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.
Eur J Pharm Sci ; 182: 106371, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36621615

RESUMEN

Topical therapies for chronic skin diseases suffer from a low patient compliance due to the inconvenient treatment regimens of available products. Dissolvable microneedles (MN) with modified release offer an interesting possibility to increase the compliance by acting as a depot in the skin and thereby decreasing the dosing frequency. Furthermore, the bioavailability can be increased significantly by bypassing the barrier of the skin by the direct penetration of the MN into the skin. In this study the depot effect and skin penetration of an innovative dissolvable MN patch was assessed by insertion in ex vivo human skin and in vivo using minipigs. The MN patches are based on biodegradable polymers and the active pharmaceutical ingredients calcipotriol (Calci) and betamethasone-17-21-dipropionate (BDP) used to treat psoriasis. Using computed tomography (CT) and Coherent anti-Stokes Raman scattering (CARS) microscopy it was possible to visualize the skin penetration and follow the morphology of the MN as function of time in the skin. The depot effect was assessed by studying the modified in vitro release in an aqueous buffer and by comparing the drug release of a single application of a patch both ex vivo and in vivo to daily application of a marketed oleogel containing the same active pharmaceutical ingredients. The CT and CARS images showed efficient penetration of the MN patches into the upper dermis and a slow swelling process of the drug containing tip over a period of 8 days. Furthermore, CARS demonstrated that it can be used as a noninvasive technique with potential applicability in clinical settings. The in vitro release studies show a release of 54% over a time period of 30 days. The pharmacological relevance of MNs was confirmed in human skin explants and in vivo after single application and showed a similar response on calcipotriol and BDP mediated signaling events compared to daily application of the active oleogel. Altogether it was demonstrated that the MN can penetrate the skin and have the potential to provide a depot effect.


Asunto(s)
Agujas , Piel , Animales , Humanos , Porcinos , Preparaciones Farmacéuticas/metabolismo , Liberación de Fármacos , Porcinos Enanos , Piel/metabolismo , Administración Cutánea , Sistemas de Liberación de Medicamentos/métodos
2.
Pharm Res ; 37(12): 243, 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-33188482

RESUMEN

PURPOSE: To investigate the difference in clinical efficacy in AD patients between two topical PDE4 inhibitors using dermal open flow microperfusion and cAMP as a pharmacodynamic read-out in fresh human skin explants. METHODS: Clinical formulations were applied to intact or barrier disrupted human skin explants and both skin biopsy samples and dermal interstitial fluid was sampled for measuring drug concentration. Furthermore, cAMP levels were determined in the skin biopsies as a measure of target engagement. RESULTS: Elevated cAMP levels were observed with LEO 29102 while no evidence of target engagement was obtained with LEO 39652. In barrier impaired skin the dISF concentration of LEO 29102 was 2100 nM while only 33 nM for LEO 39652. For both compounds the concentrations measured in skin punch biopsies were 7-33-fold higher than the dISF concentrations. CONCLUSIONS: Low unbound drug concentration in dISF in combination with minimal target engagement of LEO 39652 in barrier impaired human skin explants supports that lack of clinical efficacy of LEO 39652 in AD patients is likely due to insufficient drug availability at the target. We conclude that dOFM together with a pharmacodynamic target engagement biomarker are strong techniques for establishing skin PK/PD relations and that skin biopsies should be used with caution.


Asunto(s)
Acetamidas/farmacocinética , Dermatitis Atópica/metabolismo , Líquido Extracelular/metabolismo , Microdiálisis , Inhibidores de Fosfodiesterasa 4/farmacocinética , Piridinas/farmacocinética , Absorción Cutánea , Piel/metabolismo , Acetamidas/administración & dosificación , Acetamidas/química , Administración Cutánea , Biopsia , Células Cultivadas , Ensayos Clínicos Fase II como Asunto , AMP Cíclico/metabolismo , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/patología , Composición de Medicamentos , Estabilidad de Medicamentos , Humanos , Queratinocitos/metabolismo , Inhibidores de Fosfodiesterasa 4/administración & dosificación , Inhibidores de Fosfodiesterasa 4/química , Piridinas/administración & dosificación , Piridinas/química , Piel/efectos de los fármacos , Piel/patología , Equivalencia Terapéutica
3.
Dermatol Ther (Heidelb) ; 6(4): 599-626, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27503482

RESUMEN

INTRODUCTION: Ingenol mebutate gel (Picato®, LEO Pharma A/S) is approved for the field treatment of actinic keratosis and is characterized by high sustained clearance of actinic lesions. The inherent propensity of ingenol mebutate towards chemical rearrangement necessitates refrigeration of the final product. We sought to identify novel ingenol derivatives with enhanced chemical stability and similar or improved in vitro potency and in vivo efficacy. METHODS: A number of ingenol esters were synthesized with full regiocontrol from ingenol. Chemical stability was determined in aqueous buffer at physiological pH and hydroalcoholic gel at lower pH. Acute cytotoxicity was determined in HeLa or HSC-5 cells. Keratinocyte proliferation, viability and caspase 3/7 activation was measured in primary epidermal keratinocytes. Relative gene expression levels were determined by real-time quantitative PCR. Evaluation of in vivo tumor ablating potential was performed in the murine B16 melanoma mouse model and in the UV-induced skin carcinogenesis model in hairless SKH-1 mice following topical treatment for two consecutive days with test compounds formulated at 0.1% in a hydroalcoholic gel. RESULTS: This work resulted in the identification of ingenol disoxate (LEO 43204) displaying increased stability in a clinically relevant formulation and in aqueous buffer with minimal pH-dependent acyl migration degradation. Ingenol disoxate exhibited a significantly higher cytotoxic potency relative to ingenol mebutate. Likewise, cell growth arrest in normal human keratinocyte was more potently induced by ingenol disoxate, which was accompanied by protein kinase C dependent transcription of markers of keratinocyte differentiation. Most notably, ingenol disoxate possessed a superior antitumor effect in a B16 mouse melanoma model and significantly increased median survival time relative to ingenol mebutate. A significant effect on tumor ablation was also observed in a murine model of ultraviolet irradiation-induced skin carcinogenesis. CONCLUSION: These data illustrate that the favorable in vitro and in vivo pharmacological properties driving ingenol mebutate efficacy are either preserved or improved in ingenol disoxate. In combination with improved chemical stability to potentially facilitate storage of the final product at ambient temperatures, these features support further development of ingenol disoxate as a convenient and efficacious treatment modality of non-melanoma skin cancers. FUNDING: LEO Pharma A/S.

4.
Bioorg Med Chem Lett ; 24(1): 54-60, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24332494

RESUMEN

Ingenol 3-benzoates were investigated with respect to chemical stability, pro-inflammatory effects, cell death induction and PKCδ activation. A correlation between structure, chemical stability and biological activity was found and compared to ingenol mebutate (ingenol 3-angelate) used for field treatment of actinic keratosis. We also provided further support for involvement of PKCδ for induction of oxidative burst and cytokine release. Molecular modeling and dynamics calculations corroborated the essential interactions between key compounds and C1 domain of PKCδ.


Asunto(s)
Antineoplásicos/farmacología , Benzoatos/farmacología , Diterpenos/farmacología , Queratosis Actínica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzoatos/síntesis química , Benzoatos/química , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Diterpenos/síntesis química , Diterpenos/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Queratosis Actínica/metabolismo , Queratosis Actínica/patología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Relación Estructura-Actividad
5.
Bioorg Med Chem Lett ; 23(20): 5624-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23993332

RESUMEN

Ingenol mebutate is the active ingredient in Picato® a new drug for the treatment of actinic keratosis. A number of derivatives related to ingenol mebutate were prepared by chemical synthesis from ingenol with the purpose of investigating the SAR and potency in assays relating to pro-inflammatory effects (induction of PMN oxidative burst and keratinocyte cytokine release), the potential of cell death induction, as well as the chemical stability. By modifications of the ingenol scaffold several prerequisites for activity were identified. The chemical stability of the compounds could be linked to an acyl migration mechanism. We were able to find analogues of ingenol mebutate with comparable in vitro properties. Some key features for potent and more stable ingenol derivatives have been identified.


Asunto(s)
Antineoplásicos/síntesis química , Diterpenos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular , Diterpenos/uso terapéutico , Diterpenos/toxicidad , Humanos , Interleucina-8/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratosis Actínica/tratamiento farmacológico , Queratosis Actínica/metabolismo , Leucocitos Mononucleares/metabolismo , Melanoma/patología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Drugs Dermatol ; 11(10): 1181-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23134983

RESUMEN

We investigated the proposed necrotic mechanism of ingenol mebutate, a natural compound with anti-cancer properties in human keratinocytes, the human squamous cell carcinoma cell line HSC-5, and HeLa cervix carcinoma cells. Topical application of a clinical dose of ingenol mebutate 0.05% (1.15 mM) gel to human reconstituted full-thickness skin equivalents strongly reduced epidermal, but not dermal viability. Ingenol mebutate showed cytotoxic potency between 200-300 M on normal and cancer cells. When keratinocytes were induced to differentiate, they became significantly less sensitive to ingenol mebutate and half-maximal induction of cell death required more than 300 M ingenol mebutate. Cytotoxic concentrations of ingenol mebutate caused rupture of the mitochondrial network within minutes paralleled by cytosolic calcium release in all cells. Subsequently, plasma membrane integrity was lost as seen by propidium uptake into the cells. This was in sharp contrast to lysis of cells with low concentrations of the detergent Triton X-100 that permeabilized the plasma membrane within minutes without affecting organelle morphology. Buffering of intracellular calcium and inhibition of the mitochondrial permeability transition pore reduced the cytotoxic effect of ingenol mebutate in cancer cells, but not in normal keratinocytes. However, these inhibitors could not prevent cell death subsequent to prolonged incubation. Our findings reveal that ingenol mebutate does not mediate cytotoxicity by a simple lytic, necrotic mechanism, but activates distinct processes involving multiple cell organelles in a cell-type and differentiation-dependent manner. These data improve our understanding of ingenol mebutate-target cell interactions and offer new insights relevant to the removal of aberrant cells in human skin.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Diterpenos/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Piel/patología , Calcio/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Detergentes/farmacología , Células HeLa , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Mitocondrias/efectos de los fármacos , Necrosis , Octoxinol/farmacología , Piel/efectos de los fármacos
7.
Epilepsy Behav ; 16(2): 281-7, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19716343

RESUMEN

Animal studies have shown endocrine changes after levetiracetam treatment. The present study investigated reproductive and sexual function in patients with epilepsy (aged 18-45) treated with levetiracetam (LEV: 30 men/26 women), carbamazepine (CBZ: 63 men/30 women), or lamotrigine (LTG: 37 men/40 women) monotherapy and in healthy controls (36 men/44 women). In women, no endocrine changes were observed during LEV treatment, whereas steroid hormone-binding globulin levels were greater and progesterone levels lower in women using CBZ. Dehydroepiandrosterone sulfate levels were higher and androstenedione levels lower in LTG-treated women. Arizona Sexual Experience Scale scores, which were significantly lower in females using LTG or LEV, suggesting they have better sexual function than CBZ users and controls. In men, no drug-specific hormonal pattern was observed after LEV treatment. Male patients in all treatment groups had lower androstenedione and free testosterone. Those using CBZ had lower free androgen indices and dehydroepiandrosterone sulfate levels, and higher steroid hormone-binding globulin, follicle-stimulating hormone, and luteinizing hormone levels. Arizona Sexual Experience Scale scores for men were similar in all groups. In conclusion, LEV treatment apparently has no drug-specific sexual or endocrine side effects in men or women in this age group.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Epilepsia/fisiopatología , Hormonas Esteroides Gonadales/metabolismo , Adolescente , Adulto , Factores de Edad , Carbamazepina/uso terapéutico , Estudios Transversales , Femenino , Gonadotropinas/sangre , Humanos , Inmunoensayo/métodos , Lamotrigina , Levetiracetam , Masculino , Persona de Mediana Edad , Piracetam/análogos & derivados , Piracetam/uso terapéutico , Factores Sexuales , Encuestas y Cuestionarios , Testosterona/sangre , Triazinas/uso terapéutico , Adulto Joven
8.
Cell Signal ; 19(3): 646-57, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17052891

RESUMEN

Biochemical evidence indicates that TGF-beta-activated kinase 1 (TAK1), a key modulator of the inflammatory response, exists in a complex with various adaptor proteins including the TAK1 binding protein 1 (TAB1). However, the physiological importance of TAB1 in TAK1 activation, and in the subsequent induction of proinflammatory mediators, remains unclear. In this study, a critical role for TAK1 in IL-1alpha or TNFalpha stimulated MAPK and NFkappaB activation was confirmed by inhibition of the nuclear accumulation of NFkappaB p65 and phosphorylated forms of c-Jun and p38 following siRNA mediated TAK1 silencing. These effects were associated with significant reductions in IL-1alpha stimulated levels of secreted IL-6, IL-8, MCP-1 and GM-CSF. In contrast, IL-1alpha or TNFalpha dependent cellular redistribution of NFkappaB p65 and phosphorylated c-Jun and p38 was not affected by 80% siRNA mediated knockdown of TAB1 protein levels. Interestingly, IL-6, IL-8 and GM-CSF release from TAB1 siRNA transfected cells was significantly reduced following IL-1alpha treatment, but was unchanged after TNFalpha stimulation, suggesting differential roles for TAB1 in IL-1alpha and TNFalpha signalling pathways. These findings may imply an as yet unidentified role for TAB1 in the inflammatory response, which is independent of the activation of classical TAK1 associated signalling cascades.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Citocinas/metabolismo , Interleucina-1alfa/farmacología , Quinasas Quinasa Quinasa PAM/metabolismo , Activación Enzimática , Células HeLa , Humanos , ARN Interferente Pequeño/metabolismo
9.
Methods Enzymol ; 414: 348-63, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17110202

RESUMEN

This chapter describes a robust high-content cellular screening assay to simultaneously analyze the spatiotemporal activation of three different kinase-associated signaling pathways involving NF-kappaB, JNK, and p38, all of which are closely implicated in proliferative and proinflammatory responses. Signal transduction is dependent on the translocation of NF-kappaB p65 and phosphorylated c-Jun and p38 from the cytosol to the nucleus, and fluorescent immunolabeling was used to monitor changes in their cellular distribution. Cellular screening, data acquisition, and data interpretation were conducted on the ArrayScan HCS Reader (Cellomics Inc., Pittsburgh, PA). Assay adaptation to various cellular systems is feasible when sufficient separation of the nuclear and cytosolic compartment can be achieved and if cell adhesion properties permit proper attachment to the culture plates. Substitution of NF-kappaB p65 and phosphorylated forms of c-Jun and p38 as targets to analyze other translocating components is possible and is limited primarily by antibody specificity and the risk of fluorescent bleed-through between emission channels. Because assay validity is particularly confounded by inadequate spectral separation of the detection dyes in multicolor labeling assays, means of eliminating or counterbalancing staining artifacts are illustrated. Also, protocol parameter settings important for imaging and image processing are described, including object identification, image exposure settings, separation of cytosolic and nuclear regions, number of cells sufficient for analysis, and the use of gating thresholds critical for cell sorting and subpopulation analysis. This assay is a useful tool to investigate the interplay between signaling pathways and the mode of action, potency, and selectivity of compound inhibition of specific target molecules in a cellular context.


Asunto(s)
Inflamación/patología , Microscopía Fluorescente/métodos , Núcleo Celular/metabolismo , Citosol/metabolismo , Fragmentación del ADN , Células HeLa , Humanos , FN-kappa B/metabolismo , Transporte de Proteínas , Reproducibilidad de los Resultados , Transducción de Señal , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Assay Drug Dev Technol ; 3(3): 261-71, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15971988

RESUMEN

High content cellular screening assays are useful tools to investigate the interplay between signaling pathways and offer valuable platforms to determine the mode of action, potency, and selectivity of potential drug candidates in a biological setting. We describe a cell-based multiplex fluorescent imaging assay that permits concurrent detection and quantification of the distribution of nuclear factor kappaB (NFkappaB) p65/RelA and phosphorylated forms of p38 and c-Jun between the cytosol and nucleus. Cellular screening, data acquisition, and data interpretation were conducted on the ArrayScan HCS Reader (Cellomics Inc., Pittsburgh, PA). A significant window between untreated and interleukin-1alpha (IL-1alpha) stimulated HeLa cells for all three targets was achieved with low variability. Staining specificity was confirmed with blocking peptides and pharmacological inhibition of p38, c-Jun-N-terminal kinase (JNK), and inhibitory kappaB kinase 2, and channel bleed-through was eliminated or counterbalanced by the use of fixed exposure times together with careful reporter channel selection. The JNK inhibitor SP600125 was used as a demonstration compound because in addition to inhibiting nuclear accumulation of phosphorylated c-Jun it reduced nuclear translocation of phosphorylated p38 and NFkappaB p65/RelA in a dose-dependent manner, indicating a lack of SP600125 selectivity. This was supported by RNA interference where co-transfection of small interfering RNA targeting both JNK1 and JNK2, to limit signaling redundancy, significantly inhibited IL-1alpha-stimulated translocation of phosphorylated c-Jun without altering phosphorylated p38 and NFkappaB p65/RelA redistribution. This image analysis application is a valuable and information-rich screening tool to investigate compound selectivity and/or cross-talk between key signaling pathways involved in the inflammatory response.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Inflamación/fisiopatología , Interleucina-1/farmacología , Transducción de Señal/fisiología , Antracenos/farmacología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Fluorescencia , Técnica del Anticuerpo Fluorescente/instrumentación , Células HeLa , Humanos , Imidazoles/farmacología , Immunoblotting , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Piridinas/farmacología , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Tecnología Farmacéutica/métodos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...