Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharm Pharm Sci ; 27: 12398, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38577255

RESUMEN

Bioequivalence (BE) studies are considered the standard for demonstrating that the performance of a generic drug product in the human body is sufficiently similar to that of its comparator product. The objective of this article is to describe the recommendations from participating Bioequivalence Working Group for Generics (BEWGG) members of the International Pharmaceutical Regulators Programme (IPRP) regarding the conduct and acceptance criteria for BE studies of immediate release solid oral dosage forms. A survey was conducted among BEWGG members regarding their BE recommendations and requirements related to study subjects, study design, sample size, single or multiple dose administration, study conditions (fasting or fed), analyte to be measured, selection of product strength, drug content, handling of endogenous substances, BE acceptance criteria, and additional design aspects. All members prefer conducting single dose cross-over designed studies in healthy subjects with a minimum of 12 subjects and utilizing the parent drug data to assess BE. However, differences emerged among the members when the drug's pharmacokinetics and pharmacodynamics become more complex, such that the study design (e.g., fasting versus fed conditions) and BE acceptance criteria (e.g., highly variable drugs, narrow therapeutic index drugs) may be affected. The survey results and discussions were shared with the ICH M13 Expert Working Group (EWG) and played an important role in identifying and analyzing gaps during the harmonization process. The draft ICH M13A guideline developed by the M13 EWG was endorsed by ICH on 20 December 2022, under Step 2.


Asunto(s)
Medicamentos Genéricos , Proyectos de Investigación , Humanos , Equivalencia Terapéutica
2.
Child Health Nurs Res ; 28(1): 70-81, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35172082

RESUMEN

PURPOSE: This study aimed to develop and evaluate the effectiveness of an educational program on developmental positioning (EPDP) for nurses in neonatal intensive care units (NICUs). METHODS: The study utilized a non-equivalent control group pretestposttest design. Sixty NICU nurses were recruited from two university hospitals in Daejeon, South Korea. The EPDP consisted of a 7-week program: 3 weeks of education and practice, followed by 4 weeks of encouragement messages using social networking services. Developmental positioning (DP) posters and DP aids were also provided during the intervention period. The intervention group (n=30) received the EPDP, but not the control group. The data were analyzed using the x2 test, the Fisher exact test, the independent t-test, and repeated-measures analysis of variance. RESULTS: Participants' knowledge (t=7.49, p<.001), attitudes (t=1.99, p=.001), self-efficacy (t=2.99, p=.004), performance of DP (t=2.98, p=.004) and Infant Positioning Assessment Tool (IPAT) scores (F=29.50, p<.001) were significantly higher in the intervention group than in the control group. CONCLUSION: The EPDP can be an effective and useful program for improving the performance of DP among NICU nurses by increasing their knowledge, attitudes, and self-efficacy of DP. However, further research involving various NICU settings is needed to gather more empirical evidence.

3.
Leuk Res ; 60: 103-108, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28772205

RESUMEN

The aberrant activation of Wnt/ß-catenin signaling is involved in the development of multiple myeloma; thus, this signaling pathway is a potential target for the development of therapeutics for this malignancy. Here, we performed cell-based chemical screening and found that CGK012, a pyranocoumarin compound, suppressed the Wnt3a-CM-mediated activation of ß-catenin response transcription. CGK012 induced ß-catenin phosphorylation at Ser33/Ser37/Thr41, leading to proteasomal degradation and reducing the level of intracellular ß-catenin. Furthermore, CGK012 consistently decreased the amount of ß-catenin and repressed the expression of cyclin D1, c-myc, and axin-2 (downstream target genes of ß-catenin) in RPMI-8226 multiple myeloma cells. In addition, CGK012 inhibited the proliferation of RPMI-8226 cells and promoted apoptosis, as indicated by the increase in the population of Annexin V-FITC-stained cells and caspase-3/7 activity. These findings suggest that CGK012 could exert antiproliferative activity against multiple myeloma cells by attenuating the Wnt/ß-catenin pathway; thus, it may have potential as a therapeutic agent for multiple myeloma treatment.


Asunto(s)
Antineoplásicos/farmacología , Carbamatos/farmacología , Proliferación Celular/efectos de los fármacos , Cumarinas/farmacología , Mieloma Múltiple/patología , Piranocumarinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Fosforilación , Piranocumarinas/uso terapéutico
4.
Bioorg Med Chem Lett ; 26(15): 3529-32, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27329797

RESUMEN

We synthesized (+)-decursin derivatives substituted with cinnamoyl- and phenyl propionyl groups originating from (+)-CGK062 and screened them using a cell-based assay to detect relative luciferase reporter activity. Of this series, compound 8b, in which a 3-acetoxy cinnamoyl group was introduced, most potently inhibited (97.0%) the Wnt/ß-catenin pathway. Specifically, compound 8b dose-dependently inhibited Wnt3a-induced expression of the ß-catenin response transcription (CRT) and increased ß-catenin degradation in HEK293 reporter cells. Furthermore, compound 8b suppressed expression of the downstream ß-catenin target genes cyclin D1 and c-myc and suppressed PC3 cell growth in a concentration-dependent manner.


Asunto(s)
Benzopiranos/farmacología , Butiratos/farmacología , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Benzopiranos/síntesis química , Benzopiranos/química , Butiratos/síntesis química , Butiratos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
5.
Inflammation ; 38(4): 1649-61, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25743565

RESUMEN

Inhibition of high-mobility group box 1 (HMGB1) protein and restoration of endothelial integrity are emerging as attractive therapeutic strategies in the management of sepsis. Here, new five structurally related 3'-N-substituted carbazole derivatives were examined for their effects on lipopolysaccharide (LPS)-mediated or cecal ligation and puncture (CLP)-mediated release of HMGB1 and on modulation of HMGB1-mediated inflammatory responses. We accessed this question by monitoring the effects of posttreatment carbazole derivatives on LPS- and CLP-mediated release of HMGB1 and HMGB1-mediated regulation of proinflammatory responses in human umbilical vein endothelial cells (HUVECs) and septic mice. The new 3'-N-substituted carbazole derivatives 1-5 inhibited the release of HMGB1 and downregulated HMGB1-dependent inflammatory responses in human endothelial cells. New compounds also inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. In addition, treatment with each compound reduced CLP-induced release of HMGB1 and sepsis-related mortality and pulmonary injury in mice. These results indicate that the new 3'-N-substituted carbazole derivatives could be candidate therapeutic agents for various severe vascular inflammatory diseases owing to their inhibition of the HMGB1 signaling pathway.


Asunto(s)
Antiinfecciosos Locales/química , Antiinfecciosos Locales/uso terapéutico , Carbazoles/química , Carbazoles/uso terapéutico , Sepsis/tratamiento farmacológico , Animales , Antiinfecciosos Locales/farmacología , Carbazoles/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/metabolismo , Sepsis/patología
6.
Mar Drugs ; 13(1): 543-57, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25603347

RESUMEN

The tumor suppressor, p53, plays an essential role in the cellular response to stress through regulating the expression of genes involved in cell cycle arrest, apoptosis and autophagy. Here, we used a cell-based reporter system for the detection of p53 response transcription to identify the marine sponge metabolites, ilimaquinone and ethylsmenoquinone, as activators of the p53 pathway. We demonstrated that ilimaquinone and ethylsmenoquinone efficiently stabilize the p53 protein through promotion of p53 phosphorylation at Ser15 in both HCT116 and RKO colon cancer cells. Moreover, both compounds upregulate the expression of p21WAF1/CIP1, a p53-dependent gene, and suppress proliferation of colon cancer cells. In addition, ilimaquinone and ethylsmenoquinone induced G2/M cell cycle arrest and increased caspase-3 cleavage and the population of cells that positively stained with Annexin V-FITC, both of which are typical biochemical markers of apoptosis. Furthermore, autophagy was elicited by both compounds, as indicated by microtubule-associated protein 1 light chain 3 (LC3) puncta formations and LC3-II turnover in HCT116 cells. Our findings suggest that ilimaquinone and ethylsmenoquinone exert their anti-cancer activity by activation of the p53 pathway and may have significant potential as chemo-preventive and therapeutic agents for human colon cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Benzoquinonas/farmacología , Células HCT116/efectos de los fármacos , Quinonas/farmacología , Sesquiterpenos/farmacología , Proteína p53 Supresora de Tumor/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Fase G2/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos
7.
Mar Drugs ; 12(6): 3231-44, 2014 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-24879546

RESUMEN

Deregulation of Wnt/ß-catenin signaling promotes the development of a broad range of human cancers, including multiple myeloma, and is thus a potential target for the development of therapeutics for this disease. Here, we used a cell-based reporter system to demonstrate that ilimaquinone and ethylsmenoquinone (formerly smenorthoquinone), sesquiterpene-quinones from a marine sponge, inhibited ß-catenin response transcription induced with Wnt3a-conditioned medium, by down-regulating the level of intracellular ß-catenin. Pharmacological inhibition of glycogen synthase kinase-3ß did not abolish the ilimaquinone and ethylsmenoquinone-mediated ß-catenin down-regulation. Degradation of ß-catenin was consistently found in RPMI-8226 multiple myeloma cells after ilimaquinone and ethylsmenoquinone treatment. Ilimaquinone and ethylsmenoquinone repressed the expression of cyclin D1, c-myc, and axin-2, which are ß-catenin/T-cell factor-dependent genes, and inhibited the proliferation of multiple myeloma cells. In addition, ilimaquinone and ethylsmenoquinone significantly induced G0/G1 cell cycle arrest and apoptosis in RPMI-8266 cells. These findings suggest that ilimaquinone and ethylsmenoquinone exert their anti-cancer activity by blocking the Wnt/ß-catenin pathway and have significant potential as therapies for multiple myeloma.


Asunto(s)
Benzoquinonas/farmacología , Mieloma Múltiple/tratamiento farmacológico , Poríferos/metabolismo , Quinonas/farmacología , Sesquiterpenos/farmacología , Animales , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzoquinonas/aislamiento & purificación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mieloma Múltiple/patología , Quinonas/aislamiento & purificación , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Sesquiterpenos/aislamiento & purificación , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/genética , beta Catenina/metabolismo
8.
Eur J Pharmacol ; 733: 81-9, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24695376

RESUMEN

This study was designed to investigate the antiproliferative effect of a novel naphthoquinone derivative, 2-undecylsulfonyl-5,8-dimethoxy-1,4-naphthoquinone (2-undecylsulfonyl-DMNQ), on platelet-derived growth factor (PDGF)-stimulated vascular smooth muscle cells (VSMCs) and examine the possible molecular mechanism of its antiproliferative action. 2-Undecylsulfonyl-DMNQ significantly inhibited PDGF-stimulated cell number and DNA synthesis, and arrested the PDGF-stimulated progression through G0/G1 to S phase of cell cycle supported by the suppression of pRb phosphorylation and cyclin D1/E, CDK2/4 and PCNA expressions. 2-Undecylsulfonyl-DMNQ dose-dependently inhibited the PDGF-stimulated phosphorylation of phospholipase Cγ (PLCγ), protein kinase B (Akt/PKB), signal transducers and activators of transcription 3 (STAT3) and extracellular signal-regulated kinase 1/2 (ERK 1/2). In addition, 2-undecylsulfonyl-DMNQ inhibited PDGF-induced PDGF receptor ß (PDGF-Rß) dimerization and the phosphorylation of Tyr(579/581), Tyr(716), Tyr(751) and Tyr(1021) in PDGF-Rß. However, 2-undecylsulfonyl-DMNQ has no antiproliferative effect on epidermal growth factor (EGF)- or fetal bovine serum (FBS)-stimulated VSMCs. In conclusion, these findings suggest that the antiproliferative effects of 2-undecylsulfonyl-DMNQ on PDGF-stimulated VSMCs are due to the blockade of receptor dimerization and autophosphorylation on specific tyrosine residues of PDGF-Rß, which resulted in the subsequent suppression of signaling cascades and a cell cycle arrest. Our observation may explain an important mechanism to block the integration of multiple signals generated by growth factor receptor activation for prevention of VSMC proliferation in cardiovascular diseases.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Naftoquinonas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Sulfonas/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , ADN/biosíntesis , Immunoblotting , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Naftoquinonas/química , Fosforilación , Multimerización de Proteína , Ratas , Transducción de Señal/efectos de los fármacos , Sulfonas/química
9.
Korean J Physiol Pharmacol ; 17(3): 203-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23776396

RESUMEN

As the abnormal proliferation of vascular smooth muscle cells (VSMCs) plays a critical role in the development of atherosclerosis and vascular restenosis, a candidate drug with antiproliferative properties is needed. We investigated the antiproliferative action and underlying mechanism of a newly synthesized naphthoquinone derivative, 5,8-dimethoxy-2-nonylamino-naphthalene-1,4-dione (2-nonylamino-DMNQ), using VSMCs treated with platelet-derived growth factor (PDGF). 2-Nonylamino-DMNQ inhibited proliferation and cell number of VSMCs induced by PDGF, but not epidermal growth factor (EGF), in a concentration-dependent manner without any cytotoxicity. This derivative suppressed PDGF-induced [(3)H]-thymidine incorporation, cell cycle progression from G0/G1 to S phase, and the phosphorylation of phosphor-retinoblastoma protein (pRb) as well as the expression of cyclin E/D, cyclin-dependent kinase (CDK) 2/4, and proliferating cell nuclear antigen (PCNA). Importantly, 2-nonylamino-DMNQ inhibited the phosphorylation of PDGF receptorß(PDGF-Rß) enhanced by PDGF at Tyr(579), Tyr(716), Tyr(751), and Tyr(1021) residues. Subsequently, 2-nonylamino-DMNQ inhibited PDGF-induced phosphorylation of STAT3, ERK1/2, Akt, and PLCγ1. Therefore, our results indicate that 2-nonylamino-DMNQ inhibits PDGF-induced VSMC proliferation by blocking PDGF-Rß autophosphorylation, and subsequently PDGF-Rß-mediated downstream signaling pathways.

10.
Chem Commun (Camb) ; 47(38): 10668-70, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21887422

RESUMEN

Photochromic compound-conjugated fluorescent fullerene-silica nanoparticles prepared by the reverse-microemulsion method was utilized for photoswitchable cellular imaging by repeatable irradiation of ultraviolet and visible light.


Asunto(s)
Colorantes Fluorescentes/química , Fulerenos/química , Nanopartículas/química , Dióxido de Silicio/química , Rayos Ultravioleta , Emulsiones/química , Células HeLa , Humanos , Microscopía Confocal , Nanopartículas/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...