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.

  • Article
  • Published:

Targeted zwitterionic near-infrared fluorophores for improved optical imaging

Abstract

The signal-to-background ratio (SBR) is the key determinant of sensitivity, detectability and linearity in optical imaging. As signal strength is often constrained by fundamental limits, background reduction becomes an important approach for improving the SBR. We recently reported that a zwitterionic near-infrared (NIR) fluorophore, ZW800-1, exhibits low background. Here we show that this fluorophore provides a much-improved SBR when targeted to cancer cells or proteins by conjugation with a cyclic RGD peptide, fibrinogen or antibodies. ZW800-1 outperforms the commercially available NIR fluorophores IRDye800-CW and Cy5.5 in vitro for immunocytometry, histopathology and immunoblotting and in vivo for image-guided surgery. In tumor model systems, a tumor-to-background ratio of 17.2 is achieved at 4 h after injection of ZW800-1 conjugated to cRGD compared to ratios of 5.1 with IRDye800-CW and 2.7 with Cy5.5. Our results suggest that introducing zwitterionic properties into targeted fluorophores may be a general strategy for improving the SBR in diagnostic and therapeutic applications.

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

Access options

Buy this article

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

Figure 1: Targeted NIR fluorophores and an improved SBR during cell-based assays.
Figure 2: NIR fluorophore–conjugated antibodies and an improved SBR during histopathological analysis.
Figure 3: Improved in vivo optical imaging using zwitterionic, NIR-fluorescent, targeted small molecules and proteins.
Figure 4

Similar content being viewed by others

References

  1. Frangioni, J.V. In vivo near-infrared fluorescence imaging. Curr. Opin. Chem. Biol. 7, 626–634 (2003).

    Article  CAS  Google Scholar 

  2. Gioux, S., Choi, H.S. & Frangioni, J.V. Image-guided surgery using invisible near-infrared light: fundamentals of clinical translation. Mol. Imaging 9, 237–255 (2010).

    Article  CAS  Google Scholar 

  3. Te Velde, E.A., Veerman, T., Subramaniam, V. & Ruers, T. The use of fluorescent dyes and probes in surgical oncology. Eur. J. Surg. Oncol. 36, 6–15 (2010).

    Article  CAS  Google Scholar 

  4. Ballou, B. et al. Tumor labeling in vivo using cyanine-conjugated monoclonal antibodies. Cancer Immunol. Immunother. 41, 257–263 (1995).

    Article  CAS  Google Scholar 

  5. Ballou, B. et al. Cyanine fluorochrome-labeled antibodies in vivo: assessment of tumor imaging using Cy3, Cy5, Cy5.5, and Cy7. Cancer Detect. Prev. 22, 251–257 (1998).

    Article  CAS  Google Scholar 

  6. Ye, Y. & Chen, X. Integrin targeting for tumor optical imaging. Theranostics 1, 102–126 (2011).

    Article  CAS  Google Scholar 

  7. Tanaka, E. et al. Real-time intraoperative assessment of the extrahepatic bile ducts in rats and pigs using invisible near-infrared fluorescent light. Surgery 144, 39–48 (2008).

    Article  Google Scholar 

  8. Kobayashi, H., Ogawa, M., Alford, R., Choyke, P.L. & Urano, Y. New strategies for fluorescent probe design in medical diagnostic imaging. Chem. Rev. 110, 2620–2640 (2010).

    Article  CAS  Google Scholar 

  9. Tung, C.H., Bredow, S., Mahmood, U. & Weissleder, R. Preparation of a cathepsin D sensitive near-infrared fluorescence probe for imaging. Bioconjug. Chem. 10, 892–896 (1999).

    Article  CAS  Google Scholar 

  10. Achilefu, S. et al. Novel bioactive and stable neurotensin peptide analogues capable of delivering radiopharmaceuticals and molecular beacons to tumors. J. Med. Chem. 46, 3403–3411 (2003).

    Article  CAS  Google Scholar 

  11. Lee, S. et al. A near-infrared-fluorescence-quenched gold-nanoparticle imaging probe for in vivo drug screening and protease activity determination. Angew. Chem. Int. Edn Engl. 47, 2804–2807 (2008).

    Article  CAS  Google Scholar 

  12. Lee, S., Park, K., Kim, K., Choi, K. & Kwon, I.C. Activatable imaging probes with amplified fluorescent signals. Chem. Commun. (Camb.) 36, 4250–4260 (2008).

    Article  Google Scholar 

  13. Kobayashi, T. et al. Highly activatable and rapidly releasable caged fluorescein derivatives. J. Am. Chem. Soc. 129, 6696–6697 (2007).

    Article  CAS  Google Scholar 

  14. Urano, Y. et al. Selective molecular imaging of viable cancer cells with pH-activatable fluorescence probes. Nat. Med. 15, 104–109 (2009).

    Article  CAS  Google Scholar 

  15. Choi, H.S. et al. Renal clearance of quantum dots. Nat. Biotechnol. 25, 1165–1170 (2007).

    Article  CAS  Google Scholar 

  16. Choi, H.S. et al. Design considerations for tumour-targeted nanoparticles. Nat. Nanotechnol. 5, 42–47 (2010).

    Article  CAS  Google Scholar 

  17. Choi, H.S. et al. Rapid translocation of nanoparticles from the lung airspaces to the body. Nat. Biotechnol. 28, 1300–1303 (2010).

    Article  CAS  Google Scholar 

  18. Choi, H.S. & Frangioni, J.V. Nanoparticles for biomedical imaging: fundamentals of clinical translation. Mol. Imaging 9, 291–310 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Choi, H.S. et al. Synthesis and in vivo fate of zwitterionic near-infrared fluorophores. Angew. Chem. Int. Edn Engl. 50, 6258–6263 (2011).

    Article  CAS  Google Scholar 

  20. Hyun, H. et al. cGMP-compatible preparative scale synthesis of near-infrared fluorophores. Contrast Media Mol. Imaging 7, 516–524 (2012).

    Article  CAS  Google Scholar 

  21. Adams, J.C. & Watt, F.M. Expression of β1, β3, β4, and β5 integrins by human epidermal keratinocytes and non-differentiating keratinocytes. J. Cell Biol. 115, 829–841 (1991).

    Article  CAS  Google Scholar 

  22. Ohnishi, S., Garfein, E.S., Karp, S.J. & Frangioni, J.V. Radiolabeled and near-infrared fluorescent fibrinogen derivatives create a system for the identification and repair of obscure gastrointestinal bleeding. Surgery 140, 785–792 (2006).

    Article  Google Scholar 

  23. Colton, I.J., Carbeck, J.D., Rao, J. & Whitesides, G.M. Affinity capillary electrophoresis: a physical-organic tool for studying interactions in biomolecular recognition. Electrophoresis 19, 367–382 (1998).

    Article  CAS  Google Scholar 

  24. Gitlin, I., Gudiksen, K.L. & Whitesides, G.M. Effects of surface charge on denaturation of bovine carbonic anhydrase. ChemBioChem 7, 1241–1250 (2006).

    Article  CAS  Google Scholar 

  25. Bourré, L., Giuntini, F., Eggleston, I.M., Wilson, M. & MacRobert, A.J. Protoporphyrin IX enhancement by 5-aminolaevulinic acid peptide derivatives and the effect of RNA silencing on intracellular metabolism. Br. J. Cancer 100, 723–731 (2009).

    Article  Google Scholar 

  26. Zwaal, R.F., Comfurius, P. & van Deenen, L.L. Membrane asymmetry and blood coagulation. Nature 268, 358–360 (1977).

    Article  CAS  Google Scholar 

  27. Frangioni, J.V. New technologies for human cancer imaging. J. Clin. Oncol. 26, 4012–4021 (2008).

    Article  Google Scholar 

  28. Rasmussen, F. Renal clearance: species differences and similarities. Vet. Res. Commun. 7, 301–306 (1983).

    Article  CAS  Google Scholar 

  29. Reagan-Shaw, S., Nihal, M. & Ahmad, N. Dose translation from animal to human studies revisited. FASEB J. 22, 659–661 (2008).

    Article  CAS  Google Scholar 

  30. Kelley, K.W., Curtis, S.E., Marzan, G.T., Karara, H.M. & Anderson, C.R. Body surface area of female swine. J. Anim. Sci. 36, 927–930 (1973).

    Article  CAS  Google Scholar 

  31. Humblet, V., Misra, P. & Frangioni, J.V. An HPLC/mass spectrometry platform for the development of multimodality contrast agents and targeted therapeutics: prostate-specific membrane antigen small molecule derivatives. Contrast Media Mol. Imaging 1, 196–211 (2006).

    Article  CAS  Google Scholar 

  32. Chen, X., Conti, P.S. & Moats, R.A. In vivo near-infrared fluorescence imaging of integrin αvβ3 in brain tumor xenografts. Cancer Res. 64, 8009–8014 (2004).

    Article  CAS  Google Scholar 

  33. Troyan, S.L. et al. The FLARE™ intraoperative near-infrared fluorescence imaging system: a first-in-human clinical trial in breast cancer sentinel lymph node mapping. Ann. Surg. Oncol. 16, 2943–2952 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

We thank S. Gioux, R. Oketokoun and A. Stockdale for assistance with development of the FLARE (Fluorescence-Assisted Resection and Exploration) imaging system and software. We also thank D. Burrington Jr. for manuscript editing and E. Trabucchi for administrative assistance. This study was supported by the following grants from the US National Institutes of Health: National Cancer Institute Bioengineering Research Partnership grant #R01-CA-115296 (J.V.F.) and National Institute of Biomedical Imaging and Bioengineering grants #R01-EB-010022 and #R01-EB-011523 (both to H.S.C. and J.V.F.); this study was also supported by the Dana Foundation Program in Brain and Immuno-Imaging (H.S.C.). S.H.K. was supported by a WCU Program (R31-20029) from the Korea Ministry of Education, Science and Technology (KMEST).

Author information

Authors and Affiliations

Authors

Contributions

H.S.C., S.L.G., S.H.K., Y.A., J.H.L., H.H., G.P., Y.X., F.L., S.B. and M.H. performed the experiments. H.S.C., S.L.G. and J.V.F. reviewed, analyzed and interpreted the data. H.S.C., S.L.G. and J.V.F. wrote the paper. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to John V Frangioni.

Ethics declarations

Competing interests

FLARE technology is owned by Beth Israel Deaconess Medical Center, a teaching hospital of Harvard Medical School. It has been licensed to the FLARE Foundation, a nonprofit organization focused on promoting the dissemination of medical imaging technology for research and clinical use. J.V.F. is the founder and chairman of the FLARE Foundation. The Beth Israel Deaconess Medical Center will receive royalties for the sale of FLARE Technology. J.V.F. has elected to surrender post-market royalties to which he would otherwise be entitled as inventor, and has elected to donate pre-market proceeds to the FLARE Foundation. J.V.F. has started three for-profit companies, Curadel, Curadel Medical Devices and Curadel In Vivo Diagnostics, which may someday be nonexclusive sublicensees of FLARE technology.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6, Supplementary Methods and Supplementary Table 1 (PDF 2382 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Choi, H., Gibbs, S., Lee, J. et al. Targeted zwitterionic near-infrared fluorophores for improved optical imaging. Nat Biotechnol 31, 148–153 (2013). https://doi.org/10.1038/nbt.2468

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2468

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer