1,167
Views
4
CrossRef citations to date
0
Altmetric
Review

Evolving approaches on measurements and applications of intracellular free drug concentration and Kpuu in drug discovery

, &
Pages 733-746 | Received 07 Mar 2021, Accepted 24 May 2021, Published online: 22 Jul 2021

References

  • Smith DA, Di L, Kerns EH. The effect of plasma protein binding on in vivo efficacy: misconceptions in drug discovery. Nat Rev Drug Discov. 2010;9(12):929–939.
  • Scott DO, Ghosh A, Di L, et al. Passive Drug Permeation through Membranes and Cellular Distribution. Pharmcological Research. 2017;117:94–102.
  • Riccardi K, Li Z, Brown Janice A, et al. Determination of Unbound Partition Coefficient and in Vitro-in Vivo Extrapolation for SLC13A Transporter-Mediated Uptake. Drug Metab Dispos. 2016;44(10):1633–1642.
  • Orozco CC, Atkinson K, Ryu S, et al. Structural attributes influencing unbound tissue distribution. Eur J Med Chem. 2020;185:111813.
  • Riccardi K, Lin J, Li Z, et al. Novel Method to Predict In Vivo Liver-to-Plasma Kpuu for OATP Substrates Using Suspension Hepatocytes. Drug Metab Dispos. 2017;45(5):576–580.
  • Assmus F, Houston JB, Galetin A. Incorporation of lysosomal sequestration in the mechanistic model for prediction of tissue distribution of basic drugs. Eur J Pharm Sci. 2017;109:419–430.
  • Blouin A, Bolender RP, Weibel ER. Distribution of organelles and membranes between hepatocytes and nonhepatocytes in the rat liver parenchyma. A stereological study. Journal of Cell Biology. 1977;72(2):441–455.
  • Naim B, Brumfeld V, Kapon R, et al. Passive and Facilitated Transport in Nuclear Pore Complexes Is Largely Uncoupled. Journal of Biological Chemistry. 2007;282(6):3881–3888.
  • Timney BL, Raveh B, Mironska R, et al. Simple rules for passive diffusion through the nuclear pore complex. J. Cell Biol. 2016;215(1):57–76.
  • Gilmore IS, Heiles S, Pieterse CL. Metabolic Imaging at the Single-Cell Scale: recent Advances in Mass Spectrometry Imaging. Annu. Rev. Anal. Chem. 2019;12(1):201–224.
  • Sepp K, Lee M, Bluntzer MTJ, et al. Utilizing Stimulated Raman Scattering Microscopy To Study Intracellular Distribution of Label-Free Ponatinib in Live Cells. J Med Chem. 2020;63(5):2028–2034.
  • El-Mashtoly SF. Drug Distribution in Living Cells via Label-Free Molecular Fingerprint. J Med Chem. 2020;63(7):3472–3474.
  • Elgart V, Lin J-R, Loscalzo J. Determinants of drug-target interactions at the single cell level. PLOS Computational Biology. 2018, 14, e1006601/1-e1006601/23.(12):e1006601.
  • Riccardi K, Ryu S, Lin J, et al. Comparison of species and cell-type differences in fraction unbound of liver tissues, hepatocytes, and cell lines. Drug Metab Dispos. 2018;46(4):415–S4.
  • Riccardi K, Ryu S, Tess D, et al. Comparison of Fraction Unbound Between Liver Homogenate and Hepatocytes at 4°C. AAPS J. 2020;22(4):91.
  • Yoshikado T, Toshimoto K, Nakada T, et al. Comparison of methods for estimating unbound intracellular-to-medium concentration ratios in rat and human hepatocytes using statins. Drug Metab Dispos. 2017;45(7):779–789.
  • Shitara Y, Maeda K, Ikejiri K, et al. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34(1):45–78.
  • Izumi S, Nozaki Y, Komori T, et al. Comparison of the Predictability of Human Hepatic Clearance for Organic Anion Transporting Polypeptide Substrate Drugs Between Different In Vitro–In Vivo Extrapolation Approaches. J Pharm Sci. 2017;106(9):2678–2687.
  • Trunkle C, Lechner C, Korr D, et al. Concentration Dependence of the Unbound Partition Coefficient Kpuu and Its Application to Correct for Exposure-Related Discrepancies between Biochemical and Cellular Potency of KAT6A Inhibitors. Drug Metab Dispos. 2020;48(7):553–562.
  • Yabe Y, Galetin A, Houston JB. Kinetic characterization of rat hepatic uptake of 16 actively transported drugs. Drug Metab Dispos. 2011; 39(10):1808-1814.
  • Ryu S, Tess D, Chang G, et al. Evaluation of Fraction Unbound Across 7 Tissues of 5 Species. J. Pharm. Sci. 2020;109:1178–1190.
  • Ryu S, Riccardi K, Jordan S, et al. Determination of Fraction Unbound and Unbound Partition Coefficient to Estimate Intracellular Free Drug Concentration. In: Rosania GR, Thurber GM, editors. Quantitative Analysis of Cellular Drug Transport, Disposition, and Delivery, Methods in Pharmacology and Toxicology. Springer, New York, NY; 2021;81–96.
  • Riccardi KA, Tess DA, Lin J, et al. A novel unified approach to predict human hepatic clearance for both enzyme- and transporter-mediated mechanisms using suspended human hepatocytes. Drug Metab Dispos. 2019;47(5):484–492.
  • Riede J, Camenisch G, Huwyler J, et al. Current In Vitro Methods to Determine Hepatic Kp uu : a Comparison of Their Usefulness and Limitations. J Pharm Sci. 2017;106(9):2805–2814.
  • Ryu S, Novak JJ, Patel R, et al. The impact of low temperature on fraction unbound for plasma and tissue. Biopharmaceutics & Drug Disposition. 2018;39(9):437–442.
  • Faneyte IF, Kristel PMP, Van De Vijver MJ. DeterminingMDR1/P-glycoprotein expression in breast cancer. International Journal of Cancer. 2001;93(1):114–122.
  • Lemos C, Kathmann I, Giovannetti E, et al. Cellular folate status modulates the expression of BCRP and MRP multidrug transporters in cancer cell lines from different origins. Molecular Cancer Therapeutics. 2009;8(3):655–664.
  • Oba T, Izumi H, Ito K-I. ABCB1 and ABCC11 confer resistance to eribulin in breast cancer cell lines. Oncotarget. 2016;7(43):70011–70027.
  • Di L, Breen C, Chambers R, et al. Industry Perspective on Contemporary Protein-Binding Methodologies: considerations for Regulatory Drug-Drug Interaction and Related Guidelines on Highly Bound Drugs. J. Pharm. Sci. (Philadelphia, PA, U. S.). 2017;106:3442–3452.
  • Riccardi K, Cawley S, Yates PD, et al. Plasma Protein Binding of Challenging Compounds. J Pharm Sci. 2015;104(8):2627–2636.
  • Li Z, Erion D, Maurer T. Model-Based Assessment of Plasma Citrate Flux Into the Liver: implications for NaCT as a Therapeutic Target. CPT: pharmacometrics & systems pharmacology. 2016;5(3):132–139.
  • Huard K, Gosset JR, Montgomery JI, et al. Optimization of a Dicarboxylic Series for in Vivo Inhibition of Citrate Transport by the Solute Carrier 13 (SLC13) Family. J Med Chem. 2016;59(3):1165–1175.
  • Lee KK, Workman JL. Histone acetyltransferase complexes: one size doesn’t fit all. . Nature Reviews Molecular Cell Biology. 2007;8(4):284–295.
  • Baell JB, Leaver DJ, Hermans SJ, et al. Inhibitors of histone acetyltransferases KAT6A/B induce senescence and arrest tumour growth. Nature (London, U. K.). 2018;560(7717):253–257.
  • Arthur JSC, Ley SC. Mitogen-activated protein kinases in innate immunity. . Nature Reviews Immunology. 2013;13(9):679–692.
  • Mateus A, Gordon LJ, Wayne GJ, et al. Prediction of intracellular exposure bridges the gap between target- and cell-based drug discovery. Proceedings of the National Academy of Sciences. 2017;114(30): E6231–E6239.
  • Albermann N, Schmitz-Winnenthal FH, Z’Graggen K, et al. Expression of the drug transporters MDR1/ABCB1, MRP1/ABCC1, MRP2/ABCC2, BCRP/ABCG2, and PXR in peripheral blood mononuclear cells and their relationship with the expression in intestine and liver. Biochemical Pharmacology. 2005;70(6):949–958.
  • Shola AI, Adegboye A. Polypharmacy in the elderly: the need for concern and strategies for its control. British Journal of Pharmaceutical Research. 2015;8(5):1–12.
  • Lu C, Di L. In vitro and in vivo methods to assess pharmacokinetic drug– drug interactions in drug discovery and development. Biopharmaceutics & Drug Disposition. 2020;41(1–2):3–31.
  • Saravanakumar A, Sadighi A, Ryu R, et al. Physicochemical Properties, Biotransformation, and Transport Pathways of Established and Newly Approved Medications: a Systematic Review of the Top 200 Most Prescribed Drugs vs. the FDA-Approved Drugs Between 2005 and 2016. Clinical Pharmacokinetics. 2019;58(10):1281–1294.
  • Treyer A, Ullah M, Parrott N, et al. Impact of Intracellular Concentrations on Metabolic Drug-Drug Interaction Studies. AAPS J. 2019;21(5):77.
  • Filppula AM, Parvizi R, Mateus A, et al. Improved predictions of time-dependent drug-drug interactions by determination of cytosolic drug concentrations. Scientific Reports. 2019;9(1): 1–14.
  • Di L, Whitney‐Pickett C, Umland JP, et al. Development of a new permeability assay using low-efflux MDCKII cells. J Pharm Sci. 2011;100(11):4974–4985.
  • Keefer C, Chang G, Carlo A, et al. Mechanistic insights on clearance and inhibition discordance between liver microsomes and hepatocytes when clearance in liver microsomes is higher than in hepatocytes. Eur J Pharm Sci. 2020;155:105541.
  • Di L, Keefer C, Scott DO, et al. Mechanistic insights from comparing intrinsic clearance values between human liver microsomes and hepatocytes to guide drug design. Eur J Med Chem. 2012;57:441–448.
  • Li M, Yuan H, Li N, et al. Identification of interspecies difference in efflux transporters of hepatocytes from dog, rat, monkey and human. Eur J Pharm Sci. 2008;35(1–2):114–126.
  • Billington S, Shoner S, Lee S, et al. Positron Emission Tomography Imaging of [11 C]Rosuvastatin Hepatic Concentrations and Hepatobiliary Transport in Humans in the Absence and Presence of Cyclosporin A. Clinical Pharmacology & Therapeutics. 2019;106(5):1056–1066.
  • Gormsen LC, Sundelin EI, Jensen JB, et al. In vivo imaging of human 11C-metformin in peripheral organs: dosimetry, biodistribution, and kinetic analyses. J Nucl Med. 2016;57(12):1920–1926.
  • Kaneko K-I, Tanaka M, Ishii A, et al. A Clinical Quantitative Evaluation of Hepatobiliary Transport of [11 C]Dehydropravastatin in Humans Using Positron Emission Tomography. Drug Metab Dispos. 2018;46(5):719–728.
  • Li Z, Di TS, Maurer TS. Theoretical Considerations for Direct Translation of Unbound Liver-to-Plasma Partition Coefficient from In Vitro to In Vivo. AAPS J. 2019;21(3):43.
  • Bowman CM, Benet LZ. An examination of protein binding and protein-facilitated uptake relating to in vitro-in vivo extrapolation. Eur J Pharm Sci. 2018;123:502–514.
  • Poulin P, Haddad S. Extrapolation of the Hepatic Clearance of Drugs in the Absence of Albumin In Vitro to That in the Presence of Albumin In Vivo: comparative Assessment of 2 Extrapolation Models Based on the Albumin-Mediated Hepatic Uptake Theory and Limitations and Mechanistic Insights. J. Pharm. Sci.2018;107(7):1791–1797.
  • Miyauchi S, Masuda M, Kim S-J, et al. The phenomenon of albumin-mediated hepatic uptake of organic anion transport polypeptide substrates: prediction of the in vivo uptake clearance from the in vitro uptake by isolated hepatocytes using a facilitated-dissociation model. Drug Metab Dispos. 2018;46(3):259–S4.
  • Sodhi JK, Liu S, Benet LZ. Challenging the Relevance of Unbound Tissue-to-Blood Partition Coefficient (Kpuu) on Prediction of Drug-Drug Interactions. Pharm Res. 2020;37(4):73.
  • Jones HM, Barton HA, Lai Y, et al. Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data. Drug Metab Dispos. 2012;40(5):1007–1017.
  • Li R, Barton HA, Yates PD, et al. A “middle-out” approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn. 2014;41(3):197–209.
  • Liu H, Dong K, Zhang W, et al. Prediction of brain:blood unbound concentration ratios in CNS drug discovery employing in silico and in vitro model systems. Drug Discov Today. 2018;23(7):1357–1372.
  • Liu X, Chen C. Free Drug Hypothesis for CNS Drug Candidates. In: Di L, Kerns EH, editors. Blood-Brain Barrier in Drug Discovery. Hoboken, New Jersey: John Wiley & Sons, Inc; 2015. p. 42–65.
  • Wager TT, Hou X, Verhoest PR, et al. Moving beyond Rules: the Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach To Enable Alignment of Druglike Properties. ACS Chemical Neuroscience. 2010;1(6):435–449.
  • Knight ZA, Shokat KM. Features of Selective Kinase Inhibitors. Chemistry & Biology. 2005;12(6):621–637.
  • Satyanarayana A, Kaldis P. Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene. 2009;28(33):2925–2939.
  • Schwaid AG, Cornella-Taracido I. Causes and Significance of Increased Compound Potency in Cellular or Physiological Contexts. J Med Chem. 2018;61(5):1767–1773.
  • Vasta JD, Corona CR, Wilkinson J, et al. Quantitative, Wide-Spectrum Kinase Profiling in Live Cells for Assessing the Effect of Cellular ATP on Target Engagement. Cell Chemical Biology. 2018;25(2):206–214.e11.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.