Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Mater Today Bio ; 27: 101143, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39070097

RESUMO

Recent FDA modernization act 2.0 has led to increasing industrial R&D investment in advanced in vitro 3D models such as organoids, spheroids, organ-on-chips, 3D bioprinting, and in silico approaches. Liver-related advanced in vitro models remain the prime area of interest, as liver plays a central role in drug clearance of compounds. Growing evidence indicates the importance of recapitulating the overall liver microenvironment to enhance hepatocyte maturity and culture longevity using liver-on-chips (LoC) in vitro. Hence, pharmaceutical industries have started exploring LoC assays in the two of the most challenging areas: accurate in vitro-in vivo extrapolation (IVIVE) of hepatic drug clearance and drug-induced liver injury. We examine the joint efforts of commercial chip manufacturers and pharmaceutical companies to present an up-to-date overview of the adoption of LoC technology in the drug discovery. Further, several roadblocks are identified to the rapid adoption of LoC assays in the current drug development framework. Finally, we discuss some of the underexplored application areas of LoC models, where conventional 2D hepatic models are deemed unsuitable. These include clearance prediction of metabolically stable compounds, immune-mediated drug-induced liver injury (DILI) predictions, bioavailability prediction with gut-liver systems, hepatic clearance prediction of drugs given during pregnancy, and dose adjustment studies in disease conditions. We conclude the review by discussing the importance of PBPK modeling with LoC, digital twins, and AI/ML integration with LoC.

2.
Molecules ; 29(6)2024 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-38542971

RESUMO

Understanding the final fate of nanomaterials (NMs) in the liver is crucial for their safer application. As a representative two-dimensional (2D) soft nanomaterial, graphene oxide (GO) has shown to have high potential for applications in the biomedical field, including in biosensing, drug delivery, tissue engineering, therapeutics, etc. GO has been shown to accumulate in the liver after entering the body, and thus, understanding the GO-liver interaction will facilitate the development of safer bio-applications. In this study, the hepatic clearance of two types of PEGylated GOs with different lateral sizes (s-GOs: ~70 nm and l-GOs: ~300 nm) was carefully investigated. We found that GO sheets across the hepatic sinusoidal endothelium, which then may be taken up by the hepatocytes via the Disse space. The hepatocytes may degrade GO into dot-like particles, which may be excreted via the hepatobiliary route. In combination with ICP-MS, LA-ICP-MS, and synchrotron radiation FTIR techniques, we found that more s-GO sheets in the liver were prone to be cleared via hepatobiliary excretion than l-GO sheets. A Raman imaging analysis of ID/IG ratios further indicated that both s-GO and l-GO generated more defects in the liver. The liver microsomes may contribute to GO biotransformation into O-containing functional groups, which plays an important role in GO degradation and excretion. In particular, more small-sized GO sheets in the liver were more likely to be cleared via hepatobiliary excretion than l-GO sheets, and a greater clearance of s-GO will mitigate their hepatotoxicity. These results provide a better understanding of the hepatic clearance of soft NMs, which is important in the safer-by-design of GO.


Assuntos
Grafite , Hepatite , Nanoestruturas , Humanos
3.
Cell Metab ; 36(1): 103-115.e4, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38171330

RESUMO

The folate-dependent enzyme serine hydroxymethyltransferase (SHMT) reversibly converts serine into glycine and a tetrahydrofolate-bound one-carbon unit. Such one-carbon unit production plays a critical role in development, the immune system, and cancer. Using rodent models, here we show that the whole-body SHMT flux acts to net consume rather than produce glycine. Pharmacological inhibition of whole-body SHMT1/2 and genetic knockout of liver SHMT2 elevated circulating glycine levels up to eight-fold. Stable-isotope tracing revealed that the liver converts glycine to serine, which is then converted by serine dehydratase into pyruvate and burned in the tricarboxylic acid cycle. In response to diets deficient in serine and glycine, de novo biosynthetic flux was unaltered, but SHMT2- and serine-dehydratase-mediated catabolic flux was lower. Thus, glucose-derived serine synthesis is largely insensitive to systemic demand. Instead, circulating serine and glycine homeostasis is maintained through variable consumption, with liver SHMT2 a major glycine-consuming enzyme.


Assuntos
Glicina Hidroximetiltransferase , Glicina , Glicina Hidroximetiltransferase/genética , Homeostase , Carbono , Serina
4.
Toxics ; 11(12)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38133352

RESUMO

Few studies are available on the environmental and toxicological effects of perfluoroalkyl ether carboxylic acids (PFECAs), such as GenX, which are replacing legacy PFAS in manufacturing processes. To collect initial data on the toxicity and toxicokinetics of a longer-chain PFECA, male and female Sprague Dawley rats were exposed to perfluoro-(2,5,8-trimethyl-3,6,9-trioxadodecanoic) acid (HFPO-TeA) by oral gavage for five days over multiple dose levels (0.3-335.2 mg/kg/day). Clinically, we observed mortality at doses >17 mg/kg/day and body weight changes at doses ≤17 mg/kg/day. For the 17 mg/kg/day dose level, T3 and T4 thyroid hormone concentrations were significantly decreased (p < 0.05) from controls and HFPO-TeA plasma concentrations were significantly different between sexes. Non-targeted analysis of plasma and in vitro hepatocyte assay extractions revealed the presence of another GenX oligomer, perfluoro-(2,5-dimethyl-3,6-dioxanonanoic) acid (HFPO-TA). In vitro to in vivo extrapolation (IVIVE) parameterized with in vitro toxicokinetic data predicted steady-state blood concentrations that were within seven-fold of those observed in the in vivo study, demonstrating reasonable predictivity. The evidence of thyroid hormone dysregulation, sex-based differences in clinical results and dosimetry, and IVIVE predictions presented here suggest that the replacement PFECA HFPO-TeA induces a complex and toxic exposure response in rodents.

5.
Toxics ; 11(5)2023 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-37235277

RESUMO

Concern over per- and polyfluoroalkyl substances (PFAS) has increased as more is learned about their environmental presence, persistence, and bioaccumulative potential. The limited monitoring, toxicokinetic (TK), and toxicologic data available are inadequate to inform risk across this diverse domain. Here, 73 PFAS were selected for in vitro TK evaluation to expand knowledge across lesser-studied PFAS alcohols, amides, and acrylates. Targeted methods developed using gas chromatography-tandem mass spectrometry (GC-MS/MS) were used to measure human plasma protein binding and hepatocyte clearance. Forty-three PFAS were successfully evaluated in plasma, with fraction unbound (fup) values ranging from 0.004 to 1. With a median fup of 0.09 (i.e., 91% bound), these PFAS are highly bound but exhibit 10-fold lower binding than legacy perfluoroalkyl acids recently evaluated. Thirty PFAS evaluated in the hepatocyte clearance assay showed abiotic loss, with many exceeding 60% loss within 60 min. Metabolic clearance was noted for 11 of the 13 that were successfully evaluated, with rates up to 49.9 µL/(min × million cells). The chemical transformation simulator revealed potential (bio)transformation products to consider. This effort provides critical information to evaluate PFAS for which volatility, metabolism, and other routes of transformation are likely to modulate their environmental fates.

6.
Drug Metab Rev ; 55(3): 205-238, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36971325

RESUMO

According to the free drug hypothesis (FDH), only free, unbound drug is available to interact with biological targets. This hypothesis is the fundamental principle that continues to explain the vast majority of all pharmacokinetic and pharmacodynamic processes. Under the FDH, the free drug concentration at the target site is considered the driver of pharmacodynamic activity and pharmacokinetic processes. However, deviations from the FDH are observed in hepatic uptake and clearance predictions, where observed unbound intrinsic hepatic clearance (CLint,u) is larger than expected. Such deviations are commonly observed when plasma proteins are present and form the basis of the so-called plasma protein-mediated uptake effect (PMUE). This review will discuss the basis of plasma protein binding as it pertains to hepatic clearance based on the FDH, as well as several hypotheses that may explain the underlying mechanisms of PMUE. Notably, some, but not all, potential mechanisms remained aligned with the FDH. Finally, we will outline possible experimental strategies to elucidate PMUE mechanisms. Understanding the mechanisms of PMUE and its potential contribution to clearance underprediction is vital to improving the drug development process.


Assuntos
Proteínas Sanguíneas , Hepatócitos , Humanos , Hepatócitos/metabolismo , Proteínas Sanguíneas/metabolismo , Fígado/metabolismo , Transporte Biológico , Ligação Proteica , Modelos Biológicos
7.
J Diabetes Sci Technol ; 17(4): 1016-1028, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35343255

RESUMO

OBJECTIVE: Accurate, safe glycemic management requires reliable delivery of insulin doses. Insulin can be delivered subcutaneously for action over a longer period of time. Needle-free jet injectors provide subcutaneous (SC) delivery without requiring needle use, but the volume of insulin absorbed varies due to losses associated with the delivery method. This study employs model-based methods to determine the expected proportion of active insulin present from a needle-free SC dose. METHODS: Insulin, C-peptide, and glucose assay data from a frequently sampled insulin-modified oral glucose tolerance test trial with 2U SC insulin delivery, paired with a well-validated metabolic model, predict metabolic outcomes for N = 7 healthy adults. Subject-specific nonlinear hepatic clearance profiles are modeled over time using third-order basis splines with knots located at assay times. Hepatic clearance profiles are constrained within a physiological rate of change, and relative to plasma glucose profiles. Insulin loss proportions yielding optimal insulin predictions are then identified, quantifying delivery losses. RESULTS: Optimal parameter identification suggests losses of up to 22% of the nominal 2U SC dose. The degree of loss varies between subjects and between trials on the same subject. Insulin fit accuracy improves where loss greater than 5% is identified, relative to where delivery loss is not modeled. CONCLUSIONS: Modeling shows needle-free SC jet injection of a nominal dose of insulin does not necessarily provide metabolic action equivalent to total dose, and this availability significantly varies between trials. By quantifying and accounting for variability of jet injection insulin doses, better glycemic management outcomes using SC jet injection may be achieved.


Assuntos
Insulina Regular Humana , Insulina , Adulto , Humanos , Injeções a Jato , Injeções Subcutâneas , Teste de Tolerância a Glucose
8.
Pharm Biol ; 60(1): 2266-2275, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36412560

RESUMO

CONTEXT: Zeaxanthin is a yellow­coloured dietary carotenoid widely recognized as an essential component of the macula. It exerts blue light filtering and antioxidant activities, offering eye health and vision benefits. OBJECTIVE: This study explores the oral absorption and systemic disposition of zeaxanthin from biopharmaceutical and pharmacokinetic perspectives. MATERIALS AND METHODS: In vivo intravenous (5 and 10 mg/kg) and intraportal (5 mg/kg) pharmacokinetic studies were performed to determine intrinsic tissue­blood partition coefficient, elimination pathway, and hepatic clearance, of zeaxanthin in rats. Moreover, in vitro physicochemical property test, in situ closed loop study, in vivo oral pharmacokinetic study (20 and 100 mg/kg), and in vivo lymphatic absorption study (100 mg/kg) were conducted to investigate the gut absorption properties of zeaxanthin and assess the effects of several lipids on the lymphatic absorption of zeaxanthin in rats. RESULTS: Zeaxanthin exhibited poor solubility (≤144 ng/mL) and stability (6.0-76.9% of the initial amount remained at 24 h) in simulated gut luminal fluids. Gut absorption of zeaxanthin occurred primarily in the duodenum, but the major fraction (≥84.7%) of the dose remained unabsorbed across the entire gut tract. Considerable fractions of intravenous zeaxanthin accumulated in the liver, lung, and spleen (21.3, 11.7, and 2.0%, respectively). It was found that the liver is the major eliminating organ of zeaxanthin, accounting for 53.5-90.1% of the total clearance process (hepatic extraction ratio of 0.623). DISCUSSION AND CONCLUSIONS: To our knowledge, this is the first systematic study to report factors that determine the oral bioavailability and systemic clearance of zeaxanthin.


Assuntos
Antioxidantes , Carotenoides , Animais , Ratos , Zeaxantinas/metabolismo , Disponibilidade Biológica , Carotenoides/metabolismo , Antioxidantes/metabolismo , Fígado/metabolismo
9.
Eur J Pharm Sci ; 177: 106268, 2022 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-35901930

RESUMO

In vitro to in vivo extrapolation (IVIVE), an approach for hepatic clearance (CLH) prediction used worldwide, remains controversial due to systematic underprediction. Among the various probable factors, the original assumption of the hepatic mathematical model (i.e., the well-stirred model, WSM) may become problematic, leading to the underestimation of drug CLH. Having a similar prerequisite that the well-stirred conditions are homogenous with perfectly mixed reactants, but using a different driving concentration, the modified well-stirred model (MWSM) stands apart from the WSM. However, we believe that both models should coexist so that the entire well-stirred scenario can be completely illustrated. Consequently, we collected published data from the literature and employed a logistic regression method to differentiate the optimal timing of use between WSM and MWSM in drug CLH prediction. Generally, variances adopted in the regression, including partition coefficient (logP), fraction unbound (fu), volumes of distribution at steady-state (Vss), and mean residence time (MRT), corresponded to our assumption when protein-facilitated uptake was considered. Furthermore, a new empirical approach was introduced to allow practical use of the MWSM. The results showed that this model could provide a more precise prediction compared to previous empirical approaches. Therefore, these preliminary results not only delineated a more detailed structure and mechanism of MWSM but also highlighted its necessity and potential.


Assuntos
Fígado , Modelos Biológicos , Hepatócitos , Cinética , Fígado/metabolismo , Taxa de Depuração Metabólica , Preparações Farmacêuticas/metabolismo , Ligação Proteica
10.
J Pharm Sci ; 111(11): 2939-2942, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35660027

RESUMO

This commentary seeks to clarify the relationships between conservation of mass, spatial continuity, advective mass transport, and first-order drug metabolism in the liver. It also provides additional insight into the meaning of clearance thereby proving that the Pang-Rowland equation and its extensions are incorrect. The Kochak-Benet equation has been verified and is not a rearrangement of the Pang-Rowland equation.


Assuntos
Fígado , Modelos Biológicos , Cinética , Fígado/metabolismo , Taxa de Depuração Metabólica
11.
J Diabetes Sci Technol ; 16(4): 945-954, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-33478257

RESUMO

OBJECTIVE: Model-based metabolic tests require accurate identification of subject-specific parameters from measured assays. Insulin assays are used to identify insulin kinetics parameters, such as general and first-pass hepatic clearances. This study assesses the impact of intravenous insulin boluses on parameter identification precision. METHOD: Insulin and C-peptide data from two intravenous glucose tolerance test (IVGTT) trials of healthy adults (N = 10 × 2; denoted A and B), with (A) and without (B) insulin modification, were used to identify insulin kinetics parameters using a grid search. Monte Carlo analysis (N = 1000) quantifies variation in simulation error for insulin assay errors of 5%. A region of parameter values around the optimum was identified whose errors are within variation due to assay error. A smaller optimal region indicates more precise practical identifiability. Trial results were compared to assess identifiability and precision. RESULTS: Trial B, without insulin modification, has optimal parameter regions 4.7 times larger on average than Trial A, with 1-U insulin bolus modification. Ranges of optimal parameter values between trials A and B increase from 0.04 to 0.12 min-1 for hepatic clearance and from 0.07 to 0.14 for first-pass clearance on average. Trial B's optimal values frequently lie outside physiological ranges, further indicating lack of distinct identifiability. CONCLUSIONS: A small 1-U insulin bolus improves identification of hepatic clearance parameters by providing a smaller region of optimal parameter values. Adding an insulin bolus in metabolic tests can significantly improve identifiability and outcome test precision. Assay errors necessitate insulin modification in clinical tests to ensure identifiability and precision.


Assuntos
Insulina , Modelos Biológicos , Adulto , Peptídeo C , Simulação por Computador , Teste de Tolerância a Glucose , Humanos , Insulina/metabolismo , Cinética
12.
AAPS J ; 24(1): 5, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34853928

RESUMO

This report reviews concepts related to operation of the classic parallel-tube model (PTM) for hepatic disposition and examines two recent proposals of a newly derived equation to describe hepatic clearance (CLH). It is demonstrated that the proposed equation is identical to a re-arrangement of an earlier relationship from Pang and Rowland and provides a means of calculation of intrinsic clearance (CLint,PTM) rather than CLH as posed. We further demonstrate how classic hepatic clearance models with an assumed CLint, while subject to numerous limitations, remain highly useful and necessary in both traditional pharmacokinetics (PK) and physiologically based pharmacokinetic (PBPK) modeling.


Assuntos
Fígado , Modelos Biológicos , Cinética , Fígado/metabolismo , Taxa de Depuração Metabólica
13.
Toxicology ; 463: 152954, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34543702

RESUMO

Predicting human hepatic clearance remains a fundamental challenge in both pharmaceutical drug development and toxicological assessments of environmental chemicals, with concerns about both accuracy and precision of in vitro-derived estimates. Suggested sources of these issues have included differences in experimental protocols, differences in cell sourcing, and use of a single cell type, liver parenchymal cells (hepatocytes). Here we investigate the ability of human microfluidic four-cell liver acinus microphysiology system (LAMPS) to make predictions as to hepatic clearance for seven representative compounds: Caffeine, Pioglitazone, Rosiglitazone, Terfenadine, Tolcapone, Troglitazone, and Trovafloxacin. The model, whose reproducibility was recently confirmed in an inter-lab comparison, was constructed using primary human hepatocytes or human induced pluripotent stem cell (iPSC)-derived hepatocytes and 3 human cell lines for the endothelial, Kupffer and stellate cells. We calculated hepatic clearance estimates derived from experiments using LAMPS or traditional 2D cultures and compared the outcomes with both in vivo human clinical study-derived and in vitro human hepatocyte suspension culture-derived values reported in the literature. We found that, compared to in vivo clinically-derived values, the LAMPS model with iPSC-derived hepatocytes had higher precision as compared to primary cells in suspension or 2D culture, but, consistent with previous studies in other microphysiological systems, tended to underestimate in vivo clearance. Overall, these results suggest that use of LAMPS and iPSC-derived hepatocytes together with an empirical scaling factor warrants additional study with a larger set of compounds, as it has the potential to provide more accurate and precise estimates of hepatic clearance.


Assuntos
Células Acinares/metabolismo , Fígado/metabolismo , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Hepatócitos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microfluídica/métodos , Reprodutibilidade dos Testes
14.
Biomed Pharmacother ; 141: 111855, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34229248

RESUMO

Hepatic clearance has been widely studied for over 50 yr. Many models have been developed using either theoretical or empirical tests to predict drug metabolism. The well-stirred, parallel-tube, and dispersion metabolic models have been extensively discussed. However, to our knowledge, these models cannot fully describe all relevant scenarios in hepatic clearance. We addressed this issue using the isolated perfused rat liver technique with minor modifications. Diazepam was selected to illustrate different levels of drug plasma-protein binding by changing the added concentration of human serum albumin. The free fractions of diazepam at different albumin concentrations were assayed by rapid equilibrium dialysis. The experimental data provide new insights concerning an accepted formula used to describe hepatic clearance. Regarding drug concentrations passing through the liver, the driving force concentration (CH,ss) in terms of Cin (influx in the liver) or Cout (efflux from the liver) needs to be carefully considered when determining drug hepatic and intrinsic clearances. The newly established model, termed the modified well-stirred model, which was derived from the original formula, successfully estimated hepatic drug metabolism. Using the modified well-stirred model, a theoretical driving force concentration of diazepam passing through the liver was evaluated. The model was further used to assess the predictability of in vitro to in vivo extrapolation. This study was not intended to refute the existing models, but rather to augment them using experimental data. The results stress the importance of proper calculation of dose when the drug clearance deviates from the prediction of the well-stirred model.


Assuntos
Fígado/metabolismo , Preparações Farmacêuticas/metabolismo , Albuminas/metabolismo , Algoritmos , Animais , Diálise , Diazepam/sangue , Diazepam/farmacocinética , Humanos , Masculino , Taxa de Depuração Metabólica , Modelos Teóricos , Perfusão , Ratos , Ratos Sprague-Dawley
15.
J Clin Med ; 10(12)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204545

RESUMO

Amyloid-beta (Aß) deposition in the brain is the main pathological hallmark of Alzheimer disease. Peripheral clearance of Aß may possibly also lower brain levels. Recent evidence suggested that hepatic clearance of Aß42 is impaired in liver cirrhosis. To further test this hypothesis, serum Aß42 was measured by ELISA in portal venous serum (PVS), systemic venous serum (SVS), and hepatic venous serum (HVS) of 20 patients with liver cirrhosis. Mean Aß42 level was 24.7 ± 20.4 pg/mL in PVS, 21.2 ± 16.7 pg/mL in HVS, and 19.2 ± 11.7 pg/mL in SVS. Similar levels in the three blood compartments suggested that the cirrhotic liver does not clear Aß42. Aß42 was neither associated with the model of end-stage liver disease score nor the Child-Pugh score. Patients with abnormal creatinine or bilirubin levels or prolonged prothrombin time did not display higher Aß42 levels. Patients with massive ascites and patients with large varices had serum Aß42 levels similar to patients without these complications. Serum Aß42 was negatively associated with connective tissue growth factor levels (r = -0.580, p = 0.007) and a protective role of Aß42 in fibrogenesis was already described. Diabetic patients with liver cirrhosis had higher Aß42 levels (p = 0.069 for PVS, p = 0.047 for HVS and p = 0.181 for SVS), which is in accordance with previous reports. Present analysis showed that the cirrhotic liver does not eliminate Aß42. Further studies are needed to explore the association of liver cirrhosis, Aß42 levels, and cognitive dysfunction.

16.
AAPS J ; 23(1): 24, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33438174

RESUMO

Oxycodone is an opioid analgesic that is commonly prescribed to pregnant women to treat moderate-to-severe pain. It has been shown to cross the placenta and distribute to the fetus. Oxycodone is mainly metabolized by CYP3A4 in the adult liver. Since CYP3A7 is abundantly expressed in the fetal liver and has overlapping substrate specificity with CYP3A4, we hypothesized that the fetal liver may significantly limit fetal exposure to oxycodone. This study showed that oxycodone is metabolized by CYP3A7 to noroxycodone in fetal liver microsomes (FLMs). The measured CYP3A7 expression was 191-409 pmol/mg protein in 14 FLMs, and an intersystem extrapolation factor (ISEF) for CYP3A7 was 0.016-0.066 in the panel of fetal livers using 6ß-OH-testosterone formation as the probe reaction. Noroxycodone formation in the fetal liver was predicted from formation rate by recombinant CYP3A7, CYP3A7 expression level and the established ISEF value with average fold error of 1.25. Based on the intrinsic clearance of oxycodone measured in FLM, the fetal hepatic clearance (CLh) at term was predicted to be 495 (range: 66.4-936) µL/min, a value that is > 99% lower than the predicted adult liver CLh. The predicted fetal hepatic extraction ratio was 0.0019 (range: 0.00003-0.0036). These results suggest that fetal liver metabolism does not quantitatively contribute to the total systemic clearance of oxycodone in pregnant women nor does it provide a barrier for limiting fetal exposure to oxycodone. Additionally, since CYP3A7 forms noroxycodone, an inactive metabolite, the metabolism in the fetal liver is unlikely to affect fetal opioid activity.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Feto/enzimologia , Fígado/enzimologia , Oxicodona/farmacocinética , Adulto , Ensaios Enzimáticos , Feminino , Feto/citologia , Eliminação Hepatobiliar , Humanos , Fígado/citologia , Masculino , Troca Materno-Fetal , Microssomos Hepáticos , Morfinanos/farmacocinética , Oxicodona/administração & dosagem , Placenta , Gravidez , Cultura Primária de Células , Proteínas Recombinantes/metabolismo
17.
Curr Drug Metab ; 22(7): 523-531, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33397250

RESUMO

Membrane transporters play an important role in intestinal absorption, distribution and clearance of drugs. Additionally transporters along with enzymes regulate tissue exposures (e.g. liver, kidney and brain), which are important for safety and efficacy considerations. Early identification of transporters involved guides generation of in vitro and in vivo data needed to gain mechanistic understanding on the role of transporters in organ clearance, tissue exposures and enables development of physiological-based pharmacokinetic (PBPK) models. A lot of progress has been made in developing several in vitro assay systems and mechanistic in silico models to determine kinetic parameters for transporters, which are incorporated into PBPK models. Although, intrinsic clearance and inhibition data from in vitro systems generally tend to underpredict in vivo clearance and magnitude of drug-drug interactions (DDIs), empirical scaling factors derived from a sizable dataset are often used to offset underpredictions. PBPK models are increasing used to predict the impact of transporters on intestinal absorption, clearance, victim and perpetrator DDIs prior to first in human clinical trials. The models are often refined when clinical data is available and are used to predict pharmacokinetics in untested scenarios such as the impact of polymorphisms, ontogeny, ethnicity, disease states and DDIs with other perpetrator drugs. The aim of this review is to provide an overview of (i) regulatory requirements around transporters, (ii) in vitro systems and their limitations in predicting transporter mediated drug disposition and DDIs, (iii) PBPK modelling tactics and case studies used for internal decision making and/or for regulatory submissions.


Assuntos
Vias de Eliminação de Fármacos , Interações Medicamentosas , Absorção Intestinal , Proteínas de Membrana Transportadoras/metabolismo , Farmacocinética , Animais , Humanos , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Modelos Biológicos
18.
J Pharm Sci ; 110(3): 1385-1391, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33217427

RESUMO

Bteich and coworkers recently demonstrated in a companion manuscript (J Pharm Sci 109: https://doi.org/10.1016/j.xphs.2020.07.003) that a protein-mediated hepatic uptake have occurred in an isolated perfused rat liver (IPRL) model for two drugs (Perampanel; PER and Fluoxetine; FLU) that bind extensively to the albumin (ALB) and alpha-1-acid glycoprotein (AGP). However, to our knowledge, there is no quantitative model available to predict the impact of a plasma protein-mediated hepatic uptake on the extent of hepatic clearance (CLh) for a drug binding extensively to these two proteins. Therefore, the main objective was to predict the corresponding CLh, which is an extension of the companion manuscript. The method consisted of extrapolating the intrinsic clearance from the unbound fraction measured in the perfusate or the unbound fraction extrapolated to the surface of the hepatocyte membrane by adapting an existing model of protein-mediated hepatic uptake (i.e., the fup-adjusted model) to include a binding ratio between the ALB and AGP. This new approach showed a relevant improvement compared to the free drug hypothesis particularly for FLU that showed the highest degree of ALB-mediated uptake. Overall, this study is a first step towards the development of predictive methods of CLh by considering the binding to ALB and AGP.


Assuntos
Orosomucoide , Preparações Farmacêuticas , Albuminas/metabolismo , Animais , Proteínas Sanguíneas/metabolismo , Fígado/metabolismo , Modelos Biológicos , Orosomucoide/metabolismo , Preparações Farmacêuticas/metabolismo , Ligação Proteica , Ratos
19.
J Pharm Sci ; 110(1): 517-528, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33058894

RESUMO

Pemafibrate (PMF) is highly albumin-bound (>99.8%) and a substrate for hepatic uptake transporters (OATP1B) and CYP enzymes. Here, we developed a PBPK model of PMF to capture drug-drug interactions (DDI) incurred by cyclosporine (CsA) and rifampicin (RIF), the two OATP1B inhibitors. Initial PBPK modeling of PMF utilized in vitro hepatic uptake clearance (PSinf) obtained in the absence of albumin, but failed in capturing the blood PMF pharmacokinetic (PK) profiles. Based on the results that in vitro PSinf of unbound PMF was enhanced in the presence of albumin, we applied the albumin-facilitated dissociation model and the resulting PSinf parameters improved the prediction of the blood PMF PK profiles. In refining our PBPK model toward improved prediction of the observed DDI data (PMF co-administered with single dosing of CsA or RIF; PMF following multiple RIF dosing), we adjusted the previously obtained in vivo OATP1B inhibition constants (Ki,OATP1B) of CsA or RIF for pitavastatin by correcting for substrate-dependency. We also incorporated the induction of OATP1B and CYP enzymes after multiple RIF dosing. Sensitivity analysis informed that the higher gastrointestinal absorption rate constant could further improve capturing the observed DDI data, suggesting the possible inhibition of intestinal ABC transporter(s) by CsA or RIF.


Assuntos
Preparações Farmacêuticas , Rifampina , Albuminas , Benzoxazóis , Butiratos , Ciclosporina , Interações Medicamentosas , Modelos Biológicos
20.
AAPS J ; 22(5): 120, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32914238

RESUMO

Extensive studies have been conducted to predict in vivo metabolic clearance from in vitro human liver metabolism parameters (i.e., in vitro-in vivo extrapolation (IVIVE)) with little success. Here, deriving IVIVE from first principles, we show that the product of fraction unbound in the blood and the predicted in vivo intrinsic clearance determined from hepatocyte or microsomal incubations is the lower boundary condition for in vivo hepatic clearance and the prerequisite for IVIVE predictions to be valid, regardless of extraction ratio. For 60-80% of drugs evaluated here, this product is markedly less than the in vivo measured clearance, a result that violates the lower boundary of the predictive relationship. This can only be explained by (a) suboptimal in vitro metabolic stability assay conditions, (b) significant error in the assumption that in vitro intrinsic clearance determinations will predict in vivo intrinsic clearance simply by scaling-up the amount of enzyme (in vitro incubation to in vivo liver), and/or (c) the methods of determining fraction unbound are incorrect. We further suggest that widely employed organ blood flow values underpredict the effective blood flow within the organ by approximately 2.5-fold, thus impacting IVIVE of high clearance compounds. We propose future pathways that should be investigated in terms of the relationship to experimentally measured clearance values, rather than model-dependent intrinsic clearance. IVIVE outcome can be improved by estimating the ratio of unbound drug concentration in the liver tissue to the liver plasma, examining the assumption of the free drug theory (i.e., there are no transporter effects at the blood cell membrane) and the finding that the upper limit of organ clearance may be greater than blood flow entering the organ.


Assuntos
Hepatócitos/metabolismo , Taxa de Depuração Metabólica , Modelos Teóricos , Animais , Humanos , Circulação Hepática , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA