Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Technical Report
  • Published:

Molecular imaging of lymphoid organs and immune activation by positron emission tomography with a new [18F]-labeled 2′-deoxycytidine analog

Abstract

Monitoring immune function with molecular imaging could have a considerable impact on the diagnosis and treatment evaluation of immunological disorders and therapeutic immune responses. Positron emission tomography (PET) is a molecular imaging modality with applications in cancer and other diseases. PET studies of immune function have been limited by a lack of specialized probes. We identified [18F]FAC (1-(2′-deoxy-2′-[18F]fluoroarabinofuranosyl) cytosine) by differential screening as a new PET probe for the deoxyribonucleotide salvage pathway. [18F]FAC enabled visualization of lymphoid organs and was sensitive to localized immune activation in a mouse model of antitumor immunity. [18F]FAC microPET also detected early changes in lymphoid mass in systemic autoimmunity and allowed evaluation of immunosuppressive therapy. These data support the use of [18F]FAC PET for immune monitoring and suggest a wide range of clinical applications in immune disorders and in certain types of cancer.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Identification of fluorinated deoxycytidine analogs retained in activated versus naive T cells.
Figure 2: [18F]FAC has better selectivity for lymphoid organs compared with other PET probes for nucleoside metabolism and glycolysis.
Figure 3: Increased [18F]FAC retention in spleen and lymph nodes at the peak of the primary antitumor immune response.
Figure 4: [18F]FAC microPET-CT allows visualization of increased lymphoid mass in systemic autoimmunity and can be used to monitor immunosuppressive therapeutic interventions.

Similar content being viewed by others

References

  1. Phelps, M.E. Inaugural article: positron emission tomography provides molecular imaging of biological processes. Proc. Natl. Acad. Sci. USA 97, 9226–9233 (2000).

    Article  CAS  Google Scholar 

  2. Koehne, G. et al. Serial in vivo imaging of the targeted migration of human HSV-TK–transduced antigen-specific lymphocytes. Nat. Biotechnol. 21, 405–413 (2003).

    Article  CAS  Google Scholar 

  3. Dubey, P. et al. Quantitative imaging of the T cell antitumor response by positron-emission tomography. Proc. Natl. Acad. Sci. USA 100, 1232–1237 (2003).

    Article  CAS  Google Scholar 

  4. Su, H., Forbes, A., Gambhir, S.S. & Braun, J. Quantitation of cell number by a positron emission tomography reporter gene strategy. Mol. Imaging Biol. 6, 139–148 (2004).

    Article  Google Scholar 

  5. Shu, C.J. et al. Visualization of a primary anti-tumor immune response by positron emission tomography. Proc. Natl. Acad. Sci. USA 102, 17412–17417 (2005).

    Article  CAS  Google Scholar 

  6. Su, H., Chang, D.S., Gambhir, S.S. & Braun, J. Monitoring the antitumor response of naive and memory CD8 T cells in Rag1−/− mice by positron emission tomography. J. Immunol. 176, 4459–4467 (2006).

    Article  CAS  Google Scholar 

  7. Radu, C.G., Shu, C.J., Shelly, S.M., Phelps, M.E. & Witte, O.N. Positron emission tomography with computed tomography imaging of neuroinflammation in experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 104, 1937–1942 (2007).

    Article  CAS  Google Scholar 

  8. Van Rompay, A.R., Johansson, M. & Karlsson, A. Substrate specificity and phosphorylation of antiviral and anticancer nucleoside analogues by human deoxyribonucleoside kinases and ribonucleoside kinases. Pharmacol. Ther. 100, 119–139 (2003).

    Article  CAS  Google Scholar 

  9. Griffith, D.A. & Jarvis, S.M. Nucleoside and nucleobase transport systems of mammalian cells. Biochim. Biophys. Acta 1286, 153–181 (1996).

    Article  CAS  Google Scholar 

  10. Pankiewicz, K.W. Fluorinated nucleosides. Carbohydr. Res. 327, 87–105 (2000).

    Article  CAS  Google Scholar 

  11. Gray, J.H., Owen, R.P. & Giacomini, K.M. The concentrative nucleoside transporter family, SLC28. Pflugers Arch. 447, 728–734 (2004).

    Article  CAS  Google Scholar 

  12. Baldwin, S.A. et al. The equilibrative nucleoside transporter family, SLC29. Pflugers Arch. 447, 735–743 (2004).

    Article  CAS  Google Scholar 

  13. Eriksson, S., Munch-Petersen, B., Johansson, K. & Eklund, H. Structure and function of cellular deoxyribonucleoside kinases. Cell. Mol. Life Sci. 59, 1327–1346 (2002).

    Article  CAS  Google Scholar 

  14. Nakano, Y. et al. Gemcitabine chemoresistance and molecular markers associated with gemcitabine transport and metabolism in human pancreatic cancer cells. Br. J. Cancer 96, 457–463 (2007).

    Article  CAS  Google Scholar 

  15. van der Wilt, C.L. et al. The role of deoxycytidine kinase in gemcitabine cytotoxicity. Adv. Exp. Med. Biol. 486, 287–290 (2000).

    Article  CAS  Google Scholar 

  16. Shipley, L.A. et al. Metabolism and disposition of gemcitabine, and oncolytic deoxycytidine analog, in mice, rats, and dogs. Drug Metab. Dispos. 20, 849–855 (1992).

    CAS  PubMed  Google Scholar 

  17. Shields, A.F. et al. Imaging proliferation in vivo with [F-18]FLT and positron emission tomography. Nat. Med. 4, 1334–1336 (1998).

    Article  CAS  Google Scholar 

  18. Sun, H. et al. Imaging DNA synthesis in vivo with 18F-FMAU and PET. J. Nucl. Med. 46, 292–296 (2005).

    CAS  PubMed  Google Scholar 

  19. Fefer, A., McCoy, J.L., Perk, K. & Glynn, J.P. Immunologic, virologic and pathologic studies of regression of autochthonous Moloney sarcoma virus–induced tumors in mice. Cancer Res. 28, 1577–1585 (1968).

    CAS  PubMed  Google Scholar 

  20. Schepers, K. et al. Differential kinetics of antigen-specific CD4+ and CD8+ T cell responses in the regression of retrovirus-induced sarcomas. J. Immunol. 169, 3191–3199 (2002).

    Article  CAS  Google Scholar 

  21. Morse, H.C. III et al. Abnormalities induced by the mutant gene Ipr: expansion of a unique lymphocyte subset. J. Immunol. 129, 2612–2615 (1982).

    PubMed  Google Scholar 

  22. Kelley, V.E. & Roths, J.B. Interaction of mutant lpr gene with background strain influences renal disease. Clin. Immunol. Immunopathol. 37, 220–229 (1985).

    Article  CAS  Google Scholar 

  23. McKay, L.I. & Cidlowski, J.A. Molecular control of immune/inflammatory responses: interactions between nuclear factor-κB and steroid receptor-signaling pathways. Endocr. Rev. 20, 435–459 (1999).

    CAS  PubMed  Google Scholar 

  24. Overwijk, W.W. et al. Tumor regression and autoimmunity after reversal of a functionally tolerant state of self-reactive CD8+ T cells. J. Exp. Med. 198, 569–580 (2003).

    Article  CAS  Google Scholar 

  25. Le, L.Q. et al. Mice lacking the orphan G protein–coupled receptor G2A develop a late-onset autoimmune syndrome. Immunity 14, 561–571 (2001).

    Article  CAS  Google Scholar 

  26. Hamacher, K., Coenen, H.H. & Stocklin, G. Efficient stereospecific synthesis of no-carrier-added 2-[18F]-fluoro-2-deoxy-D-glucose using aminopolyether supported nucleophilic substitution. J. Nucl. Med. 27, 235–238 (1986).

    CAS  PubMed  Google Scholar 

  27. Mangner, T.J., Klecker, R.W., Anderson, L. & Shields, A.F. Synthesis of 2′-deoxy-2′-[18F]fluoro-β-D-arabinofuranosyl nucleosides, [18F]FAU, [18F]FMAU, [18F]FBAU and [18F]FIAU, as potential PET agents for imaging cellular proliferation. Synthesis of [18F]labelled FAU, FMAU, FBAU, FIAU. Nucl. Med. Biol. 30, 215–224 (2003).

    Article  CAS  Google Scholar 

  28. Qi, J., Leahy, R.M., Cherry, S.R., Chatziioannou, A. & Farquhar, T.H. High-resolution 3D Bayesian image reconstruction using the microPET small-animal scanner. Phys. Med. Biol. 43, 1001–1013 (1998).

    Article  CAS  Google Scholar 

  29. Chow, P.L., Stout, D.B., Komisopoulou, E. & Chatziioannou, A.F. A method of image registration for small animal, multi-modality imaging. Phys. Med. Biol. 51, 379–390 (2006).

    Article  Google Scholar 

  30. Loening, A.M. & Gambhir, S.S. AMIDE: a free software tool for multimodality medical image analysis. Mol. Imaging 2, 131–137 (2003).

    Article  Google Scholar 

Download references

Acknowledgements

We are grateful to D. Stout, W. Ladno and J. Edwards for microPET imaging and to the chemists and cyclotron group for production of PET probes. We thank G. Toy, M. Riedinger, S. Quan, D. Chen, J. Wengrod, D. Goldstein and A. Tran for outstanding technical assistance. We thank J. Lee for the analysis of the microarray data, H. Su for imaging of the U87 tumors, J. Liu and C. Shen for help with biochemical analyses, J. McLaughlin (UCLA) for providing leukemic animal models for imaging studies, and J. Czernin, H. Herschman and A. Ribas for insightful discussions. We also thank B. Anderson for help with preparing the manuscript. O.N.W. is an investigator of the Howard Hughes Medical Institute. C.G.R. was supported by In Vivo Cellular and Molecular Imaging Centers Developmental Project Award, grant NIH P50 CA86306 from the National Cancer Institute at the US National Institutes of Health, by US National Cancer Institute grant 5U54 CA119347 and by Juvenile Diabetes Research Foundation Award 17-2006-870. C.G.R. acknowledges unrestricted support from Merck Research Laboratories. C.J.S. was supported by a Fred Eiserling and Judith Lengyel Graduate Doctorate Fellowship. E.N.-G. was supported by the US National Institutes of Health T32 GM08042 UCLA Medical Scientist Training Program. This research was supported in part by US Department of Energy Contract DE-FG02-06ER64249 (M.E.P.), by US National Cancer Institute grant R24CA92865 and by funds from the Samuel Waxman Cancer Research Foundation and the W.M. Keck Foundation.

Author information

Authors and Affiliations

Authors

Contributions

C.G.R., C.J.S., E.N.-G., N.S., J.R.B., M.E.P. and O.N.W. designed research; C.G.R., C.J.S., E.N.-G., N.S. and S.M.S. performed research; C.G.R., C.J.S., E.N.-G., N.S., S.M.S. and O.N.W. analyzed data; C.G.R., C.J.S. and O.N.W. wrote the paper.

Corresponding authors

Correspondence to Caius G Radu or Owen N Witte.

Ethics declarations

Competing interests

Portions of the work covered in this manuscript have been disclosed to the University of California, Los Angeles Office of Intellectual Property Administration and included in patent applications to the US Patent Office.

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–8 and Supplementary Methods (PDF 2372 kb)

Supplementary Movie 1

[18F]FAC biodistribution during the first 3 min after injection. (MOV 1899 kb)

Supplementary Movie 2

[18F]FAC biodistribution 3–10 min after injection. (MOV 2195 kb)

Supplementary Movie 3

[18F]FAC biodistribution 10–30 min after injection. (MOV 2290 kb)

Supplementary Movie 4

[18F]FAC biodistribution 30–60 min after injection. (MOV 2008 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Radu, C., Shu, C., Nair-Gill, E. et al. Molecular imaging of lymphoid organs and immune activation by positron emission tomography with a new [18F]-labeled 2′-deoxycytidine analog. Nat Med 14, 783–788 (2008). https://doi.org/10.1038/nm1724

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm1724

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing