Skip to main content

Investigation of Transporter-Mediated Drug-Drug Interactions Using PET/MRI

  • Chapter
  • First Online:
Image Fusion in Preclinical Applications

Abstract

Drug disposition consists of absorption, distribution, metabolism and excretion (ADME). For these processes, drugs and drug metabolites have to cross cellular membranes in different organs and tissues (e.g. intestine, liver, kidney, etc.). In many cases, movement of drugs and drug metabolites across cellular membranes is not just a passive diffusion-mediated process but occurs via saturable transmembrane transporters belonging either to the solute carrier (SLC) or adenosine triphosphate-binding cassette (ABC) families. When two drugs, which are recognized by the same SLC or ABC transporters, are co-administered, one drug may induce transporter inhibition/saturation and thereby change the disposition of the other drug as compared to when the drugs are administered alone. This phenomenon has been termed transporter-mediated drug-drug interaction (DDI), which is of great concern in drug development as this may impact drug safety and efficacy [1]. In many cases, changes in the activity of transmembrane transporters may lead to pronounced changes in drug tissue distribution (e.g. liver, kidneys, brain) without changes in drug plasma pharmacokinetics [2]. To assess such tissue DDIs in vivo, a methodology is needed to measure drug tissue concentration levels. The non-invasive nuclear imaging method positron emission tomography (PET) is a very powerful tool for measuring tissue distribution of drugs radiolabelled with positron-emitting radionuclides, such as carbon-11 (11C, half-life: 20.4 min) or fluorine-18 (18F, half-life: 109.8 min), in animals or humans [3, 4]. Dedicated small-animal PET systems allow to measure rodents (mice, rats) with high sensitivity and good spatial resolution. As PET does not provide anatomical information, it needs to be combined with anatomical imaging to allow for better definition of organs or tissues of interest. In this chapter, we provide a case report of how PET in combination with magnetic resonance imaging (MRI) can be used to assess transporter-mediated DDIs in different organs of the mouse.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

eBook
USD 16.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 129.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Nies AT, Schwab M, Keppler D. Interplay of conjugating enzymes with OATP uptake transporters and ABCC/MRP efflux pumps in the elimination of drugs. Expert Opin Drug Metab Toxicol. 2008;4(5):545–68.

    Article  CAS  Google Scholar 

  2. Kusuhara H, Sugiyama Y. In vitro-in vivo extrapolation of transporter-mediated clearance in the liver and kidney. Drug Metab Pharmacokinet. 2009;24(1):37–52.

    Article  CAS  Google Scholar 

  3. Langer O. Use of PET imaging to evaluate transporter-mediated drug-drug interactions. J Clin Pharmacol. 2016;56(Suppl 7):S143–56.

    Article  CAS  Google Scholar 

  4. Wagner CC, Langer O. Approaches using molecular imaging technology -- use of PET in clinical microdose studies. Adv Drug Deliver Rev. 2011;63(7):539–46.

    Article  CAS  Google Scholar 

  5. Nagy K, Toth M, Major P, Patay G, Egri G, Haggkvist J, et al. Performance evaluation of the small-animal nanoScan PET/MRI system. J Nucl Med. 2013;54(10):1825–32.

    Article  Google Scholar 

  6. Wehrl HF, Amend M, Thielcke A. Multimodal imaging and image fusion. In: Kiessling F, Pichler BJ, Hauff P, editors. Small Animal imaging basics and practical guide. Berlin: Springer International Publishing; 2017. p. 491–507.

    Chapter  Google Scholar 

  7. Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharm. 2009;158(3):693–705.

    Article  CAS  Google Scholar 

  8. Hammerman PS, Janne PA, Johnson BE. Resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clin Cancer Res. 2009;15(24):7502–9.

    Article  CAS  Google Scholar 

  9. Ling J, Johnson KA, Miao Z, Rakhit A, Pantze MP, Hamilton M, et al. Metabolism and excretion of erlotinib, a small molecule inhibitor of epidermal growth factor receptor tyrosine kinase, in healthy male volunteers. Drug Metab Dispos. 2006;34(3):420–6.

    CAS  PubMed  Google Scholar 

  10. Agarwal S, Manchanda P, Vogelbaum MA, Ohlfest JR, Elmquist WF. Function of the blood-brain barrier and restriction of drug delivery to invasive glioma cells: findings in an orthotopic rat xenograft model of glioma. Drug Metab Dispos. 2013;41(1):33–9.

    Article  CAS  Google Scholar 

  11. de Vries NA, Buckle T, Zhao J, Beijnen JH, Schellens JH, van Tellingen O. Restricted brain penetration of the tyrosine kinase inhibitor erlotinib due to the drug transporters P-gp and BCRP. Investig New Drugs. 2012;30(2):443–9.

    Article  CAS  Google Scholar 

  12. Kodaira H, Kusuhara H, Ushiki J, Fuse E, Sugiyama Y. Kinetic analysis of the cooperation of P-glycoprotein (P-gp/Abcb1) and breast cancer resistance protein (Bcrp/Abcg2) in limiting the brain and testis penetration of erlotinib, flavopiridol, and mitoxantrone. J Pharmacol Exp Ther. 2010;333(3):788–96.

    Article  CAS  Google Scholar 

  13. Loening AM, Gambhir SS. AMIDE: a free software tool for multimodality medical image analysis. Mol Imaging. 2003;2(3):131–7.

    Article  Google Scholar 

  14. Tai YC, Ruangma A, Rowland D, Siegel S, Newport DF, Chow PL, et al. Performance evaluation of the microPET focus: a third-generation microPET scanner dedicated to animal imaging. J Nucl Med. 2005;46(3):455–63.

    PubMed  Google Scholar 

  15. Takashima T, Wu C, Takashima-Hirano M, Katayama Y, Wada Y, Suzuki M, et al. Evaluation of breast cancer resistance protein function in hepatobiliary and renal excretion using PET with 11C-SC-62807. J Nucl Med. 2013;54(2):267–76.

    Article  CAS  Google Scholar 

  16. Takano A, Kusuhara H, Suhara T, Ieiri I, Morimoto T, Lee YJ, et al. Evaluation of in vivo P-glycoprotein function at the blood-brain barrier among MDR1 gene polymorphisms by using 11C-verapamil. J Nucl Med. 2006;47(9):1427–33.

    CAS  PubMed  Google Scholar 

  17. Schmid A, Schmitz J, Mannheim JG, Maier FC, Fuchs K, Wehrl HF, et al. Feasibility of sequential PET/MRI using a state-of-the-art small animal PET and a 1 T benchtop MRI. Mol Imaging Biol. 2013;15(2):155–65.

    Article  Google Scholar 

  18. Johnston RA, Rawling T, Chan T, Zhou F, Murray M. Selective inhibition of human solute carrier transporters by multikinase inhibitors. Drug Metab Dispos. 2014;42(11):1851–7.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Thomas Wanek .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Wanek, T., Traxl, A., Kuntner-Hannes, C., Langer, O. (2019). Investigation of Transporter-Mediated Drug-Drug Interactions Using PET/MRI. In: Kuntner-Hannes, C., Haemisch, Y. (eds) Image Fusion in Preclinical Applications. Springer, Cham. https://doi.org/10.1007/978-3-030-02973-9_6

Download citation

  • DOI: https://doi.org/10.1007/978-3-030-02973-9_6

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-030-02972-2

  • Online ISBN: 978-3-030-02973-9

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics