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:

Selective molecular imaging of viable cancer cells with pH-activatable fluorescence probes

Abstract

A long-term goal of cancer diagnosis is to develop tumor-imaging techniques that have sufficient specificity and sensitivity. To achieve this goal, minimizing the background signal originating from nontarget tissues is crucial. Here we achieve highly specific in vivo cancer visualization by using a newly designed targeted 'activatable' fluorescent imaging probe. This agent is activated after cellular internalization by sensing the pH change in the lysosome. Novel acidic pH–activatable probes based on the boron-dipyrromethene fluorophore were synthesized and then conjugated to a cancer-targeting monoclonal antibody. As proof of concept, ex vivo and in vivo imaging of human epidermal growth factor receptor type 2–positive lung cancer cells in mice was performed. The probe was highly specific for tumors with minimal background signal. Furthermore, because the acidic pH in lysosomes is maintained by the energy-consuming proton pump, only viable cancer cells were successfully visualized. The design concept can be widely adapted to cancer-specific, cell surface–targeting molecules that result in cellular internalization.

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: Mechanism of a cancer imaging strategy using target-specific activatable probes.
Figure 2: Development of a series of fluorescence probes for various acidic environments.
Figure 3: In vitro imaging of NIH3T3 HER2+ cells with Ph-BDP–, DiMeN-BDP–, EtMeN-BDP– or DiEtN-BDP–labeled trastuzumab.
Figure 4: The activatable probe can produce a fluorescence signal only from HER2-positive tumors and not from HER2-negative tumors, with minimal background from normal tissue.
Figure 5: The activatable probe can produce a fluorescence signal only from living HER2-positive cells and tumors and not from dead cells or fixed tumors.

Similar content being viewed by others

References

  1. Yamamoto, N. et al. Cellular dynamics visualized in live cells in vitro and in vivo by differential dual-color nuclear-cytoplasmic fluorescent-protein expression. Cancer Res. 64, 4251–4256 (2004).

    Article  CAS  Google Scholar 

  2. Yamauchi, K. et al. Induction of cancer metastasis by cyclophosphamide pretreatment of host mice: an opposite effect of chemotherapy. Cancer Res. 68, 516–520 (2008).

    Article  CAS  Google Scholar 

  3. Yang, M., Jiang, P. & Hoffman, R.M. Whole-body subcellular multicolor imaging of tumor-host interaction and drug response in real time. Cancer Res. 67, 5195–5200 (2007).

    Article  CAS  Google Scholar 

  4. Wu, A.M. & Senter, P.D. Arming antibodies: prospects and challenges for immunoconjugates. Nat. Biotechnol. 23, 1137–1146 (2005).

    Article  CAS  Google Scholar 

  5. Yarden, Y. The EGFR family and its ligands in human cancer. signalling mechanisms and therapeutic opportunities. Eur. J. Cancer 37, Suppl 4, S3–S8 (2001).

    Article  CAS  Google Scholar 

  6. Miura, T. et al. Rational design principle for modulating fluorescence properties of fluorescein-based probes by photoinduced electron transfer. J. Am. Chem. Soc. 125, 8666–8671 (2003).

    Article  CAS  Google Scholar 

  7. Ueno, T. et al. Rational principles for modulating fluorescence properties of fluorescein. J. Am. Chem. Soc. 126, 14079–14085 (2004).

    Article  CAS  Google Scholar 

  8. Tanaka, K. et al. Rational design of fluorescein-based fluorescence probes. Mechanism-based design of a maximum fluorescence probe for singlet oxygen. J. Am. Chem. Soc. 123, 2530–2536 (2001).

    Article  CAS  Google Scholar 

  9. Urano, Y. et al. Evolution of fluorescein as a platform for finely tunable fluorescence probes. J. Am. Chem. Soc. 127, 4888–4894 (2005).

    Article  CAS  Google Scholar 

  10. Gabe, Y., Urano, Y., Kikuchi, K., Kojima, H. & Nagano, T. Highly sensitive fluorescence probes for nitric oxide based on boron dipyrromethene chromophore - rational design of potentially useful bioimaging fluorescence probe. J. Am. Chem. Soc. 126, 3357–3367 (2004).

    Article  CAS  Google Scholar 

  11. Queen, C. et al. A humanized antibody that binds to the interleukin 2 receptor. Proc. Natl. Acad. Sci. USA 86, 10029–10033 (1989).

    Article  CAS  Google Scholar 

  12. Minta, A., Kao, J.P.Y. & Tsien, R.Y. Fluorescent indicators for cytosolic calcium based on rhodamine and fluorescein chromophores. J. Biol. Chem. 264, 8171–8178 (1989).

    CAS  PubMed  Google Scholar 

  13. Weissleder, R., Tung, C.H., Mahmood, U. & Bogdanov, A. Jr. In vivo imaging of tumors with proteaseactivated near-infrared fluorescent probes. Nat. Biotechnol. 17, 375–378 (1999).

    Article  CAS  Google Scholar 

  14. Maeda, H., Wu, J., Sawa, T., Matsumura, Y. & Hori, K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J. Control. Release 65, 271–284 (2000).

    Article  CAS  Google Scholar 

  15. Paradiso, A.M., Tsien, R.Y. & Machen, T.E. Na+-H+ exchange in gastric glands as measured with a cytoplasmic-trapped, fluorescent ph indicator. Proc. Natl. Acad. Sci. USA 81, 7436–7440 (1984).

    Article  CAS  Google Scholar 

  16. de Silva, A.P. et al. Signaling recognition events with fluorescent sensors and switches. Chem. Rev. 97, 1515–1566 (1997).

    Article  CAS  Google Scholar 

  17. Kollmannsberger, M., Rurack, K., Resch-Genger, U. & Daub, J. Ultrafast charge transfer in amino-substituted boron dipyrromethene dyes and its inhibition by cation complexation: a new design concept for highly sensitive fluorescent probes. J. Phys. Chem. A 102, 10211–10220 (1998).

    Article  CAS  Google Scholar 

  18. Adie, E.J. et al. A pH-sensitive fluor, CypHer™ 5, used to monitor agonist-induced G protein-coupled receptor internalization in live cells. Biotechniques 33, 1152–1154 (2002).

    Article  CAS  Google Scholar 

  19. Nwokolo, C.U., Payne-James, J.J., Silk, D.B., Misiewicz, J.J. & Loft, D.E. Palliation of malignant dysphagia by ethanol induced tumour necrosis. Gut 35, 299–303 (1994).

    Article  CAS  Google Scholar 

  20. Hama, Y. et al. A target-cell specific activatable fluorescence probe for in vivo molecular imaging of cancer based on a self-quenched avidin-rhodamine conjugate. Cancer Res. 67, 2791–2799 (2007).

    Article  CAS  Google Scholar 

  21. Kamiya, M. et al. An enzymatically activated fluorescence probe for targeted tumor imaging. J. Am. Chem. Soc. 129, 3918–3929 (2007).

    Article  CAS  Google Scholar 

  22. Hoffman, R.M. The multiple uses of fluorescent proteins to visualize cancer in vivo. Nat. Rev. Cancer 5, 796–806 (2005).

    Article  CAS  Google Scholar 

  23. Baselga, J., Norton, L., Albanell, J., Kim, Y.M. & Mendelsohn, J. Recombinant humanized anti-HER2 antibody (Herceptin) enhances the antitumor activity of paclitaxel and doxorubicin against HER2/neu overexpressing human breast cancer xenografts. Cancer Res. 58, 2825–2831 (1998).

    CAS  PubMed  Google Scholar 

  24. Gancberg, D. et al. Comparison of HER-2 status between primary breast cancer and corresponding distant metastatic sites. Ann. Oncol. 13, 1036–1043 (2002).

    Article  CAS  Google Scholar 

  25. Zidan, J. et al. Comparison of HER-2 overexpression in primary breast cancer and metastatic sites and its effect on biological targeting therapy of metastatic disease. Br. J. Cancer 93, 552–556 (2005).

    Article  CAS  Google Scholar 

  26. Mandler, R., Kobayashi, H., Hinson, E.R., Brechbiel, M.W. & Waldmann, T.A. Herceptin-geldanamycin immunoconjugates: pharmacokinetics, biodistribution, and enhanced antitumor activity. Cancer Res. 64, 1460–1467 (2004).

    Article  CAS  Google Scholar 

  27. Hama, Y., Urano, Y., Koyama, Y., Choyke, P.L. & Kobayashi, H. D-galactose receptor–targeted in vivo spectral fluorescence imaging of peritoneal metastasis using galactosamin-conjugated serum albumin-rhodamine green. J. Biomed. Opt. 12, 051501 (2007).

    Article  Google Scholar 

  28. Alencar, H. et al. Colonic adenocarcinomas: near-infrared microcatheter imaging of smart probes for early detection—study in mice. Radiology 244, 232–238 (2007).

    Article  Google Scholar 

  29. De Grand, A.M. & Frangioni, J.V. An operational near-infrared fluorescence imaging system prototype for large animal surgery. Technol. Cancer Res. Treat. 2, 553–562 (2003).

    Article  CAS  Google Scholar 

  30. Parker, C.A. & Rees, W.T. Correction of fluorescence spectra and measurement of fluorescence quantum efficiency. Analyst 85, 587–600 (1960).

    Article  CAS  Google Scholar 

  31. Koyama, Y., Hama, Y., Urano, Y., Nguyen, D.M., Choyke, P.L. & Kobayashi, H. Spectral fluorescence molecular imaging of lung metastases targeting HER2/neu. Clin. Cancer Res. 13, 2936–2945 (2007).

    Article  CAS  Google Scholar 

  32. Levenson, R.M. & Mansfield, J.R. Multispectral imaging in biology and medicine: slices of life. Cytometry A. 69, 748–758 (2006).

    Article  Google Scholar 

Download references

Acknowledgements

This study was supported in part by the Precursory Research for Embryonic Sciences and Technology from the Japan Science and Technology Agency, by research grants 19021010 and 19205021 to Y.U. and by the Intramural Research Program of the US National Institutes of Health, National Cancer Institute, Center for Cancer Research to Y.H., Y.K., T.B., P.L.C. and H.K.

Author information

Authors and Affiliations

Authors

Contributions

Y.U. and H.K. planned the projects, developed the probes, performed the in vivo experiments and wrote and edited the manuscript. D.A. and M.K. developed the probes, performed the in vivo experiments and wrote and edited the manuscript. Y.H., Y.K., T.B., T.W. and A.H. performed the in vivo experiments and wrote and edited the manuscript. T.N. and P.L.C. planned the projects and wrote and edited the manuscript.

Corresponding authors

Correspondence to Yasuteru Urano or Hisataka Kobayashi.

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–4, Supplementary Tables 1–3 and Supplementary Methods (PDF 563 kb)

Supplementary Video 1

Reversible activation by altering the pH. (AVI 4293 kb)

Supplementary Video 2

Fluorescence-guided Laparoscope (MOV 12792 kb)

Supplementary Video 3

Fluorescence-guided Laparoscope (MOV 5792 kb)

Supplementary Video 4

Fluorescence-guided Laparoscope (MOV 8635 kb)

Supplementary Video 5

pH-activatable rhodamine (MOV 2877 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Urano, Y., Asanuma, D., Hama, Y. et al. Selective molecular imaging of viable cancer cells with pH-activatable fluorescence probes. Nat Med 15, 104–109 (2009). https://doi.org/10.1038/nm.1854

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.1854

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