ArticlePublisher preview available

Prediction of the Renal Organic Anion Transporter 1 (OAT1)- Mediated Drug Interactions for LY404039, the Active Metabolite of Pomaglumetad Methionil

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Purpose The objective of this work was to demonstrate that clinical OAT1-mediated DDIs can be predicted using physiologically based pharmacokinetic (PBPK) modeling. Methods LY404039 is a metabotropic glutamate receptor 2/3 agonist and the active moiety of the prodrug pomaglumetad methionil (LY2140023). After oral administration, pomaglumetad methionil is rapidly taken up by enterocytes via PEPT1 and once absorbed, converted to LY404039 via membrane dehydropeptidase 1 (DPEP1). LY404039 is renally excreted by both glomerular filtration and active secretion and in vitro studies showed that the active secretion of LY404039 was mediated by the organic anion transporter 1 (OAT1). Both clinical and in vitro data were used to build a PBPK model to predict OAT1-mediated DDIs. Results In vitro inhibitory potencies (IC50) of the known OAT inhibitors, probenecid and ibuprofen, were determined to be 4.00 and 2.63 µM, respectively. Subsequently, clinical drug-drug interaction (DDI) study showed probenecid reduced the renal clearance of LY404039 by 30 to 40%. The PBPK bottom-up model, predicted a renal clearance that was approximately 20% lower than the observed one. The middle-out model, using an OAT1 relative activity factor (RAF) of 3, accurately reproduced the renal clearance of LY404039 and pharmacokinetic (PK) changes of LY404039 in the presence of probenecid. Conclusions OAT1- mediated DDIs can be predicted using in vitro measured IC50 and PBPK modeling. The effect of ibuprofen was predicted to be minimal (AUC ratio of 1.15) and not clinically relevant.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Pharmaceutical Research (2023) 40:2499–2511
https://doi.org/10.1007/s11095-022-03464-y
ORIGINAL RESEARCH ARTICLE
Prediction oftheRenal Organic Anion Transporter
1 (OAT1)‑ Mediated Drug Interactions forLY404039, theActive
Metabolite ofPomaglumetad Methionil
Y.AnnePak1· MariaM.Posada1· JamesBacon1· AmandaLong· WilliamAnnes1· JenniferWitcher1·
MalcolmMitchell1· RommelG.Tirona2· StephenD.Hall1· KathleenM.Hillgren1
Received: 24 May 2022 / Accepted: 16 December 2022 / Published online: 12 January 2023
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2023
Abstract
Purpose The objective of this work was to demonstrate that clinical OAT1-mediated DDIs can be predicted using physi-
ologically based pharmacokinetic (PBPK) modeling.
Methods LY404039 is a metabotropic glutamate receptor 2/3 agonist and the active moiety of the prodrug pomaglumetad methionil
(LY2140023). After oral administration, pomaglumetad methionil is rapidly taken up by enterocytes via PEPT1 and once absorbed,
converted to LY404039 via membrane dehydropeptidase 1 (DPEP1). LY404039 is renally excreted by both glomerular filtration and
active secretion and in vitro studies showed that the active secretion of LY404039 was mediated by the organic anion transporter
1 (OAT1). Both clinical and in vitro data were used to build a PBPK model to predict OAT1-mediated DDIs.
Results In vitro inhibitory potencies (IC50) of the known OAT inhibitors, probenecid and ibuprofen, were determined to be
4.00 and 2.63µM, respectively. Subsequently, clinical drug-drug interaction (DDI) study showed probenecid reduced the
renal clearance of LY404039 by 30 to 40%. The PBPK bottom-up model, predicted a renal clearance that was approximately
20% lower than the observed one. The middle-out model, using an OAT1 relative activity factor (RAF) of 3, accurately
reproduced the renal clearance of LY404039 and pharmacokinetic (PK) changes of LY404039 in the presence of probenecid.
Conclusions OAT1- mediated DDIs can be predicted using in vitro measured IC50 and PBPK modeling. The effect of ibu-
profen was predicted to be minimal (AUC ratio of 1.15) and not clinically relevant.
Keywords active transport· drug interaction· organic anion transporters· PBPK model· renal transporters
Abbreviations
AE Adverse event
AUC Area under the concentration time curve
AUC
0-∞ AUC from time 0 extrapolated to infinity
CLr Renal clearance
Cmax Maximum observed plasma concentration
DDI Drug-drug interaction
fmet Fraction of the metabolite formed
GFR Glomerular filtration rate
HEK Human embryonic kidney
HEK-VC HEK vector control
ITC International Transporter Consortium
kfmet The first order metabolite formation rate
constant
KHB Krebs Henseleit buffer
OAT Organic anion transporter
PEPT1 Peptide transporter 1
PKPD Pharmacokinetics and pharmacodynamics
PBPK Physiologically based pharmacokinetic
RAF Relative activity factor
t1/2 Elimination half-life
tlag The lag time
tmax, Time of maximum observed plasma
concentration
Introduction
Pomaglumetad methionil (LY2140023 monohydrate) is a
potent and selective prodrug of the metabotropic glutamate
receptor 2/3 agonist, LY404039, and was developed for
* Maria M. Posada
mmposada@lilly.com
1 Eli Lilly andCompany, Lilly Corporate Center, Indianapolis,
IN46285, USA
2 Division ofClinical Pharmacology, Department ofMedicine,
The University ofWestern Ontario, London, ON, Canada
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Article
Modulation of the transport-mediated active uptake by human serum albumin (HSA) for highly protein-bound substrates has been reported and improved the in vitro-to-in vivo extrapolation (IVIVE) of hepatic clearance. However, evidence for the relevance of such a phenomenon in the case of renal transporters is sparse. In this study, transport of renal organic anion transporter 1 or 3 (OAT1/3) substrates into conditionally immortalized proximal tubular epithelial cells transduced with OAT1/3 was measured in the presence and absence of 1 and 4% HSA while keeping the unbound substrate concentration constant (based on measured fraction unbound, fu,inc). In the presence of 4% HSA, the unbound intrinsic active uptake clearance (CLint,u,active) of six highly protein-bound substrates increased substantially relative to the HSA-free control (3.5- to 122-fold for the OAT1 CLint,u,active, and up to 28-fold for the OAT3 CLint,u,active). The albumin-mediated uptake effect (fold increase in CLint,u,active) was more pronounced with highly bound substrates compared to no effect seen for weakly protein-bound substrates adefovir (OAT1-specific) and oseltamivir carboxylate (OAT3-specific). The relationship between OAT1/3 CLint,u,active and fu,inc agreed with the facilitated-dissociation model; a relationship was established between the albumin-mediated fold change in CLint,u,active and fu,inc for both the OAT1 and OAT3, with implications for IVIVE modeling. The relative activity factor and the relative expression factor based on global proteomic quantification of in vitro OAT1/3 expression were applied for IVIVE of renal clearance. The inclusion of HSA improved the bottom-up prediction of the level of OAT1/3-mediated secretion and renal clearance (CLsec and CLr), in contrast to the underprediction observed with the control (HSA-free) scenario. For the first time, this study confirmed the presence of the albumin-mediated uptake effect with renal OAT1/3 transporters; the extent of the effect was more pronounced for highly protein-bound substrates. We recommend the inclusion of HSA in routine in vitro OAT1/3 assays due to considerable improvements in the IVIVE of CLsec and CLr.
Article
Physiologically-based pharmacokinetic (PBPK) modeling is a mechanistic dynamic modeling approach that can be used to predict or retrospectively describe changes in drug exposure due to drug-drug interactions. With advancements in commercially available PBPK software, PBPK DDI modeling has become a mainstream approach from early drug discovery through to late stage drug development and is often utilized to support regulatory packages for new drug applications. This minireview will briefly describe the approaches to predicting DDI utilizing PBPK and static modeling approaches, the basic model structures and features inherent to PBPK DDI models and key examples where PBPK DDI models have been used to describe complex DDI mechanisms. Future directions aimed at using PBPK models to characterize transporter-mediated DDI, predict DDI in special populations and assess the DDI potential of protein therapeutics will be discussed. A summary of the 209 PBPK DDI examples published to date in 2023 will be provided. Overall, current data and trends suggest a continued role for PBPK models in the characterization and prediction of DDI for therapeutic molecules. Significance Statement PBPK models have been a key tool in the characterization of various pharmacokinetic phenomenon, including drug-drug interactions. This minireview will highlight recent advancements and publications around PBPK DDI modeling, an important area of drug discovery and development research in light of the increasing prevalence of polypharmacology in clinical settings.
Article
Full-text available
Purpose To provide whole-body physiologically based pharmacokinetic (PBPK) models of the potent clinical organic anion transporter (OAT) inhibitor probenecid and the clinical OAT victim drug furosemide for their application in transporter-based drug-drug interaction (DDI) modeling. Methods PBPK models of probenecid and furosemide were developed in PK-Sim®. Drug-dependent parameters and plasma concentration-time profiles following intravenous and oral probenecid and furosemide administration were gathered from literature and used for model development. For model evaluation, plasma concentration-time profiles, areas under the plasma concentration–time curve (AUC) and peak plasma concentrations (Cmax) were predicted and compared to observed data. In addition, the models were applied to predict the outcome of clinical DDI studies. Results The developed models accurately describe the reported plasma concentrations of 27 clinical probenecid studies and of 42 studies using furosemide. Furthermore, application of these models to predict the probenecid-furosemide and probenecid-rifampicin DDIs demonstrates their good performance, with 6/7 of the predicted DDI AUC ratios and 4/5 of the predicted DDI Cmax ratios within 1.25-fold of the observed values, and all predicted DDI AUC and Cmax ratios within 2.0-fold. Conclusions Whole-body PBPK models of probenecid and furosemide were built and evaluated, providing useful tools to support the investigation of transporter mediated DDIs.
Article
Full-text available
Multiple doses of tenofovir disoproxil fumarate (TDF) together with emtricitabine is effective for HIV Pre‐Exposure Prophylaxis (PrEP). TDF is converted to tenofovir (TFV) in circulation, which is subsequently cleared via tubular secretion by OAT1 and OAT3. Using in vitro kinetic parameters for TFV and the OAT1 and OAT3 inhibitor probenecid, a bottom‐up PBPK model was successfully developed for the first time that accurately describes the probenecid‐TFV interaction. This model predicted an increase in TFV plasma exposure by 60%, which was within 15% of the observed clinical PK data, and a 3‐fold decrease in renal cells exposure following coadministration of a 600mg TDF dose with 2g probenecid. Compared to multiple‐dose regimens, a single‐dose PRO‐boosted TDF regimen may be effective for HIV PrEP and improve adherence and safety by minimizing TFV‐induced nephrotoxicity by reducing TFV accumulation in renal cells.
Article
Full-text available
Baricitinib, an oral selective Janus kinase 1 and 2 inhibitor, undergoes active renal tubular secretion. Baricitinib was not predicted to inhibit hepatic and renal uptake and efflux drug transporters, based on the ratio of the unbound maximum eliminating-organ inlet concentration and the in vitro half-maximal inhibitory concentrations (IC50). In vitro, baricitinib was a substrate for organic anion transporter (OAT)3, multidrug and toxin extrusion protein (MATE)2-K, P-glycoprotein (P-gp), and breast cancer resistance protein (BCRP). Probenecid, a strong OAT3 inhibitor, increased the area under the concentration-time curve from time zero to infinity (AUC[0–∞]) of baricitinib by twofold and decreased renal clearance to 69% of control in healthy subjects. Physiologically based pharmacokinetic (PBPK) modeling reproduced the renal clearance of baricitinib and the inhibitory effect of probenecid using the in vitro IC50 value of 4.4 μM. Using ibuprofen and diclofenac in vitro IC50 values of 4.4 and 3.8 μM toward OAT3, 1.2 and 1.0 AUC(0–∞) ratios of baricitinib were predicted. These predictions suggest clinically relevant drug-drug interactions (DDIs) with ibuprofen and diclofenac are unlikely.
Article
Full-text available
Despite PEPT1 being responsible for the bioavailability for a variety of drugs, there has been little study of its potential involvement in drug-drug interactions. Pomaglumetad methionil, a metabotropic glutamate 2/3 receptor agonist prodrug, utilizes PEPT1 to enhance absorption and bioavailability. In vitro studies were conducted to guide the decision to conduct a clinical drug interaction study and to inform the clinical study design. In vitro investigations determined the prodrug (LY2140023 monohydrate) is a substrate of PEPT1 with Km value of approximately 30 μM, while the active moiety (LY404039) is not a PEPT1 substrate. In addition, among the eight known PEPT1 substrates evaluated in vitro, valacyclovir was the most potent inhibitor (IC50 = 0.46 mM) of PEPT1-mediated uptake of the prodrug. Therefore, a clinical drug interaction study was conducted to evaluate the potential interaction between the prodrug and valacyclovir in healthy subjects. No effect of coadministration was observed on the pharmacokinetics of the prodrug, valacyclovir, or either of their active moieties. Although in vitro studies showed potential for the prodrug and valacyclovir interaction via PEPT1, an in vivo study showed no interaction between these two drugs. PEPT1 does not appear to easily saturate due to its high capacity and expression in the intestine. Thus, a clinical interaction at PEPT1 is unlikely even with a compound with high affinity for the transporter.
Article
Since 2016, results from physiologically based pharmacokinetic (PBPK) analyses have been routinely found in the clinical pharmacology section of regulatory applications submitted to the US Food and Drug Administration (FDA). In 2018, the Food and Drug Administration's Office of Clinical Pharmacology published a commentary summarizing the application of PBPK modeling in the submissions it received between 2008 and 2017 and its impact on prescribing information. In this commentary, we provide an update on the application of PBPK modeling in submissions received between 2018 and 2019 and highlight a few notable examples.
Article
A PBPK modelling approach was used to predict organic anion transporter (OAT) mediated drug-drug interactions involving S 44121, a substrate and an inhibitor of OAT1 and OAT3. Model predictions were then compared to the results of a clinical DDI study which was carried out to investigate the interaction of S 44121 with probenecid, tenofovir and ciprofloxacin. PBPK models were developed and qualified using existing clinical data, and inhibition constants were determined in vitro. The model predictions for S 44121 as an OAT inhibitor were similar to the results obtained from the clinical DDI study, with no interaction observed for tenofovir or ciprofloxacin in the presence of S 44121. An observed AUC ratio of 2.2 was obtained for S 44121 in the presence of probenecid, which was slightly higher than the model predicted AUC ratio of 1.6. A DDI study in the monkey was also carried out for the interaction between S 44121 and probenecid, since the monkey has previously been reported to be a good preclinical model for OAT-mediated DDI. However, this study highlighted a species difference in the major route of S 44121 elimination between monkey (mainly hepatic metabolism) and human (mainly renal excretion of unchanged drug), rendering a comparison between the two DDI studies difficult. Overall, for S 44121 the PBPK modelling approach gave a better prediction of the extent of DDI than the static predictions based on inhibitor Cmax and IC50, therefore this can be considered a potentially valuable tool within drug development.
Article
The pharmacokinetics of cetirizine, a non-sedating antihistamine, is profoundly affected by transporter-mediated membrane transport in the kidney. In this study, we aimed to investigate the transport mechanism of levocetirizine, the pharmacologically active enantiomer of cetirizine, via human organic anion transporter 4 (OAT4) expressed in the apical membrane of renal proximal tubules and the basal plasma membrane of placental syncytiotrophoblasts. In cells expressing human OAT4 under the control of tetracycline, levocetirizine uptake was increased by tetracycline treatment. On the other hand, OAT4 expression did not facilitate efflux of preloaded levocetirizine from the cells, either in the presence or absence of extracellular Cl(-). The OAT4-mediated levocetirizine uptake was concentration-dependent with a Km of 38 μM. The uptake rate of levocetirizine via OAT4 was approximately twice that of racemic cetirizine, indicating stereoselective uptake of levocetirizine. On the other hand, OAT4-mediated [(3)H]dehydroepiandrosterone sulfate uptake was inhibited by dextrocetirizine as well as levocetirizine. Overall, our findings indicate that OAT4 mediates levocetirizine uptake, but is unlikely to mediate the efflux.
Article
The impact of transporters in modulating the disposition of drugs in the liver and their passage across the gut wall has received much more attention than their role in renal excretion, despite the fact that 25–30 % of drugs are cleared predominantly by renal clearance and renal transporters contribute significantly to this process. Thus there is a need to improve the ability to predict changes in renal clearance arising from genetic variability, the impact of disease and interactions related to renal transporters. Such changes may also influence the accumulation of xenobiotics within the kidney cell leading to nephrotoxicity. Attempts to develop mechanistic, physiologically based models of renal drug elimination have been limited. This chapter outlines the features and application of a new model (Mech KiM) that links drug characteristics to knowledge of renal physiology in predicting the contributions of glomerular filtration, active and passive secretion, active and passive reabsorption and metabolism to renal elimination.
Article
To characterize the hydrolysis of the peptide prodrug, pomaglumetad methionil (LY2140023), to the active drug, LY404039, a series of in vitro studies were performed in various matrices including human intestinal, liver, kidney homogenate and human plasma. The studies were performed to determine the tissue(s) and enzyme(s) responsible for the conversion of prodrug to the active molecule, this could enable an assessment of the risk for drug interactions, an evaluation of pharmacogenomic implications, as well as the development of a PBPK model for formation of the active drug. Of the matrices examined, hydrolysis of pomaglumetad methionil was observed in intestinal and kidney homogenate preparations and in plasma, but not in liver homogenate. Clearance values calculated after applying standard scaling factors suggest the intestine and kidney as primary sites of hydrolysis. Studies with peptidase inhibitors were performed in an attempt to identify the enzyme(s) catalyzing the conversion. Near complete inhibition of LY404039 formation was observed in intestinal and kidney homogenate and human plasma with the selective dehydropeptidase1 (DPEP1) inhibitor cilastatin. Human recombinant DPEP1 was expressed and shown to catalyze the hydrolysis, which was completely inhibited by cilastatin. These studies demonstrate pomaglumetad methionil can be converted to LY404039 via one or multiple enzymes completely inhibited by cilastatin, likely DPEP1, in plasma, intestine and kidney, with the plasma and kidney involved in the clearance of circulating prodrug. These experiments define a strategy for the characterization of enzymes responsible for the metabolism of other peptide like compounds. The American Society for Pharmacology and Experimental Therapeutics.
Article
The organic anion transporter (OAT) subfamily, which constitutes roughly half of the SLC22 (solute carrier 22) transporter family, has received a great deal of attention because of its role in handling of common drugs (antibiotics, antivirals, diuretics, nonsteroidal anti-inflammatory drugs), toxins (mercury, aristolochic acid), and nutrients (vitamins, flavonoids). Oats are expressed in many tissues, including kidney, liver, choroid plexus, olfactory mucosa, brain, retina, and placenta. Recent metabolomics and microarray data from Oat1 [Slc22a6, originally identified as NKT (novel kidney transporter)] and Oat3 (Slc22a8) knockouts, as well as systems biology studies, indicate that this pathway plays a central role in the metabolism and handling of gut microbiome metabolites as well as putative uremic toxins of kidney disease. Nuclear receptors and other transcription factors, such as Hnf4α and Hnf1α, appear to regulate the expression of certain Oats in conjunction with phase I and phase II drug metabolizing enzymes. Some Oats have a strong selectivity for particular signaling molecules, including cyclic nucleotides, conjugated sex steroids, odorants, uric acid, and prostaglandins and/or their metabolites. According to the "Remote Sensing and Signaling Hypothesis," which is elaborated in detail here, Oats may function in remote interorgan communication by regulating levels of signaling molecules and key metabolites in tissues and body fluids. Oats may also play a major role in interorganismal communication (via movement of small molecules across the intestine, placental barrier, into breast milk, and volatile odorants into the urine). The role of various Oat isoforms in systems physiology appears quite complex, and their ramifications are discussed in the context of remote sensing and signaling. Copyright © 2015 the American Physiological Society.