was increased approximately 50-fold, leading to substantially decreased systemic toxicities. In addition, PTX-R showed a significant and specific accumulation in the poorly differentiated intratumor area and necrotic area. Conclusion: This method was demonstrated to be a reliable, feasible and easy-to-implement strategy to quantitatively map the absorption, distribution, metabolism and excretion (ADME) of a drug in the whole-body and tissue microregions and could therefore evaluate the tumor-targeting efficiency of anticancer drugs to predict drug efficacy and safety and provide key insights into drug disposition and mechanisms of action and resistance. Thus, this strategy could significantly facilitate the design and optimization of drugs at the early stage of drug research and development.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias/tratamiento farmacológico , Paclitaxel/farmacocinética , Profármacos/farmacocinética , Células A549 , Animales , Antineoplásicos/administración & dosificación , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Paclitaxel/administración & dosificación , Profármacos/administración & dosificación
8.
Eur J Drug Metab Pharmacokinet ; 45(2): 257-264, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31820303

RESUMEN

BACKGROUND AND OBJECTIVES: Licorice is the dried roots and rhizomes of Glycyrrhiza uralensis Fisch (Leguminosae), which is often used with paclitaxel to alleviate paclitaxel-induced pain in clinics. However, the herb-drug interaction between licorice and paclitaxel is still unknown. Our study evaluates the effects of oral licorice on the paclitaxel in rats via pharmacokinetic studies. METHODS: A simple and rapid ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method was developed to determine paclitaxel in rat. SD rats were randomly divided into 3 groups of 6 animals each as follows: two groups of rats that were pretreated with a daily gavage of licorice (3 g/kg) for 1 or 14 successive days; Control group that was administered distilled water. All rats were then intravenously administered with paclitaxel (3 mg/kg). RESULTS: The results showed that 14 days pretreatment of licorice could decrease the area under the curve (AUC0-t) (from 7483.08 ± 528.78 to 6679.12 ± 266.56 mg/L × h) (P < 0.01), and increase the total clearance (CL) (from 0.36 ± 0.02 to 0.39 ± 0.02 L/h/kg) of paclitaxel (P < 0.01). However, a single co-administration of licorice did not significantly alter the pharmacokinetic parameters of paclitaxel, such as AUC0-t (from 7483.08 ± 528.78 to 7201.24 ± 292.76 mg/L × h) (P > 0.05) and CL (from 0.36 ± 0.02 to 0.36 ± 0.01 L/h/kg) (P > 0.05). CONCLUSIONS: The results will contribute to better use of licorice in the adjunctive therapy and provide information to study the interaction between herbs and chemotherapy.


Asunto(s)
Glycyrrhiza/química , Interacciones de Hierba-Droga , Paclitaxel/farmacocinética , Extractos Vegetales/farmacología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/análisis , Antineoplásicos Fitogénicos/farmacocinética , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Masculino , Paclitaxel/administración & dosificación , Paclitaxel/análisis , Extractos Vegetales/administración & dosificación , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem
9.
Phytomedicine ; 67: 153141, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31884406

RESUMEN

BACKGROUND: Nobiletin (N), a polymethoxylated flavone from citrus fruits, enhanced anti-cancer effects of paclitaxel (PTX) in multi-drug resistance (MDR) cancer cells via inhibiting P-glycoprotein (P-gp) in our previous report. But the in vivo chemo-sensitizing effect of nobiletin is unknown. Moreover, considering the nonlinear pharmacokinetics and narrow therapeutic window of PTX, drug-drug interaction should be explored for using nobiletin with PTX together. PURPOSE: In this study, we wanted to explore whether nobiletin could affect the pharmacokinetic (PK) behavior of PTX and reverse drug resistance in vivo as well as the corresponding mechanisms. STUDY DESIGN AND METHODS: Accurate and sensitive UPLC-MS/MS method was developed for the detection of PTX, and was applied to the pharmacokinetic study in rats. In vivo anti-MDR tumor study was carried out with A549/T xenograft nude mice model. Immunohistochemistry and western blot analysis were used for evaluating the levels of P-gp, Nrf2, and AKT/ERK pathways in MDR tumors. RESULTS: Nobiletin significantly enhanced the therapeutic effects of PTX, and inhibited the MDR tumor sizes in the A549/T xenograft model, while PTX or nobiletin alone did not. We found that nobiletin increased the PTX concentrations in tumor tissues but did not affect the PK behavior of PTX. Notably, Nrf2 and phosphorylation of AKT/ERK expression in MDR tumor tissues were significantly inhibited by giving nobiletin and PTX together. However, nobiletin did not affect the expression of P-gp. CONCLUSION: Nobiletin reversed PTX resistance in MDR tumor via increasing the PTX content in the MDR tumor and inhibiting AKT/ERK/Nrf2 pathways, but without affecting the systematic exposure of PTX, indicating that nobiletin may be an effective and safe MDR tumor reversal agent.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Flavonas/farmacocinética , Paclitaxel/farmacocinética , Células A549 , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Cromatografía Liquida , Flavonas/administración & dosificación , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Desnudos , Paclitaxel/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Sci Rep ; 9(1): 14881, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31619730

RESUMEN

Patients with advanced ovarian cancer develop recurrence despite initial treatment response to standard treatment of surgery and intravenous/intraperitoneal (IP) chemotherapy, partly due to a limited peritoneal exposure time of chemotherapeutics. Paclitaxel-loaded genipin-crosslinked gelatin microspheres (PTX-GP-MS) are evaluated for the treatment of microscopic peritoneal carcinomatosis and prevention of recurrent disease. The highest drug load (39.2 µg PTX/mg MS) was obtained by immersion of GP-MS in aqueous PTX nanosuspension (PTXnano-GP-MS) instead of ethanolic PTX solution (PTXEtOH-GP-MS). PTX release from PTX-GP-MS was prolonged. PTXnano-GP-MS displayed a more controlled release compared to a biphasic release from PTXEtOH-GP-MS. Anticancer efficacy of IP PTX-GP-MS (PTXEtOH-GP-MS, D = 7.5 mg PTX/kg; PTXnano-GP-MS D = 7.5 and 35 mg PTX/kg), IP nanoparticular albumin-bound PTX (D = 35 mg PTX/kg) and controls (0.9% NaCl, blank GP-MS) was evaluated in a microscopic peritoneal carcinomatosis xenograft mouse model. PTXnano-GP-MS showed superior anticancer efficacy with significant increased survival time, decreased peritoneal carcinomatosis index score and ascites incidence. However, prolonged PTX release over 14 days from PTXnano-GP-MS caused drug-related toxicity in 27% of high-dosed PTXnano-GP-MS-treated mice. Dose simulations for PTXnano-GP-MS demonstrated an optimal survival without drug-induced toxicity in a range of 7.5-15 mg PTX/kg. Low-dosed PTXnano-GP-MS can be a promising IP drug delivery system to prevent recurrent ovarian cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinoma Epitelial de Ovario/terapia , Portadores de Fármacos , Gelatina/química , Ovario/efectos de los fármacos , Paclitaxel/farmacología , Neoplasias Peritoneales/prevención & control , Abdomen/patología , Animales , Antineoplásicos Fitogénicos/farmacocinética , Carcinoma Epitelial de Ovario/mortalidad , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos/métodos , Evaluación Preclínica de Medicamentos , Liberación de Fármacos , Femenino , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones , Ratones Desnudos , Microesferas , Ovario/patología , Paclitaxel/farmacocinética , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/patología , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Biomater Sci ; 7(10): 4060-4074, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31475710

RESUMEN

Combined photothermal-chemotherapy guided by multimodal imaging is a promising strategy for cancer diagnosis and treatment. Multifunctional nanoparticles, such as those comprising organic and inorganic compounds, have been extensively investigated for combined photothermal-chemotherapy; however, their application is still limited by their potential long-term toxicity and lack of contrast properties. To solve these problems, in this study, a new type of multifunctional nanoparticle for combined photothermal-chemotherapy guided by dual-modality imaging was prepared with endogenous melanin by multistep emulsification to enhance tumor ablation. The nanoparticles were coated with poly(lactide-co-glycolic acid) (PLGA) and loaded with paclitaxel (PTX), encapsulated melanin and perfluoropentane (PFP). The materials in the nanoparticles were endogenous, ensuring high stability, biocompatibility, and biosafety. Nanoparticles irradiated with a laser, which induced their phase transformation into microbubbles, exhibited high photothermal conversion efficiency, thereby achieving photoacoustic (PA)/ultrasound (US) dual-modality imaging to determine tumor location, boundary, and size and to monitor drug distribution. Furthermore, optical droplet vaporization (ODV) of the nanoparticles could trigger the release of PTX; thus, these nanoparticles are a useful drug carrier. In vivo and in vitro experiments revealed that a strong synergistic antitumor effect was achieved by combining the photothermal properties of the nanoparticles with a chemotherapy drug. Importantly, the cavitation, thermoelastic expansion, and sonoporation caused by the phase transformation of the nanoparticles could directly damage the tumors. These processes also promoted the release, penetration and absorption of the drug, further enhancing the effect of combined photothermal-chemotherapy on tumor suppression. Therefore, the multifunctional nanoparticles prepared in this study provide a new strategy of using endogenous materials for controlled near-infrared (NIR)-responsive drug release and combined photothermal-chemotherapy guided by multimodal imaging.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Fluorocarburos/administración & dosificación , Melaninas/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias/terapia , Paclitaxel/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Animales , Antineoplásicos Fitogénicos/farmacocinética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Preparaciones de Acción Retardada/administración & dosificación , Femenino , Fluorocarburos/farmacocinética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Melaninas/farmacocinética , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Paclitaxel/farmacocinética , Técnicas Fotoacústicas , Fototerapia , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacocinética , Distribución Tisular , Ultrasonografía
12.
J Pharm Biomed Anal ; 174: 728-733, 2019 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-31299453

RESUMEN

Xiao-Ai-Ping injection (XAP) has been shown to be clinically effective in treatment of gastric carcinoma, liver cancer and lung cancer, when it was combined with anticancer drug paclitaxel (PTX). To analyze the effect of XAP on the pharmacokinetics of PTX, a liquid chromatography-tandem mass spectroscopy (LCMS/MS) assay method was developed and validated to quantify PTX simultaneously and its main metabolite 3'-p-hydroxypaclitaxel (C3'-OHP) in rat plasma. PTX and C3'-OHP were quantified using positive MRM mode. The analysis method was validated for specificity, recovery, carry-over, accuracy, precision, sample stability and dilution integrity under various storage conditions. The pharmacokinetic parameters were determined in rats after tail intravenous administration of 6 mg/mL PTX in the absence (control group) or presence of intraperitoneal administration of 10 mL/kg、20 mL/kg XAP (study groups). Compared to control group, the area under the plasma concentration-time curve (AUC) of PTX and C3'-OHP in study groups increased significantly following consecutive administration with XAP for 10 days. In conclusion, pretreatment with XAP enhanced the exposure of PTX and C3'-OHP. There would be herb-drug interaction happening between XAP and PTX in rats.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Interacciones de Hierba-Droga , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/sangre , Antineoplásicos Fitogénicos/farmacocinética , Calibración , Cromatografía Liquida , Femenino , Modelos Lineales , Paclitaxel/sangre , Control de Calidad , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem
13.
Artículo en Inglés | MEDLINE | ID: mdl-31276955

RESUMEN

In spite of having a remarkable anti tumor activity against a wide variety of cancers, the clinical effectiveness of the major chemotherapeutic drug paclitaxel is often limited by the issues of drug resistance that hampers the therapeutic effectiveness of the drug. The combination of proton pump inhibitor with paclitaxel is an effective approach to overcome therapeutic resistance caused by the acidic microenvironment (Warburg effect) in tumor. In the present study a new simple, precise and selective liquid chromatography tandem mass spectrometry method was developed for quantification of paclitaxel and lansoprazole using esomeprazole as an internal standard and applied for the pharmacokinetic study of investigational paclitaxel - lansoprazole loaded PLGA nanoparticles. The developed method quantifies both the drugs simultaneously irrespective of their dissimilar stability concerns. The detection was exercised with multiple reaction-monitoring mode in positive ionization that yielded highly intense response of parent-product (m/z) transition pair 854.4 → 286.1, 370.1 → 251.9 and 346 → 198 for paclitaxel, lansoprazole and Esomeprazole respectively. The chromatographic separation was achieved using phenomenex Kinetex 5 µ C18 100A 50 × 3.0 mm column and a gradient mobile phase combination of ammonium acetate in deionized water (pH 6.8, 2 mM, w/v) and acetonitrile spiked with formic acid (1:1000, v/v ). This method showed good linearity over a concentration range of 10-320 ng/mL and 100-3200 ng/mL with correlation coefficient (R2) 0.98 and 0.94 for paclitaxel and lansoprazole respectively. Using liquid liquid extraction process both the drugs were extracted from rat plasma. The intra- and inter-day precision and accuracy values were within the variability limits and both the analytes were found to be stable throughout the freeze-thaw, auto-sampler, bench top and long term stability studies. The liquid chromatography tandem mass spectrometry method was successfully validated in accordance with United States Food and Drug administration guidelines and the results were within the acceptable limits. The liquid chromatography tandem mass spectrometry method was successfully utilized for the pharmacokinetic investigation of experimental paclitaxel - lansoprazole loaded PLGA nanoparticles in rat plasma.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Lansoprazol/sangre , Paclitaxel/sangre , Espectrometría de Masas en Tándem/métodos , Animales , Composición de Medicamentos/métodos , Evaluación Preclínica de Medicamentos , Femenino , Lansoprazol/química , Lansoprazol/farmacocinética , Masculino , Paclitaxel/química , Paclitaxel/farmacocinética , Ratas
14.
Theranostics ; 9(9): 2505-2525, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31131050

RESUMEN

The delivery of therapeutics into tumors remains a challenge in nanoparticle-mediated drug delivery. However, effective therapies such as photothermal therapy (PTT) are limited by quick systemic clearance and non-specific biodistribution. Anti-tumor strategies tailored to accommodate both tumor accumulation/retention and cellular internalization under a single platform would be a promising strategy. This work demonstrates a hierarchical activating strategy that would exhibit enhanced circulation and rapid tumor-tropism as well as facilitate tumor penetration, followed by tumor-specific drug release to realize trackable photothermal/chemotherapy. Methods: We engineered a lithocholic acid-conjugated disulfide-linked polyethyleneimine micelle (LAPMi) loaded with paclitaxel (LAPMi-PTX, L), followed by the electrostatic adsorption of indocyanine green (ICG, I) on LAPMI-PTX and subsequently coated them with thermosensitive DPPC and DSPE-PEG-NH2 lipids (L), producing Lipid/ICG/LAPMi-PTX (LIL-PTX) nanoparticles (NPs). The characteristics of NPs, including physicochemical characterization, photothermal & pH responsiveness, cell uptake, tumor spheroid penetration, anti-tumor efficacy and hierarchical activation of LIL-PTX NPs were investigated in vitro and in vivo by using CT26 cell line. The anti-metastatic potential of LIL-PTX NPs were demonstrated using 4T1 orthotopic tumor model. Results: The NPs synthesized possessed charge switchability in the mildly acidic pH, and were laser- and pH-responsive. Dual stimuli-responsive nature of LIL-PTX NPs improved the disposition of therapeutics to the tumor, reflected by enhanced intracellular uptake, tumor spheroid penetration and in vitro cytotoxicity studies. LIL-PTX NPs readily switched its surface charge from neutral to positive upon reaching the tumor milieu, thus resulting in rapid tumor tropism and accumulation. Under near-infrared laser irradiation, the thermosensitive lipids on LIL-PTX NPs were deshielded, and the tumor-penetrating LAPMi-PTX was subsequently exposed to the tumor milieu, thus resulting in enhanced intracellular internalization. Next, LAPMi-PTX evaded the endo-lysosomes, thereby releasing the PTX through the degradation of LAPMi mediated by intracellular GSH in the tumor. LIL-PTX NPs significantly improved the therapy by eradicating primary tumors completely and suppressing their subsequent lung metastasis. Conclusion: The improved therapeutic index is due to enhanced passive targeting by rapid tumor-tropic accumulation and tumor penetration by laser-driven exposure of LAPMi, thereby improving the therapeutic delivery for image-guided photothermal/chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Quimioterapia/métodos , Hipertermia Inducida/métodos , Neoplasias Experimentales/terapia , Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Ratones Endogámicos BALB C , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Radioterapia Guiada por Imagen/métodos , Resultado del Tratamiento
15.
AAPS PharmSciTech ; 20(5): 198, 2019 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127389

RESUMEN

In this work, several normal, oil-in-water (o/w) microemulsions (MEs) were prepared using peppermint essential oil, jojoba oil, trans-anethole, and vitamin E as oil phases to test their capacity to load paclitaxel (PTX). Initially, pseudo-ternary partial phase diagrams were constructed in order to find the normal microemulsion region using d-α-tocopherol polyethylene glycol 1000 succinate (TPGS-1000) as surfactant and isobutanol (iso-BuOH) as co-surfactant. Selected ME formulations were loaded with PTX reaching concentrations of 0.6 mg mL-1 for the peppermint oil and trans-anethole MEs, while for the vitamin E and jojoba oil MEs, the maximum concentration was 0.3 mg mL-1. The PTX-loaded MEs were stable according to the results of heating-cooling cycles and mechanical force (centrifugation) test. Particularly, drug release profile for the PTX-loaded peppermint oil ME (MEPP) showed that ∼ 90% of drug was released in the first 48 h. Also, MEPP formulation showed 70% and 90% viability reduction on human cervical cancer (HeLa) cells after 24 and 48 h of exposure, respectively. In addition, HeLa cell apoptosis was confirmed by measuring caspase activity and DNA fragmentation. Results showed that the MEPP sample presented a major pro-apoptotic capability by comparing with the unloaded PTX ME sample.


Asunto(s)
Antineoplásicos Fitogénicos/síntesis química , Apoptosis/efectos de los fármacos , Citotoxinas/síntesis química , Nanosferas/química , Paclitaxel/síntesis química , Aceites de Plantas/síntesis química , Antineoplásicos Fitogénicos/farmacocinética , Apoptosis/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citotoxinas/farmacocinética , Relación Dosis-Respuesta a Droga , Liberación de Fármacos , Células HeLa , Humanos , Mentha piperita , Paclitaxel/farmacocinética , Aceites de Plantas/farmacocinética , Polietilenglicoles/síntesis química , Polietilenglicoles/farmacocinética , Tensoactivos/síntesis química , Tensoactivos/farmacocinética , Vitamina E/síntesis química , Vitamina E/farmacocinética
16.
Nanoscale ; 11(12): 5474-5488, 2019 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-30855625

RESUMEN

Clinical chemotherapy for cancer is limited by the physiological barrier of tumors, resulting in low drug delivery to tumors, poor efficacy of drugs and inability to block tumor metastasis. Here we developed an intelligent switchable nitric oxide (NO)-releasing nanoparticle, IPH-NO, which loads a photosensitizer (IR780) and the chemotherapy drug paclitaxel (PTX) into NO donor-S-nitrosated human serum albumin (HSA-NO). NO exhibits two effects based on its concentration: enhancement of chemotherapy by increasing the enhanced permeability and retention (EPR) effect at low concentrations and direct killing of cancer cells at high concentrations. IPH-NO can slowly release NO in the presence of glutathione to boost tumor vascular permeability and improve drug accumulation. Near-infrared light irradiation was utilized to induce a quick release of NO that can directly kill cancer cells at high concentrations. This combination of phototherapy and NO gas therapy activated by NIR together with chemotherapy showed significant effects in tumor inhibition. Furthermore, IPH-NO blocked tumor metastasis by inhibiting epithelial mesenchymal transition. PH-NO provides a novel strategy to control NO release at tumor site for drug accumulation and combination therapies, consequently potentiating the anticancer efficacy and inhibiting tumor metastasis.


Asunto(s)
Nanomedicina , Nanopartículas/química , Óxido Nítrico/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glutatión/química , Semivida , Humanos , Hipotermia Inducida , Rayos Infrarrojos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Paclitaxel/química , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Albúmina Sérica Humana/química , Oxígeno Singlete/análisis
17.
Clin Transl Oncol ; 21(9): 1260-1269, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30761508

RESUMEN

BACKGROUND: In some patients with peritoneal carcinomatosis, we could perform the cytoreductive surgery and the HIPEC procedure by a complete laparoscopic approach to avoid morbidity. We consider that using laparoscopic approach for performing peritoneal carcinomatosis cytoreductive surgery and HIPEC with closed CO2 recirculation technique is possible and safe, with equal efficacy to conventional methods and hemodynamic complications. OBJECTIVE: Monitoring the effectiveness of the drug distribution in a laparoscopic ctoreductive and HIPEC surgery group with CO2 recirculation respect to a closed and open HIPEC group METHODS: Porcine model that included fifteen mini-pigs. Five pigs were operated with laparoscopic approach performing a pelvic and retroperitoneal lymphadenectomy. They later received a total laparoscopic closed HIPEC with CO2 recirculation (G1). Group 2 (G2): five pigs operated by an open cytoreductive surgery and closed HIPEC technique. Group 3 (G3): five animals in which an open cytoreductive surgery and an open HIPEC technique was performed. Blood and peritoneal determinations were realized after recirculation of the drug, at 60 min using chromatographic analysis. RESULTS: G1-G2: phrenic right peritoneum, p: 0.46. Phrenic left peritoneum, p: 0.46. Pelvic peritoneum, p: 0.17. Serum paclitaxel: p: 0.01. G1-G3: phrenic right peritoneum, p: 0.34. Phrenic left peritoneum, p: 0.34. Pelvic peritoneum, p: 0.17. Serum paclitaxel G1-G3, p: 0.02. CONCLUSIONS: A total laparoscopic approach for ctoreductive surgery and closed HIPEC with CO2 recirculation may be safe and feasible. In our experimental model there was no significant difference in tissue drug distribution respect the conventional techniques and there was a less toxicity because the serum drug concentration was significantly lower with laparoscopic approach respect the other groups.


Asunto(s)
Quimioterapia del Cáncer por Perfusión Regional , Procedimientos Quirúrgicos de Citorreducción/métodos , Hipertermia Inducida , Laparoscopía/métodos , Paclitaxel/administración & dosificación , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/terapia , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Terapia Combinada , Femenino , Escisión del Ganglio Linfático/métodos , Paclitaxel/farmacocinética , Neoplasias Pélvicas/metabolismo , Neoplasias Pélvicas/patología , Neoplasias Pélvicas/terapia , Neoplasias Peritoneales/patología , Porcinos , Distribución Tisular
18.
ACS Appl Mater Interfaces ; 10(45): 38700-38714, 2018 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-30360090

RESUMEN

Photothermal therapy is a promising approach for antitumor application although regrettably restricted by available photothermal agents. Physical entrapment of organic near-infrared dyes into nanosystems was extensively studied to reverse the dilemma. However, problems still remained, such as drug bursting and leakage. We developed here an amphiphilic prodrug conjugate by chemically modifying indocyanine green derivative (ICG-COOH) and paclitaxel (PTX) to hyaluronic acid (HA) backbone for integration of photothermal-chemotherapy and specific tumor imaging. The prepared ICG-HA-PTX conjugates could self-assemble into nanomicelles to improve the stability and reduce systemic toxicity of the therapeutic agents. The high local concentration of ICG-COOH in nanomicelles resulted in fluorescence self-quenching, leading to no fluorescence signal being detected in circulation. When the nanomicelles reached the tumor site via electron paramagnetic resonance effect and HA-mediated active targeting, the overexpressed esterase in tumor cells ruptured the ester linkage between drugs and HA, achieving tumor-targeted therapy and specific imaging. A series of in vitro and in vivo experiments demonstrated that the easily prepared ICG- HA-PTX nanomicelles with high stability, smart release behavio r, and excellent tumor targeting ability showed formidable synergy in tumor inhibition, which provided new thoughts in developing an organic near-infrared-dye-based multifunctional delivery system for tumor theranostics.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Hipertermia Inducida/métodos , Verde de Indocianina/administración & dosificación , Nanoestructuras/administración & dosificación , Paclitaxel/administración & dosificación , Fototerapia/métodos , Profármacos/administración & dosificación , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Verde de Indocianina/química , Verde de Indocianina/farmacocinética , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Micelas , Células 3T3 NIH , Nanoestructuras/química , Imagen Óptica/métodos , Paclitaxel/química , Paclitaxel/farmacocinética , Profármacos/química , Profármacos/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mater Sci Eng C Mater Biol Appl ; 92: 547-553, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184781

RESUMEN

We developed a magnetic solid lipid nanoparticles formulation of paclitaxel (PTX-loaded MSLNs) via emulsification-diffusion method. The physicochemical characterization of PTX-loaded MSLNs was performed by AFM, DLS, determination of entrapment efficiency (EE) and drug loading (DL), DSC, VSM, and physical stability. The in vitro effect of temperature and pulsed magnetic hyperthermia on drug release were studied. PTX-loaded MSLNs had a particle diameter around 250 nm with a narrow size distribution, spherical morphology, EE of 67.3 ±â€¯1.2% and a DL of 17.1 ±â€¯0.4 µg/mg. A decrease of the melting point of the lipid was observed following the preparation of the MSLNs. A threefold increase in the in vitro drug release rate was seen when temperature was raised from 25 to 43 °C. The lipid coating of MPs confer a temperature-dependent drug release and magnetic hyperthermia was used to trigger controlled PTX release from MSLNs.


Asunto(s)
Hipertermia Inducida , Lípidos/química , Campos Magnéticos , Nanopartículas/análisis , Paclitaxel , Paclitaxel/química , Paclitaxel/farmacocinética
20.
Gynecol Oncol ; 150(3): 478-486, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30068487

RESUMEN

PURPOSE: Preclinical studies performed in our laboratory have shown that high-dose selenium inhibits the development of carboplatin drug resistance in an ovarian cancer mouse xenograft model. Based on these data, as well as the potential serious toxicities of supranutritional doses of selenium, a phase I trial of a combination of selenium/carboplatin/paclitaxel was designed to determine the maximum tolerated dose, safety, and effects of selenium on carboplatin pharmacokinetics in the treatment of chemo-naive women with gynecologic cancers. Correlative studies were performed to identify gene targets of selenium. METHODS: Chemo-naïve patients with gynecologic malignancy received selenious acid IV on day 1 followed by carboplatin IV and paclitaxel IV on day 3. A standard 3 + 3 dose-escalating design was used for addition of selenium to standard dose chemotherapy. Concentrations of selenium in plasma and carboplatin in plasma ultrafiltrate were analyzed. RESULTS: Forty-five patients were enrolled and 291 treatment cycles were administered. Selenium was administered as selenious acid to 9 cohorts of patients with selenium doses ranging from 50 µg to 5000 µg. Grade 3/4 toxicities included neutropenia (66.7%), febrile neutropenia (2.2%), pain (20.0%), infection (13.3%), neurologic (11.1%), and pulmonary adverse effects (11.1%). The maximum tolerated dose of selenium was not reached. Selenium had no effect on carboplatin pharmacokinetics. Correlative studies showed post-treatment downregulation of RAD51AP1, a protein involved in DNA repair, in both cancer cell lines and patient tumors. CONCLUSION: Overall, the addition of selenium to carboplatin/paclitaxel chemotherapy is safe and well tolerated, and does not alter carboplatin pharmacokinetics. A 5000 µg dose of elemental selenium as selenious acid is suggested as the dose to be evaluated in a phase II trial.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carboplatino/farmacocinética , Línea Celular Tumoral , Neutropenia Febril Inducida por Quimioterapia/etiología , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/sangre , Humanos , Infecciones/inducido químicamente , Enfermedades Pulmonares/inducido químicamente , Dosis Máxima Tolerada , Persona de Mediana Edad , Enfermedades del Sistema Nervioso/inducido químicamente , Paclitaxel/administración & dosificación , Paclitaxel/farmacocinética , Proteínas de Unión al ARN , Criterios de Evaluación de Respuesta en Tumores Sólidos , Ácido Selenioso/administración & dosificación , Ácido Selenioso/farmacocinética , Selenio/sangre , Selenoproteína P/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA