Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Alzheimers Dement ; 19(6): 2287-2297, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36454709

RESUMEN

INTRODUCTION: A quantitative model of Alzheimer's disease (AD) based on the amyloid/tau/neurodegeneration biomarker framework (Q-ATN model) was developed to sequentially link amyloid positron emission tomography (PET), tau PET, medial temporal cortical thickness, and clinical outcome (Clinical Dementia Rating - Sum of Boxes; CDR-SB). METHODS: Published data and biologically plausible mechanisms were used to construct, calibrate, and validate the model. Clinical trial simulations were performed for different anti-amyloid antibodies, including a 5-year simulation of subcutaneous gantenerumab treatment. RESULTS: The simulated time-course of biomarkers and CDR-SB was consistent with natural history studies and described the effects of several anti-amyloid antibodies observed in trials with positive and negative (or non-significant) outcomes. The 5-year simulation predicts that the beneficial effects of continued anti-amyloid treatment should increase markedly over time. DISCUSSION: The Q-ATN model offers a novel approach for linking amyloid PET to CDR-SB, and provides theoretical support for the potential clinical benefit of anti-amyloid therapy. HIGHLIGHTS: A semi-mechanistic model was developed to link amyloid/tau/neurodegeneration biomarkers to clinical outcome (Q-ATN model). The Q-ATN model describes the disease progression seen in natural history studies. Model simulations agree well with mean data from the aducanumab EMERGE study. A 5-year simulation of gantenerumab predicts greater benefit with longer treatment.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Amiloide , Tomografía de Emisión de Positrones , Biomarcadores , Proteínas Amiloidogénicas , Péptidos beta-Amiloides , Proteínas tau
2.
Mol Pharm ; 17(2): 695-709, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31876425

RESUMEN

Therapeutic antibodies administered intravitreally are the current standard of care to treat retinal diseases. The ocular half-life (t1/2) is a key determinant of the duration of target suppression. To support the development of novel, longer-acting drugs, a reliable determination of t1/2 is needed together with an improved understanding of the factors that influence it. A model-based meta-analysis was conducted in humans and nonclinical species (rat, rabbit, monkey, and pig) to determine consensus values for the ocular t1/2 of IgG antibodies and Fab fragments. Results from multiple literature and in-house pharmacokinetic studies are presented within a mechanistic framework that assumes diffusion-controlled drug elimination from the vitreous. Our analysis shows, both theoretically and experimentally, that the ocular t1/2 increases in direct proportion to the product of the hydrodynamic radius of the macromolecule (3.0 nm for Fab and 5.0 nm for IgG) and the square of the radius of the vitreous globe, which varies approximately 24-fold from the rat to the human. Interspecies differences in the proportionality factors are observed and discussed in mechanistic terms. In addition, mathematical formulae are presented that allow prediction of the ocular t1/2 for molecules of interest. The utility of these formulae is successfully demonstrated in case studies of aflibercept, brolucizumab, and PEGylated Fabs, where the predicted ocular t1/2 values are found to be in reasonable agreement with the experimental data available for these molecules.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Productos Biológicos/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Inmunoglobulina G/administración & dosificación , Inyecciones Intravítreas/métodos , Receptores de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Productos Biológicos/farmacocinética , Difusión , Semivida , Haplorrinos , Humanos , Hidrodinámica , Conejos , Ratas , Proteínas Recombinantes de Fusión/farmacocinética , Enfermedades de la Retina/tratamiento farmacológico , Porcinos , Distribución Tisular , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/metabolismo
3.
Mol Pharm ; 15(7): 2770-2784, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29734810

RESUMEN

Neovascular age-related macular degeneration (wet AMD) results from the pathological angiogenesis of choroidal capillaries, which leak fluid within or below the macular region of the retina. The current standard of care for treating wet AMD utilizes intravitreal injections of anti-VEGF antibodies or antibody fragments to suppress ocular vascular endothelial growth factor (VEGF) levels. While VEGF suppression has been demonstrated in wet AMD patients by serial measurements of free-VEGF concentrations in aqueous humor samples, it is presumed that anti-VEGF molecules also permeate across the inner limiting membrane (ILM) of the retina as well as the retinal pigmented epithelium (RPE) and suppress VEGF levels in the retina and/or choroidal regions. The latter effects are inferred from serial optical coherence tomography (OCT) measurements of fluid in the retinal and sub-retinal spaces. In order to gain theoretical insights to the dynamics of retinal levels of free-VEGF following intravitreal injection of anti-VEGF molecules, we have extended our previous two-compartment pharmacokinetic/pharmacodynamic (PK/PD) model of ranibizumab-VEGF suppression in vitreous and aqueous humors to a three-compartment model that includes the retinal compartment. In the new model, reference values for the macromolecular permeability coefficients between retina and vitreous ( pILM) and between retina and choroid ( pRPE) were estimated from PK data obtained in rabbit. With these values, the three-compartment model was used to re-analyze the aqueous humor levels of free-VEGF obtained in wet AMD patients treated with ranibizumab and to compare them to the simulated retinal levels of free-VEGF, including the observed variability in PK and PD. We have also used the model to explore the impact of varying pILM and pRPE to assess the case in which an anti-VEGF molecule is impermeable to the ILM and to assess the potential effects of AMD pathology on the RPE barrier. Our simulations show that, for the reference values of pILM and pRPE, the simulated duration of VEGF suppression in the retina is approximately 50% shorter than the observed duration of VEGF suppression in the aqueous humor, a finding that may explain the short duration of suppressed disease activity in the "high anti-VEGF demand" patients reported by Fauser and Muether ( Br. J. Ophthalmol. 2016, 100, 1494-1498 ). At 10-fold lower values of pRPE, the durations of VEGF suppression in the retina and aqueous humor are comparable. Lastly we have used the model to explore the impact of dose and binding parameters on the duration and depth of VEGF suppression in the aqueous and retinal compartments. Our simulations with the three-compartment PK/PD model provide new insights into inter-patient variability in response to anti-VEGF therapy and offer a mechanistic framework for developing treatment regimens and molecules that may prolong the duration of retinal VEGF suppression.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Ranibizumab/farmacología , Retina/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Degeneración Macular Húmeda/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Humor Acuoso/efectos de los fármacos , Humor Acuoso/metabolismo , Humanos , Inyecciones Intravítreas , Modelos Biológicos , Ranibizumab/uso terapéutico , Retina/efectos de los fármacos , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Cuerpo Vítreo/efectos de los fármacos , Cuerpo Vítreo/metabolismo , Degeneración Macular Húmeda/patología
4.
J Lipid Res ; 58(7): 1325-1337, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28442497

RESUMEN

We developed an in silico mathematical model of retinal cholesterol (Ch) dynamics (RCD) to quantify the physiological rate of Ch turnover in the rod outer segment (ROS), the lipoprotein transport mechanisms by which Ch enters and leaves the outer retina, and the rates of drusen growth and macrophage-mediated clearance in dry age-related macular degeneration. Based on existing experimental data and mechanistic hypotheses, we estimated the Ch turnover rate in the ROS to be 1-6 pg/mm2/min, dependent on the rate of Ch recycling in the outer retina, and found comparable rates for LDL receptor-mediated endocytosis of Ch by the retinal pigment epithelium (RPE), ABCA1-mediated Ch transport from the RPE to the outer retina, ABCA1-mediated Ch efflux from the RPE to the choroid, and the secretion of 70 nm ApoB-Ch particles from the RPE. The drusen growth rate is predicted to increase from 0.7 to 4.2 µm/year in proportion to the flux of ApoB-Ch particles. The rapid regression of drusen may be explained by macrophage-mediated clearance if the macrophage density reaches ∼3,500 cells/mm2 The RCD model quantifies retinal Ch dynamics and suggests that retinal Ch turnover and recycling, ApoB-Ch particle efflux, and macrophage-mediated clearance may explain the dynamics of drusen growth and regression.


Asunto(s)
Colesterol/metabolismo , Simulación por Computador , Degeneración Macular/metabolismo , Retina/metabolismo , Transporte Biológico , Humanos , Degeneración Macular/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo
5.
Mol Pharm ; 14(8): 2690-2696, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28631484

RESUMEN

Intravitreally (IVT) injected macromolecules for the treatment of age-related macular degeneration must permeate through the inner limiting membrane (ILM) into the retina and through the retinal pigment epithelium (RPE) to enter the choroid. A quantitative understanding of intraocular transport mechanisms, elimination pathways, and the effect of molecular size is currently incomplete. We present a semimechanistic, 3-compartment (retina, vitreous, and aqueous) pharmacokinetic (PK) model, expressed using linear ordinary differential equations (ODEs), to describe the molecular concentrations following a single IVT injection. The model was fit to experimental rabbit data, with Fab, Fc, IgG, and IgG null antibodies and antibody fragments, to estimate key ocular pharmacokinetic parameters. The model predicts an ocular half-life, t1/2, which is the same for all compartments and dependent on the hydrodynamic radius (Rh) of the respective molecules, consistent with observations from the experimental data. Estimates of the permeabilities of the RPE and ILM are derived for Rh values ranging from 2.5 to 4.9 nm, and are found to be in good agreement with ex-vivo measurements from bovine eyes. We show that the ratio of these permeabilities largely determines the ratio of the molecular concentrations in the retina and vitreal compartments and their dependence on Rh. The model further provides estimates for the ratio of fluxes corresponding to the elimination pathways from the eye, i.e., aqueous humor to retina/choroid, which increase from 5:1 to 7:1 as Rh decreases. Our semimechanistic model provides a quantitative framework for interpreting ocular PK and the effects of molecule size on rate-determining parameters. We have shown that intraocular permeabilities can be reasonably estimated from 3-compartment ocular PK data and can determine how these parameters influence the half-life, retinal permeation, and elimination of intravitreally injected molecules from the eye.


Asunto(s)
Anticuerpos/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Anticuerpos/administración & dosificación , Coroides/metabolismo , Inmunoglobulina G/metabolismo , Inyecciones Intravítreas , Modelos Teóricos , Conejos , Cuerpo Vítreo/metabolismo
6.
J Lipid Res ; 57(1): 46-55, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26522778

RESUMEN

The recent failures of cholesteryl ester transport protein inhibitor drugs to decrease CVD risk, despite raising HDL cholesterol (HDL-C) levels, suggest that pharmacologic increases in HDL-C may not always reflect elevations in reverse cholesterol transport (RCT), the process by which HDL is believed to exert its beneficial effects. HDL-modulating therapies can affect HDL properties beyond total HDL-C, including particle numbers, size, and composition, and may contribute differently to RCT and CVD risk. The lack of validated easily measurable pharmacodynamic markers to link drug effects to RCT, and ultimately to CVD risk, complicates target and compound selection and evaluation. In this work, we use a systems pharmacology model to contextualize the roles of different HDL targets in cholesterol metabolism and provide quantitative links between HDL-related measurements and the associated changes in RCT rate to support target and compound evaluation in drug development. By quantifying the amount of cholesterol removed from the periphery over the short-term, our simulations show the potential for infused HDL to treat acute CVD. For the primary prevention of CVD, our analysis suggests that the induction of ApoA-I synthesis may be a more viable approach, due to the long-term increase in RCT rate.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , HDL-Colesterol/metabolismo , Hipolipemiantes/farmacología , Apolipoproteína A-I/biosíntesis , Apolipoproteína A-I/efectos de los fármacos , Apolipoproteína A-I/metabolismo , Transporte Biológico , Biomarcadores/sangre , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , Colesterol/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Humanos , Lipoproteínas HDL/metabolismo , Modelos Biológicos , Quinazolinas/farmacología , Quinazolinonas , Factores de Riesgo , Regulación hacia Arriba
7.
Mol Pharm ; 13(9): 2941-50, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-26726925

RESUMEN

Intravitreal injection of anti-VEGF (vascular endothelial growth factor) antibodies or antibody fragments has been shown to be a highly effective treatment for neovascular age-related macular degeneration (wet AMD). The ocular half-life (t1/2) of these large molecules, determined in ocular fluids or derived from serum levels, varies with molecular size and is larger in humans than in preclinical animal species. The high affinity binding of VEGF to these molecules lowers the free concentration of VEGF and reduces its occupancy on VEGF receptors in ocular tissues. To understand the biophysical determinants of t1/2 for anti-VEGF antibodies and the time-course of VEGF in ocular fluids, we developed a mechanistic model of intravitreal pharmacokinetics (IVT PK) for anti-VEGF antibodies and combined it with a mechanistic model of the pharmacodynamics (RVR PD) of VEGF suppression by ranibizumab, an anti-VEGF recombinant, humanized monoclonal antibody fragment (Fab). Our IVT PK model predicts that the ocular t1/2 of a large molecule will be approximately four-times the calculated value of its vitreous diffusion time (Tdiff), defined as rvit(2)/6D, where rvit is the radius of the vitreous chamber in that species (modeled as a sphere), and D is the diffusion coefficient of the molecule in physiological saline at 37 °C obtained from the Stokes-Einstein relation. This prediction is verified from a compilation of data and calculations on various large molecules in the human, monkey, rabbit, and rat and is consistent with the reported t1/2 values of ranibizumab in humans (mean value 7.9 days) and the calculated Tdiff of 1.59 days. Our RVR PD model is based on the publication of Saunders et al. (Br. J. Ophthalmol. 2015, 99, 1554-1559) who reported data on the time-course of VEGF levels in aqueous humor samples obtained from 31 patients receiving ranibizumab treatment for wet AMD and developed a compartmental mathematical model to describe the VEGF suppression profiles. We modified Saunders' model with the known 2:1 stoichiometry of ranibizumab-VEGF binding and included the association and dissociation kinetics of the binding reactions. Using the RVR PD model, we reanalyzed Saunders' data to estimate the in vivo dissociation constant (KD) between ranibizumab and VEGF. Our analysis demonstrates the delicate interrelationship between the in vivo KD value and the intravitreal half-life and yields an in vivo KD estimate that is appreciably larger than the in vitro KD estimates reported in the literature. Potential explanations for the difference between the in vivo and in vitro KD values, which appear to reflect the different methodologies and experimental conditions, are discussed. We conclude that the combined mechanistic model of IVT PK and RVR PD provides a useful framework for simulating the effects of dose, KD, and the molecular weight of VEGF-binding molecules on the duration of VEGF suppression.


Asunto(s)
Ranibizumab/farmacocinética , Ranibizumab/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Degeneración Macular Húmeda/tratamiento farmacológico , Degeneración Macular Húmeda/metabolismo , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacocinética , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Haplorrinos , Humanos , Inyecciones Intravítreas , Cinética , Modelos Teóricos , Conejos , Ranibizumab/administración & dosificación , Ratas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
8.
PLoS Comput Biol ; 10(3): e1003509, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24625468

RESUMEN

High-density lipoprotein (HDL) is believed to play an important role in lowering cardiovascular disease (CVD) risk by mediating the process of reverse cholesterol transport (RCT). Via RCT, excess cholesterol from peripheral tissues is carried back to the liver and hence should lead to the reduction of atherosclerotic plaques. The recent failures of HDL-cholesterol (HDL-C) raising therapies have initiated a re-examination of the link between CVD risk and the rate of RCT, and have brought into question whether all target modulations that raise HDL-C would be atheroprotective. To help address these issues, a novel in-silico model has been built to incorporate modern concepts of HDL biology, including: the geometric structure of HDL linking the core radius with the number of ApoA-I molecules on it, and the regeneration of lipid-poor ApoA-I from spherical HDL due to remodeling processes. The ODE model has been calibrated using data from the literature and validated by simulating additional experiments not used in the calibration. Using a virtual population, we show that the model provides possible explanations for a number of well-known relationships in cholesterol metabolism, including the epidemiological relationship between HDL-C and CVD risk and the correlations between some HDL-related lipoprotein markers. In particular, the model has been used to explore two HDL-C raising target modulations, Cholesteryl Ester Transfer Protein (CETP) inhibition and ATP-binding cassette transporter member 1 (ABCA1) up-regulation. It predicts that while CETP inhibition would not result in an increased RCT rate, ABCA1 up-regulation should increase both HDL-C and RCT rate. Furthermore, the model predicts the two target modulations result in distinct changes in the lipoprotein measures. Finally, the model also allows for an evaluation of two candidate biomarkers for in-vivo whole-body ABCA1 activity: the absolute concentration and the % lipid-poor ApoA-I. These findings illustrate the potential utility of the model in drug development.


Asunto(s)
Biomarcadores/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Transportador 1 de Casete de Unión a ATP/metabolismo , Algoritmos , Teorema de Bayes , Transporte Biológico , Calibración , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Biología Computacional/métodos , Femenino , Regulación de la Expresión Génica , Humanos , Masculino
10.
JAMA ; 314(6): 570-81, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26262795

RESUMEN

IMPORTANCE: Testosterone use in older men is increasing, but its long-term effects on progression of atherosclerosis are unknown. OBJECTIVE: To determine the effect of testosterone administration on subclinical atherosclerosis progression in older men with low or low-normal testosterone levels. DESIGN, SETTING, AND PARTICIPANTS: Testosterone's Effects on Atherosclerosis Progression in Aging Men (TEAAM) was a placebo-controlled, double-blind, parallel-group randomized trial involving 308 men 60 years or older with low or low-normal testosterone levels (100-400 ng/dL; free testosterone <50 pg/mL), recruited at 3 US centers. Recruitment took place between September 2004 and February 2009; the last participant completed the study in May 2012. INTERVENTIONS: One hundred fifty-six participants were randomized to receive 7.5 g of 1% testosterone and 152 were randomized to receive placebo gel packets daily for 3 years. The dose was adjusted to achieve testosterone levels between 500 and 900 ng/dL. MAIN OUTCOMES AND MEASURES: Coprimary outcomes included common carotid artery intima-media thickness and coronary artery calcium; secondary outcomes included sexual function and health-related quality of life. RESULTS: Baseline characteristics were similar between groups: patients were a mean age of 67.6 years; 42% had hypertension; 15%, diabetes; 15%, cardiovascular disease; and 27%, obesity. The rate of change in intima-media thickness was 0.010 mm/year in the placebo group and 0.012 mm/year in the testosterone group (mean difference adjusted for age and trial site, 0.0002 mm/year; 95% CI, -0.003 to 0.003, P = .89). The rate of change in the coronary artery calcium score was 41.4 Agatston units/year in the placebo group and 31.4 Agatston units/year in the testosterone group (adjusted mean difference, -10.8 Agatston units/year; 95% CI, -45.7 to 24.2; P = .54). Changes in intima-media thickness or calcium scores were not associated with change in testosterone levels among individuals assigned to receive testosterone. Sexual desire, erectile function, overall sexual function scores, partner intimacy, and health-related quality of life did not differ significantly between groups. Hematocrit and prostate-specific antigen levels increased more in testosterone group. CONCLUSIONS AND RELEVANCE: Among older men with low or low-normal testosterone levels, testosterone administration for 3 years vs placebo did not result in a significant difference in the rates of change in either common carotid artery intima-media thickness or coronary artery calcium nor did it improve overall sexual function or health-related quality of life. Because this trial was only powered to evaluate atherosclerosis progression, these findings should not be interpreted as establishing cardiovascular safety of testosterone use in older men. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00287586.


Asunto(s)
Aterosclerosis/inducido químicamente , Grosor Intima-Media Carotídeo , Testosterona/efectos adversos , Anciano , Calcio/análisis , Vasos Coronarios/química , Progresión de la Enfermedad , Método Doble Ciego , Estado de Salud , Humanos , Hipertensión , Masculino , Persona de Mediana Edad , Obesidad , Calidad de Vida , Disfunciones Sexuales Fisiológicas/complicaciones , Disfunciones Sexuales Fisiológicas/etiología , Testosterona/sangre , Testosterona/deficiencia , Testosterona/uso terapéutico , Resultado del Tratamiento
11.
N Engl J Med ; 363(2): 109-22, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20592293

RESUMEN

BACKGROUND: Testosterone supplementation has been shown to increase muscle mass and strength in healthy older men. The safety and efficacy of testosterone treatment in older men who have limitations in mobility have not been studied. METHODS: Community-dwelling men, 65 years of age or older, with limitations in mobility and a total serum testosterone level of 100 to 350 ng per deciliter (3.5 to 12.1 nmol per liter) or a free serum testosterone level of less than 50 pg per milliliter (173 pmol per liter) were randomly assigned to receive placebo gel or testosterone gel, to be applied daily for 6 months. Adverse events were categorized with the use of the Medical Dictionary for Regulatory Activities classification. The data and safety monitoring board recommended that the trial be discontinued early because there was a significantly higher rate of adverse cardiovascular events in the testosterone group than in the placebo group. RESULTS: A total of 209 men (mean age, 74 years) were enrolled at the time the trial was terminated. At baseline, there was a high prevalence of hypertension, diabetes, hyperlipidemia, and obesity among the participants. During the course of the study, the testosterone group had higher rates of cardiac, respiratory, and dermatologic events than did the placebo group. A total of 23 subjects in the testosterone group, as compared with 5 in the placebo group, had cardiovascular-related adverse events. The relative risk of a cardiovascular-related adverse event remained constant throughout the 6-month treatment period. As compared with the placebo group, the testosterone group had significantly greater improvements in leg-press and chest-press strength and in stair climbing while carrying a load. CONCLUSIONS: In this population of older men with limitations in mobility and a high prevalence of chronic disease, the application of a testosterone gel was associated with an increased risk of cardiovascular adverse events. The small size of the trial and the unique population prevent broader inferences from being made about the safety of testosterone therapy. (ClinicalTrials.gov number, NCT00240981.)


Asunto(s)
Enfermedades Cardiovasculares/inducido químicamente , Testosterona/efectos adversos , Administración Cutánea , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Prueba de Esfuerzo , Geles , Humanos , Hiperlipidemias/complicaciones , Hipertensión/complicaciones , Estimación de Kaplan-Meier , Modelos Logísticos , Masculino , Fuerza Muscular/efectos de los fármacos , Obesidad/complicaciones , Factores de Riesgo , Testosterona/sangre , Testosterona/deficiencia , Testosterona/uso terapéutico , Caminata
12.
Clin Chem ; 59(6): 949-58, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23426429

RESUMEN

BACKGROUND: HDL size and composition vary among individuals and may be associated with cardiovascular disease and diabetes. We investigated the theoretical relationship between HDL size and composition using an updated version of the spherical model of lipoprotein structure proposed by Shen et al. (Proc Natl Acad Sci U S A 1977;74:837-41.) and compared its predictions with experimental data from the Women's Health Study (WHS). METHODS: The Shen model was updated to predict the relationship between HDL diameter and the ratio of HDL-cholesterol (HDL-C) to apolipoprotein A-I (ApoA-I) plasma concentrations (HDL-C/ApoA-I ratio). In the WHS (n = 26 772), nuclear magnetic resonance spectroscopy (NMR) was used to measure the mean HDL diameter (d(mean,NMR)) and particle concentration (HDL-P); HDL-C and ApoA-I (mg/dL) were measured by standardized assays. RESULTS: The updated Shen model predicts a quasilinear increase of HDL diameter with the HDL-C/ApoA-I ratio, consistent with the d(mean,NMR) values from WHS, which ranged between 8.0 and 10.8 nm and correlated positively with the HDL-C/ApoA-I ratio (r = 0.608, P < 2.2 × 10(-16)). The WHS data were further described by a linear regression equation: d(WHS) = 4.66 nm + 12.31(HDL-C/Apo-I), where d(WHS) is expressed in nanometers. The validity of this equation for estimating HDL size was assessed with data from cholesteryl ester transfer protein deficiency and pharmacologic inhibition. We also illustrate how HDL-P can be estimated from the HDL size and ApoA-I concentration. CONCLUSIONS: This study provides a large-scale experimental examination of the updated Shen model. The results offer new insights into HDL structure, composition and remodeling and suggest that the HDL-C/ApoA-I ratio might be a readily available biomarker for estimating HDL size and HDL-P.


Asunto(s)
Apolipoproteína A-I/química , HDL-Colesterol/química , Lipoproteínas HDL/química , Modelos Teóricos , Tamaño de la Partícula , Femenino , Humanos , Espectroscopía de Resonancia Magnética , Salud de la Mujer
13.
J Pharmacokinet Pharmacodyn ; 40(2): 129-42, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23307170

RESUMEN

HbA1c is the most commonly used biomarker for the adequacy of glycemic management in diabetic patients and a surrogate endpoint for anti-diabetic drug approval. In spite of an empirical description for the relationship between average glucose (AG) and HbA1c concentrations, obtained from the A1c-derived average glucose (ADAG) study by Nathan et al., a model for the non-steady-state relationship is still lacking. Using data from the ADAG study, we here develop such models that utilize literature information on (patho)physiological processes and assay characteristics. The model incorporates the red blood cell (RBC) aging description, and uses prior values of the glycosylation rate constant (KG), mean RBC life-span (LS) and mean RBC precursor LS obtained from the literature. Different hypothesis were tested to explain the observed non-proportional relationship between AG and HbA1c. Both an inverse dependence of LS on AG and a non-specificity of the National Glycohemoglobin Standardization Program assay used could well describe the data. Both explanations have mechanistic support and could be incorporated, alone or in combination, in models allowing prediction of the time-course of HbA1c changes associated with changes in AG from, for example dietary or therapeutic interventions, and vice versa, to infer changes in AG from observed changes in HbA1c. The selection between the alternative mechanistic models require gathering of new information.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Hemoglobina Glucada/metabolismo , Diabetes Mellitus/sangre , Eritrocitos/metabolismo , Glicosilación , Humanos
14.
Ann Intern Med ; 157(10): 681-91, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23165659

RESUMEN

BACKGROUND: Erectile dysfunction and low testosterone levels frequently occur together. OBJECTIVE: To determine whether addition of testosterone to sildenafil therapy improves erectile response in men with erectile dysfunction and low testosterone levels. DESIGN: Randomized, double-blind, parallel, placebo-controlled trial. (ClinicalTrials.gov registration number: NCT00512707) SETTING: Outpatient academic research center. PARTICIPANTS: Men aged 40 to 70 years with scores of 25 or less for the erectile function domain (EFD) of the International Index of Erectile Function, total testosterone levels less than 11.45 nmol/L (<330 ng/dL), or free testosterone levels less than 173.35 pmol/L (<50 pg/mL). INTERVENTION: Sildenafil dose was optimized, and 140 participants were then randomly assigned to 14 weeks of daily transdermal gel that contained 10-g testosterone for 70 participants and placebo for the remaining 70 participants. All participants were included in the primary analysis, although 10 in the testosterone group and 12 in the placebo group did not complete the study. RESULTS: At baseline, the 2 groups had similar EFD scores. Administration of sildenafil alone was associated with a substantial increase in EFD score (mean, 7.7 [95% CI, 6.5 to 8.8]), but change in EFD score after randomization did not differ between the groups (difference, 2.2 [CI, -0.8 to 5.1]; P = 0.150). The findings were similar for other domains of sexual function in younger men, more obese men, and men with lower baseline testosterone levels or an inadequate response to sildenafil alone. Frequency of adverse events was similar for testosterone and placebo groups. LIMITATION: Whether testosterone could improve erectile function without sildenafil was not studied. CONCLUSION: Sildenafil plus testosterone was not superior to sildenafil plus placebo in improving erectile function in men with erectile dysfunction and low testosterone levels. PRIMARY FUNDING SOURCE: National Institute of Child Health and Human Development.


Asunto(s)
Andrógenos/uso terapéutico , Disfunción Eréctil/tratamiento farmacológico , Terapia de Reemplazo de Hormonas , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Piperazinas/uso terapéutico , Sulfonas/uso terapéutico , Testosterona/uso terapéutico , Administración Cutánea , Adulto , Anciano , Andrógenos/administración & dosificación , Andrógenos/efectos adversos , Coito , Método Doble Ciego , Quimioterapia Combinada , Disfunción Eréctil/sangre , Geles , Humanos , Masculino , Persona de Mediana Edad , Orgasmo , Erección Peniana , Inhibidores de Fosfodiesterasa 5/administración & dosificación , Inhibidores de Fosfodiesterasa 5/efectos adversos , Piperazinas/administración & dosificación , Purinas/administración & dosificación , Purinas/uso terapéutico , Calidad de Vida , Citrato de Sildenafil , Sulfonas/administración & dosificación , Testosterona/administración & dosificación , Testosterona/efectos adversos , Testosterona/sangre
15.
JAMA ; 307(9): 931-9, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22396515

RESUMEN

CONTEXT: Steroid 5α-reductase inhibitors are used to treat benign prostatic hyperplasia and androgenic alopecia, but the role of 5α-dihydrotestosterone (DHT) in mediating testosterone's effects on muscle, sexual function, erythropoiesis, and other androgen-dependent processes remains poorly understood. OBJECTIVE: To determine whether testosterone's effects on muscle mass, strength, sexual function, hematocrit level, prostate volume, sebum production, and lipid levels are attenuated when its conversion to DHT is blocked by dutasteride (an inhibitor of 5α-reductase type 1 and 2). DESIGN, SETTING, AND PATIENTS: The 5α-Reductase Trial was a randomized controlled trial of healthy men aged 18 to 50 years comparing placebo plus testosterone enthanate with dutasteride plus testosterone enanthate from May 2005 through June 2010. INTERVENTIONS: Eight treatment groups received 50, 125, 300, or 600 mg/wk of testosterone enanthate for 20 weeks plus placebo (4 groups) or 2.5 mg/d of dutasteride (4 groups). MAIN OUTCOME MEASURES: The primary outcome was change in fat-free mass; secondary outcomes: changes in fat mass, muscle strength, sexual function, prostate volume, sebum production, and hematocrit and lipid levels. RESULTS: A total of 139 men were randomized; 102 completed the 20-week intervention. Men assigned to dutasteride were similar at baseline to those assigned to placebo. The mean fat-free mass gained by the dutasteride groups was 0.6 kg (95% CI, -0.1 to 1.2 kg) when receiving 50 mg/wk of testosterone enanthate, 2.6 kg (95% CI, 0.9 to 4.3 kg) for 125 mg/wk, 5.8 kg (95% CI, 4.8 to 6.9 kg) for 300 mg/wk, and 7.1 kg (95% CI, 6.0 to 8.2 kg) for 600 mg/wk. The mean fat-free mass gained by the placebo groups was 0.8 kg (95% CI, -0.1 to 1.7 kg) when receiving 50 mg/wk of testosterone enanthate, 3.5 kg (95% CI, 2.1 to 4.8 kg) for 125 mg/wk, 5.7 kg (95% CI, 4.8 to 6.5 kg) for 300 mg/wk, and 8.1 kg (95% CI, 6.7 to 9.5 kg) for 600 mg/wk. The dose-adjusted differences between the dutasteride and placebo groups for fat-free mass were not significant (P = .18). Changes in fat mass, muscle strength, sexual function, prostate volume, sebum production, and hematocrit and lipid levels did not differ between groups. CONCLUSION: Changes in fat-free mass in response to graded testosterone doses did not differ in men in whom DHT was suppressed by dutasteride from those treated with placebo, indicating that conversion of testosterone to DHT is not essential for mediating its anabolic effects on muscle. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00493987.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Adiposidad/efectos de los fármacos , Azaesteroides/farmacología , Fuerza Muscular/efectos de los fármacos , Reproducción/efectos de los fármacos , Testosterona/análogos & derivados , Testosterona/metabolismo , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Adulto , Índice de Masa Corporal , Método Doble Ciego , Dutasterida , Hematócrito , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Próstata/anatomía & histología , Próstata/efectos de los fármacos , Sebo/efectos de los fármacos , Sebo/metabolismo , Testosterona/administración & dosificación , Testosterona/fisiología , Resultado del Tratamiento
16.
Alzheimers Dement (N Y) ; 8(1): e12306, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35676943

RESUMEN

Introduction: Amyloid-related imaging abnormalities with edema/effusion (ARIA-E) are commonly observed with anti-amyloid therapies in Alzheimer's disease. We developed a semi-mechanistic, in silico model to understand the time course of ARIA-E and its dose dependency. Methods: Dynamic and statistical analyses of data from 112 individuals that experienced ARIA-E in the open-label extension of SCarlet RoAD (a study of gantenerumab in participants with prodromal Alzheimer's disease) and Marguerite RoAD (as study of Gantenerumab in participants with mild Alzheimer's disease) studies were used for model building. Gantenerumab pharmacokinetics, local amyloid removal, disturbance and repair of the vascular wall, and ARIA-E magnitude were represented in the novel vascular wall disturbance (VWD) model of ARIA-E. Results: The modeled individual-level profiles provided a good representation of the observed pharmacokinetics and time course of ARIA-E magnitude. ARIA-E dynamics were shown to depend on the interplay between drug-mediated amyloid removal and intrinsic vascular repair processes. Discussion: Upon further refinement and validation, the VWD model could inform strategies for dosing and ARIA monitoring in individuals with an ARIA-E history.

17.
Steroids ; 74(6): 512-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19321131

RESUMEN

In humans, testosterone (T), dihydrotestosterone (DHT), estradiol (E2), estrone (E1) and cortisol (C) bind to the serum proteins sex hormone-binding globulin (SHBG), albumin (Alb) and corticosteroid-binding globulin (CBG). Equilibrium dialysis is considered to be the "gold standard" for measuring the free concentrations of these steroids but is technically difficult and not widely available. Based on a mathematical model of the 5-ligand/3-protein binding equilibria, we developed a novel spreadsheet method for calculating the free and bioavailable (free+Alb-bound) concentrations of each steroid in terms of the total steroid and protein concentrations. The model uses 15 association constants K(SHBG-X), K(Alb-X), and K(CBG-X) (X=T, DHT, E2, E1 and C) that have been estimated from a systematic review of published binding studies. The computation of the free and bioavailable concentrations uses an iterative numerical method that can be readily programmed on a spreadsheet. The method is illustrated with six examples corresponding to young men (YM), old men (OM), obese men (Ob M), young women (YM), pregnant women in the 3rd trimester (Preg T3) and oophorectomized women on oral conjugated equine estrogens (CEE). The resulting free hormone concentrations for YM and YW fall within the normal references ranges obtained by equilibrium dialysis for all five hormones. The model also accounts for the competitive binding effects of high estrogen levels on the free T levels in Preg T3. This novel spreadsheet method provides a "user-friendly" approach for estimating the free concentrations of circulating sex hormones and cortisol in men and women.


Asunto(s)
Dihidrotestosterona/sangre , Estradiol/sangre , Estrona/sangre , Hidrocortisona/sangre , Cómputos Matemáticos , Testosterona/sangre , Adulto , Anciano , Algoritmos , Femenino , Humanos , Masculino , Ovariectomía , Embarazo , Tercer Trimestre del Embarazo/sangre , Valores de Referencia , Reproducibilidad de los Resultados
18.
J Clin Endocrinol Metab ; 93(5): 1702-10, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18303079

RESUMEN

OBJECTIVE: Our objective was to compare the effects of oral vs. transdermal estrogen therapy on C-reactive protein (CRP), IL-6, E- and P-selectin, intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule-1, serum amyloid A, transferrin, prealbumin, IGF-I, SHBG, thyroxine-binding globulin (TBG), and cortisol-binding globulin (CBG) in naturally menopausal women. DESIGN: This was a randomized, open-label crossover clinical trial. A 6-wk withdrawal from prior hormone therapy (baseline) was followed in randomized order by 12-wk oral conjugated equine estrogens (CEEs) (0.625 mg/d) and 12-wk transdermal estradiol (E2) (0.05 mg/d), with oral micronized progesterone (100 mg/d) given continuously during both regimens. RESULTS: A total of 27 women enrolled, and 25 completed both treatment periods. Nine parameters changed significantly during oral CEE (median percent change from baseline; P value): CRP (192%; P <0.001); E-selectin (-16.3%; P = 0.003); P-selectin (-15.3%; P = 0.012); ICAM-1 (-5%; P = 0.015); transferrin (5.3%; P = 0.024); IGF-I (-30.5%; P < 0.001); SHBG (113%; P < 0.001); TBG (38%; P < 0.001); and CBG (20%; P < 0.001). With transdermal E2, only three parameters changed significantly and to a lesser degree: ICAM-1 (-2.1%; P = 0.04); IGF-I (-12.5%; P < 0.001); and SHBG (2.6%; P = 0.042). During oral CEE the intrasubject changes in CRP correlated strongly with the changes in serum amyloid A (r = 0.805; P < 0.001), and were only weakly associated with the changes in SHBG (r = 0.248; nonsignificant), TBG (0.430; P = 0.031), and CBG (r = 0.072; nonsignificant). The log-log relationship between CRP and IL-6 observed at baseline showed a parallel shift during oral CEE, suggesting an amplified hepatic response or a greater sensitivity to IL-6 stimulation. CONCLUSION: Compared with oral CEE, transdermal E2 exerts minimal effects on CRP and the other inflammation and hepatic parameters.


Asunto(s)
Proteína C-Reactiva/análisis , Estradiol/administración & dosificación , Estrógenos Conjugados (USP)/farmacología , Menopausia/sangre , Administración Cutánea , Adulto , Anciano , Índice de Masa Corporal , Estudios Cruzados , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/sangre , Interleucina-6/sangre , Persona de Mediana Edad , Prealbúmina/análisis , Globulina de Unión a Hormona Sexual/análisis , Transferrina/análisis , Molécula 1 de Adhesión Celular Vascular/sangre
19.
Menopause ; 14(6): 985-94, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17507833

RESUMEN

OBJECTIVE: To compare the changes induced by oral versus transdermal estrogen therapy on the total and free serum concentrations of testosterone (T), thyroxine (T4), and cortisol (C) and the concentrations of their serum binding globulins sex hormone-binding globulin, thyroxine-binding globulin, and cortisol-binding globulin in naturally menopausal women. DESIGN: Randomized, open-label, crossover. Interventions included a 6-week withdrawal from previous hormone therapy (baseline), followed in randomized order by 12 weeks of oral conjugated equine estrogens (CEE) (0.625 mg/d) and 12 weeks of transdermal estradiol (TD E2) (0.05 mg/d), with oral micronized progesterone (100 mg/d) given continuously during both transdermal estrogen therapy regimens. RESULTS: Twenty-seven women were enrolled in the study, and 25 completed both treatment periods. The mean(SD) percentage changes from baseline of sex hormone-binding globulin, total T, and free T with oral CEE were +132.1% (74.5%), +16.4% (43.8%), and -32.7% (25.9%), respectively, versus +12.0% (25.1%), +1.2% (43.7%), and +1.0% (45.0%) with TD E2. The mean (SD) percentage changes of thyroxine-binding globulin, total T4, and free T4 with oral CEE were +39.9% (20.1%), +28.4% (29.2%), and -10.4% (22.3%), respectively, versus +0.4% (11.1%), -0.7% (16.5%), and +0.2% (26.6%) with TD E2. The mean (SD) percentage changes of cortisol-binding globulin, total C, and free C with oral CEE were +18.0% (19.5%), +29.2% (46.3%), and +50.4% (126.5%), respectively, versus -2.2% (11.3%), -6.7% (30.8%), and +1.8% (77.1%) with TD E2. Concentrations of all hormones and binding globulins were significantly different (P < or = 0.003) during administration of oral versus transdermal estrogen therapy, except for free T4 and free C. CONCLUSIONS: Compared with oral CEE, TD E2 exerts minimal effects on the total and free concentrations of T, T4, and C and their binding proteins.


Asunto(s)
Terapia de Reemplazo de Estrógeno , Estrógenos Conjugados (USP)/administración & dosificación , Progesterona/administración & dosificación , Administración Cutánea , Administración Oral , Adulto , Anciano , Proteínas Portadoras/sangre , Estudios Cruzados , Femenino , Humanos , Menopausia , Persona de Mediana Edad , Testosterona/sangre , Tiroxina/sangre , Resultado del Tratamiento
20.
J Clin Endocrinol Metab ; 91(11): 4669-75, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16912120

RESUMEN

BACKGROUND: Recently we found that testosterone levels are higher in older men than young men receiving exogenous testosterone. We hypothesized that older men have lower apparent testosterone metabolic clearance rates (aMCR-T) that contribute to higher testosterone levels. OBJECTIVE: The objective of the study was to compare aMCR-T in older and young men and identify predictors of aMCR-T. METHODS: Sixty-one younger (19-35 yr) and 60 older (59-75 yr) men were given a monthly GnRH agonist and weekly testosterone enanthate (TE) (25, 50, 125, 300, or 600 mg) for 5 months. Estimated aMCR-T was calculated from the amount of TE delivered weekly and trough serum testosterone concentrations, corrected for real-time absorption kinetics from the im testosterone depot. RESULTS: Older men had lower total (316 +/- 13 vs. 585 +/- 26 ng/dl, P < 0.00001) and free testosterone (4 +/- 0.1 vs. 6 +/- 0.3 ng/dl, P < 0.00001) and higher SHBG (52 +/- 3 vs. 33 +/- 2 nmol/liter, P < 0.00001) than younger men at baseline. Total and free testosterones increased with TE dose and were higher in older men than young men in the 125-, 300-, and 600-mg dose groups. aMCR-T was lower in older men than young men (1390 +/- 69 vs. 1821 +/- 102 liter/d, P = 0.006). aMCR-T correlated negatively with age (P = 0.0007), SHBG (P = 0.046), and total testosterone during treatment (P = 0.02) and percent body fat at baseline (P = 0.01) and during treatment (P = 0.004). aMCR-T correlated positively with lean body mass at baseline (P = 0.03) and during treatment (P = 0.01). In multiple regression models, significant predictors of aMCR-T included lean body mass (P = 0.008), percent fat mass (P = 0.009), and SHBG (P = 0.001). CONCLUSIONS: Higher testosterone levels in older men receiving TE were associated with an age-related decrease in apparent testosterone metabolic clearance rates. Body composition and SHBG were significant predictors of aMCR-T.


Asunto(s)
Gonadotropinas/agonistas , Hipogonadismo/tratamiento farmacológico , Testosterona/análogos & derivados , Adulto , Factores de Edad , Anciano , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Terapia de Reemplazo de Hormonas , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Cooperación del Paciente/estadística & datos numéricos , Globulina de Unión a Hormona Sexual/análisis , Testosterona/sangre , Testosterona/farmacocinética , Testosterona/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA