Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Kidney Int ; 105(6): 1212-1220, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38514000

RESUMEN

Accurate assessment of the glomerular filtration rate (GFR) is crucial for researching kidney disease in rats. Although validation of methods that assess GFR is crucial, large-scale comparisons between different methods are lacking. Both transcutaneous GFR (tGFR) and a newly developed estimated GFR (eGFR) equation by our group provide a low-invasive approach enabling repeated measurements. The tGFR is a single bolus method using FITC-labeled sinistrin to measure GFR based on half-life of the transcutaneous signal, whilst the eGFR is based on urinary sinistrin clearance. Here, we retrospectively compared tGFR, using both 1- and 3- compartment models (tGFR_1c and tGFR_3c, respectively) to the eGFR in a historic cohort of 43 healthy male rats and 84 male rats with various models of chronic kidney disease. The eGFR was on average considerably lower than tGFR-1c and tGFR-3c (mean differences 855 and 216 µL/min, respectively) and only 20 and 47% of measurements were within 30% of each other, respectively. The relative difference between eGFR and tGFR was highest in rats with the lowest GFR. Possible explanations for the divergence are problems inherent to tGFR, such as technical issues with signal measurement, description of the signal kinetics, and translation of half-life to tGFR, which depends on distribution volume. The unknown impact of isoflurane anesthesia used in determining mGFR remains a limiting factor. Thus, our study shows that there is a severe disagreement between GFR measured by tGFR and eGFR, stressing the need for more rigorous validation of the tGFR and possible adjustments to the underlying technique.


Asunto(s)
Modelos Animales de Enfermedad , Tasa de Filtración Glomerular , Insuficiencia Renal Crónica , Animales , Masculino , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/orina , Insuficiencia Renal Crónica/diagnóstico , Ratas , Riñón/fisiopatología , Ratas Sprague-Dawley , Estudios Retrospectivos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Fluoresceína-5-Isotiocianato/administración & dosificación , Reproducibilidad de los Resultados , Eliminación Renal/fisiología , Fluoresceínas , Oligosacáridos
2.
Eur J Pharm Sci ; 189: 106553, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37532063

RESUMEN

HSK7653, an oral dipeptidyl peptidase-4 inhibitor administered every 2 weeks, is a candidate for the treatment of type 2 diabetes mellitus. The major elimination pathway of HSK7653 in vivo is renal excretion, and hepatic metabolism and fecal excretion of unchanged compound contribute less to the systemic clearance of HSK7653. Considering the disposition characteristics and the potential indication population of HSK7653, evaluating the HSK7653 exposure in patients with renal impairment and geriatric populations is a prerequisite for bringing more benefits to the patients. Here, a PBPK model was developed based on in vitro experimental results, such as dissolution, permeability, and metabolism, and the in vivo renal clearance, to evaluate the effects of physiological factors and food on HSK7653 exposure in specific populations, including adult and elder individuals with renal impairment and geriatric populations. Simulation results showed that the AUC of HSK7653 increased by 46%, 82%, and 129% in adult patients with mild, moderate, and severe renal impairment, and by 56%, 78%, and 101% in patients aged 65-75, 75-85 and 85-95 years, respectively. The AUC increased in the range of 62%-83%, 98%-133%, and 153%-195% in elderly patients (65-95 years) with mild, moderate, and severe renal impairment, respectively. Moreover, two different absorption model development methods (dissolution profile method and the diffusion layer model method) predicted that food had no effect on the exposure of the same simulated population. Since the predicted AUC of HSK7653 at the 10 mg dose in various specific populations was still within the relatively flat results of the exposure-response analysis, the 10 mg dose of HSK7653 was first used to explore the exposure in the renal impairment population (CTR20221952).


Asunto(s)
Diabetes Mellitus Tipo 2 , Inhibidores de la Dipeptidil-Peptidasa IV , Adulto , Anciano , Humanos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Eliminación Renal/fisiología , Inhibidores de la Dipeptidil-Peptidasa IV/farmacocinética , Administración Oral , Comprimidos , Modelos Biológicos , Simulación por Computador
3.
Pharm Res ; 38(11): 1873-1887, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34750720

RESUMEN

AIM: The pharmacokinetic (PK) properties of cyclosporine (CsA) in renal transplant recipients are patient- and time-dependent. Knowledge of this time-related variability is necessary to maintain or achieve CsA target exposure. Here, we aimed to identify factors explaining variabilities in CsA PK properties and characterize time-varying clearance (CL/F) by performing a comprehensive analysis of CsA PK factors using population PK (popPK) modeling of long-term follow-up data from our institution. METHODS: In total, 3674 whole-blood CsA concentrations from 183 patients who underwent initial renal transplantation were analyzed using nonlinear mixed-effects modeling. The effects of potential covariates were selected according to a previous study and well-accepted theoretical mechanisms. Model-informed individualized therapeutic regimens were also evaluated. RESULTS: A two-compartment model adequately described the data and the estimated mean CsA CL/F was 32.6 L h-1 (relative standard error: 5%). Allometrically scaled body size, hematocrit (HCT) level, CGC haplotype carrier status, and postoperative time may contribute to CsA PK variability. The CsA bioavailability in patients receiving a prednisolone dose (PD) of 80 mg was 20.6% lower than that in patients receiving 20 mg. A significant decrease (52.6%) in CL/F was observed as the HCT increased from 10.5% to 60.5%. The CL/F of the non-CGC haplotype carrier was 14.4% lower than that of the CGC haplotype carrier at 3 months post operation. CONCLUSIONS: By monitoring body size, HCT, PD, and CGC haplotype, changes in CsA CL/F over time could be predicted. Such information could be used to optimize CsA therapy. CsA dose adjustments should be considered in different postoperative periods.


Asunto(s)
Inhibidores de la Calcineurina/farmacocinética , Ciclosporina/farmacocinética , Rechazo de Injerto/prevención & control , Trasplante de Riñón/efectos adversos , Modelos Biológicos , Adolescente , Adulto , Aloinjertos/metabolismo , Disponibilidad Biológica , Variación Biológica Poblacional , Inhibidores de la Calcineurina/administración & dosificación , Ciclosporina/administración & dosificación , Femenino , Estudios de Seguimiento , Rechazo de Injerto/inmunología , Humanos , Riñón/metabolismo , Fallo Renal Crónico/sangre , Fallo Renal Crónico/cirugía , Masculino , Persona de Mediana Edad , Eliminación Renal/fisiología , Adulto Joven
4.
Drug Metab Dispos ; 49(12): 1109-1117, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34625435

RESUMEN

Linerixibat, an oral small-molecule ileal bile acid transporter inhibitor under development for cholestatic pruritus in primary biliary cholangitis, was designed for minimal absorption from the intestine (site of pharmacological action). This study characterized the pharmacokinetics, absorption, metabolism, and excretion of [14C]-linerixibat in humans after an intravenous microtracer concomitant with unlabeled oral tablets and [14C]-linerixibat oral solution. Linerixibat exhibited absorption-limited flip-flop kinetics: longer oral versus intravenous half-life (6-7 hours vs. 0.8 hours). The short intravenous half-life was consistent with high systemic clearance (61.9 l/h) and low volume of distribution (16.3 l). In vitro studies predicted rapid hepatic clearance via cytochrome P450 3A4 metabolism, which predicted human hepatic clearance within 1.5-fold. However, linerixibat was minimally metabolized in humans after intravenous administration: ∼80% elimination via biliary/fecal excretion (>90%-97% as unchanged parent) and ∼20% renal elimination by glomerular filtration (>97% as unchanged parent). Absolute oral bioavailability of linerixibat was exceedingly low (0.05%), primarily because of a very low fraction absorbed (0.167%; fraction escaping first-pass gut metabolism (fg) ∼100%), with high hepatic extraction ratio (77.0%) acting as a secondary barrier to systemic exposure. Oral linerixibat was almost entirely excreted (>99% recovered radioactivity) in feces as unchanged and unabsorbed linerixibat. Consistent with the low oral fraction absorbed and ∼20% renal recovery of intravenous [14C]-linerixibat, urinary elimination of orally administered radioactivity was negligible (<0.04% of dose). Linerixibat unequivocally exhibited minimal gastrointestinal absorption and oral systemic exposure. Linerixibat represents a unique example of high CYP3A4 clearance in vitro but nearly complete excretion as unchanged parent drug via the biliary/fecal route. SIGNIFICANCE STATEMENT: This study conclusively established minimal absorption and systemic exposure to orally administered linerixibat in humans. The small amount of linerixibat absorbed was eliminated efficiently as unchanged parent drug via the biliary/fecal route. The hepatic clearance mechanism was mispredicted to be mediated via cytochrome P450 3A4 metabolism in vitro rather than biliary excretion of unchanged linerixibat in vivo.


Asunto(s)
Administración Intravenosa , Administración Oral , Proteínas Portadoras/antagonistas & inhibidores , Eliminación Hepatobiliar , Glicoproteínas de Membrana/antagonistas & inhibidores , Metilaminas/farmacocinética , Eliminación Renal , Tiazepinas/farmacocinética , Adulto , Disponibilidad Biológica , Fármacos Gastrointestinales/farmacocinética , Voluntarios Sanos , Eliminación Hepatobiliar/efectos de los fármacos , Eliminación Hepatobiliar/fisiología , Humanos , Absorción Intestinal , Masculino , Tasa de Depuración Metabólica , Eliminación Renal/efectos de los fármacos , Eliminación Renal/fisiología , Resultado del Tratamiento
5.
Pharm Res ; 38(7): 1139-1156, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34254223

RESUMEN

PURPOSE: A dataset of fraction excreted unchanged in the urine (fe) values was developed and used to evaluate the ability of preclinical animal species to predict high urinary excretion, and corresponding poor metabolism, in humans. METHODS: A literature review of fe values in rats, dogs, and monkeys was conducted for all Biopharmaceutics Drug Disposition Classification System (BDDCS) Class 3 and 4 drugs (n=352) and a set of Class 1 and 2 drugs (n=80). The final dataset consisted of 202 total fe values for 135 unique drugs. Human and animal data were compared through correlations, two-fold analysis, and binary classifications of high (fe ≥30%) versus low (<30%) urinary excretion in humans. Receiver Operating Characteristic curves were plotted to optimize animal fe thresholds. RESULTS: Significant correlations were found between fe values for each animal species and human fe (p<0.05). Sixty-five percent of all fe values were within two-fold of human fe with animals more likely to underpredict human urinary excretion as opposed to overpredict. Dogs were the most reliable predictors of human fe of the three animal species examined with 72% of fe values within two-fold of human fe and the greatest accuracy in predicting human fe ≥30%. ROC determined thresholds of ≥25% in rats, ≥19% in dogs, and ≥10% in monkeys had improved accuracies in predicting human fe of ≥30%. CONCLUSIONS: Drugs with high urinary excretion in animals are likely to have high urinary excretion in humans. Animal models tend to underpredict the urinary excretion of unchanged drug in humans.


Asunto(s)
Evaluación Preclínica de Medicamentos , Eliminación Renal/fisiología , Animales , Conjuntos de Datos como Asunto , Perros , Haplorrinos , Humanos , Curva ROC , Ratas , Especificidad de la Especie
6.
Drug Metab Dispos ; 49(10): 929-937, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34315779

RESUMEN

Pregnancy can significantly change the pharmacokinetics of drugs, including those renally secreted by organic anion transporters (OATs). Quantifying these changes in pregnant women is logistically and ethically challenging. Hence, predicting the in vivo plasma renal secretory clearance (CLsec) and renal CL (CLrenal) of OAT drugs in pregnancy is important to design correct dosing regimens of OAT drugs. Here, we first quantified the fold-change in renal OAT activity in pregnant versus nonpregnant individual using available selective OAT probe drug CLrenal data (training dataset; OAT1: tenofovir, OAT2: acyclovir, OAT3: oseltamivir carboxylate). The fold-change in OAT1 activity during the 2nd and 3rd trimester was 2.9 and 1.0 compared with nonpregnant individual, respectively. OAT2 activity increased 3.1-fold during the 3rd trimester. OAT3 activity increased 2.2, 1.7 and 1.3-fold during the 1st, 2nd, and 3rd trimester, respectively. Based on these data, we predicted the CLsec, CLrenal and total clearance ((CLtotal) of drugs in pregnancy, which are secreted by multiple OATs (verification dataset; amoxicillin, pravastatin, cefazolin and ketorolac, R-ketorolac, S-ketorolac). Then, the predicted clearances (CLs) were compared with the observed values. The predicted/observed CLsec, CLrenal, and CLtotal of drugs in pregnancy of all verification drugs were within 0.80-1.25 fold except for CLsec of amoxicillin in the 3rd trimester (0.76-fold) and cefazolin in the 2nd trimester (1.27-fold). Overall, we successfully predicted the CLsec, CLrenal, and CLtotal of drugs in pregnancy that are renally secreted by multiple OATs. This approach could be used in the future to adjust dosing regimens of renally secreted OAT drugs which are administered to pregnant women. SIGNIFICANCE STATEMENT: To the authors' knowledge, this is the first report to successfully predict renal secretory clearance and renal clearance of multiple OAT substrate drugs during pregnancy. The data presented here could be used in the future to adjust dosing regimens of renally secreted OAT drugs in pregnancy. In addition, the mechanistic approach used here could be extended to drugs transported by other renal transporters.


Asunto(s)
Transporte Biológico Activo/fisiología , Relación Dosis-Respuesta a Droga , Transportadores de Anión Orgánico , Farmacocinética , Eliminación Renal/fisiología , Biotransformación/fisiología , Cálculo de Dosificación de Drogas , Femenino , Células HEK293 , Humanos , Tasa de Depuración Metabólica , Transportadores de Anión Orgánico/clasificación , Transportadores de Anión Orgánico/metabolismo , Preparaciones Farmacéuticas/clasificación , Preparaciones Farmacéuticas/metabolismo , Embarazo , Trimestres del Embarazo/efectos de los fármacos , Trimestres del Embarazo/metabolismo , Reproducibilidad de los Resultados
7.
Drug Metab Dispos ; 49(8): 629-637, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074729

RESUMEN

Exposure to the environmental pollutant cadmium is ubiquitous, as it is present in cigarette smoke and the food supply. Over time, cadmium enters and accumulates in the kidneys, where it causes tubular injury. The breast cancer resistance protein (BCRP, ATP-Binding Cassette G2 ABCG2) is an efflux transporter that mediates the urinary secretion of pharmaceuticals and toxins. The ABCG2 genetic variant Q141K exhibits altered membrane trafficking that results in reduced efflux of BCRP substrates. Here, we sought to 1) evaluate the in vitro and in vivo ability of BCRP to transport cadmium and protect kidney cells from toxicity and 2) determine whether this protection is impaired by the Q141K variant. Cadmium concentrations, cellular stress, and toxicity were quantified in human embryonic kidney 293 cells expressing an empty vector (EV), BCRP wild-type (WT), or variant (Q141K) gene. Treatment with CdCl2 resulted in greater accumulation of cadmium and apoptosis in EV cells relative to WT cells. Exposure to CdCl2 induced expression of stress-related genes and proteins including MT-1A/MT-2A, NAD(P)H quinone dehydrogenase 1, and heme oxygenase-1 to a higher extent in EV cells compared with WT cells. Notably, the Q141K variant protected against CdCl2-induced activation of stress genes and cytotoxicity, but this protection was to a lesser magnitude than observed with WT BCRP. Lastly, concentrations of cadmium in the kidneys of Bcrp knockout mice were 40% higher than in WT mice, confirming that cadmium is an in vivo substrate of BCRP. In conclusion, BCRP prevents the accumulation of cadmium and protects against toxicity, a response that is impaired by the Q141K variant. SIGNIFICANCE STATEMENT: The breast cancer resistance protein transporter lowers cellular accumulation of the toxic heavy metal cadmium. This protective function is partially attenuated by the Q141K genetic variant in the ABCG2 gene.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Cadmio/farmacocinética , Riñón , Proteínas de Neoplasias , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico Activo/genética , Cadmio/toxicidad , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/fisiopatología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polimorfismo de Nucleótido Simple , Factores Protectores , Eliminación Renal/fisiología
8.
AAPS J ; 23(4): 86, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34142253

RESUMEN

Serum creatinine (sCr) is a commonly measured biomarker to estimate glomerular filtration rate (GFR) and therefore widely used as a covariate in population pharmacokinetic models of renally excreted drugs. In neonates, sCr dynamically changes during the first few weeks after birth. Missing covariates are a common problem in pharmacokinetic modeling of neonates due to the limited availability of blood sampling in number and volume. The objective of this work is to develop a parsimonious population model describing time courses of sCr in neonates with the intent to be incorporated into pharmacokinetic models of various drugs where sCr values are sparse or missing. The data for model development consisted of sCr measurements in 1080 newborns with a gestational age of 24-42 weeks. The model is based on a pharmacokinetic model of sCr that involves GFR, backflow of creatinine from the renal tubules, and urinary flow. Gestational age is the only covariate explaining between-subject variability of sCr. The model adequately describes distinct features of the sCr time course such as a peak and decline to a plateau. For a neonate with a GA of 35 weeks, the typical value of sCr at birth was 0.584 mg/dL, the peak (0.794 mg/dL) occurred 2.3 days after birth, to reach a plateau of 0.255 mg/dL approximately after 24.7 days. Model simulations reveal that in neonates with a similar postnatal age, sCr decreases with increasing GA. In summary, our model is designed to be a part of full random effects pharmacokinetic models where sCr is a significant covariate.


Asunto(s)
Creatinina/sangre , Tasa de Filtración Glomerular/fisiología , Modelos Biológicos , Eliminación Renal/fisiología , Variación Biológica Poblacional , Peso al Nacer/fisiología , Creatinina/metabolismo , Conjuntos de Datos como Asunto , Femenino , Edad Gestacional , Humanos , Recién Nacido , Unidades de Cuidado Intensivo Neonatal , Masculino , Valores de Referencia , Estudios Retrospectivos
9.
Drug Metab Dispos ; 49(8): 619-628, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34011533

RESUMEN

Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that is currently in clinical development for the treatment of heart failure. The absorption and disposition of [14C]OM (60 µCi) were studied after a single intravenous infusion (35 mg over 1 hour) or oral solution dose (35 mg) in 14 healthy male subjects. Mean recovery of the administered [14C]OM dose was 85.1% and 86.5% over 336 hours for the intravenous and oral routes, respectively. After intravenous dosing, 47.8% and 37.3% of the dose was recovered in urine and feces, respectively; after oral dosing, 48.6% and 38.0% was recovered in urine and feces, respectively. Unchanged OM accounted for a minor percentage of radioactivity in urine (mean 7.7% of dose) and feces (mean 4.1% of dose) across all subjects. The major metabolites recovered in urine and feces were M3 (decarbamoylation product) and sequential metabolite M4 (lactam of M3), which accounted for means of 26.5% and 11.6% of the administered dose, respectively. The CYP4 family of enzymes was primarily responsible for the formation of M3 based on in vitro studies. Other metabolic pathways accounted for 14.9% of the administered dose. In pooled plasma, OM, M3, and M4 accounted for 83.8%, 6.0%, and 3.3% of the total [14C]OM-related materials. No other plasma metabolites constituted more than 3% of the administered dose. The bioavailability for OM solution was 93.5% after rapid and extensive absorption. SIGNIFICANCE STATEMENT: This study characterized the absorption and disposition of OM, a novel small molecule being developed for the treatment of heart failure. OM was primarily cleared through metabolism by the CYP4 family through oxidative cleavage of a terminal carbamate moiety that resembles hydrolysis.


Asunto(s)
Familia 4 del Citocromo P450/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Eliminación Hepatobiliar/fisiología , Absorción Intestinal/fisiología , Eliminación Renal/fisiología , Urea/análogos & derivados , Administración Intravenosa , Administración Oral , Adulto , Disponibilidad Biológica , Biotransformación , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/farmacocinética , Voluntarios Sanos , Humanos , Infusiones Intravenosas , Masculino , Urea/administración & dosificación , Urea/farmacocinética
10.
Clin Transl Sci ; 14(5): 1906-1916, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33982438

RESUMEN

Nausea, vomiting, and renal injury are the common adverse effects associated with cisplatin. Cisplatin is excreted via the multidrug and toxin release (MATE) transporter, and the involvement of the MATE transporter in cisplatin-induced kidney injury has been reported. The MATE transporter is also involved in the excretion of ondansetron, but the effects of 5-HT3 receptor antagonists used clinically for cisplatin-induced renal injury have not been elucidated. Therefore, the aim of this study was to investigate the effects of 5-HT3 receptor antagonists in a mouse model of cisplatin-induced kidney injury and to validate the results using medical big data analysis of more than 1.4 million reports and a survey of 3000 hospital medical records. The concomitant use of a first-generation 5-HT3 receptor antagonist (ondansetron, granisetron, or ramosetron) significantly increased cisplatin accumulation in the kidneys and worsened renal damage. Conversely, the concomitant use of palonosetron had no effect on renal function compared with the use of cisplatin alone. Furthermore, an analysis of data from the US Food and Drug Administration Adverse Event Reporting System and retrospective medical records revealed that the combination treatment of cisplatin and a first-generation 5-HT3 receptor antagonist significantly increased the number of reported renal adverse events compared with the combination treatment of cisplatin and a second-generation 5-HT3 receptor antagonist. These results suggest that compared with the first-generation antagonists, second-generation 5-HT3 receptor antagonists do not worsen cisplatin-induced acute kidney injury. The findings should be validated in a prospective controlled trial before implementation in clinical practice.


Asunto(s)
Lesión Renal Aguda/patología , Cisplatino/efectos adversos , Náusea/tratamiento farmacológico , Antagonistas del Receptor de Serotonina 5-HT3/administración & dosificación , Vómitos/tratamiento farmacológico , Lesión Renal Aguda/inducido químicamente , Anciano , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Cisplatino/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Granisetrón/administración & dosificación , Granisetrón/efectos adversos , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Masculino , Ratones , Persona de Mediana Edad , Náusea/inducido químicamente , Ondansetrón/administración & dosificación , Ondansetrón/efectos adversos , Proteínas de Transporte de Catión Orgánico/metabolismo , Palonosetrón/administración & dosificación , Palonosetrón/efectos adversos , Eliminación Renal/fisiología , Estudios Retrospectivos , Antagonistas del Receptor de Serotonina 5-HT3/efectos adversos , Vómitos/inducido químicamente
11.
Clin Transl Sci ; 14(5): 1689-1704, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33982447

RESUMEN

An impaired renal function, including acute and chronic kidney disease and end-stage renal disease, can be the result of aging, certain disease conditions, the use of some medications, or as a result of smoking. In patients with renal impairment (RI), the pharmacokinetics (PKs) of drugs or drug metabolites may change and result in increased safety risks or decreased efficacy. In order to make specific dose recommendations in the label of drugs for patients with RI, a clinical trial may have to be conducted or, when not feasible, modeling and simulations approaches, such as population PK modeling or physiologically-based PK modelling may be applied. This tutorial aims to provide an overview of the global regulatory landscape and a practical guidance for successfully designing and conducting clinical RI trials or, alternatively, on applying modeling and simulation tools to come to a dose recommendation for patients with RI in the most efficient manner.


Asunto(s)
Lesión Renal Aguda/complicaciones , Relación Dosis-Respuesta a Droga , Fallo Renal Crónico/complicaciones , Riñón/fisiopatología , Modelos Biológicos , Lesión Renal Aguda/fisiopatología , Ensayos Clínicos como Asunto , Simulación por Computador , Tasa de Filtración Glomerular/fisiología , Humanos , Fallo Renal Crónico/fisiopatología , Eliminación Renal/fisiología
12.
AAPS J ; 23(3): 65, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33948771

RESUMEN

To date, information on the ontogeny of renal transporters is limited. Here, we propose to estimate the in vivo functional ontogeny of transporters using a combined population pharmacokinetic (popPK) and physiology-based pharmacokinetic (PBPK) modeling approach called popPBPK. Clavulanic acid and amoxicillin were used as probes for glomerular filtration, combined glomerular filtration, and active secretion through OAT1,3, respectively. The predictive value of the estimated OAT1,3 ontogeny function was assessed by PBPK predictions of renal clearance (CLR) of other OAT1,3 substrates: cefazolin and piperacillin. Individual CLR post-hoc values, obtained from a published popPK model on the concomitant use of clavulanic acid and amoxicillin in critically ill children between 1 month and 15 years, were used as dependent variables in the popPBPK analysis. CLR was re-parameterized according to PBPK principles, resulting in the estimation of OAT1,3-mediated intrinsic clearance (CLint,OAT1,3,invivo) and its ontogeny. CLint,OAT1,3,invivo ontogeny was described by a sigmoidal function, reaching half of adult level around 7 months of age, comparable to findings based on renal transporter-specific protein expression data. PBPK-based CLR predictions including this ontogeny function were reasonably accurate for piperacillin in a similar age range (2.5 months-15 years) as well as for cefazolin in neonates as compared to published data (%RMSPE of 21.2 and 22.8%, respectively and %PE within ±50%). Using this novel approach, we estimated an in vivo functional ontogeny profile for CLint,OAT1,3,invivo that yields accurate CLR predictions for different OAT1,3 substrates across different ages. This approach deserves further study on functional ontogeny of other transporters.


Asunto(s)
Riñón/metabolismo , Modelos Biológicos , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Eliminación Renal/fisiología , Adolescente , Amoxicilina/administración & dosificación , Amoxicilina/farmacocinética , Variación Biológica Poblacional , Cefazolina/administración & dosificación , Cefazolina/farmacocinética , Niño , Preescolar , Ácido Clavulánico/administración & dosificación , Ácido Clavulánico/farmacocinética , Interacciones Farmacológicas , Tasa de Filtración Glomerular/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Piperacilina/administración & dosificación , Piperacilina/farmacocinética
13.
Medicine (Baltimore) ; 100(18): e25577, 2021 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-33950934

RESUMEN

INTRODUCTION: Brugada syndrome may be unmasked by non-antiarrhythmic pharmaceuticals or drugs. Lacosamide is an antiepileptic agent with a novel mechanism of sodium channel inhibition and has the potential to cause cardiac sodium channel blockade. PATIENT CONCERNS: In this report, we describe the case of patient with a history of a seizure disorder who presented with Brugada I electrocardiogram morphology in the setting of septicemia. DIAGNOSIS: Brugada I electrocardiogram morphology was unmasked by lacosamide antiepileptic monotherapy. INTERVENTIONS: Lacosamide therapy was discontinued. OUTCOMES: Normalization of the electrocardiogram and resolution of Brugada morphology occurred on hospital day 1. CONCLUSION: Caution should be exercised in the use of lacosamide in those at risk for conduction delay, or in combination therapy with medications that impair renal clearance, metabolism of lacosamide, or that display inherent sodium channel blocking properties.


Asunto(s)
Anticonvulsivantes/efectos adversos , Síndrome de Brugada/inducido químicamente , Epilepsia/tratamiento farmacológico , Lacosamida/efectos adversos , Sepsis/complicaciones , Bloqueadores del Canal de Sodio Activado por Voltaje/efectos adversos , Anciano de 80 o más Años , Anticonvulsivantes/farmacocinética , Síndrome de Brugada/diagnóstico , Electrocardiografía , Humanos , Lacosamida/farmacocinética , Masculino , Eliminación Renal/fisiología , Sepsis/fisiopatología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacocinética
14.
Nutrients ; 13(4)2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33916877

RESUMEN

The western dietary pattern is known for its frequent meals rich in saturated fat and protein, resulting in a postprandial state for a large part of the day. Therefore, our aim was to investigate the postprandial glucose and lipid metabolism in response to high (HP) or normal (NP) protein, high-fat hypercaloric diet and to identify early biomarkers of protein intake and hepatic lipid accumulation. In a crossover design, 17 healthy subjects were randomly assigned to consume a HP or NP hypercaloric diet for two weeks. In parallel, a control group (CD; n = 10) consumed a weight-maintaining control diet. Biomarkers of postprandial lipid and glucose metabolism were measured in 24 h urine and in plasma before and following a meal challenge. The metabolic profile of urine but not plasma, showed increased excretion of 13C, carnitine and short chain acyl-carnitines after adaptation to the HP diet. Urinary excretion of decatrienoylcarnitine and octenoylcarnitine increased after adaptation to the NP diet. Our results suggest that the higher excretion of short-chain urinary acyl-carnitines could facilitate the elimination of excess fat of the HP diet and thereby reduce hepatic fat accumulation previously reported, whereas the higher excretion medium-chains acyl-carnitine could be early biomarkers of hepatic lipid accumulation.


Asunto(s)
Carnitina/análogos & derivados , Dieta Alta en Grasa/efectos adversos , Dieta Rica en Proteínas/efectos adversos , Dieta Occidental/efectos adversos , Síndrome Metabólico/diagnóstico , Adulto , Biomarcadores/orina , Carnitina/metabolismo , Carnitina/orina , Estudios Cruzados , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/metabolismo , Proteínas en la Dieta/metabolismo , Ingestión de Energía/fisiología , Femenino , Glucosa/metabolismo , Voluntarios Sanos , Humanos , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Síndrome Metabólico/orina , Periodo Posprandial/fisiología , Eliminación Renal/fisiología , Adulto Joven
15.
Ther Drug Monit ; 43(4): 455-458, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33908408
16.
AAPS J ; 23(3): 53, 2021 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-33839974

RESUMEN

Vancomycin is an effective but potentially nephrotoxic antibiotic commonly used for severe infections. Dosing guidelines for vancomycin in obese children and adolescents with or without renal impairment are currently lacking. This study describes the pharmacokinetics of vancomycin in a large pediatric cohort with varying degrees of obesity and renal function to design practical dosing guidelines for this population. A multi-center retrospective population pharmacokinetic study was conducted using data from patients aged 1-18 years who received >1 dose of vancomycin and had ≥1 vancomycin concentration measured between January 2006 and December 2012. Besides pharmacokinetic data, age, gender, body weight, creatinine clearance (CLcr, bedside Schwartz equation), ward, race, and neutropenic status were collected. Population pharmacokinetic analysis and simulations were performed using NONMEM7.4. A total of 1892 patients (5524 samples) were included, with total body weight (TBW) ranging 6-188 kg (1344 normal weight, 247 overweight, and 301 obese patients) and CLcr down to 8.6 mL/min/1.73 m2. The two-compartment model, with clearance (CL) significantly increasing with TBW and CLcr, central and peripheral volume of distribution and inter-compartmental clearance increasing with TBW, performed well for all age, weight, and renal function ranges. A dosing guideline is proposed that integrates body weight and CLcr resulting in effective and safe exposures across all ages, body weight, and renal functions in the pediatric population. We have characterized the full pharmacokinetic profile of vancomycin in obese children and adolescents aged 1-18 years and propose a practical dosing guideline that integrates both body weight and renal function.


Asunto(s)
Cálculo de Dosificación de Drogas , Enfermedades Renales/fisiopatología , Obesidad/metabolismo , Eliminación Renal/fisiología , Vancomicina/administración & dosificación , Adolescente , Peso Corporal/fisiología , Niño , Preescolar , Femenino , Tasa de Filtración Glomerular , Humanos , Lactante , Masculino , Obesidad/diagnóstico , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Vancomicina/farmacocinética
17.
Pharmacol Res Perspect ; 9(2): e00734, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33689224

RESUMEN

The primary aim of this study was to examine the effect of severe renal impairment (SRI) on the pharmacokinetics of lemborexant, a dual orexin receptor antagonist indicated for the treatment of insomnia. A phase 1 multicenter, single-dose, open-label, parallel-group study was conducted in subjects with SRI not requiring dialysis (estimated glomerular filtration rate 15-29 ml/min/1.73 m2 ; n = 8) compared with demographically matched healthy subjects with normal renal function (n = 8). Plasma levels of lemborexant and its metabolites were measured over 240 h following a single oral 10-mg dose administered in the morning. Relative to subjects with normal renal function, lemborexant maximum plasma concentration (Cmax ) was similar, whereas area under the plasma concentration-time curve from zero to time of last quantifiable concentration (AUC(0-t) ) and AUC from zero to infinity (AUC(0-inf) ) were about 1.5-fold higher in subjects with SRI. The geometric mean ratios (90% confidence interval) were 104.8 (77.4-142.0), 150.5 (113.2-200.3), and 149.8 (113.1-198.6) for Cmax , AUC(0-t) , and AUC(0-inf) , respectively. In both groups, the median lemborexant time to Cmax (tmax ) was 1 h, and the mean unbound fraction of lemborexant was ~7%. For the M4, M9, and M10 metabolites, Cmax was reduced ~20% and exposure (AUC(0-t) and AUC(0-inf) ) was ~1.4- to 1.5-fold higher in subjects with SRI versus healthy subjects; tmax was delayed ~1.5-2 h for M4 and M10. All treatment-emergent adverse events were mild or moderate. Lemborexant pharmacokinetics were not sufficiently altered to warrant a dose adjustment for subjects with renal impairment.


Asunto(s)
Antagonistas de los Receptores de Orexina/farmacocinética , Piridinas/farmacocinética , Pirimidinas/farmacocinética , Eliminación Renal/fisiología , Insuficiencia Renal/metabolismo , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Adolescente , Adulto , Anciano , Área Bajo la Curva , Femenino , Tasa de Filtración Glomerular/fisiología , Semivida , Humanos , Masculino , Persona de Mediana Edad , Antagonistas de los Receptores de Orexina/administración & dosificación , Antagonistas de los Receptores de Orexina/efectos adversos , Piridinas/administración & dosificación , Piridinas/efectos adversos , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/fisiopatología , Índice de Severidad de la Enfermedad , Adulto Joven
18.
Clin Pharmacol Ther ; 110(4): 897-908, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33763856

RESUMEN

The transgender adult population is growing globally, but clinical pharmacology has lagged behind other areas of transgender medicine. Medical care for transgender adults may include long-term testosterone or estrogen treatment to align secondary sex characteristics with gender identity. Clinicians often use drug-drug interaction data from the general adult population to predict medication disposition or safety among transgender adults. However, this approach does not address the complex pharmacodynamic effects of hormone therapy in transgender adults. In this review, we critically examine sex-related and gender-related differences in clinical pharmacology and apply these data to discuss current gaps in transgender medicine.


Asunto(s)
Andrógenos/farmacología , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Estrógenos/farmacología , Glucuronosiltransferasa/efectos de los fármacos , Personas Transgénero , Andrógenos/uso terapéutico , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Estrógenos/uso terapéutico , Femenino , Glucuronosiltransferasa/metabolismo , Humanos , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Masculino , Farmacología Clínica , Eliminación Renal/efectos de los fármacos , Eliminación Renal/fisiología , Factores Sexuales , Testosterona/uso terapéutico , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
19.
AAPS J ; 23(3): 47, 2021 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-33744999

RESUMEN

Food intake influences the pharmacokinetics of orally administered drugs by altering drug absorption, metabolism, and excretion. A drug which is mainly excreted into urine as parent drug is usually highly water-soluble and metabolically stable. Food intake is not expected to significantly affect its extent of oral absorption, metabolism, and excretion. Therefore, we hypothesize that an orally administered drug with significant renal excretion should not have a dramatic food effect (FE). To test our hypothesis, we summarized the FE for orally administered immediate-release (IR) and modified-release (MR) formulations approved by the US FDA from 1998 to 2019, focusing on drugs undergoing significant renal excretion. Totally, 98 active pharmaceutical ingredients (APIs) in IR formulations and 34 APIs in MR formulations were selected. The results demonstrate that the area-under-the-curve (AUC) for IR drug products with fur_unchanged_po > 10% is unlikely to be affected by food, although the peak plasma concentration (Cmax) may increase or decrease by up to 50%. Compared with IR drug products with fur_unchanged_po > 10%, MR drug products with fur_unchanged_po > 10% tend to have more significant FE. Although our proposed approach cannot substitute a clinical FE study, it could be a useful addition to early drug development to get an initial sense of the potential for FE for a drug candidate.


Asunto(s)
Interacciones Alimento-Droga , Absorción Intestinal/fisiología , Modelos Biológicos , Eliminación Renal/fisiología , Administración Oral , Área Bajo la Curva , Humanos , Estados Unidos
20.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-33573145

RESUMEN

Renal ischemia and reperfusion (I/R) injury is the most common cause of acute kidney injury (AKI). Pathogenesis of postischemic AKI involves hemodynamic changes, oxidative stress, inflammation process, calcium ion overloading, apoptosis and necrosis. Up to date, therapeutic approaches to treat AKI are extremely limited. Thus, the aim of this study was to evaluate the effects of hyperbaric oxygen (HBO) preconditioning on citoprotective enzyme, heme oxygenase-1 (HO-1), pro-apoptotic Bax and anti-apoptotic Bcl-2 proteins expression, in postischemic AKI induced in normotensive Wistar and spontaneously hypertensive rats (SHR). The animals were randomly divided into six experimental groups: SHAM-operated Wistar rats (W-SHAM), Wistar rats with induced postischemic AKI (W-AKI) and Wistar group with HBO preconditioning before AKI induction (W-AKI + HBO). On the other hand, SHR rats were also divided into same three groups: SHR-SHAM, SHR-AKI and SHR-AKI + HBO. We demonstrated that HBO preconditioning upregulated HO-1 and anti-apoptotic Bcl-2 protein expression, in both Wistar and SH rats. In addition, HBO preconditioning improved glomerular filtration rate, supporting by significant increase in creatinine, urea and phosphate clearances in both rat strains. Considering our results, we can also say that even in hypertensive conditions, we can expect protective effects of HBO preconditioning in experimental model of AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Hemo Oxigenasa (Desciclizante)/metabolismo , Oxigenoterapia Hiperbárica/métodos , Hipertensión/complicaciones , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/orina , Animales , Creatinina/metabolismo , Creatinina/orina , Modelos Animales de Enfermedad , Humanos , Hipertensión/fisiopatología , Hipertensión/terapia , Riñón/irrigación sanguínea , Riñón/patología , Riñón/fisiopatología , Masculino , Oxígeno/administración & dosificación , Fosfatos/metabolismo , Fosfatos/orina , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Eliminación Renal/fisiología , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Daño por Reperfusión/orina , Regulación hacia Arriba , Urea/metabolismo , Urea/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA