10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Delineating the Role of Various Factors in Renal Disposition of Digoxin through Application of Physiologically Based Kidney Model to Renal Impairment Populations

      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

          Development of submodels of organs within physiologically-based pharmacokinetic (PBPK) principles and beyond simple perfusion limitations may be challenging because of underdeveloped in vitro-in vivo extrapolation approaches or lack of suitable clinical data for model refinement. However, advantage of such models in predicting clinical observations in divergent patient groups is now commonly acknowledged. Mechanistic understanding of altered renal secretion in renal impairment is one area that may benefit from such models, despite knowledge gaps in renal pathophysiology. In the current study, a PBPK kidney model was developed for digoxin, accounting for the roles of organic anion transporting peptide 4C1 (OATP4C1) and P-glycoprotein (P-gp) in its tubular secretion, with the aim to investigate the impact of age and renal impairment (moderate to severe) on renal drug disposition. Initial PBPK simulations based on changes in glomerular filtration rate (GFR) underestimated the observed reduction in digoxin renal excretion clearance (CL R) in subjects with moderately impaired renal function relative to healthy. Reduction in either proximal tubule cell number or the OATP4C1 abundance in the mechanistic kidney model successfully predicted 59% decrease in digoxin CL R, in particular when these changes were proportional to reduction in GFR. In contrast, predicted proximal tubule concentration of digoxin was only sensitive to changes in the transporter expression/ million proximal tubule cells. Based on the mechanistic modeling, reduced proximal tubule cellularity and OATP4C1 abundance, and inhibition of OATP4C1-mediated transport, are proposed as possible causes of reduced digoxin renal secretion in renally impaired patients.

          Abstract

          Related collections

          Most cited references64

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

          Prediction of creatinine clearance from serum creatinine.

          A formula has been developed to predict creatinine clearance (Ccr) from serum creatinine (Scr) in adult males: (see article)(15% less in females). Derivation included the relationship found between age and 24-hour creatinine excretion/kg in 249 patients aged 18-92. Values for Ccr were predicted by this formula and four other methods and the results compared with the means of two 24-hour Ccr's measured in 236 patients. The above formula gave a correlation coefficient between predicted and mean measured Ccr's of 0.83; on average, the difference predicted and mean measured values was no greater than that between paired clearances. Factors for age and body weight must be included for reasonable prediction.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Chronic hypoxia and tubulointerstitial injury: a final common pathway to end-stage renal failure.

            Recent studies emphasize the role of chronic hypoxia in the tubulointerstitium as a final common pathway to end-stage renal failure. When advanced, tubulointerstitial damage is associated with the loss of peritubular capillaries. Associated interstitial fibrosis impairs oxygen diffusion and supply to tubular and interstitial cells. Hypoxia of tubular cells leads to apoptosis or epithelial-mesenchymal transdifferentiation. This in turn exacerbates fibrosis of the kidney and subsequent chronic hypoxia, setting in train a vicious cycle whose end point is ESRD. A number of mechanisms that induce tubulointerstitial hypoxia at an early stage have been identified. Glomerular injury and vasoconstriction of efferent arterioles as a result of imbalances in vasoactive substances decrease postglomerular peritubular capillary blood flow. Angiotensin II not only constricts efferent arterioles but, via its induction of oxidative stress, also hampers the efficient utilization of oxygen in tubular cells. Relative hypoxia in the kidney also results from increased metabolic demand in tubular cells. Furthermore, renal anemia hinders oxygen delivery. These factors can affect the kidney before the appearance of significant pathologic changes in the vasculature and predispose the kidney to tubulointerstitial injury. Therapeutic approaches that target the chronic hypoxia should prove effective against a broad range of renal diseases. Current modalities include the improvement of anemia with erythropoietin, the preservation of peritubular capillary blood flow by blockade of the renin-angiotensin system, and the use of antioxidants. Recent studies have elucidated the mechanism of hypoxia-induced transcription, namely that prolyl hydroxylase regulates hypoxia-inducible factor. This has given hope for the development of novel therapeutic approaches against this final common pathway.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions.

              A key component of whole body physiologically based pharmacokinetic (WBPBPK) models is the tissue-to-plasma water partition coefficients (Kpu's). The predictability of Kpu values using mechanistically derived equations has been investigated for 7 very weak bases, 20 acids, 4 neutral drugs and 8 zwitterions in rat adipose, bone, brain, gut, heart, kidney, liver, lung, muscle, pancreas, skin, spleen and thymus. These equations incorporate expressions for dissolution in tissue water and, partitioning into neutral lipids and neutral phospholipids. Additionally, associations with acidic phospholipids were incorporated for zwitterions with a highly basic functionality, or extracellular proteins for the other compound classes. The affinity for these cellular constituents was determined from blood cell data or plasma protein binding, respectively. These equations assume drugs are passively distributed and that processes are nonsaturating. Resultant Kpu predictions were more accurate when compared to published equations, with 84% as opposed to 61% of the predicted values agreeing with experimental values to within a factor of 3. This improvement was largely due to the incorporation of distribution processes related to drug ionisation, an issue that is not addressed in earlier equations. Such advancements in parameter prediction will assist WBPBPK modelling, where time, cost and labour requirements greatly deter its application. (c) 2006 Wiley-Liss, Inc. and the American Pharmacists Association
                Bookmark

                Author and article information

                Journal
                J Pharmacol Exp Ther
                J. Pharmacol. Exp. Ther
                jpet
                J Pharmacol Exp Ther
                JPET
                The Journal of Pharmacology and Experimental Therapeutics
                The American Society for Pharmacology and Experimental Therapeutics (Bethesda, MD )
                0022-3565
                1521-0103
                March 2017
                March 2017
                March 2017
                : 360
                : 3
                : 484-495
                Affiliations
                [1]Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (D.S., A.G., A.R.-H.); DMPK, Oncology iMed, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, United Kingdom (C.R.J.); and Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, United Kingdom (A.R.-H.)
                Author notes
                Address correspondence to: Amin Rostami-Hodjegan, Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK. E-mail: amin.rostami@ 123456manchester.ac.uk
                [1]

                Current afiliation: Heptares Therapeutics Limited, BioPark, Welwyn Garden City, Hertfordshire, UK.

                Article
                JPET_237438
                10.1124/jpet.116.237438
                5370399
                28057840
                e439b4de-f7d0-4f98-be5d-888a70359099
                Copyright © 2017 by The Author(s)

                This is an open access article distributed under the CC BY Attribution 4.0 International license.

                History
                : 25 August 2016
                : 20 December 2016
                Page count
                Figures: 9, Tables: 4, Equations: 2, References: 73, Pages: 12
                Categories
                Metabolism, Transport, and Pharmacogenomics
                Custom metadata
                v1

                Comments

                Comment on this article