Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Physiologically Based Pharmacokinetic Modeling of Transporter-Mediated Hepatic Disposition of Imaging Biomarker Gadoxetate in Rats

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

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

          Related collections

          Most cited references66

          • Record: found
          • Abstract: not found
          • Article: not found

          Physiological parameters in laboratory animals and humans.

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Estimating kinetic parameters from dynamic contrast-enhanced T(1)-weighted MRI of a diffusable tracer: standardized quantities and symbols.

            We describe a standard set of quantity names and symbols related to the estimation of kinetic parameters from dynamic contrast-enhanced T(1)-weighted magnetic resonance imaging data, using diffusable agents such as gadopentetate dimeglumine (Gd-DTPA). These include a) the volume transfer constant K(trans) (min(-1)); b) the volume of extravascular extracellular space (EES) per unit volume of tissue v(e) (0 < v(e) < 1); and c) the flux rate constant between EES and plasma k(ep) (min(-1)). The rate constant is the ratio of the transfer constant to the EES (k(ep) = K(trans)/v(e)). Under flow-limited conditions K(trans) equals the blood plasma flow per unit volume of tissue; under permeability-limited conditions K(trans) equals the permeability surface area product per unit volume of tissue. We relate these quantities to previously published work from our groups; our future publications will refer to these standardized terms, and we propose that these be adopted as international standards.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              HIGH-YIELD PREPARATION OF ISOLATED RAT LIVER PARENCHYMAL CELLS

              A new technique employing continuous recirculating perfusion of the rat liver in situ, shaking of the liver in buffer in vitro, and filtration of the tissue through nylon mesh, results in the conversion of about 50% of the liver into intact, isolated parenchymal cells. The perfusion media consist of: (a) calcium-free Hanks' solution containing 0.05% collagenase and 0.10% hyaluronidase, and (b) magnesium and calcium-free Hanks' solution containing 2 mM ethylenediaminetetraacetate. Biochemical and morphologic studies indicate that the isolated cells are viable. They respire in a medium containing calcium ions, synthesize glucose from lactate, are impermeable to inulin, do not stain with trypan blue, and retain their structural integrity. Electron microscopy of biopsies taken during and after perfusion reveals that desmosomes are quickly cleaved. Hemidesmosome-containing areas of the cell membrane invaginate and appear to pinch off and migrate centrally. Tight and gap junctions, however, persist on the intact, isolated cells, retaining small segments of cytoplasm from formerly apposing parenchymal cells. Cells which do not retain tight and gap junctions display swelling of Golgi vacuoles and vacuoles in the peripheral cytoplasm. Cytoplasmic vacuolization in a small percentage of cells and potassium loss are the only indications of cell injury detected. By other parameters measured, the isolated cells are comparable to normal hepatic parenchymal cells in situ in appearance and function.
                Bookmark

                Author and article information

                Journal
                Mol Pharm
                Mol Pharm
                mp
                mpohbp
                Molecular Pharmaceutics
                American Chemical Society
                1543-8384
                1543-8392
                20 July 2021
                02 August 2021
                : 18
                : 8
                : 2997-3009
                Affiliations
                []Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester , Manchester M13 9PL, U.K.
                []Division of Medical Physics, University of Leeds , Leeds LS2 9JT, U.K.
                [§ ]MR & CT Contrast Media Research, Bayer AG , Berlin 13342, Germany
                []Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield , Sheffield S10 2TN, U.K.
                Author notes
                [* ]Email: aleksandra.galetin@ 123456manchester.ac.uk . Phone: +44-161-2756886.
                Author information
                https://orcid.org/0000-0001-9144-3824
                https://orcid.org/0000-0002-3933-5217
                Article
                10.1021/acs.molpharmaceut.1c00206
                8397403
                34283621
                2bd55225-eeba-44c9-a479-d3b1475031d8
                © 2021 The Authors. Published by American Chemical Society

                Permits the broadest form of re-use including for commercial purposes, provided that author attribution and integrity are maintained ( https://creativecommons.org/licenses/by/4.0/).

                History
                : 15 March 2021
                : 28 June 2021
                : 21 June 2021
                Funding
                Funded by: Horizon 2020 Framework Programme, doi 10.13039/100010661;
                Award ID: NA
                Funded by: Innovative Medicines Initiative, doi 10.13039/501100010767;
                Award ID: 116106
                Funded by: European Federation of Pharmaceutical Industries and Associations, doi 10.13039/100013322;
                Award ID: NA
                Categories
                Article
                Custom metadata
                mp1c00206
                mp1c00206

                Pharmacology & Pharmaceutical medicine
                gadoxetate,imaging biomarker,drug transporters,physiologically based pharmacokinetic model,hepatobiliary excretion,drug−drug interactions,quantitative translation

                Comments

                Comment on this article