Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Front Oncol ; 13: 1010563, 2023.
Article in English | MEDLINE | ID: mdl-36890818

ABSTRACT

Introduction: Alterations in expression and activity of human receptor tyrosine kinases (RTKs) are associated with cancer progression and in response to therapeutic intervention. Methods: Thus, protein abundance of 21 RTKs was assessed in 15 healthy and 18 cancerous liver samples [2 primary and 16 colorectal cancer liver metastasis (CRLM)] matched with non-tumorous (histologically normal) tissue, by a validated QconCAT-based targeted proteomic approach. Results: It was demonstrated, for the first time, that the abundance of EGFR, INSR, VGFR3 and AXL, is lower in tumours relative to livers from healthy individuals whilst the opposite is true for IGF1R. EPHA2 was upregulated in tumour compared with histologically normal tissue surrounding it. PGFRB levels were higher in tumours relative to both histologically normal tissue surrounding tumour and tissues taken from healthy individuals. The abundances of VGFR1/2, PGFRA, KIT, CSF1R, FLT3, FGFR1/3, ERBB2, NTRK2, TIE2, RET, and MET were, however, comparable in all samples. Statistically significant, but moderate correlations were observed (Rs > 0.50, p < 0.05) for EGFR with INSR and KIT. FGFR2 correlated with PGFRA and VGFR1 with NTRK2 in healthy livers. In non-tumorous (histologically normal) tissues from cancer patients, there were correlations between TIE2 and FGFR1, EPHA2 and VGFR3, FGFR3 and PGFRA (p < 0.05). EGFR correlated with INSR, ERBB2, KIT and EGFR, and KIT with AXL and FGFR2. In tumours, CSF1R correlated with AXL, EPHA2 with PGFRA, and NTRK2 with PGFRB and AXL. Sex, liver lobe and body mass index of donors had no impact on the abundance of RTKs, although donor age showed some correlations. RET was the most abundant of these kinases in non-tumorous tissues (~35%), while PGFRB was the most abundant RTK in tumours (~47%). Several correlations were also observed between the abundance of RTKs and proteins relevant to drug pharmacokinetics (enzymes and transporters). Discussion: DiscussionThis study quantified perturbation to the abundance of several RTKs in cancer and the value generated in this study can be used as input to systems biology models defining liver cancer metastases and biomarkers of its progression.

2.
Clin Pharmacokinet ; 62(3): 457-480, 2023 03.
Article in English | MEDLINE | ID: mdl-36752991

ABSTRACT

BACKGROUND AND OBJECTIVE: Mechanistic static and dynamic physiologically based pharmacokinetic models are used in clinical drug development to assess the risk of drug-drug interactions (DDIs). Currently, the use of mechanistic static models is restricted to screening DDI risk for an investigational drug, while dynamic physiologically based pharmacokinetic models are used for quantitative predictions of DDIs to support regulatory filing. As physiologically based pharmacokinetic model development by sponsors as well as a review of models by regulators require considerable resources, we explored the possibility of using mechanistic static models to support regulatory filing, using representative cases of successful physiologically based pharmacokinetic submissions to the US Food and Drug Administration under different classes of applications. METHODS: Drug-drug interaction predictions with mechanistic static models were done for representative cases in the different classes of applications using the same data and modelling workflow as described in the Food and Drug Administration clinical pharmacology reviews. We investigated the hypothesis that the use of unbound average steady-state concentrations of modulators as driver concentrations in the mechanistic static models should lead to the same conclusions as those from physiologically based pharmacokinetic modelling for non-dynamic measures of DDI risk assessment such as the area under the plasma concentration-time curve ratio, provided the same input data are employed for the interacting drugs. RESULTS: Drug-drug interaction predictions of area under the plasma concentration-time curve ratios using mechanistic static models were mostly comparable to those reported in the Food and Drug Administration reviews using physiologically based pharmacokinetic models for all representative cases in the different classes of applications. CONCLUSIONS: The results reported in this study should encourage the use of models that best fit an intended purpose, limiting the use of physiologically based pharmacokinetic models to those applications that leverage its unique strengths, such as what-if scenario testing to understand the effect of dose staggering, evaluating the role of uptake and efflux transporters, extrapolating DDI effects from studied to unstudied populations, or assessing the impact of DDIs on the exposure of a victim drug with concurrent mechanisms. With this first step, we hope to trigger a scientific discussion on the value of a routine comparison of the two methods for regulatory submissions to potentially create a best practice that could help identify examples where the use of dynamic changes in modulator concentrations could make a difference to DDI risk assessment.


Subject(s)
Filing , Models, Biological , Humans , Drug Interactions , Pharmaceutical Preparations
3.
Xenobiotica ; 52(7): 661-668, 2022 Jul.
Article in English | MEDLINE | ID: mdl-36190773

ABSTRACT

Prediction of rat, dog, monkey, and human volume of distribution (VDss) by Rodgers-Lukacova model was evaluated using a data set of more than 100 compounds.The prediction accuracy was best for humans followed by monkeys and dogs with 59, 52, and 41% of compounds within 2-fold, respectively.The accuracy of predictions in preclinical species was indicative of the human situation. This was particularly true for monkeys, where 87% of the compounds that were predicted within 2-fold in monkeys were also predicted within 2-fold in humans.The model's tendency to underestimate VDss was higher in rats and dogs compared to humans and monkeys for all ion classes but zwitterions. Hence, correction of human predictions using prediction errors in rats and dogs resulted in overestimation of VDss.The model had a similar degree of underestimation in humans and monkeys. Correction using monkeys improved the accuracy of the human estimate, especially for basic and zwitterion compounds.A strategy is proposed based on the accuracy of prediction in monkey and monkey scalars for prediction and prospective assessment of the accuracy of human VDss.


Subject(s)
Prospective Studies , Humans , Animals , Dogs , Rats , Haplorhini
4.
Br J Clin Pharmacol ; 88(4): 1811-1823, 2022 02.
Article in English | MEDLINE | ID: mdl-34599518

ABSTRACT

AIMS: This study aims to quantify drug-metabolising enzymes, transporters, receptor tyrosine kinases (RTKs) and protein markers (involved in pathways affected in cancer) in pooled healthy, histologically normal and matched cancerous liver microsomes from colorectal cancer liver metastasis (CRLM) patients. METHODS: Microsomal fractionation was performed and pooled microsomes were prepared. Global and accurate mass and retention time liquid chromatography-mass spectrometry proteomics were used to quantify proteins. A QconCAT (KinCAT) for the quantification of RTKs was designed and applied for the first time. Physiologically based pharmacokinetic (PBPK) simulations were performed to assess the contribution of altered abundance of drug-metabolising enzymes and transporters to changes in pharmacokinetics. RESULTS: Most CYPs and UGTs were downregulated in histologically normal relative to healthy samples, and were further reduced in cancer samples (up to 54-fold). The transporters, MRP2/3, OAT2/7 and OATP2B1/1B3/1B1 were downregulated in CRLM. Application of abundance data in PBPK models for substrates with different attributes indicated substantially lower (up to 13-fold) drug clearance when using cancer-specific instead of default parameters in cancer population. Liver function markers were downregulated, while inflammation proteins were upregulated (by up to 76-fold) in cancer samples. Various pharmacodynamics markers (e.g. RTKs) were altered in CRLM. Using global proteomics, we examined proteins in pathways relevant to cancer (such as metastasis and desmoplasia), including caveolins and collagen chains, and confirmed general over-expression of such pathways. CONCLUSION: This study highlights impaired drug metabolism, perturbed drug transport and altered abundance of cancer markers in CRLM, demonstrating the importance of population-specific abundance data in PBPK models for cancer.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Elimination Routes , Humans , Liver/metabolism , Liver Neoplasms/drug therapy , Membrane Transport Proteins/metabolism , Proteomics/methods
5.
CPT Pharmacometrics Syst Pharmacol ; 10(6): 622-632, 2021 06.
Article in English | MEDLINE | ID: mdl-34080804

ABSTRACT

Merck KGaA observed slight differences in the dissolution of Concor® (bisoprolol) batches over the years. The purpose of this work was to assess the impact of in vitro dissolution on the simulated pharmacokinetics of bisoprolol using in vitro-in vivo relationship established with available in vitro dissolution and corresponding plasma concentrations-time data for several bisoprolol batches. A mechanistic absorption model/physiologically based pharmacokinetics model linked with a biopharmaceutics tool such as dissolution testing, namely, physiologically based biopharmaceutics modeling (PBBM), can be valuable in determining a dissolution "safe space." A PBBM for bisoprolol was built using in vitro, in silico, and clinical data. We evaluated potential influences of variability in dissolution of bisoprolol batches on its clinical performance through PBBM and virtual bioequivalence (BE) trials. We demonstrated that in vitro dissolution was not critical for the clinical performance of bisoprolol over a wide range of tested values. Based on virtual BE trials, safe space expansion was explored using hypothetical dissolution data. A formulation with in vitro dissolution reaching 70% dissolved in 15 min and 79.5% in 30 min was shown to be BE to classical fast dissolution of bisoprolol (>85% within 15 min), as point estimates and 90% confidence intervals of the maximum plasma concentration and area under the concentration-time curve were within the BE limits (0.8-1.25).


Subject(s)
Antihypertensive Agents , Bisoprolol , Models, Biological , Administration, Intravenous , Administration, Oral , Adult , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/blood , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacokinetics , Biopharmaceutics , Bisoprolol/administration & dosage , Bisoprolol/blood , Bisoprolol/chemistry , Bisoprolol/pharmacokinetics , Clinical Trials as Topic , Drug Liberation , Fasting/metabolism , Healthy Volunteers , Humans , Male , Therapeutic Equivalency
6.
Drug Metab Dispos ; 49(7): 563-571, 2021 07.
Article in English | MEDLINE | ID: mdl-33980603

ABSTRACT

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.


Subject(s)
Antinematodal Agents/pharmacokinetics , Colorectal Neoplasms/pathology , Liver Neoplasms/physiopathology , Liver/metabolism , Models, Biological , Adult , Aged , Aged, 80 and over , Antinematodal Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Dose-Response Relationship, Drug , Female , Hepatectomy , Hepatobiliary Elimination , Humans , Liver/pathology , Liver/surgery , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Male , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Middle Aged
7.
Clin Pharmacol Ther ; 110(2): 297-310, 2021 08.
Article in English | MEDLINE | ID: mdl-33270249

ABSTRACT

The predictive performance of physiologically-based pharmacokinetics (PBPK) models for pharmacokinetics (PK) in renal impairment (RI) and hepatic impairment (HI) populations was evaluated using clinical data from 29 compounds with 106 organ impairment study arms were collected from 19 member companies of the International Consortium for Innovation and Quality in Pharmaceutical Development. Fifty RI and 56 HI study arms with varying degrees of organ insufficiency along with control populations were evaluated. For RI, the area under the curve (AUC) ratios of RI to healthy control were predicted within twofold of the observed ratios for > 90% (N = 47/50 arms). For HI, > 70% (N = 43/56 arms) of the hepatically impaired to healthy control AUC ratios were predicted within twofold. Inaccuracies, typically overestimation of AUC ratios, occurred more in moderate and severe HI. PBPK predictions can help determine the need and timing of organ impairment study. It may be suitable for predicting the impact of RI on PK of drugs predominantly cleared by metabolism with varying contribution of renal clearance. PBPK modeling may be used to support mild impairment study waivers or clinical study design.


Subject(s)
Drug Industry/organization & administration , Kidney Diseases/metabolism , Liver Diseases/metabolism , Models, Biological , Pharmacokinetics , Area Under Curve , Computer Simulation , Dose-Response Relationship, Drug , Drug Industry/standards , Humans , Severity of Illness Index
8.
Eur J Pharm Sci ; 152: 105431, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32562690

ABSTRACT

Solubility is one of the key parameters that is optimized during drug discovery to ensure sufficient drug concentration in systemic circulation and to achieve the desired pharmacological response. We recently reported the application of PBPK analysis of early clinical pharmacokinetic data to identify drugs whose absorption are truly limited by solubility. In this work, we selected ten anticancer drugs that exhibit poor in vitro solubility to explore the utility of this approach to identify solubility-limited absorption based on rat pharmacokinetic data and compare the findings to human data. Oral rat pharmacokinetic studies were performed at the body weight-scaled doses of the model drugs' human food effect studies, and analyzed using a top-down PBPK modeling approach. A good correlation of solubility-limited absorption in rat and human was observed. These results allow an early identification of drugs with truly solubility-limited absorption, with the potential to guide decisions and save valuable resources in drug development.


Subject(s)
Drug Development , Models, Biological , Administration, Oral , Animals , Humans , Rats , Solubility
9.
Drug Discov Today ; 25(5): 909-919, 2020 05.
Article in English | MEDLINE | ID: mdl-31981792

ABSTRACT

High-quality dose predictions based on a good understanding of target engagement is one of the main translational goals in drug development. Here, we systematically evaluate active human dose predictions for 15 Merck KGaA/EMD Serono assets spanning several modalities and therapeutic areas. Using case studies, we illustrate the value of adhering to the translational best practices of having an exposure-response relationship in an appropriate animal model; having validated, translatable pharmacodynamic (PD) biomarkers measurable in Phase I populations in the right tissue; having a deeper understanding of biology; and capturing uncertainties in predictions. Given the gap in publications on the subject, we believe that the learnings from this unique diverse data set, which are generic to the industry, will trigger actions to improve future predictions.


Subject(s)
Dose-Response Relationship, Drug , Animals , Biomarkers/metabolism , Drug Development/methods , Drug Industry/methods , Humans
10.
Clin Pharmacol Ther ; 107(3): 650-661, 2020 03.
Article in English | MEDLINE | ID: mdl-31608434

ABSTRACT

Poor aqueous solubility and dissolution of drug candidates drive key decisions on lead series optimization during drug discovery, on formulation optimization, and clinical studies planning during drug development. The interpretation of the in vivo relevance of early pharmaceutical profiling is often confounded by the multiple factors affecting oral systemic exposure. There is growing evidence that in vitro drug solubility may underestimate the true in vivo solubility and lead to drug misclassification. Based on 10 poorly water-soluble tyrosine kinase inhibitors, this paper demonstrates the use of physiologically-based pharmacokinetic (PK) analysis in combination with early clinical PK data to identify drugs whose absorption is truly limited by solubility in vivo and, therefore, expected to exhibit food effect. Our study supports a totality of evidence approach using early clinical data to guide decisions on conducting drug interaction studies with food and acid-reducing agents.


Subject(s)
Food-Drug Interactions , Models, Biological , Protein Kinase Inhibitors/administration & dosage , Administration, Oral , Chemistry, Pharmaceutical/methods , Drug Development/methods , Humans , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Solubility , Water/chemistry
11.
Clin Pharmacokinet ; 58(11): 1355-1371, 2019 11.
Article in English | MEDLINE | ID: mdl-31236775

ABSTRACT

When scientifically well-founded, the mechanistic basis of physiologically based pharmacokinetic (PBPK) models can help reduce the uncertainty and increase confidence in extrapolations outside the studied scenarios or studied populations. However, it is not always possible to establish mechanistically credible PBPK models. Requirements to establishing confidence in PBPK models, and challenges to meeting these requirements, are presented in this article. Parameter non-identifiability is the most challenging among the barriers to establishing confidence in PBPK models. Using case examples of small molecule drugs, this article examines the use of hypothesis testing to overcome parameter non-identifiability issues, with the objective of enhancing confidence in the mechanistic basis of PBPK models and thereby improving the quality of predictions that are meant for internal decisions and regulatory submissions. When the mechanistic basis of a PBPK model cannot be established, we propose the use of simpler models or evidence-based approaches.


Subject(s)
Models, Biological , Pharmacokinetics , Humans
12.
CPT Pharmacometrics Syst Pharmacol ; 8(9): 685-695, 2019 09.
Article in English | MEDLINE | ID: mdl-31215774

ABSTRACT

Regulatory agencies currently recommend itraconazole (ITZ) as a strong cytochrome P450 3A (CYP3A) inhibitor for clinical drug-drug interaction (DDI) studies. This work by an International Consortium for Innovation and Quality in Pharmaceutical Development working group (WG) is to develop and verify a mechanistic ITZ physiologically-based pharmacokinetic model and provide recommendations for optimal DDI study design based on model simulations. To support model development and verification, in vitro and clinical PK data for ITZ and its metabolites were collected from WG member companies. The model predictions of ITZ DDIs with seven different CYP3A substrates were within the guest criteria for 92% of area under the concentration-time curve ratios and 95% of maximum plasma concentration ratios, thus verifying the model for DDI predictions. The verified model was used to simulate various clinical DDI study scenarios considering formulation, duration of dosing, dose regimen, and food status to recommend the optimal design for maximal inhibitory effect by ITZ.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Itraconazole/pharmacokinetics , Area Under Curve , Drug Dosage Calculations , Drug Interactions , Food-Drug Interactions , Humans , Itraconazole/pharmacology , Models, Statistical
13.
CPT Pharmacometrics Syst Pharmacol ; 8(4): 220-229, 2019 04.
Article in English | MEDLINE | ID: mdl-30762304

ABSTRACT

Physiologically-based pharmacokinetic models are increasingly applied for pediatric dose selection along with traditional methods such as allometry and population pharmacokinetic models. We report a retrospective evaluation of the three methods. Pediatric population pharmacokinetic models sourced from literature for a subset of eight compounds were used to predict clearances for children < 2 years when they were within the modeled age range (interpolation, N = 11) or including those outside the modeled age range (interpolation and extrapolation, N = 18). Pediatric/adult clearance ratios were evaluated with a strict performance criterion of 0.8-1.25 and with twofold criteria. For children > 2 years, 58-75% of the clinical studies (N = 10) met the strict criteria, and > 80% of the clinical studies were predicted within twofold by all three methods. For children < 2 years, physiologically-based pharmacokinetic, allometry with age-dependent exponents, and pediatric population pharmacokinetic models predict 54%, 82%, and 64% within twofold of the observed, respectively.


Subject(s)
Models, Biological , Pharmaceutical Preparations/administration & dosage , Adult , Child Development , Clinical Decision-Making , Clinical Studies as Topic , Drug Dosage Calculations , Humans , Infant , Infant, Newborn , Metabolic Clearance Rate , Pharmacokinetics , Practice Guidelines as Topic , Retrospective Studies
14.
Clin Pharmacol Ther ; 104(1): 88-110, 2018 07.
Article in English | MEDLINE | ID: mdl-29315504

ABSTRACT

This work provides a perspective on the qualification and verification of physiologically based pharmacokinetic (PBPK) platforms/models intended for regulatory submission based on the collective experience of the Simcyp Consortium members. Examples of regulatory submission of PBPK analyses across various intended applications are presented and discussed. European Medicines Agency (EMA) and US Food and Drug Administration (FDA) recent draft guidelines regarding PBPK analyses and reporting are encouraging, and to advance the use and acceptability of PBPK analyses, more clarity and flexibility are warranted.


Subject(s)
Computer Simulation , Drug Approval , Models, Biological , Pharmacokinetics , Europe , Humans , United States , United States Food and Drug Administration
15.
Biopharm Drug Dispos ; 38(3): 163-186, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28152562

ABSTRACT

Extensive gut metabolism is often associated with the risk of low and variable bioavailability. The prediction of the fraction of drug escaping gut wall metabolism as well as transporter-mediated secretion (Fg ) has been challenged by the lack of appropriate preclinical models. The purpose of this study is to compare the performance of models that are widely employed in the pharmaceutical industry today to estimate Fg and, based on the outcome, to provide recommendations for the prediction of human Fg during drug discovery and early drug development. The use of in vitro intrinsic clearance from human liver microsomes (HLM) in three mechanistic models - the ADAM, Qgut and Competing Rates - was evaluated for drugs whose metabolism is dominated by CYP450s, assuming that the effect of transporters is negligible. The utility of rat as a model for human Fg was also explored. The ADAM, Qgut and Competing Rates models had comparable prediction success (70%, 74%, 69%, respectively) and bias (AFE = 1.26, 0.74 and 0.81, respectively). However, the ADAM model showed better accuracy compared with the Qgut and Competing Rates models (RMSE =0.20 vs 0.30 and 0.25, respectively). Rat is not a good model (prediction success =32%, RMSE =0.48 and AFE = 0.44) as it seems systematically to under-predict human Fg . Hence, we would recommend the use of rat to identify the need for Fg assessment, followed by the use of HLM in simple models to predict human Fg . © 2017 Merck KGaA. Biopharmaceutics & Drug Disposition Published by John Wiley & Sons, Ltd.


Subject(s)
Gastrointestinal Tract/metabolism , Models, Biological , Pharmaceutical Preparations/metabolism , Animals , Biological Availability , Biological Transport , Humans , Microsomes, Liver/metabolism
16.
AAPS J ; 18(3): 589-604, 2016 05.
Article in English | MEDLINE | ID: mdl-26964996

ABSTRACT

Quantifying the multiple processes which control and modulate the extent of oral bioavailability for drug candidates is critical to accurate projection of human pharmacokinetics (PK). Understanding how gut wall metabolism and hepatic elimination factor into first-pass clearance of drugs has improved enormously. Typically, the cytochrome P450s, uridine 5'-diphosphate-glucuronosyltransferases and sulfotransferases, are the main enzyme classes responsible for drug metabolism. Knowledge of the isoforms functionally expressed within organs of first-pass clearance, their anatomical topology (e.g. zonal distribution), protein homology and relative abundances and how these differ across species is important for building models of human metabolic extraction. The focus of this manuscript is to explore the parameters influencing bioavailability and to consider how well these are predicted in human from animal models or from in vitro to in vivo extrapolation. A unique retrospective analysis of three AstraZeneca molecules progressed to first in human PK studies is used to highlight the impact that species differences in gut wall metabolism can have on predicted human PK. Compared to the liver, pharmaceutical research has further to go in terms of adopting a common approach for characterisation and quantitative prediction of intestinal metabolism. A broad strategy is needed to integrate assessment of intestinal metabolism in the context of typical DMPK activities ongoing within drug discovery programmes up until candidate drug nomination.


Subject(s)
Gastrointestinal Tract/metabolism , Intestinal Absorption/physiology , Models, Animal , Models, Biological , Pharmaceutical Preparations/metabolism , Animals , Biological Availability , Cytochrome P-450 Enzyme System/metabolism , Drug Evaluation, Preclinical/methods , Forecasting , Gastrointestinal Tract/drug effects , Humans , Intestinal Absorption/drug effects , Pharmaceutical Preparations/administration & dosage
17.
Clin Pharmacokinet ; 55(6): 673-96, 2016 06.
Article in English | MEDLINE | ID: mdl-26895020

ABSTRACT

Intestinal metabolism can limit oral bioavailability of drugs and increase the risk of drug interactions. It is therefore important to be able to predict and quantify it in drug discovery and early development. In recent years, a plethora of models-in vivo, in situ and in vitro-have been discussed in the literature. The primary objective of this review is to summarize the current knowledge in the quantitative prediction of gut-wall metabolism. As well as discussing the successes of current models for intestinal metabolism, the challenges in the establishment of good preclinical models are highlighted, including species differences in the isoforms; regional abundances and activities of drug metabolizing enzymes; the interplay of enzyme-transporter proteins; and lack of knowledge on enzyme abundances and availability of empirical scaling factors. Due to its broad specificity and high abundance in the intestine, CYP3A is the enzyme that is frequently implicated in human gut metabolism and is therefore the major focus of this review. A strategy to assess the impact of gut wall metabolism on oral bioavailability during drug discovery and early development phases is presented. Current gaps in the mechanistic understanding and the prediction of gut metabolism are highlighted, with suggestions on how they can be overcome in the future.


Subject(s)
Intestinal Absorption/physiology , Models, Biological , ATP-Binding Cassette Transporters/metabolism , Animals , Animals, Genetically Modified , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Enterocytes/enzymology , Food-Drug Interactions , Glucuronosyltransferase/metabolism , Humans , Hydrogen-Ion Concentration , Intestinal Mucosa/metabolism , Metabolic Clearance Rate , Models, Animal , Pharmacokinetics , Sulfotransferases/metabolism
18.
Drug Metab Dispos ; 41(12): 2033-46, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23918667

ABSTRACT

A growing awareness of the risks associated with extensive intestinal metabolism has triggered an interest in developing robust methods for its quantitative assessment. This study explored the utility of intestinal S9 fractions, human liver microsomes, and recombinant cytochromes P450 to quantify CYP3A-mediated intestinal extraction in humans for a selection of marketed drugs that are predominantly metabolized by CYP3A4. A simple competing rates model is used to estimate the fraction of drug escaping gut wall metabolism (fg) from in vitro intrinsic clearance in humans. The fg values extrapolated from the three in vitro systems used in this study, together with literature-derived fg from human intestinal microsomes, were validated against fg extracted from human in vivo pharmacokinetic (PK) profiles using a generic whole-body physiologically-based pharmacokinetic (PBPK) model. The utility of the rat as a model for human CYP3A-mediated intestinal metabolism was also evaluated. Human fg from PBPK compares well with that from the grapefruit juice method, justifying its use for the evaluation of human in vitro systems. Predictive performance of all human in vitro systems was comparable [root mean square error (RMSE) = 0.22-0.27; n = 10]. Rat fg derived from in vivo PK profiles using PBPK has the lowest RMSE (0.19; n = 11) for the prediction of human fg for the selected compounds, most of which have a fraction absorbed close to 1. On the basis of these evaluations, the combined use of fg from human in vitro systems and rats is recommended for the estimation of CYP3A4-mediated intestinal metabolism in lead optimization and preclinical development phases.


Subject(s)
Intestinal Absorption/physiology , Intestinal Mucosa/metabolism , Pharmaceutical Preparations/metabolism , Animals , Cytochrome P-450 CYP3A/metabolism , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Female , Humans , Male , Microsomes/metabolism , Microsomes, Liver/metabolism , Middle Aged , Models, Biological , Rats , Rats, Sprague-Dawley
19.
Drug Metab Dispos ; 40(8): 1495-507, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22566536

ABSTRACT

Simcyp, a population-based simulator, is widely used for evaluating drug-drug interaction (DDI) risks in healthy and disease populations. We compare the prediction performance of Simcyp with that of mechanistic static models using different types of inhibitor concentrations, with the aim of understanding their strengths/weaknesses and recommending the optimal use of tools in drug discovery/early development. The inclusion of an additional term in static equations to consider the contribution of hepatic first pass to DDIs (AUCR(hfp)) has also been examined. A second objective was to assess Simcyp's estimation of variability associated with DDIs. The data set used for the analysis comprises 19 clinical interactions from 11 proprietary compounds. Except for gut interaction parameters, all other input data were identical for Simcyp and static models. Static equations using an unbound average steady-state systemic inhibitor concentration (I(sys)) and a fixed fraction of gut extraction and neglecting gut extraction in the case of induction interactions performed better than Simcyp (84% compared with 58% of the interactions predicted within 2-fold). Differences in the prediction outcomes between the static and dynamic models are attributable to differences in first-pass contribution to DDI. The inclusion of AUCR(hfp) in static equations leads to systematic overprediction of interaction, suggesting a limited role for hepatic first pass in determining inhibition-based DDIs for our data set. Our analysis supports the use of static models when elimination routes of the victim compound and the role of gut extraction for the victim and/or inhibitor in humans are not well defined. A fixed variability of 40% of predicted mean area under the concentration-time curve ratio is recommended.


Subject(s)
Drug Discovery , Drug Interactions , Models, Theoretical , Area Under Curve
20.
Curr Opin Drug Discov Devel ; 12(4): 509-18, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19562647

ABSTRACT

Physiologically based pharmacokinetics (PBPK) models are increasingly being used in the lead optimization (LO) process. Although there are currently few literature reports of the application of PBPK, the scope of PBPK modeling is expanding and there is a steady increase in the number of publications in this field. Recent publications covering four important areas of the application of PBPK modeling in LO have been reviewed.


Subject(s)
Computer Simulation , Drug Discovery/methods , Models, Biological , Pharmaceutical Preparations , Pharmacokinetics , Physiology , Animals , Drug Interactions , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...