1
|
Lane TR, Urbina F, Zhang X, Fye M, Gerlach J, Wright SH, Ekins S. Machine Learning Models Identify New Inhibitors for Human OATP1B1. Mol Pharm 2022; 19:4320-4332. [PMID: 36269563 PMCID: PMC9873312 DOI: 10.1021/acs.molpharmaceut.2c00662] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The uptake transporter OATP1B1 (SLC01B1) is largely localized to the sinusoidal membrane of hepatocytes and is a known victim of unwanted drug-drug interactions. Computational models are useful for identifying potential substrates and/or inhibitors of clinically relevant transporters. Our goal was to generate OATP1B1 in vitro inhibition data for [3H] estrone-3-sulfate (E3S) transport in CHO cells and use it to build machine learning models to facilitate a comparison of seven different classification models (Deep learning, Adaboosted decision trees, Bernoulli naïve bayes, k-nearest neighbors (knn), random forest, support vector classifier (SVC), logistic regression (lreg), and XGBoost (xgb)] using ECFP6 fingerprints to perform 5-fold, nested cross validation. In addition, we compared models using 3D pharmacophores, simple chemical descriptors alone or plus ECFP6, as well as ECFP4 and ECFP8 fingerprints. Several machine learning algorithms (SVC, lreg, xgb, and knn) had excellent nested cross validation statistics, particularly for accuracy, AUC, and specificity. An external test set containing 207 unique compounds not in the training set demonstrated that at every threshold SVC outperformed the other algorithms based on a rank normalized score. A prospective validation test set was chosen using prediction scores from the SVC models with ECFP fingerprints and were tested in vitro with 15 of 19 compounds (84% accuracy) predicted as active (≥20% inhibition) showed inhibition. Of these compounds, six (abamectin, asiaticoside, berbamine, doramectin, mobocertinib, and umbralisib) appear to be novel inhibitors of OATP1B1 not previously reported. These validated machine learning models can now be used to make predictions for drug-drug interactions for human OATP1B1 alongside other machine learning models for important drug transporters in our MegaTrans software.
Collapse
Affiliation(s)
- Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Xiaohong Zhang
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Margret Fye
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Jacob Gerlach
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| | - Stephen H. Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510 Raleigh, NC 27606, USA
| |
Collapse
|
2
|
Wright SH, Secomb TW. Novel method for kinetic analysis applied to transport by the uniporter OCT2. Am J Physiol Renal Physiol 2022; 323:F370-F387. [PMID: 35862650 PMCID: PMC9423780 DOI: 10.1152/ajprenal.00106.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/22/2022] Open
Abstract
The kinetics of solute transport shed light on the roles these processes play in cellular physiology, and the absolute values of the kinetic parameters that quantitatively describe transport are increasingly used to model its impact on drug clearance. However, accurate assessment of transport kinetics is challenging. Although most carrier-mediated transport is adequately described by the Michaelis-Menten equation, its use presupposes that the rates of uptake used in the analysis of maximal rates of transport (Jmax) and half-saturation constants (Kt) reflect true unidirectional rates of influx from known concentrations of substrate. Most experimental protocols estimate the initial rate of transport from net substrate accumulation determined at a single time point (typically between 0.5 and 5 min) and assume it reflects unidirectional influx. However, this approach generally results in systematic underestimates of Jmax and overestimates of Kt; the former primarily due to the unaccounted impact of efflux of accumulated substrate, and the latter due to the influence of unstirred water layers. Here, we describe the bases of these time-dependent effects and introduce a computational model that analyzes the time course of net substrate uptake at several concentrations to calculate Jmax and Kt for unidirectional influx, taking into account the influence of unstirred water layers and mediated efflux. This method was then applied to calculate the kinetics of transport of 1-methyl-4-phenylpryridinium and metformin by renal organic cation transporter 2 as expressed in cultured Chinese hamster ovary cells.NEW & NOTEWORTHY Here, we describe a mathematical model that uses the time course of net substrate uptake into cells from several increasing concentrations to calculate unique kinetic parameters [maximal rates of transport (Jmax) and half-saturation constants (Kt)] of the process. The method is the first to take into consideration the common complicating factors of unstirred layers and carrier-mediated efflux in the experimental determination of transport kinetics.
Collapse
Affiliation(s)
- Stephen H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Timothy W Secomb
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|
3
|
Hau RK, Tash JS, Georg GI, Wright SH, Cherrington NJ. Physiological Characterization of the Transporter-Mediated Uptake of the Reversible Male Contraceptive H2-Gamendazole Across the Blood-Testis Barrier. J Pharmacol Exp Ther 2022; 382:299-312. [PMID: 35779861 PMCID: PMC9426764 DOI: 10.1124/jpet.122.001195] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/14/2022] [Indexed: 11/22/2022] Open
Abstract
The blood-testis barrier (BTB) is formed by a tight network of Sertoli cells (SCs) to limit the movement of reproductive toxicants from the blood into the male genital tract. Transporters expressed at the basal membranes of SCs also influence the disposition of drugs across the BTB. The reversible, nonhormonal contraceptive, H2-gamendazole (H2-GMZ), is an indazole carboxylic acid analog that accumulates over 10 times more in the testes compared with other organs. However, the mechanism(s) by which H2-GMZ circumvents the BTB are unknown. This study describes the physiologic characteristics of the carrier-mediated process(es) that permit H2-GMZ and other analogs to penetrate SCs. Uptake studies were performed using an immortalized human SC line (hT-SerC) and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Uptake of H2-GMZ and four analogs followed Michaelis-Menten transport kinetics (one analog exhibited poor penetration). H2-GMZ uptake was strongly inhibited by indomethacin, diclofenac, MK-571, and several analogs. Moreover, H2-GMZ uptake was stimulated by an acidic extracellular pH, reduced at basic pHs, and independent of extracellular Na+, K+, or Cl- levels, which are intrinsic characteristics of OATP-mediated transport. Therefore, the characteristics of H2-GMZ transport suggest that one or more OATPs may be involved. However, endogenous transporter expression in wild-type Chinese hamster ovary (CHO), Madin-Darby canine kidney (MDCK), and human embryonic kidney-293 (HEK-293) cells limited the utility of heterologous transporter expression to identify a specific OATP transporter. Altogether, characterization of the transporters involved in the flux of H2-GMZ provides insight into the selectivity of drug disposition across the human BTB to understand and overcome the pharmacokinetic and pharmacodynamic difficulties presented by this barrier. SIGNIFICANCE STATEMENT: Despite major advancements in female contraceptives, male alternatives, including vasectomy, condom usage, and physical withdrawal, are antiquated and the widespread availability of nonhormonal, reversible chemical contraceptives is nonexistent. Indazole carboxylic acid analogs such as H2-GMZ are promising new reversible, antispermatogenic drugs that are highly effective in rodents. This study characterizes the carrier-mediated processes that permit H2-GMZ and other drugs to enter Sertoli cells and the observations made here will guide the development of drugs that effectively circumvent the BTB.
Collapse
Affiliation(s)
- Raymond K Hau
- Department of Pharmacology and Toxicology, College of Pharmacy (R.K.H., N.J.C.), and Department of Physiology, College of Medicine (S.H.W.), The University of Arizona, Tucson, Arizona; Department of Molecular and Integrative Physiology, KU School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas (J.S.T.); Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, The University of Minnesota, Minneapolis, Minnesota (G.I.G.)
| | - Joseph S Tash
- Department of Pharmacology and Toxicology, College of Pharmacy (R.K.H., N.J.C.), and Department of Physiology, College of Medicine (S.H.W.), The University of Arizona, Tucson, Arizona; Department of Molecular and Integrative Physiology, KU School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas (J.S.T.); Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, The University of Minnesota, Minneapolis, Minnesota (G.I.G.)
| | - Gunda I Georg
- Department of Pharmacology and Toxicology, College of Pharmacy (R.K.H., N.J.C.), and Department of Physiology, College of Medicine (S.H.W.), The University of Arizona, Tucson, Arizona; Department of Molecular and Integrative Physiology, KU School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas (J.S.T.); Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, The University of Minnesota, Minneapolis, Minnesota (G.I.G.)
| | - Stephen H Wright
- Department of Pharmacology and Toxicology, College of Pharmacy (R.K.H., N.J.C.), and Department of Physiology, College of Medicine (S.H.W.), The University of Arizona, Tucson, Arizona; Department of Molecular and Integrative Physiology, KU School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas (J.S.T.); Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, The University of Minnesota, Minneapolis, Minnesota (G.I.G.)
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, College of Pharmacy (R.K.H., N.J.C.), and Department of Physiology, College of Medicine (S.H.W.), The University of Arizona, Tucson, Arizona; Department of Molecular and Integrative Physiology, KU School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas (J.S.T.); Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, The University of Minnesota, Minneapolis, Minnesota (G.I.G.)
| |
Collapse
|
4
|
Zhang X, Wright SH. Transport Turnover Rates for Human OCT2 and MATE1 Expressed in Chinese Hamster Ovary Cells. Int J Mol Sci 2022; 23:ijms23031472. [PMID: 35163393 PMCID: PMC8836179 DOI: 10.3390/ijms23031472] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
MATE1 (multidrug and toxin extruder 1) and OCT2 (organic cation transporter 2) play critical roles in organic cation excretion by the human kidney. The transporter turnover rate (TOR) is relevant to understanding both their transport mechanisms and interpreting the in vitro-in vivo extrapolation (IVIVE) required for physiologically-based pharmacokinetic (PBPK) modeling. Here, we use a quantitative western blot method to determine TORs for MATE1 and OCT2 proteins expressed in CHO cells. MATE1 and OCT2, each with a C-terminal V-5 epitope tag, were cell surface biotinylated and the amount of cell surface MATE1 and OCT2 protein was quantified by western analysis, using standard curves for the V5 epitope. Cell surface MATE1 and OCT2 protein represented 25% and 24%, respectively, of the total expression of these proteins in CHO cells. The number of cell surface transporters was ~55 fmol cm-2 for MATE1 and ~510 fmol cm-2 for OCT2. Dividing these values into the different Jmax values for transport of MPP, metformin, and atenolol mediated by MATE1 and OCT2 resulted in calculated TOR values (±SE, n = 4) of 84.0 ± 22.0 s-1 and 2.9 ± 0.6 s-1; metformin, 461.0 ± 121.0 s-1 and 12.6 ± 2.4 s-1; atenolol, 118.0 ± 31.0 s-1, respectively. These values are consistent with the TOR values determined for a variety of exchangers (NHEs), cotransporters (SGLTs, Lac permease), and uniporters (GLUTs, ENTs).
Collapse
|
5
|
Martinez-Guerrero L, Zhang X, Zorn KM, Ekins S, Wright SH. Cationic Compounds with SARS-CoV-2 Antiviral Activity and Their Interaction with Organic Cation Transporter/Multidrug and Toxin Extruder Secretory Transporters. J Pharmacol Exp Ther 2021; 379:96-107. [PMID: 34253645 PMCID: PMC9006906 DOI: 10.1124/jpet.121.000619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022] Open
Abstract
In the wake of the COVID-19 pandemic, drug repurposing has been highlighted for rapid introduction of therapeutics. Proposed drugs with activity against SARS-CoV-2 include compounds with positive charges at physiologic pH, making them potential targets for the organic cation secretory transporters of kidney and liver, i.e., the basolateral organic cation transporters, OCT1 and OCT2; and the apical multidrug and toxin extruders, MATE1 and MATE2-K. We selected several compounds proposed to have in vitro activity against SARS-CoV-2 (chloroquine, hydroxychloroquine, quinacrine, tilorone, pyronaridine, cetylpyridinium, and miramistin) to test their interaction with OCT and MATE transporters. We used Bayesian machine learning models to generate predictions for each molecule with each transporter and also experimentally determined IC50 values for each compound against labeled substrate transport into CHO cells that stably expressed OCT2, MATE1, or MATE2-K using three structurally distinct substrates (atenolol, metformin and 1-methyl-4-phenylpyridinium) to assess the impact of substrate structure on inhibitory efficacy. For the OCTs substrate identity influenced IC50 values, although the effect was larger and more systematic for OCT2. In contrast, inhibition of MATE1-mediated transport was largely insensitive to substrate identity. Unlike MATE1, inhibition of MATE2-K was influenced, albeit modestly, by substrate identity. Maximum unbound plasma concentration/IC50 ratios were used to identify potential clinical DDI recommendations; all the compounds interacted with the OCT/MATE secretory pathway, most with sufficient avidity to represent potential DDI issues for secretion of cationic drugs. This should be considered when proposing cationic agents as repurposed antivirals. SIGNIFICANCE STATEMENT: Drugs proposed as potential COVID-19 therapeutics based on in vitro activity data against SARS-CoV-2 include compounds with positive charges at physiological pH, making them potential interactors with the OCT/MATE renal secretory pathway. We tested seven such molecules as inhibitors of OCT1/2 and MATE1/2-K. All the compounds blocked transport activity regardless of substrate used to monitor activity. Suggesting that plasma concentrations achieved by normal clinical application of the test agents could be expected to influence the pharmacokinetics of selected cationic drugs.
Collapse
Affiliation(s)
- Lucy Martinez-Guerrero
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Xiaohong Zhang
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Kimberley M Zorn
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Sean Ekins
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Stephen H Wright
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona (L.M.-G., X.Z., S.H.W.), and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| |
Collapse
|
6
|
Ciarimboli G. Regulation Mechanisms of Expression and Function of Organic Cation Transporter 1. Front Pharmacol 2021; 11:607613. [PMID: 33732143 PMCID: PMC7959823 DOI: 10.3389/fphar.2020.607613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
The organic cation transporter 1 (OCT1) belongs together with OCT2 and OCT3 to the solute carrier family 22 (SLC22). OCTs are involved in the movement of organic cations through the plasma membrane. In humans, OCT1 is mainly expressed in the sinusoidal membrane of hepatocytes, while in rodents, OCT1 is strongly represented also in the basolateral membrane of renal proximal tubule cells. Considering that organic cations of endogenous origin are important neurotransmitters and that those of exogenous origin are important drugs, these transporters have significant physiological and pharmacological implications. Because of the high expression of OCTs in excretory organs, their activity has the potential to significantly impact not only local but also systemic concentration of their substrates. Even though many aspects governing OCT function, interaction with substrates, and pharmacological role have been extensively investigated, less is known about regulation of OCTs. Possible mechanisms of regulation include genetic and epigenetic modifications, rapid regulation processes induced by kinases, regulation caused by protein–protein interaction, and long-term regulation induced by specific metabolic and pathological situations. In this mini-review, the known regulatory processes of OCT1 expression and function obtained from in vitro and in vivo studies are summarized. Further research should be addressed to integrate this knowledge to known aspects of OCT1 physiology and pharmacology.
Collapse
Affiliation(s)
- Giuliano Ciarimboli
- Experimental Nephrology, Medicine Clinic D, Münster University Hospital, Münster, Germany
| |
Collapse
|
7
|
Da Silva A, Chretien B, Alexandre J, Emile G. Abemaciclib‐induced reversible grade 4 nephrotoxicity. Breast J 2020; 26:2329-2330. [DOI: 10.1111/tbj.13980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Angélique Da Silva
- Breast Cancer Unit Centre François Baclesse Institut Normand du Sein Caen France
| | - Basile Chretien
- Department of Pharmacology Caen University Hospital Caen France
- Pharmacovigilance Regional Center Caen University Hospital Caen France
| | - Joachim Alexandre
- Department of Pharmacology Caen University Hospital Caen France
- Pharmacovigilance Regional Center Caen University Hospital Caen France
| | - George Emile
- Breast Cancer Unit Centre François Baclesse Institut Normand du Sein Caen France
| |
Collapse
|
8
|
Organic Cation Transporters in Human Physiology, Pharmacology, and Toxicology. Int J Mol Sci 2020; 21:ijms21217890. [PMID: 33114309 PMCID: PMC7660683 DOI: 10.3390/ijms21217890] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
Individual cells and epithelia control the chemical exchange with the surrounding environment by the fine-tuned expression, localization, and function of an array of transmembrane proteins that dictate the selective permeability of the lipid bilayer to small molecules, as actual gatekeepers to the interface with the extracellular space. Among the variety of channels, transporters, and pumps that localize to cell membrane, organic cation transporters (OCTs) are considered to be extremely relevant in the transport across the plasma membrane of the majority of the endogenous substances and drugs that are positively charged near or at physiological pH. In humans, the following six organic cation transporters have been characterized in regards to their respective substrates, all belonging to the solute carrier 22 (SLC22) family: the organic cation transporters 1, 2, and 3 (OCT1–3); the organic cation/carnitine transporter novel 1 and 2 (OCTN1 and N2); and the organic cation transporter 6 (OCT6). OCTs are highly expressed on the plasma membrane of polarized epithelia, thus, playing a key role in intestinal absorption and renal reabsorption of nutrients (e.g., choline and carnitine), in the elimination of waste products (e.g., trimethylamine and trimethylamine N-oxide), and in the kinetic profile and therapeutic index of several drugs (e.g., metformin and platinum derivatives). As part of the Special Issue Physiology, Biochemistry, and Pharmacology of Transporters for Organic Cations, this article critically presents the physio-pathological, pharmacological, and toxicological roles of OCTs in the tissues in which they are primarily expressed.
Collapse
|
9
|
Wright SH. Molecular and cellular physiology of organic cation transporter 2. Am J Physiol Renal Physiol 2019; 317:F1669-F1679. [PMID: 31682169 DOI: 10.1152/ajprenal.00422.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Organic cation transporters play a critical role in mediating the distribution of cationic pharmaceuticals. Indeed, organic cation transporter (OCT)2 is the initial step in the renal secretion of organic cations and consequently plays a defining role in establishing the pharmacokinetics of many cationic drugs. Although a hallmark of OCTs is their broad selectivity, this characteristic also makes them targets for unwanted, adverse drug-drug interactions (DDIs), making them a focus for efforts to develop models of ligand interaction that could predict and preempt these adverse interactions. This review discusses the molecular characteristics of these transporters as well as the evidence that established the OCTs as key players in the distribution of organic cations. However, the primary focus is the present understanding of the complexity of ligand interaction with OCTs, particularly OCT2, including evidence for the presence of multiple ligand-binding sites and the influence of substrate structure on the affinity of the transporter for inhibitory ligands. This leads to a discussion of the complexities associated with the development of protocols for assessing the inhibitory potential of new molecular entities to perpetrate unwanted DDIs, the criteria that should be considered in the interpretation of the results of such protocols, and the challenges associated with development of models capable of predicting unwanted DDIs.
Collapse
Affiliation(s)
- Stephen H Wright
- Department of Physiology, University of Arizona, Tucson, Arizona
| |
Collapse
|