Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 716
Filtrar
1.
Respir Physiol Neurobiol ; 296: 103825, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34808585

RESUMEN

Lung delivery for glucocorticoids (GCs) is very low and depends on the system used. Exogenous pulmonary surfactant (EPS) is a promising tool to transporting GCs efficiently to the airways. We developed a new formulation of EPS with Budesonide (BUD) incorporated into EPS membranes (EPS-BUD) to improve lung delivery of BUD. We evaluated the biodistribution and pharmacokinetic of the transported BUD by intra-tracheal instillation of EPS-BUD in healthy rats. Aqueous suspension of Budesonide was used as control. Budesonide and its esters present in trachea, kidneys and lungs were determined by HPLC. The delivery of BUD in lung for EPS-BUD group was 75 % of total instilled and only 35 % for the control group. BUD was rapidly internalized in pneumocytes and a high proportion of Budesonide esters and persistent concentrations of active free BUD were found for up to 6 h after instillation. The new EPS-BUD formulation developed significantly improves the deposition and increases the permanence of BUD in lung.


Asunto(s)
Budesonida/farmacocinética , Glucocorticoides/farmacocinética , Pulmón/efectos de los fármacos , Surfactantes Pulmonares/farmacocinética , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Ratas
2.
Drug Metab Dispos ; 49(10): 919-928, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34426410

RESUMEN

Many women take drugs during their pregnancy to treat a variety of clinical conditions. To optimize drug efficacy and reduce fetal toxicity, it is important to determine or predict fetal drug exposure throughout pregnancy. Previously, we developed and verified a maternal-fetal physiologically based pharmacokinetic (m-f PBPK) model to predict fetal Kp,uu (unbound fetal plasma AUC/unbound maternal plasma AUC) of drugs that passively cross the placenta. Here, we used in vitro transport studies in Transwell, in combination with our m-f PBPK model, to predict fetal Kp,uu of drugs that are effluxed by placental P-glycoprotein (P-gp)-namely, dexamethasone, betamethasone, darunavir, and lopinavir. Using Transwell, we determined the efflux ratio of these drugs in hMDR1-MDCKcP-gpKO cells, in which human P-gp was overexpressed and the endogenous P-gp was knocked out. Then, using the proteomics-informed efflux ratio-relative expressive factor approach, we predicted the fetal Kp,uu of these drugs at term. Finally, to verify our predictions, we compared them with the observed in vivo fetal Kp,uu at term. The latter was estimated using our m-f PBPK model and published fetal [umbilical vein (UV)]/maternal plasma drug concentrations obtained at term (UV/maternal plasma). Fetal Kp,uu predictions for dexamethasone (0.63), betamethasone (0.59), darunavir (0.17), and lopinavir (0.08) were successful, as they fell within the 90% confidence interval of the corresponding in vivo fetal Kp,uu (0.30-0.66, 0.29-0.71, 0.11-0.22, 0.04-0.19, respectively). This is the first demonstration of successful prediction of fetal Kp,uu of P-gp drug substrates from in vitro studies. SIGNIFICANCE STATEMENT: For the first time, using in vitro studies in cells, this study successfully predicted human fetal Kp,uu of P-gp substrate drugs. This success confirms that the m-f PBPK model, combined with the ER-REF approach, can successfully predict fetal drug exposure to P-gp substrates. This success provides increased confidence in the use of the ER-REF approach, combined with the m-f PBPK model, to predict fetal Kp,uu of drugs (transported by P-gp or other transporters), both at term and at earlier gestational ages.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Feto , Glucocorticoides , Inhibidores de la Proteasa del VIH , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Perros , Resistencia a Múltiples Medicamentos , Femenino , Feto/efectos de los fármacos , Feto/metabolismo , Edad Gestacional , Glucocorticoides/sangre , Glucocorticoides/farmacocinética , Inhibidores de la Proteasa del VIH/sangre , Inhibidores de la Proteasa del VIH/farmacocinética , Humanos , Células de Riñón Canino Madin Darby , Modelos Biológicos , Valor Predictivo de las Pruebas , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/prevención & control
3.
J Med Chem ; 64(16): 11958-11971, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34378927

RESUMEN

Glucocorticoids (GCs) are widely used to treat a variety of autoimmune and inflammatory diseases; however, systemic delivery of GCs is associated with side effects that affect essentially every organ system, reflecting the nearly ubiquitous expression of the glucocorticoid receptor (GR). Targeted delivery of GCs to diseased tissues using antibody-glucocorticoid conjugates (GC-ADCs) offers a therapeutic alternative to overcome these adverse effects. Herein, we describe novel classes of GCs that exhibited greater potency than dexamethasone and budesonide, a 100-fold selectivity toward the GR over other nuclear receptors, and no in vitro safety liability in pharmacology assays (hERG, AMES) and that demonstrated a substantial reduction in tumor necrosis factor-α (TNF-α) release in mice challenged with lipopolysaccharide (LPS). The site-specific conjugated GC-ADCs via cathepsin-cleavable linkers were highly stable in plasma and specifically released GCs in antigen-positive cells, suggesting that these novel GCs can serve as ADC payloads to treat autoimmune and inflammatory diseases.


Asunto(s)
Budesonida/análogos & derivados , Budesonida/uso terapéutico , Glucocorticoides/uso terapéutico , Inmunoconjugados/uso terapéutico , Inflamación/tratamiento farmacológico , Animales , Budesonida/metabolismo , Budesonida/farmacocinética , Catepsina B/metabolismo , Glucocorticoides/síntesis química , Glucocorticoides/metabolismo , Glucocorticoides/farmacocinética , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Inmunoconjugados/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Estructura Molecular , Receptores de Glucocorticoides/metabolismo , Receptores de Prolactina/inmunología , Relación Estructura-Actividad
4.
CPT Pharmacometrics Syst Pharmacol ; 10(9): 1057-1070, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34273255

RESUMEN

We have previously developed a maternal-fetal physiologically-based pharmacokinetic (m-f PBPK) model to dynamically predict (and verify) fetal-maternal exposure to drugs that passively diffuse across the placenta. Here, we extended the application of this model to dynamically predict fetal exposure to drugs which are effluxed by placental P-glycoprotein, namely the antenatal corticosteroids (ACS; dexamethasone [DEX], and betamethasone [BET]). To do so, we estimated both the placental P-gp mediated efflux clearance (CL) and the passive diffusion CL of the ACS. The efficacy and toxicity of the currently used maternal ACS dosing regimens to prevent neonatal respiratory distress syndrome could be improved by altering their dosing regimens. Therefore, to illustrate the utility of our m-f PBPK model, we used it to design alternative dosing regimens of DEX and BET that could potentially improve their efficacy and reduce their toxicity. The redesigned dosing regimens are convenient to administer, maintain maternal-fetal exposure (area under the concentration-time curve [AUC]) or maximum plasma concentration (Cmax ) or both (DEX and BET) or minimize maternal exposure while maintaining fetal drug plasma concentrations above the minimum therapeutic threshold of 1 ng/ml for 48 h (BET only; based on efficacy data in sheep). To our knowledge, this is the first study to dynamically predict fetal plasma concentrations of placental P-gp effluxed drugs. Our approach and our m-f PBPK model could be used in the future to predict maternal-fetal exposure to any drug and to design alternative dosing regimens of the drug.


Asunto(s)
Glucocorticoides/administración & dosificación , Modelos Biológicos , Placenta/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/prevención & control , Animales , Área Bajo la Curva , Betametasona/administración & dosificación , Betametasona/farmacocinética , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Feto/metabolismo , Glucocorticoides/farmacocinética , Humanos , Recién Nacido , Intercambio Materno-Fetal/fisiología , Embarazo , Atención Prenatal/métodos , Ovinos
5.
Anal Bioanal Chem ; 413(16): 4363-4371, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34002273

RESUMEN

Corticosteroids as budesonide can be effective in reducing topic inflammation processes in different organs. Therapeutic use of budesonide in respiratory diseases, like asthma, chronic obstructive pulmonary disease, and allergic rhinitis is well known. However, the pulmonary distribution of budesonide is not well understood, mainly due to the difficulties in tracing the molecule in lung samples without the addition of a label. In this paper, we present a matrix-assisted laser desorption/ionization mass spectrometry imaging protocol that can be used to visualize the pulmonary distribution of budesonide administered to a surfactant-depleted adult rabbit. Considering that budesonide is not easily ionized by MALDI, we developed an on-tissue derivatization method with Girard's reagent P followed by ferulic acid deposition as MALDI matrix. Interestingly, this sample preparation protocol results as a very effective strategy to raise the sensitivity towards not only budesonide but also other corticosteroids, allowing us to track its distribution and quantify the drug inside lung samples.


Asunto(s)
Budesonida/farmacocinética , Glucocorticoides/farmacocinética , Pulmón/metabolismo , Animales , Budesonida/administración & dosificación , Budesonida/análisis , Glucocorticoides/administración & dosificación , Glucocorticoides/análisis , Indicadores y Reactivos , Conejos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Esteroides/administración & dosificación , Esteroides/análisis , Esteroides/farmacocinética
6.
Pulm Med ; 2021: 8881895, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815843

RESUMEN

Both asthma and chronic obstructive pulmonary disease (COPD) are inflammatory chronic respiratory conditions with high rates of morbidity and mortality worldwide. The objectives of this review are to briefly describe the pathophysiology and epidemiology of asthma and COPD, discuss guideline recommendations for uncontrolled disease, and review a new generic option for the treatment of asthma and COPD. Although mild forms of these diseases may be controlled with as-needed pharmacotherapy, uncontrolled or persistent asthma and moderate or severe COPD uncontrolled by bronchodilators with elevated eosinophilia or frequent exacerbations may require intervention with combination therapy with inhaled corticosteroids (ICS) and long-acting beta agonists (LABAs), according to international guidelines. Fixed-dose combinations of ICS/LABA are commonly prescribed for both conditions, with fluticasone propionate (FP) and salmeterol forming a cornerstone of many treatment plans. An oral inhalation powder containing the combination of FP and salmeterol has been available as Advair Diskus® in the United States for almost 20 years, and the first and only substitutable generic version of this product has recently been approved for use: Wixela™ Inhub™. Bioequivalence of Wixela Inhub and Advair Diskus has been established. Furthermore, the Inhub inhaler was shown to be robust and easy to use, suggesting that Wixela Inhub may provide an alternative option to Advair Diskus for patients with asthma or COPD requiring intervention with an ICS/LABA.


Asunto(s)
Asma , Broncodilatadores , Combinación Fluticasona-Salmeterol , Glucocorticoides , Enfermedad Pulmonar Obstructiva Crónica , Administración por Inhalación , Asma/tratamiento farmacológico , Asma/epidemiología , Asma/fisiopatología , Broncodilatadores/farmacocinética , Broncodilatadores/uso terapéutico , Medicamentos Genéricos/farmacocinética , Medicamentos Genéricos/uso terapéutico , Combinación Fluticasona-Salmeterol/farmacocinética , Combinación Fluticasona-Salmeterol/uso terapéutico , Glucocorticoides/farmacocinética , Glucocorticoides/uso terapéutico , Necesidades y Demandas de Servicios de Salud/estadística & datos numéricos , Humanos , Nebulizadores y Vaporizadores , Polvos/farmacocinética , Polvos/uso terapéutico , Guías de Práctica Clínica como Asunto , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Nivel de Atención , Equivalencia Terapéutica
7.
ACS Appl Mater Interfaces ; 13(18): 21076-21086, 2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-33913684

RESUMEN

The nontargeted distribution and uncontrolled in vivo release of drugs impede their efficacy in the treatment of rheumatoid arthritis (RA). Delivering drugs to arthritic joints and releasing drugs on demand are a feasible solution to achieve the effective treatment of RA. In this paper, we report a facile method to assemble dual-stimuli responsive polymeric micelles from polyethylene glycol-phenylboric acid-triglycerol monostearate (PEG-PBA-TGMS, PPT) conjugates with the aim of delivering dexamethasone (Dex) to arthritic joints and controlling the release of Dex by inflammatory stimuli. We show that the release of Dex from the PPT micelles is accelerated in response to acidic pH and overexpressed matrix metalloproteinases. In an adjuvant-induced arthritis model, the PPT micelles preferentially accumulate in arthritic joints and show an excellent therapeutic efficacy after being intravenously administrated. Our results highlight the potential of the dual stimuli-responsive micelles as a promising therapeutic option for the effective treatment of inflammatory diseases.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Dexametasona/administración & dosificación , Portadores de Fármacos , Glucocorticoides/administración & dosificación , Micelas , Polímeros/química , Animales , Materiales Biocompatibles , Dexametasona/farmacocinética , Dexametasona/uso terapéutico , Liberación de Fármacos , Glucocorticoides/farmacocinética , Glucocorticoides/uso terapéutico , Hemólisis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Distribución Tisular
8.
Exp Eye Res ; 206: 108534, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33722510

RESUMEN

Uveitis is the inflammation of uveal tract comprising of iris, ciliary body and choroid. Blood ocular barriers maintaining the homeostasis of eye breach during uveitis, leads to high risk for sight-threatening complications. The purpose of this study was to compare the anti-inflammatory activity enabled by two diverse pharmacological agents (prednisolone and dapsone) using their effect on aqueous humor proteome. Wistar rats of either sex (150-200g) were used and randomly divided into various groups. Normal group was injected with 0.1ml normal saline (NS), endotoxin (LPS) (200 µg/0.1ml NS) was injected into endotoxin induced inflammatory groups followed by 0.1% dapsone and 1% prednisolone treatment in endotoxin induced uveitis (EIU) groups, respectively. Aqueocentesis was performed post 24 hour inflammation and samples were subjected for clinical parameter evaluation, cytokine analysis as well as global proteomic analysis using High-resolution mass spectrometer. Following which spectrum analysis, production spectra of peptides were matched against R. Norvegicus Protein Database (Uniport) using Proteome Discoverer (v2.2). Upon clinical evaluation, the anterior segment images post dapsone and prednisolone treatment have shown marked decrease in hyperaemia, miosis and iridial vessels vasodilation in rat eyes as compared to inflammation group. The result of cytokine analysis revealed 0.1% dapsone and prednisolone both significantly decreased the TNF-α levels. HRMS studies analysis expressed 140, 160, 158 and 141 proteins unique to normal, EIU, Dapsone and prednisolone group respectively. To conclude aqueous humor pharmacoproteomic revealed the anti-inflammatory activity of the dapsone comparable to the prednisolone treatment in endotoxin induced uveitis. The topical dapsone may be used as an alternative therapeutic option in treating uveitis without elevating intraocular pressure.


Asunto(s)
Humor Acuoso/metabolismo , Dapsona/farmacocinética , Prednisolona/farmacocinética , Proteómica , Uveítis Anterior/tratamiento farmacológico , Administración Tópica , Animales , Antiinfecciosos/administración & dosificación , Antiinfecciosos/farmacocinética , Dapsona/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacocinética , Masculino , Prednisolona/administración & dosificación , Ratas , Ratas Wistar , Uveítis Anterior/metabolismo
9.
Biopharm Drug Dispos ; 42(5): 191-203, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33638217

RESUMEN

The pharmacokinetic (PK) parameters of dexamethasone (DEX) in 11 species were collected from the literature and clearances (CL) assessed by basic allometric methods, and concentration-time course profiles were fitted using two PK models incorporating physiological or allometric scaling. Plots of log CL vs. log body weights (BW) correlated reasonably with R2  = 0.91, with a maximum ratio of actual to fitted CL of 6 (for pig). A minimal physiologically-based pharmacokinetic (mPBPK) model containing blood and two lumped tissue compartments and integrated utilization of physiological parameters was compared to an allometric two-compartment model (a2CM). The plasma PK profiles of DEX from 11 species were analyzed jointly, with the mPBPK model having conserved partition coefficients (Kp ), physiologic blood and tissue volumes, and species-specific CL values. The DEX PK profiles were reasonably captured by the mPBPK model for 9 of 11 species in the joint analysis with three fitted parameters (besides CL) including an overall tissue-to-plasma partition coefficient of 1.07. The a2CM with distribution CL and central and peripheral volumes scaled allometrically fitted the plasma concentration profiles similarly but required a total of six parameters (besides CL). Overall, the literature reported that DEX CL values exhibit moderate variability (mean = 0.64 L/h/kg; coefficient of variation = 105%), but distribution parameters were largely conserved across most species.


Asunto(s)
Antiinflamatorios/farmacocinética , Dexametasona/farmacocinética , Glucocorticoides/farmacocinética , Modelos Biológicos , Animales , Antiinflamatorios/sangre , Dexametasona/sangre , Glucocorticoides/sangre , Humanos , Especificidad de la Especie
10.
Int J Pharm ; 595: 120241, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33484917

RESUMEN

Inhaled ciclesonide (CIC), a corticosteroid used to treat asthma that is also being investigated for the treatment of corona virus disease 2019, hydrolyzes to desisobutyryl-ciclesonide (des-CIC) followed by reversible esterification when exposed to fatty acids in lungs. While previous studies have described the distribution and metabolism of the compounds after inhalation, spatial localization in the lungs remains unclear. We visualized two-dimensional spatial localization of CIC and its metabolites in rat lungs after administration of a single dose of a CIC aerosol (with the mass median aerodynamic diameter of 0.918-1.168 µm) using desorption electrospray ionization-time of flight mass spectrometry imaging (DESI-MSI). In the analysis, CIC, des-CIC, and des-CIC-oleate were imaged in frozen lung sections at high spatial and mass resolutions in negative-ion mode. MSI revealed the coexistence of CIC, des-CIC, and des-CIC-oleate on the airway epithelium, and the distribution of des-CIC and des-CIC-oleate in peripheral lung regions. In addition, a part of CIC independently localized on the airway epithelium. These results suggest that distribution of CIC and its metabolites in lungs is related to both the intended delivery of aerosols to pulmonary alveoli and peripheral regions, and the potential deposition of CIC particles on the airway epithelium.


Asunto(s)
Glucocorticoides/administración & dosificación , Glucocorticoides/farmacocinética , Pulmón/diagnóstico por imagen , Pulmón/metabolismo , Pregnenodionas/administración & dosificación , Pregnenodionas/farmacocinética , Espectrometría de Masa por Ionización de Electrospray/métodos , Administración por Inhalación , Aerosoles/química , Animales , Células Epiteliales/metabolismo , Glucocorticoides/sangre , Pregnenodionas/sangre , Pregnenodionas/metabolismo , Alveolos Pulmonares/metabolismo , Ratas , Ratas Sprague-Dawley , Distribución Tisular , Tratamiento Farmacológico de COVID-19
11.
Pharm Res ; 37(12): 245, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33215336

RESUMEN

PURPOSE: The purpose of this study is to show how the Ocular Compartmental Absorption & Transit (OCAT™) model in GastroPlus® can be used to characterize ocular drug pharmacokinetic performance in rabbits for ointment formulations. METHODS: A newly OCAT™ model developed for fluorometholone, as well as a previously verified model for dexamethasone, were used to characterize the aqueous humor (AH) concentration following the administration of multiple ointment formulations to rabbit. The model uses the following parameters: application surface area (SA), a fitted application time, and the fitted Higuchi release constant to characterize the rate of passage of the active pharmaceutical ingredient from the ointment formulations into the tears in vivo. RESULTS: Parameter sensitivity analysis was performed to understand the impact of ointment formulation changes on ocular exposure. While application time was found to have a significant impact on the time of maximal concentration in AH, both the application SA and the Higuchi release constant significantly influenced both the maximum concentration and the ocular exposure. CONCLUSIONS: This initial model for ointment ophthalmic formulations is a first step to better understand the interplay between physiological factors and ophthalmic formulation physicochemical properties and their impact on in vivo ocular drug pharmacokinetic performance in rabbits.


Asunto(s)
Dexametasona/farmacocinética , Ojo/metabolismo , Fluorometolona/farmacocinética , Glucocorticoides/farmacocinética , Modelos Biológicos , Absorción Ocular , Administración Oftálmica , Animales , Humor Acuoso/metabolismo , Simulación por Computador , Dexametasona/administración & dosificación , Fluorometolona/administración & dosificación , Glucocorticoides/administración & dosificación , Pomadas , Conejos
12.
J Sport Rehabil ; 30(4): 538-544, 2020 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-33120356

RESUMEN

CONTEXT: Low current intensity iontophoresis treatments have increased skin perfusion over 700% from baseline potentially altering drug clearance from or diffusion to the targeted area. OBJECTIVE: To determine the effects of a preceding 10-minute ice massage on subcutaneous dexamethasone sodium phosphate (Dex-P) concentration and skin perfusion during and after a 4-mA iontophoresis treatment. DESIGN: Controlled laboratory study. SETTING: Research laboratory. PATIENTS OR OTHER PARTICIPANTS: Twenty-four participants (male = 12, female = 12; age = 25.6 [4.5] y, height = 173.9 [8.51] cm, mass = 76.11 [16.84] kg). INTERVENTION(S): Participants were randomly assigned into 2 groups: (1) pretreatment 10-minute ice massage and (2) no pretreatment ice massage. Treatment consisted of an 80-mA·minute (4 mA, 20 min) Dex-P iontophoresis treatment. Microdialysis probes (3 mm deep in the forearm) were used to assess Dex-P, dexamethasone (Dex), and its metabolite (Dex-Met) concentrations. Skin perfusion was measured using laser Doppler flowmetry. MAIN OUTCOME MEASURE(S): Microdialysis samples were collected at baseline, at conclusion of treatment, and every 20 minutes posttreatment for 60 minutes. Samples were analyzed to determine Dex-Total (Dex-Total = Dex-P + Dex + Dex-Met). Skin perfusion was calculated as a percentage change from baseline. A mixed-design analysis of variance was used to determine Dex-Total and skin perfusion difference between groups overtime. RESULTS: There was no difference between groups (P = .476), but [Dex-Total] significantly increased over the course of the iontophoresis and posttreatment time (P < .001). Dex-P was measured in 18 of 24 participants with a mean concentration of 0.67 (1.09) µg/mL. Skin perfusion was significantly greater in the no ice treatment group (P = .002). Peak skin perfusion reached 27.74% (47.49%) and 117.39% (103.45%) from baseline for the ice and no ice groups, respectively. CONCLUSIONS: Ice massage prior to iontophoresis does not alter the tissue [Dex-Total] even with less skin perfusion.


Asunto(s)
Crioterapia/métodos , Dexametasona/análogos & derivados , Glucocorticoides/administración & dosificación , Iontoforesis/métodos , Masaje/métodos , Adulto , Análisis de Varianza , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Femenino , Glucocorticoides/farmacocinética , Humanos , Hielo , Masculino , Microdiálisis , Piel/metabolismo , Factores de Tiempo
13.
Carbohydr Polym ; 248: 116724, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32919548

RESUMEN

In this study, the ability of different beta-cyclodextrins to facilitate homogeneous dispersion of triamcinolone acetonide (TA) into chitosan membranes is assessed. Drug loading was assessed through atomic force microscopy (AFM), scanning electron microscopy (MEV-FEG), and X-ray diffraction analyses. Drug interactions with the co-polymer were investigated with Fourier transform infrared spectroscopy, thermal analyses. Swelling assay, and in vitro drug release experiment were used to assess TA release behavior. Undispersed particles of drug were observed to remain in the simple chitosan membranes. Hydroxypropyl-ß-cyclodextrin enabled the dispersion of TA into chitosan membranes and subsequent sustained drug release. In addition, the membrane performance as a drug delivery device is improved by adding specified amounts of the co-solvent triethanolamine. The experimental data presented in this study confirm the utility of our novel and alternative approach for obtaining a promising device for slow and controlled release of glucocorticoids, such as triamcinolone acetonide, for topical ulcerations.


Asunto(s)
Corticoesteroides/administración & dosificación , Quitosano/química , Preparaciones de Acción Retardada/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , beta-Ciclodextrinas/química , Corticoesteroides/química , Corticoesteroides/farmacocinética , Química Farmacéutica/métodos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Glucocorticoides/administración & dosificación , Glucocorticoides/química , Glucocorticoides/farmacocinética , Membranas Artificiales , Microscopía de Fuerza Atómica , Microscopía Electrónica de Rastreo , Polímeros/química , Solubilidad , Solventes/química , Espectroscopía Infrarroja por Transformada de Fourier , Triamcinolona/administración & dosificación , Triamcinolona/química , Triamcinolona/farmacocinética , Difracción de Rayos X
14.
J Ocul Pharmacol Ther ; 36(6): 404-409, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32678687

RESUMEN

Purpose: To determine the effect of particle size and viscosity of suspensions on topical ocular bioavailability of budesonide, a corticosteroid drug. Methods: Budesonide microparticle and nanoparticle (MP and NP) suspensions were prepared with or without homogenization and microfluidization. Using different grades of hydroxyl propyl methyl cellulose, low viscosity NP (NP-LV) and low and high viscosity MP (MP-LV and MP-HV) were prepared. Suspensions were characterized for particle size, viscosity, and osmolality. Budesonide suspensions were administered topically to rabbits and aqueous humor was collected and analyzed for budesonide. Budesonide Cmax, tmax, and the area under the concentration time curve (AUC (0-6h)) values were determined. The geometric mean ratio of AUC and bioequivalence was evaluated using a bootstrap method. Results: The particle sizes for NP and MP were ∼700 and 2,000 nm. The viscosities for low and HV formulations were ∼5 and 50 cP. The geometric mean budesonide Cmax values for the suspensions NP-LV, MP-LV, and MP-HV were 0.22, 0.22, and 0.31 µg/g, tmax values were 0.67, 0.60 and 0.53 h, and AUC0-6h values were 0.72, 0.53, and 0.95 µg h/g, respectively. Bootstrap analysis indicated that the 90% confidence intervals of the geometric mean ratio of AUC0-6h values were 1.00-1.74 (MP-HV vs. NP-LV), 0.57-0.96 (MP-LV vs. NP-LV), and 0.45-0.70 (MP-LV vs. MP-HV). Conclusions: The 3 budesonide suspensions assessed in this study were not bioequivalent. Results suggested that an increase in viscosity improves the bioavailability of budesonide from the microsuspension formulation.


Asunto(s)
Budesonida/farmacocinética , Composición de Medicamentos/métodos , Glucocorticoides/farmacocinética , Viscosidad/efectos de los fármacos , Administración Tópica , Animales , Área Bajo la Curva , Disponibilidad Biológica , Budesonida/administración & dosificación , Budesonida/efectos adversos , Sistemas de Liberación de Medicamentos/métodos , Glucocorticoides/administración & dosificación , Glucocorticoides/efectos adversos , Derivados de la Hipromelosa/química , Masculino , Modelos Animales , Nanopartículas/administración & dosificación , Nanopartículas/química , Tamaño de la Partícula , Conejos , Suspensiones/administración & dosificación , Suspensiones/química , Equivalencia Terapéutica
15.
Drug Metab Dispos ; 48(9): 811-818, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32601175

RESUMEN

Blood and multitissue concentration-time profiles for dexamethasone (DEX), a synthetic corticosteroid, were measured in male rats after subcutaneous bolus and infusion dosing. A physiologically based pharmacokinetics (PBPK) model was applied for 12 measured tissues. Tissue partition coefficients (K p ) and metabolic clearance were assessed from infusion studies. Blood cell to plasma partitioning (0.664) and plasma free fraction (0.175) for DEX were found to be moderate. DEX was extensively partitioned into liver (K p = 6.76), whereas the calculated K p values of most tissues ranged between 0.1 and 1.5. Despite the moderate lipophilicity of DEX (log P = 1.8), adipose exhibited very limited distribution (K p = 0.17). Presumably due to P-glycoprotein-mediated efflux, DEX concentrations were very low in brain compared with its expected high permeability. Infusion studies yielded K p values from male and female rats at steady state that were similar. In silico K p values calculated for different tissues by using GastroPlus software were similar to in vivo values except for adipose and liver. Glucocorticoid receptors are found in diverse tissues, and these PBPK modeling results may help provide exposure profiles driving pharmacodynamic effects of DEX. SIGNIFICANCE STATEMENT: Our physiologically based pharmacokinetics model describes the experimentally determined tissue and plasma dexamethasone (DEX) pharmacokinetics (PK) profiles in rats reasonably well. This model can serve for further investigation of DEX tissue distribution in rats as the PK driving force for PD effects in different tissues. No major sex differences were found for DEX tissue distribution. Knowledge gained in this study may be translatable to higher-order species including humans.


Asunto(s)
Dexametasona/farmacocinética , Glucocorticoides/farmacocinética , Modelos Biológicos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Simulación por Computador , Dexametasona/administración & dosificación , Evaluación Preclínica de Medicamentos , Femenino , Glucocorticoides/administración & dosificación , Infusiones Subcutáneas , Masculino , Modelos Animales , Ratas , Factores Sexuales , Distribución Tisular
16.
Clin Pharmacol Ther ; 108(5): 1026-1035, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32394434

RESUMEN

Despite antenatal corticosteroids therapy, respiratory distress syndrome (RDS) is still a leading cause of neonatal morbidity and mortality in premature newborns. To date, the relationship between in utero fetal drug exposure and occurrence of RDS remains poorly evaluated. This study aims to describe the pharmacokinetics of betamethasone in pregnant women and to evaluate the transplacental drug transfer and administration scheme for the prevention of RDS. Pregnant women > 27 weeks' gestation and who received at least a single dose of betamethasone for prevention of RDS were enrolled. Maternal, cord blood, and amniotic fluid betamethasone time-courses were analyzed using the Monolix software. A total of 220 maternal blood, 56 cord blood, and 26 amniotic fluid samples were described by a two-compartment model with two effect compartments linked by rate transfer constants. Apparent clearances and volumes of distribution parameters were allometrically scaled for a 70 kg third trimester pregnant woman. The impact of a twin pregnancy was found to increase maternal clearance by 28%. Using a fetal-to-mother exposure ratio, the median (95% confidence interval (CI)) transplacental transfer of betamethasone was estimated to 35% (95% CI 0.11-0.67). After adjustment for gestational age and twin pregnancy, RDS was found to be associated to the time spent in utero below quantifiable concentrations (i.e., < 1 ng/mL): odds ratio of 1.10 (95% CI 1.01-1.19) per day increase (P < 0.05). Trying to take into account both efficacy and safety, we simulated different dosing schemes in order to maintain a maximum of fetuses above 1 ng/mL without exceeding the total standard dose.


Asunto(s)
Betametasona/análogos & derivados , Glucocorticoides/farmacocinética , Intercambio Materno-Fetal , Síndrome de Dificultad Respiratoria del Recién Nacido/prevención & control , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Adolescente , Adulto , Líquido Amniótico/metabolismo , Betametasona/administración & dosificación , Betametasona/sangre , Betametasona/farmacocinética , Cálculo de Dosificación de Drogas , Monitoreo de Drogas , Femenino , Sangre Fetal/metabolismo , Francia , Edad Gestacional , Glucocorticoides/administración & dosificación , Glucocorticoides/sangre , Humanos , Recién Nacido , Inyecciones Intramusculares , Masculino , Modelos Biológicos , Farmacogenética , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Embarazo , Estudios Prospectivos , Síndrome de Dificultad Respiratoria del Recién Nacido/diagnóstico , Síndrome de Dificultad Respiratoria del Recién Nacido/fisiopatología , Adulto Joven
17.
Drug Test Anal ; 12(8): 1087-1101, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32436346

RESUMEN

Corticosteroids are potent anti-inflammatory drugs and as such are commonly administered to performance and racehorses. The objectives of the current study were to describe blood and urine concentrations and the pharmacokinetics and effects on cortisol and inflammatory mediator concentrations, following intravenous and oral administration to 12 exercised horses. Horses received an intravenous administration of 40 mg of dexamethasone sodium phosphate and 20 mg of dexamethasone tablets with a 4 week washout in between administrations. Blood and urine samples were collected prior to and for up to 96 hours post drug administration. Whole blood samples were collected at various time points and challenged with lipopolysaccharide or calcium ionophore to induce ex vivo synthesis of eicosanoids. The concentrations of dexamethasone and eicosanoids were measured using LC-MS/MS and the concentrations from both routes of administration fit simultaneously using a three-compartment pharmacokinetic model. A turnover model with inhibition of Kin gave an adequate fit to the dexamethasone-cortisol PKPD data. Serum and urine dexamethasone concentrations were at the limit of quantitation at 96 and 48 hours post administration, respectively. The volume of distribution, systemic clearance, and terminal half-life was 0.907 L/kg, 7.89 mL/h/kg, and 1.34 h, respectively. The IC50 for cortisol suppression was 0.007 ng/mL. Stimulation of dexamethasone treated blood with lipopolysaccharide and calcium ionophore resulted in an inhibition of inflammatory biomarker production for a prolonged period of time post drug administration. The results of this study suggest that dexamethasone has a prolonged anti-inflammatory effect following intravenous or oral administration to horses.


Asunto(s)
Antiinflamatorios/administración & dosificación , Dexametasona/análogos & derivados , Glucocorticoides/administración & dosificación , Modelos Biológicos , Administración Intravenosa , Administración Oral , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Cromatografía Liquida/métodos , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Dexametasona/farmacología , Glucocorticoides/farmacocinética , Glucocorticoides/farmacología , Semivida , Caballos , Inflamación/tratamiento farmacológico , Mediadores de Inflamación/metabolismo , Concentración 50 Inhibidora , Espectrometría de Masas en Tándem/métodos , Factores de Tiempo , Distribución Tisular
18.
Clin Exp Dermatol ; 45(5): 549-554, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32410250

RESUMEN

BACKGROUND: Wet dressings combined with topical corticosteroids are beneficial for patients with generalized and refractory dermatosis; however, to our knowledge, serum levels after topical corticosteroid absorption during intensive therapy have not been reported previously. AIM: To examine serum levels of triamcinolone acetonide (TAC) after topical corticosteroid application during intensive wet-dressing therapy. METHODS: We performed a retrospective study of adult patients admitted for inpatient wet-dressing therapy from 7 November 2015 to 24 June 2016. Data were collected on sex, age, body surface area, TAC serum levels, number of wet-dressing changes after 24 and 48 h, and type of wet dressing. RESULTS: In total, 29 patients (14 men, 15 women) were assessed. Median [interquartile range (IQR)] age was 57 years (51.5-67.0 years) and involved body surface area was 1.98 m2 (1.88-2.15) m2 . Before the 24-hour blood draw, patients had received 1-3 dressing changes. Median (IQR) TAC level at 24 h was 0.33 µg/dL (0.20-0.58 µg/dL), with no significant difference noted between the number of dressing changes and TAC serum level. At 48 h, results of a serum TAC test were available for 22 patients with 2-6 dressing changes. Mean (IQR) serum level was 0.30 µg/dL (0.30-0.87 µg/dL). For each additional dressing change, there was an estimated 0.21 µg/dL increase in TAC serum level (95% CI 0.11-0.31; P < 0.001). TAC serum level was not significantly associated with sex, age, body surface area or dressing type. CONCLUSIONS: Intensive, inpatient wet-dressing therapy is associated with detectable TAC serum levels. However, we suspect that topical TAC has a primarily local therapeutic effect on the skin.


Asunto(s)
Vendajes , Glucocorticoides/sangre , Enfermedades de la Piel/tratamiento farmacológico , Triamcinolona Acetonida/sangre , Administración Tópica , Anciano , Femenino , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacocinética , Hospitalización , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Triamcinolona Acetonida/administración & dosificación , Triamcinolona Acetonida/farmacocinética
19.
J Athl Train ; 55(6): 628-635, 2020 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-32320285

RESUMEN

CONTEXT: The effect of ultrasound frequency on phonophoresis drug delivery in humans is unknown. OBJECTIVE: To determine if a low (45-kHz) or high (1-MHz) frequency delivered a higher dexamethasone (Dex) concentration through the skin. DESIGN: Controlled laboratory study. SETTING: Laboratory. PATIENTS OR OTHER PARTICIPANTS: A total of 40 healthy men between the ages of 18 and 45 years (age = 23.1 ± 2.6 years, height = 176.1 ± 7.2 cm, mass = 88.5 ± 19.4 kg, posterior calf subcutaneous thickness measured using musculoskeletal ultrasound imaging = 0.6 ± 0.2 cm). INTERVENTION(S): Participants were randomly assigned to 1 of 4 groups (ultrasound frequency at microdialysis probe depth): (1) 45-kHz frequency at 1 mm, (2) 45-kHz frequency at 4 mm, (3) 1-MHz frequency at 1 mm, or (4) 1-MHz frequency at 4 mm (n = 10 in each group). Three linear microdialysis probes were inserted at the desired tissue depth. We rubbed dexamethasone sodium phosphate (Dex-P) into the skin and then applied a 15-minute phonophoresis treatment. MAIN OUTCOME MEASURE(S): Dialysate was collected during the treatment and 60 minutes posttreatment and analyzed for Dex-P, Dex, and the metabolite form of Dex. The sum of the 3 analytes was calculated as total dexamethasone (Dex-total), and differences between the 45-kHz and 1-MHz treatment groups were determined by a repeated-measures analysis of variance. RESULTS: At 1 mm, 3 (30%) participants in the 45-kHz and 4 (40%) participants in the 1-MHz group had measurable levels of Dex-P. Total dexamethasone increased after the treatment ceased, independent of ultrasound frequency (P < .001), with a trend of the 45-kHz treatment to produce a greater increase in drug concentration (P = .006). At 4 mm, 5 (50%) participants in the 45-kHz and 1 (10%) participant in the 1-MHz group had measurable levels of Dex-P. We observed no difference in Dex-total concentration between treatment groups at 4 mm (P = .72). CONCLUSIONS: Phonophoresis provided a mechanism for Dex-total delivery at the 1- and 4-mm tissue depths. However, the effectiveness of the ultrasound frequencies varied between the 2 measured tissue depths.


Asunto(s)
Dexametasona , Fonoforesis/métodos , Adulto , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Disponibilidad Biológica , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Monitoreo de Drogas/métodos , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacocinética , Voluntarios Sanos , Humanos , Masculino , Evaluación de Resultado en la Atención de Salud , Factores de Tiempo
20.
Artículo en Inglés | MEDLINE | ID: mdl-32202079

RESUMEN

Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects 0.5-1% of the world population. Current treatments include on one hand non-steroidal anti-inflammatory drugs and glucocorticoids (GCs) for treating pain and on the other hand disease-modifying anti-rheumatic drugs such as methotrexate, Janus kinase inhibitors or biologics such as antibodies targeting mainly cytokine expression. More recently, nucleic acids such as siRNA, miRNA, or anti-miRNA have shown strong potentialities for the treatment of RA. This review discusses the way nanomedicines can target GCs and nucleic acids to inflammatory sites, increase drug penetration within inflammatory cells, achieve better subcellular distribution and finally protect drugs against degradation. For GCs such a targeting effect would allow the treatment to be more effective at lower doses and to reduce the administration frequency as well as to induce much fewer side-effects. In the case of nucleic acids, particularly siRNA, knocking down proteins involved in RA, could importantly be facilitated using nanomedicines. Finally, the combination of both siRNA and GCs in the same carrier allowed for the same cell to target both the GCs receptor as well as any other signaling pathway involved in RA. Nanomedicines appear to be very promising for the delivery of conventional and novel drugs in RA therapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures.


Asunto(s)
Antirreumáticos , Artritis Reumatoide/tratamiento farmacológico , Glucocorticoides , Nanomedicina , Ácidos Nucleicos , Animales , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapéutico , Glucocorticoides/farmacocinética , Glucocorticoides/uso terapéutico , Humanos , Ratones , Ácidos Nucleicos/farmacocinética , Ácidos Nucleicos/uso terapéutico , ARN Interferente Pequeño , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...