Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Drug Metab Dispos ; 51(12): 1591-1606, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37751998

RESUMO

Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.


Assuntos
Aldeído Oxidase , Fígado , Humanos , Aldeído Oxidase/metabolismo , Taxa de Depuração Metabólica , Fígado/metabolismo , Hepatócitos/metabolismo , Microssomos Hepáticos/metabolismo
2.
Ther Drug Monit ; 45(6): 743-753, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37315152

RESUMO

BACKGROUND: Routine therapeutic drug monitoring (TDM) relies heavily on measuring trough drug concentrations. Trough concentrations are affected not only by drug bioavailability and clearance, but also by various patient and disease factors and the volume of distribution. This often makes interpreting differences in drug exposure from trough data challenging. This study aimed to combine the advantages of top-down analysis of therapeutic drug monitoring data with bottom-up physiologically-based pharmacokinetic (PBPK) modeling to investigate the effect of declining renal function in chronic kidney disease (CKD) on the nonrenal intrinsic metabolic clearance ( CLint ) of tacrolimus as a case example. METHODS: Data on biochemistry, demographics, and kidney function, along with 1167 tacrolimus trough concentrations for 40 renal transplant patients, were collected from the Salford Royal Hospital's database. A reduced PBPK model was developed to estimate CLint for each patient. Personalized unbound fractions, blood-to-plasma ratios, and drug affinities for various tissues were used as priors to estimate the apparent volume of distribution. Kidney function based on the estimated glomerular filtration rate ( eGFR ) was assessed as a covariate for CLint using the stochastic approximation of expectation and maximization method. RESULTS: At baseline, the median (interquartile range) eGFR was 45 (34.5-55.5) mL/min/1.73 m 2 . A significant but weak correlation was observed between tacrolimus CLint and eGFR (r = 0.2, P < 0.001). The CLint declined gradually (up to 36%) with CKD progression. Tacrolimus CLint did not differ significantly between stable and failing transplant patients. CONCLUSIONS: Kidney function deterioration in CKD can affect nonrenal CLint for drugs that undergo extensive hepatic metabolism, such as tacrolimus, with critical implications in clinical practice. This study demonstrates the advantages of combining prior system information (via PBPK) to investigate covariate effects in sparse real-world datasets.


Assuntos
Transplante de Rim , Insuficiência Renal Crônica , Humanos , Tacrolimo/uso terapêutico , Tacrolimo/farmacocinética , Imunossupressores/uso terapêutico , Imunossupressores/farmacocinética , Insuficiência Renal Crônica/tratamento farmacológico , Taxa de Filtração Glomerular
3.
Drug Metab Dispos ; 49(7): 563-571, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33980603

RESUMO

In vitro-in vivo extrapolation (IVIVE) linked with physiologically based pharmacokinetics (PBPK) modeling is used to predict the fates of drugs in patients. Ideally, the IVIVE-PBPK models should incorporate systems information accounting for characteristics of the specific target population. There is a paucity of such scaling factors in cancer, particularly microsomal protein per gram of liver (MPPGL) and cytosolic protein per gram of liver (CPPGL). In this study, cancerous and histologically normal liver tissue from 16 patients with colorectal liver metastasis were fractionated to microsomes and cytosol. Protein content was measured in homogenates, microsomes, and cytosol. The loss of microsomal protein during fractionation was accounted for using corrections based on NADPH cytochrome P450 reductase activity in different matrices. MPPGL was significantly lower in cancerous tissue (24.8 ± 9.8 mg/g) than histologically normal tissue (39.0 ± 13.8 mg/g). CPPGL in cancerous tissue was 42.1 ± 12.9 mg/g compared with 56.2 ± 16.9 mg/g in normal tissue. No correlations between demographics (sex, age, and body mass index) and MPPGL or CPPGL were apparent in the data. The generated scaling factors together with assumptions regarding the relative volumes of cancerous versus noncancerous tissue were used to simulate plasma exposure of drugs with different extraction ratios. The PBPK simulations revealed a substantial difference in drug exposure (area under the curve), up to 3.3-fold, when using typical scaling factors (healthy population) instead of disease-related parameters in cancer population. These indicate the importance of using population-specific scalars in IVIVE-PBPK for different disease states. SIGNIFICANCE STATEMENT: Accuracy in predicting the fate of drugs from in vitro data using IVIVE-PBPK depends on using correct scaling factors. The values for two of such scalars, namely microsomal and cytosolic protein per gram of liver, is not known in patients with cancer. This study presents, for the first time, scaling factors from cancerous and matched histologically normal livers. PBPK simulations of various metabolically cleared drugs demonstrate the necessity of population-specific scaling for model-informed precision dosing in oncology.


Assuntos
Antinematódeos/farmacocinética , Neoplasias Colorretais/patologia , Neoplasias Hepáticas/fisiopatologia , Fígado/metabolismo , Modelos Biológicos , Adulto , Idoso , Idoso de 80 Anos ou mais , Antinematódeos/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Relação Dose-Resposta a Droga , Feminino , Hepatectomia , Eliminação Hepatobiliar , Humanos , Fígado/patologia , Fígado/cirurgia , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Masculino , Taxa de Depuração Metabólica , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade
4.
Mol Pharm ; 18(8): 2997-3009, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34283621

RESUMO

Physiologically based pharmacokinetic (PBPK) models are increasingly used in drug development to simulate changes in both systemic and tissue exposures that arise as a result of changes in enzyme and/or transporter activity. Verification of these model-based simulations of tissue exposure is challenging in the case of transporter-mediated drug-drug interactions (tDDI), in particular as these may lead to differential effects on substrate exposure in plasma and tissues/organs of interest. Gadoxetate, a promising magnetic resonance imaging (MRI) contrast agent, is a substrate of organic-anion-transporting polypeptide 1B1 (OATP1B1) and multidrug resistance-associated protein 2 (MRP2). In this study, we developed a gadoxetate PBPK model and explored the use of liver-imaging data to achieve and refine in vitro-in vivo extrapolation (IVIVE) of gadoxetate hepatic transporter kinetic data. In addition, PBPK modeling was used to investigate gadoxetate hepatic tDDI with rifampicin i.v. 10 mg/kg. In vivo dynamic contrast-enhanced (DCE) MRI data of gadoxetate in rat blood, spleen, and liver were used in this analysis. Gadoxetate in vitro uptake kinetic data were generated in plated rat hepatocytes. Mean (%CV) in vitro hepatocyte uptake unbound Michaelis-Menten constant (Km,u) of gadoxetate was 106 µM (17%) (n = 4 rats), and active saturable uptake accounted for 94% of total uptake into hepatocytes. PBPK-IVIVE of these data (bottom-up approach) captured reasonably systemic exposure, but underestimated the in vivo gadoxetate DCE-MRI profiles and elimination from the liver. Therefore, in vivo rat DCE-MRI liver data were subsequently used to refine gadoxetate transporter kinetic parameters in the PBPK model (top-down approach). Active uptake into the hepatocytes refined by the liver-imaging data was one order of magnitude higher than the one predicted by the IVIVE approach. Finally, the PBPK model was fitted to the gadoxetate DCE-MRI data (blood, spleen, and liver) obtained with and without coadministered rifampicin. Rifampicin was estimated to inhibit active uptake transport of gadoxetate into the liver by 96%. The current analysis highlighted the importance of gadoxetate liver data for PBPK model refinement, which was not feasible when using the blood data alone, as is common in PBPK modeling applications. The results of our study demonstrate the utility of organ-imaging data in evaluating and refining PBPK transporter IVIVE to support the subsequent model use for quantitative evaluation of hepatic tDDI.


Assuntos
Meios de Contraste/farmacocinética , Gadolínio DTPA/farmacocinética , Fígado/diagnóstico por imagem , Fígado/metabolismo , Imageamento por Ressonância Magnética/métodos , Rifampina/farmacocinética , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Biomarcadores/metabolismo , Células Cultivadas , Meios de Contraste/administração & dosagem , Meios de Contraste/metabolismo , Interações Medicamentosas , Gadolínio DTPA/administração & dosagem , Gadolínio DTPA/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Masculino , Modelos Animais , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/metabolismo , Ratos , Rifampina/administração & dosagem , Rifampina/metabolismo
5.
Drug Metab Dispos ; 48(12): 1271-1282, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32978222

RESUMO

In vitro to in vivo extrapolation (IVIVE) enables prediction of in vivo clinical outcomes related to drug exposure in various populations from in vitro data. Prudent IVIVE requires scalars specific to the biologic characteristics of the system in each population. This study determined experimentally for the first time scalars in liver samples from patients with varying degrees of cirrhosis. Microsomal and cytosolic fractions were extracted from 13 noncirrhotic and 32 cirrhotic livers (six mild, 13 moderate, and 13 severe, based on Child-Pugh score). Fractional protein content was determined, and cytochrome P450 reductase activity was used to correct for microsomal protein loss. Although the median microsomal protein per gram liver (MPPGL) in mild, moderate, and severe cirrhosis (26.2, 32.4, and 30.8 mg⋅g-1, respectively) seemed lower than control livers (36.6 mg⋅g-1), differences were not statistically significant (Kruskal-Wallis test, P > 0.05). Corresponding values for cytosolic protein per gram liver were 88.2, 67.9, 62.2, and 75.4 (mg⋅g-1) for mild, moderate, and severe cirrhosis and control livers, respectively, with statistically lower values for severe versus controls (Mann-Whitney P = 0.006). Cirrhosis associated with cancer showed lower MPPGL (24.8 mg⋅g-1) than cirrhosis associated with cholestasis (38.3 mg⋅g-1, P = 0.003). Physiologically based pharmacokinetic simulations with disease-specific scalars captured cirrhosis impact on exposure to alfentanil, metoprolol, midazolam, and ethinylestradiol. These experimentally-determined scalars should alleviate the need for indirect scaling using functional liver volume. Scaling factors in cirrhosis might be a reflection of the etiology rather than the disease severity. Hence, bundling various cirrhotic conditions under the same umbrella when predicting hepatic impairment impact should be revisited. SIGNIFICANCE STATEMENT: Cirrhosis-specific scalars required for extrapolation from microsomal or cytosolic in vitro systems to liver tissue are lacking. These scalars can help in predicting drug clearance and selection of dosage regimens for cirrhosis populations. Attempts to consider potential changes have been empirical and ignored the potential impact of the cause of cirrhosis. We obtained experimental values for these scalars for the first time and assessed their impact on predicted exposure to various substrate drugs using physiologically-based pharmacokinetics simulations.


Assuntos
Eliminação Hepatobiliar/fisiologia , Cirrose Hepática/fisiopatologia , Fígado/metabolismo , Administração Intravenosa , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Voluntários Saudáveis , Humanos , Fígado/fisiopatologia , Cirrose Hepática/diagnóstico , Masculino , Microssomos Hepáticos , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Adulto Jovem
6.
J Pharmacol Exp Ther ; 368(2): 157-168, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30413628

RESUMO

In vitro-in vivo extrapolation (IVIVE) of renal excretory clearance (CLR) using the physiologically based kidney models can provide mechanistic insight into the interplay of multiple processes occurring in the renal tubule; however, the ability of these models to capture quantitatively the impact of perturbed conditions (e.g., urine flow, urine pH changes) on CLR has not been fully evaluated. In this work, we aimed to assess the predictability of the effect of urine flow and urine pH on CLR and tubular drug concentrations (selected examples). Passive diffusion clearance across the nephron tubule membrane was scaled from in vitro human epithelial cell line Caco-2 permeability data by nephron tubular surface area to predict the fraction reabsorbed and the CLR of caffeine, chloramphenicol, creatinine, dextroamphetamine, nicotine, sulfamethoxazole, and theophylline. CLR values predicted using mechanistic kidney model at a urinary pH of 6.2 and 7.4 resulted in prediction bias of 2.87- and 3.62-fold, respectively. Model simulations captured urine flow-dependent CLR, albeit with minor underprediction of the observed magnitude of change. The relationship between drug solubility, urine flow, and urine pH, illustrated in simulated intratubular concentrations of acyclovir and sulfamethoxazole, agreed with clinical data on tubular precipitation and crystal-induced acute kidney injury. This study represents the first systematic evaluation of the ability of the mechanistic kidney model to capture the impact of urine flow and urine pH on CLR and drug tubular concentrations with the aim of facilitating refinement of IVIVE-based mechanistic prediction of renal excretion.


Assuntos
Taxa de Depuração Metabólica/fisiologia , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Eliminação Renal/fisiologia , Micção/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Testes de Função Renal/métodos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Masculino , Taxa de Depuração Metabólica/efeitos dos fármacos , Eliminação Renal/efeitos dos fármacos , Micção/efeitos dos fármacos , Adulto Jovem
7.
Mol Pharm ; 16(11): 4551-4562, 2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31525064

RESUMO

Indoxyl sulfate (IxS), a highly albumin-bound uremic solute, accumulates in chronic kidney disease (CKD) due to reduced renal clearance. This study was designed to specifically investigate the role of human serum albumin (HSA) in IxS renal secretion via organic anion transporter 1 (OAT1) in a microfluidic system and subsequently apply quantitative translation of in vitro data to predict extent of change in IxS renal clearance in CKD stage IV relative to healthy. Conditionally immortalized human proximal tubule epithelial cells overexpressing OAT1 were incubated with IxS (5-200 µM) in the HSA-free medium or in the presence of either HSA or CKD-modified HSA. IxS uptake in the presence of HSA resulted in more than 20-fold decrease in OAT1 affinity (Km,u) and 37-fold greater in vitro unbound intrinsic clearance (CLint,u) versus albumin-free condition. In the presence of CKD-modified albumin, Km,u increased four-fold and IxS CLint,u decreased almost seven-fold relative to HSA. Fold-change in parameters exceeded differences in IxS binding between albumin conditions, indicating additional mechanism and facilitating role of albumin in IxS OAT1-mediated uptake. Quantitative translation of IxS in vitro OAT1-mediated CLint,u predicted a 60% decrease in IxS renal elimination as a result of CKD, in agreement with the observed data (80%). The findings of the current study emphasize the role of albumin in IxS transport via OAT1 and explored the impact of modifications in albumin on renal excretion via active secretion in CKD. For the first time, this study performed quantitative translation of transporter kinetic data generated in a novel microfluidic in vitro system to a clinically relevant setting. Knowledge gaps and future directions in quantitative translation of renal drug disposition from microphysiological systems are discussed.


Assuntos
Transporte Biológico/fisiologia , Indicã/metabolismo , Insuficiência Renal Crônica/metabolismo , Albumina Sérica Humana/metabolismo , Linhagem Celular , Humanos , Túbulos Renais Proximais/metabolismo , Cinética , Proteínas de Membrana Transportadoras/metabolismo , Microfluídica , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo
8.
J Pharmacol Exp Ther ; 360(3): 484-495, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28057840

RESUMO

Development of submodels of organs within physiologically-based pharmacokinetic (PBPK) principles and beyond simple perfusion limitations may be challenging because of underdeveloped in vitro-in vivo extrapolation approaches or lack of suitable clinical data for model refinement. However, advantage of such models in predicting clinical observations in divergent patient groups is now commonly acknowledged. Mechanistic understanding of altered renal secretion in renal impairment is one area that may benefit from such models, despite knowledge gaps in renal pathophysiology. In the current study, a PBPK kidney model was developed for digoxin, accounting for the roles of organic anion transporting peptide 4C1 (OATP4C1) and P-glycoprotein (P-gp) in its tubular secretion, with the aim to investigate the impact of age and renal impairment (moderate to severe) on renal drug disposition. Initial PBPK simulations based on changes in glomerular filtration rate (GFR) underestimated the observed reduction in digoxin renal excretion clearance (CLR) in subjects with moderately impaired renal function relative to healthy. Reduction in either proximal tubule cell number or the OATP4C1 abundance in the mechanistic kidney model successfully predicted 59% decrease in digoxin CLR, in particular when these changes were proportional to reduction in GFR. In contrast, predicted proximal tubule concentration of digoxin was only sensitive to changes in the transporter expression/ million proximal tubule cells. Based on the mechanistic modeling, reduced proximal tubule cellularity and OATP4C1 abundance, and inhibition of OATP4C1-mediated transport, are proposed as possible causes of reduced digoxin renal secretion in renally impaired patients.


Assuntos
Digoxina/farmacocinética , Rim , Insuficiência Renal , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Taxa de Filtração Glomerular/efeitos dos fármacos , Taxa de Filtração Glomerular/fisiologia , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/fisiopatologia , Taxa de Depuração Metabólica/efeitos dos fármacos , Taxa de Depuração Metabólica/fisiologia , Modelos Biológicos , Transportadores de Ânions Orgânicos/metabolismo , Eliminação Renal/efeitos dos fármacos , Eliminação Renal/fisiologia , Insuficiência Renal/metabolismo , Insuficiência Renal/fisiopatologia
9.
Drug Metab Dispos ; 45(5): 556-568, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28270564

RESUMO

In vitro-in vivo extrapolation of drug metabolism data obtained in enriched preparations of subcellular fractions rely on robust estimates of physiologically relevant scaling factors for the prediction of clearance in vivo. The purpose of the current study was to measure the microsomal and cytosolic protein per gram of kidney (MPPGK and CPPGK) in dog and human kidney cortex using appropriate protein recovery marker and evaluate functional activity of human cortex microsomes. Cytochrome P450 (CYP) content and glucose-6-phosphatase (G6Pase) activity were used as microsomal protein markers, whereas glutathione-S-transferase activity was a cytosolic marker. Functional activity of human microsomal samples was assessed by measuring mycophenolic acid glucuronidation. MPPGK was 33.9 and 44.0 mg/g in dog kidney cortex, and 41.1 and 63.6 mg/g in dog liver (n = 17), using P450 content and G6Pase activity, respectively. No trends were noted between kidney, liver, and intestinal scalars from the same animals. Species differences were evident, as human MPPGK and CPPGK were 26.2 and 53.3 mg/g in kidney cortex (n = 38), respectively. MPPGK was 2-fold greater than the commonly used in vitro-in vivo extrapolation scalar; this difference was attributed mainly to tissue source (mixed kidney regions versus cortex). Robust human MPPGK and CPPGK scalars were measured for the first time. The work emphasized the importance of regional differences (cortex versus whole kidney-specific MPPGK, tissue weight, and blood flow) and a need to account for these to improve assessment of renal metabolic clearance and its extrapolation to in vivo.


Assuntos
Citosol/metabolismo , Córtex Renal/metabolismo , Microssomos/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Citosol/química , Cães , Feminino , Glucose-6-Fosfatase/metabolismo , Humanos , Córtex Renal/química , Masculino , Microssomos/química , Especificidade da Espécie
10.
J Pharm Sci ; 113(6): 1664-1673, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38417790

RESUMO

Accounting for variability in plasma protein binding of drugs is an essential input to physiologically-based pharmacokinetic (PBPK) models of special populations. Prediction of fraction unbound in plasma (fu) in such populations typically considers changes in plasma protein concentration while assuming that the binding affinity remains unchanged. A good correlation between predicted vs observed fu data reported for various drugs in a given special population is often used as a justification for such predictive methods. However, none of these analyses evaluated the prediction of the fold-change in fu in special populations relative to the reference population. This would be a more appropriate assessment of the predictivity, analogous to drug-drug interactions. In this study, predictive performance of the single protein binding model was assessed by predicting fu for alpha-1-acid glycoprotein and albumin bound drugs in hepatic impairment, renal impairment, paediatric, elderly, patients with inflammatory disease, and in different ethnic groups for a dataset of >200 drugs. For albumin models, the concordance correlation coefficients for predicted fu were >0.90 for 16 out of 17 populations with sub-groups, indicating strong agreement between predicted and observed values. In contrast, concordance correlation coefficients for predicted fold-change in fu for the same dataset were <0.38 for all populations and sub-groups. Trends were similar for alpha-1-acid glycoprotein models. Accordingly, the predictions of fu solely based on changes in protein concentrations in plasma cannot explain the observed values in some special populations. We recommend further consideration of the impact of changes in special populations to endogenous substances that competitively bind to plasma proteins, and changes in albumin structure due to posttranslational modifications. PBPK models of special populations for highly bound drugs should preferably use measured fu data to ensure reliable prediction of drug exposure or compare predicted unbound drug exposure between populations knowing that these will not be sensitive to changes in fu.


Assuntos
Proteínas Sanguíneas , Modelos Biológicos , Ligação Proteica , Humanos , Proteínas Sanguíneas/metabolismo , Preparações Farmacêuticas/metabolismo , Preparações Farmacêuticas/sangue , Orosomucoide/metabolismo , Idoso , Criança , Farmacocinética
11.
Clin Pharmacol Ther ; 114(6): 1243-1253, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37620246

RESUMO

Monitoring endogenous biomarkers is increasingly used to evaluate transporter-mediated drug-drug interactions (DDIs) in early drug development and may be applied to elucidate changes in transporter activity in disease. 4-pyridoxic acid (PDA) has been identified as the most sensitive plasma endogenous biomarker of renal organic anion transporters (OAT1/3). Increase in PDA baseline concentrations was observed after administration of probenecid, a strong clinical inhibitor of OAT1/3 and also in patients with chronic kidney disease (CKD). The aim of this study was to develop and verify a physiologically-based pharmacokinetic (PBPK) model of PDA, to predict the magnitude of probenecid DDI and predict the CKD-related changes in PDA baseline. The PBPK model for PDA was first developed in healthy population, building on from previous population pharmacokinetic modeling, and incorporating a mechanistic kidney model to consider OAT1/3-mediated renal secretion. Probenecid PBPK model was adapted from the Simcyp database and re-verified to capture its dose-dependent pharmacokinetics (n = 9 studies). The PBPK model successfully predicted the PDA plasma concentrations, area under the curve, and renal clearance in healthy subjects at baseline and after single/multiple probenecid doses. Prospective simulations in severe CKD predicted successfully the increase in PDA plasma concentration relative to healthy (within 2-fold of observed data) after accounting for 60% increase in fraction unbound in plasma and additional 50% decline in OAT1/3 activity beyond the decrease in glomerular filtration rate. The verified PDA PBPK model supports future robust evaluation of OAT1/3 DDI in drug development and increases our confidence in predicting exposure and renal secretion in patients with CKD.


Assuntos
Ácido Piridóxico , Insuficiência Renal Crônica , Humanos , Probenecid/farmacologia , Insuficiência Renal Crônica/tratamento farmacológico , Rim , Interações Medicamentosas , Biomarcadores , Modelos Biológicos
12.
Eur J Pharm Sci ; 182: 106375, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36626943

RESUMO

Biologics are a fast-growing therapeutic class, with intertwined pharmacokinetics and pharmacodynamics, affected by the abundance and function of the FcRn receptor. While many investigators assume adequacy of classical models, such as allometry, for pharmacokinetic characterization of biologics, advocates of physiologically-based pharmacokinetics (PBPK) propose consideration of known systems parameters that affect the fate of biologics to enable a priori predictions, which go beyond allometry. The aim of this study was to deploy a systems-informed modelling approach to predict the disposition of Fc-containing biologics. We used global proteomics to quantify the FcRn receptor [p51 and ß2-microglobulin (B2M) subunits] in 167 samples of human tissue (liver, intestine, kidney and skin) and assessed covariates of its expression. FcRn p51 subunit was highest in liver relative to other tissues, and B2M was 1-2 orders of magnitude more abundant than FcRn p51 across all sets. There were no sex-related differences, while higher expression was confirmed in neonate liver compared with adult liver. Trends of expression in liver and kidney indicated a moderate effect of body mass index, which should be confirmed in a larger sample size. Expression of FcRn p51 subunit was approximately 2-fold lower in histologically normal liver tissue adjacent to cancer compared with healthy liver. FcRn mRNA in plasma-derived exosomes correlated moderately with protein abundance in matching liver tissue, opening the possibility of use as a potential clinical tool. Predicted effects of trends in FcRn abundance in healthy and disease (cancer and psoriasis) populations using trastuzumab and efalizumab PBPK models were in line with clinical observations, and global sensitivity analysis revealed endogenous IgG plasma concentration and tissue FcRn abundance as key systems parameters influencing exposure to Fc-conjugated biologics.


Assuntos
Produtos Biológicos , Adulto , Recém-Nascido , Humanos , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/análise , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo , Fígado/metabolismo
13.
Pharmaceutics ; 15(3)2023 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-36986758

RESUMO

Gadoxetate, a magnetic resonance imaging (MRI) contrast agent, is a substrate of organic-anion-transporting polypeptide 1B1 and multidrug resistance-associated protein 2. Six drugs, with varying degrees of transporter inhibition, were used to assess gadoxetate dynamic contrast enhanced MRI biomarkers for transporter inhibition in rats. Prospective prediction of changes in gadoxetate systemic and liver AUC (AUCR), resulting from transporter modulation, were performed by physiologically-based pharmacokinetic (PBPK) modelling. A tracer-kinetic model was used to estimate rate constants for hepatic uptake (khe), and biliary excretion (kbh). The observed median fold-decreases in gadoxetate liver AUC were 3.8- and 1.5-fold for ciclosporin and rifampicin, respectively. Ketoconazole unexpectedly decreased systemic and liver gadoxetate AUCs; the remaining drugs investigated (asunaprevir, bosentan, and pioglitazone) caused marginal changes. Ciclosporin decreased gadoxetate khe and kbh by 3.78 and 0.09 mL/min/mL, while decreases for rifampicin were 7.20 and 0.07 mL/min/mL, respectively. The relative decrease in khe (e.g., 96% for ciclosporin) was similar to PBPK-predicted inhibition of uptake (97-98%). PBPK modelling correctly predicted changes in gadoxetate systemic AUCR, whereas underprediction of decreases in liver AUCs was evident. The current study illustrates the modelling framework and integration of liver imaging data, PBPK, and tracer-kinetic models for prospective quantification of hepatic transporter-mediated DDI in humans.

14.
Clin Pharmacol Ther ; 112(3): 643-652, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35569107

RESUMO

There is growing evidence that active tubular secretory clearance (CLs ) may not decline proportionally with the glomerular filtration rate (GFR) in chronic kidney disease (CKD), leading to the overestimation of renal clearance (CLr ) when using solely GFR to approximate disease effect on renal elimination. The clinical pharmacokinetic data of 33 renally secreted OAT1/3 substrates were collated to investigate the impact of mild, moderate, and severe CKD on CLr , tubular secretion and protein binding (fu,p ). The fu,p of the collated substrates ranged from 0.0026 to 1.0 in healthy populations; observed CKD-related increase in the fu,p (up to 2.7-fold) of 8 highly bound substrates (fu,p ≤ 0.2) was accounted for in the analysis. Use of prediction equation based on disease-related changes in albumin resulted in underprediction of the CKD-related increase in fu,p of highly bound substrates, highlighting the necessity to measure protein binding in severe CKD. The critical analysis of clinical data for 33 OAT1/3 probes established that decrease in OAT1/3 activity proportional to the changes in GFR was insufficient to recapitulate effects of severe CKD on unbound tubular secretion clearance. OAT1/3-mediated CLs was estimated to decline by an additional 50% relative to the GFR decline in severe CKD, whereas change in active secretion in mild and moderate CKD was proportional to GFR. Consideration of this additional 50% decline in OAT1/3-mediated CLs is recommended for physiologically-based pharmacokinetic models and dose adjustment of OAT1/3 substrates in severe CKD, especially for substrates with high contribution of the active secretion to CLr .


Assuntos
Transportadores de Ânions Orgânicos , Insuficiência Renal Crônica , Taxa de Filtração Glomerular , Humanos , Rim/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Eliminação Renal , Insuficiência Renal Crônica/metabolismo
15.
Clin Pharmacol Ther ; 112(3): 615-626, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35652251

RESUMO

Coproporphyrin I (CPI) is an endogenous biomarker of organic anion transporting polypeptide 1B transporter (OATP1B). CPI plasma baseline was reported to increase with severity of chronic kidney disease (CKD). Further, ratio of CPI area under the plasma concentration-time curve (AUCR) in the presence/absence of OATP1B inhibitor rifampin was higher in patients with CKD compared with healthy participants, in contrast to pitavastatin (a clinical OATP1B probe). This study investigated mechanism(s) contributing to altered CPI baseline in patients with CKD by extending a previously developed physiologically-based pharmacokinetic (PBPK) model to this patient population. CKD-related covariates were evaluated in a stepwise manner on CPI fraction unbound in plasma (fu,p ), OATP1B-mediated hepatic uptake clearance (CLactive ), renal clearance (CLR ), and endogenous synthesis (ksyn ). The CPI model successfully recovered increased baseline and rifampin-mediated AUCR in patients with CKD by accounting for the following disease-related changes: 13% increase in fu,p , 29% and 39% decrease in CLactive in mild and moderate to severe CKD, respectively, decrease in CLR proportional to decline in glomerular filtration rate, and 27% decrease in ksyn in severe CKD. Almost complete decline in CPI renal elimination in severe CKD increased its fraction transported by OATP1B, rationalizing differences in the CPI-rifampin interaction observed between healthy participants and patients with CKD. In conclusion, mechanistic modeling performed here supports CKD-related decrease in OATP1B function to inform prospective PBPK modeling of OATP1B-mediated drug-drug interaction in these patients. Monitoring of CPI allows detection of CKD-drug interaction risk for OATP1B drugs with combined hepatic and renal elimination which may be underestimated by extrapolating the interaction risk based on pitavastatin data in healthy participants.


Assuntos
Coproporfirinas , Transportadores de Ânions Orgânicos , Insuficiência Renal Crônica , Biomarcadores , Coproporfirinas/análise , Interações Medicamentosas , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado , Transportadores de Ânions Orgânicos/metabolismo , Estudos Prospectivos , Insuficiência Renal Crônica/diagnóstico , Rifampina/farmacologia
16.
AAPS J ; 24(1): 13, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34907479

RESUMO

Dosing guidance is often lacking for chronic kidney disease (CKD) due to exclusion of such patients from pivotal clinical trials. Physiologically based pharmacokinetic (PBPK) modelling supports model-informed dosing when clinical data are lacking, but application of these approaches to patients with impaired renal function is not yet at full maturity. In the current study, a ganciclovir PBPK model was developed for patients with normal renal function and extended to CKD population. CKD-related changes in tubular secretion were explored in the mechanistic kidney model and implemented either as proportional or non-proportional decline relative to GFR. Crystalluria risk was evaluated in different clinical settings (old age, severe CKD and low fluid intake) by simulating ganciclovir medullary collecting duct (MCD) concentrations. The ganciclovir PBPK model captured observed changes in systemic pharmacokinetic endpoints in mild-to-severe CKD; these trends were evident irrespective of assumed pathophysiological mechanism of altered active tubular secretion in the model. Minimal difference in simulated ganciclovir MCD concentrations was noted between young adult and geriatric populations with normal renal function and urine flow (1 mL/min), with lower concentrations predicted for severe CKD patients. High crystalluria risk was identified at reduced urine flow (0.1 mL/min) as simulated ganciclovir MCD concentrations exceeded its solubility (2.6-6 mg/mL), irrespective of underlying renal function. The analysis highlighted the importance of appropriate distribution of virtual subjects' systems data in CKD populations. The ganciclovir PBPK model illustrates the ability of this translational tool to explore individual and combined effects of age, urine flow, and renal impairment on local drug renal exposure.


Assuntos
Ganciclovir , Insuficiência Renal , Idoso , Simulação por Computador , Humanos , Rim , Modelos Biológicos , Adulto Jovem
17.
Clin Pharmacol Ther ; 110(5): 1389-1400, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34390491

RESUMO

The applications of translational modeling of local drug concentrations in various organs had a sharp increase over the last decade. These are part of the model-informed drug development initiative, adopted by the pharmaceutical industry and promoted by drug regulatory agencies. With respect to the kidney, the models serve as a bridge for understanding animal vs. human observations related to renal drug disposition and any consequential adverse effects. However, quantitative data on key drug-metabolizing enzymes and transporters relevant for predicting renal drug disposition are limited. Using targeted and global quantitative proteomics, we determined the abundance of multiple enzymes and transporters in 20 human kidney cortex samples. Nine enzymes and 22 transporters were quantified (8 for the first time in the kidneys). In addition, > 4,000 proteins were identified and used to form an open database. CYP2B6, CYP3A5, and CYP4F2 showed comparable, but generally low expression, whereas UGT1A9 and UGT2B7 levels were the highest. Significant correlation between abundance and activity (measured by mycophenolic acid clearance) was observed for UGT1A9 (Rs = 0.65, P = 0.004) and UGT2B7 (Rs = 0.70, P = 0.023). Expression of P-gp ≈ MATE-1 and OATP4C1 transporters were high. Strong intercorrelations were observed between several transporters (P-gp/MRP4, MRP2/OAT3, and OAT3/OAT4); no correlation in expression was apparent for functionally related transporters (OCT2/MATEs). This study extends our knowledge of pharmacologically relevant proteins in the kidney cortex, with implications on more prudent use of mechanistic kidney models under the general framework of quantitative systems pharmacology and toxicology.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Córtex Renal/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Proteômica/métodos , Sistema Enzimático do Citocromo P-450/genética , Bases de Dados Factuais , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Rim/metabolismo , Cinética , Proteínas de Membrana Transportadoras/genética , UDP-Glucuronosiltransferase 1A
18.
Clin Pharmacokinet ; 60(9): 1187-1199, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33840062

RESUMO

BACKGROUND: Endogenous biomarkers are promising tools to assess transporter-mediated drug-drug interactions early in humans. METHODS: We evaluated on a common and validated in vitro system the selectivity of 4-pyridoxic acid (PDA), homovanillic acid (HVA), glycochenodeoxycholate-3-sulphate (GCDCA-S) and taurine towards different renal transporters, including multidrug resistance-associated protein, and assessed the in vivo biomarker sensitivity towards the strong organic anion transporter (OAT) inhibitor probenecid at 500 mg every 6 h to reach close to complete OAT inhibition. RESULTS: PDA and HVA were substrates of the OAT1/2/3, OAT4 (PDA only) and multidrug resistance-associated protein 4; GCDCA-S was more selective, having affinity only towards OAT3 and multidrug resistance-associated protein 2. Taurine was not a substrate of any of the investigated transporters under the in vitro conditions tested. Plasma exposure of PDA and HVA significantly increased and the renal clearance of GCDCA-S, PDA and HVA decreased; the magnitude of these changes was comparable to those of known clinical OAT probe substrates. PDA and GCDCA-S were the most promising endogenous biomarkers of the OAT pathway activity: PDA plasma exposure was the most sensitive to probenecid inhibition, and, in contrast, GCDCA-S was the most sensitive OAT biomarker based on renal clearance, with higher selectivity towards the OAT3 transporter. CONCLUSIONS: The current findings illustrate a clear benefit of measuring PDA plasma exposure during phase I studies when a clinical drug candidate is suspected to be an OAT inhibitor based on in vitro data. Subsequently, combined monitoring of PDA and GCDCA-S in both urine and plasma is recommended to tease out the involvement of OAT1/3 in the inhibition interaction. CLINICAL TRIAL REGISTRATION: EudraCT number: 2016-003923-49.


Assuntos
Proteína 1 Transportadora de Ânions Orgânicos , Preparações Farmacêuticas , Biomarcadores , Interações Medicamentosas , Células HEK293 , Humanos , Rim , Transportadores de Ânions Orgânicos Sódio-Independentes
19.
CPT Pharmacometrics Syst Pharmacol ; 9(12): 695-706, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33049120

RESUMO

Elevated serum creatinine (SCr ) caused by the inhibition of renal transporter(s) may be misinterpreted as kidney injury. The interpretation is more complicated in patients with chronic kidney disease (CKD) due to altered disposition of creatinine and renal transporter inhibitors. A clinical study was conducted in 17 patients with CKD (estimated glomerular filtration rate 15-59 mL/min/1.73 m2 ); changes in SCr were monitored during trimethoprim treatment (100-200 mg/day), administered to prevent recurrent urinary infection, relative to the baseline level. Additional SCr -interaction data with trimethoprim, cimetidine, and famotidine in patients with CKD were collated from the literature. Our published physiologically-based creatinine model was extended to predict the effect of the CKD on SCr and creatinine-drug interaction. The creatinine-CKD model incorporated age/sex-related differences in creatinine synthesis, CKD-related glomerular filtration deterioration; change in transporter activity either proportional or disproportional to glomerular filtration rate (GFR) decline were explored. Optimized models successfully recovered baseline SCr from 64 patients with CKD (geometric mean fold-error of 1.1). Combined with pharmacokinetic models of inhibitors, the creatinine model was used to simulate transporter-mediated creatinine-drug interactions. Use of inhibitor unbound plasma concentrations resulted in 66% of simulated SCr interaction data within the prediction limits, with cimetidine interaction significantly underestimated. Assuming that transporter activity deteriorates disproportional to GFR decline resulted in higher predicted sensitivity to transporter inhibition in patients with CKD relative to healthy patients, consistent with sparse clinical data. For the first time, this novel modelling approach enables quantitative prediction of SCr in CKD and delineation of the effect of disease and renal transporter inhibition in this patient population.


Assuntos
Creatinina/sangue , Inibidores do Citocromo P-450 CYP2C8/farmacocinética , Insuficiência Renal Crônica/sangue , Trimetoprima/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Cimetidina/farmacocinética , Simulação por Computador , Inibidores do Citocromo P-450 CYP1A2/farmacocinética , Inibidores do Citocromo P-450 CYP2C8/administração & dosagem , Inibidores do Citocromo P-450 CYP2C8/uso terapêutico , Interações Medicamentosas , Famotidina/farmacocinética , Feminino , Taxa de Filtração Glomerular/fisiologia , Antagonistas dos Receptores H2 da Histamina/farmacocinética , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Trimetoprima/administração & dosagem , Trimetoprima/uso terapêutico , Infecções Urinárias/tratamento farmacológico , Infecções Urinárias/prevenção & controle
20.
CPT Pharmacometrics Syst Pharmacol ; 9(5): 282-293, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32410382

RESUMO

Creatinine is widely used as a biomarker of glomerular filtration, and, hence, renal function. However, transporter-mediated secretion also contributes to its renal clearance, albeit to a lesser degree. Inhibition of these transporters causes transient serum creatinine elevation, which can be mistaken as impaired renal function. The current study developed mechanistic models of creatinine kinetics within physiologically based framework accounting for multiple transporters involved in creatinine renal elimination, assuming either unidirectional or bidirectional-OCT2 transport (driven by electrochemical gradient). Robustness of creatinine models was assessed by predicting creatinine-drug interactions with 10 perpetrators; performance evaluation accounted for 5% intra-individual variability in serum creatinine. Models showed comparable predictive performances of the maximum steady-state effect regardless of OCT2 directionality assumptions. However, only the bidirectional-OCT2 model successfully predicted the minimal effect of ranitidine. The dynamic nature of models provides clear advantage to static approaches and most advanced framework for evaluating interplay between multiple processes in creatinine renal disposition.


Assuntos
Biomarcadores/metabolismo , Creatinina/sangue , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Transporte Biológico/fisiologia , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Testes de Função Renal , Transportador 2 de Cátion Orgânico/metabolismo , Preparações Farmacêuticas/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA