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:

Selective in vivo metabolic cell-labeling-mediated cancer targeting

Abstract

Distinguishing cancer cells from normal cells through surface receptors is vital for cancer diagnosis and targeted therapy. Metabolic glycoengineering of unnatural sugars provides a powerful tool to manually introduce chemical receptors onto the cell surface; however, cancer-selective labeling still remains a great challenge. Herein we report the design of sugars that can selectively label cancer cells both in vitro and in vivo. Specifically, we inhibit the cell-labeling activity of tetraacetyl-N-azidoacetylmannosamine (Ac4ManAz) by converting its anomeric acetyl group to a caged ether bond that can be selectively cleaved by cancer-overexpressed enzymes and thus enables the overexpression of azido groups on the surface of cancer cells. Histone deacetylase and cathepsin L-responsive acetylated azidomannosamine, one such enzymatically activatable Ac4ManAz analog developed, mediated cancer-selective labeling in vivo, which enhanced tumor accumulation of a dibenzocyclooctyne–doxorubicin conjugate via click chemistry and enabled targeted therapy against LS174T colon cancer, MDA-MB-231 triple-negative breast cancer and 4T1 metastatic breast cancer in mice.

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: Design of sugars for controlled metabolic labeling.
Figure 2: Replacing the C1–OAc of Ac4ManAz with a cleavable ether bond–enabled controlled metabolic cell labeling in vitro and in vivo.
Figure 3: DCL-AAM-mediated cancer-selective labeling in vitro.
Figure 4: DCL-AAM mediated cancer-selective labeling in vivo.
Figure 5: DCL-AAM-mediated tumor labeling improved antitumor efficacy of DBCO–drug conjugate against primary LS174T colon tumor and MDA-MB-231 triple-negative breast tumor models.
Figure 6: DCL-AAM-mediated tumor labeling improved anticancer efficacy of DBCO–drug conjugate against 4T1 lung metastases.

Similar content being viewed by others

References

  1. Hynes, R.O. Integrins: a family of cell surface receptors. Cell 48, 549–554 (1987).

    Article  CAS  PubMed  Google Scholar 

  2. Aruffo, A., Stamenkovic, I., Melnick, M., Underhill, C.B. & Seed, B. CD44 is the principal cell surface receptor for hyaluronate. Cell 61, 1303–1313 (1990).

    Article  CAS  PubMed  Google Scholar 

  3. Wright, S.D., Ramos, R.A., Tobias, P.S., Ulevitch, R.J. & Mathison, J.C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 249, 1431–1433 (1990).

    Article  CAS  PubMed  Google Scholar 

  4. Nusser, Z. et al. Cell type and pathway dependence of synaptic AMPA receptor number and variability in the hippocampus. Neuron 21, 545–559 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Cull-Candy, S., Brickley, S. & Farrant, M. NMDA receptor subunits: diversity, development and disease. Curr. Opin. Neurobiol. 11, 327–335 (2001).

    Article  CAS  PubMed  Google Scholar 

  6. Raff, M.C. et al. Cell-type-specific markers for distinguishing and studying neurons and the major classes of glial cells in culture. Brain Res. 174, 283–308 (1979).

    Article  CAS  PubMed  Google Scholar 

  7. Liaw, D. et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat. Genet. 16, 64–67 (1997).

    Article  CAS  PubMed  Google Scholar 

  8. Mombaerts, P. et al. Spontaneous development of inflammatory bowel disease in T cell receptor mutant mice. Cell 75, 274–282 (1993).

    Article  CAS  PubMed  Google Scholar 

  9. Puffenberger, E.G. et al. A missense mutation of the endothelin-B receptor gene in multigenic Hirschsprung's disease. Cell 79, 1257–1266 (1994).

    Article  CAS  PubMed  Google Scholar 

  10. Slamon, D.J. et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 344, 783–792 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Vogel, C.L. et al. Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J. Clin. Oncol. 20, 719–726 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Romond, E.H. et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N. Engl. J. Med. 353, 1673–1684 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. Tang, L. et al. Aptamer-functionalized, ultra-small, monodisperse silica nanoconjugates for targeted dual-modal imaging of lymph nodes with metastatic tumors. Angew. Chem. Int. Ed. Engl. 51, 12721–12726 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Keefe, A.D., Pai, S. & Ellington, A. Aptamers as therapeutics. Nat. Rev. Drug Discov. 9, 537–550 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Santra, S., Kaittanis, C., Santiesteban, O.J. & Perez, J.M. Cell-specific, activatable, and theranostic prodrug for dual-targeted cancer imaging and therapy. J. Am. Chem. Soc. 133, 16680–16688 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Slamon, D.J. et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244, 707–712 (1989).

    Article  CAS  PubMed  Google Scholar 

  17. Goldhirsch, A. et al. 2 years versus 1 year of adjuvant trastuzumab for HER2-positive breast cancer (HERA): an open-label, randomised controlled trial. Lancet 382, 1021–1028 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Hudziak, R.M., Schlessinger, J. & Ullrich, A. Increased expression of the putative growth factor receptor p185HER2 causes transformation and tumorigenesis of NIH 3T3 cells. Proc. Natl. Acad. Sci. USA 84, 7159–7163 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hartley, J.L., Temple, G.F. & Brasch, M.A. DNA cloning using in vitro site-specific recombination. Genome Res. 10, 1788–1795 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Prescher, J.A., Dube, D.H. & Bertozzi, C.R. Chemical remodelling of cell surfaces in living animals. Nature 430, 873–877 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Prescher, J.A. & Bertozzi, C.R. Chemistry in living systems. Nat. Chem. Biol. 1, 13–21 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Lee, S. et al. Chemical tumor-targeting of nanoparticles based on metabolic glycoengineering and click chemistry. ACS Nano 8, 2048–2063 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Koo, H. et al. Bioorthogonal copper-free click chemistry in vivo for tumor-targeted delivery of nanoparticles. Angew. Chem. Int. Ed. Engl. 51, 11836–11840 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Xie, R., Hong, S., Feng, L., Rong, J. & Chen, X. Cell-selective metabolic glycan labeling based on ligand-targeted liposomes. J. Am. Chem. Soc. 134, 9914–9917 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Xie, R. et al. Targeted imaging and proteomic analysis of tumor-associated glycans in living animals. Angew. Chem. Int. Ed. Engl. 53, 14082–14086 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Xie, R. et al. In vivo metabolic labeling of sialoglycans in the mouse brain by using a liposome-assisted bioorthogonal reporter strategy. Proc. Natl. Acad. Sci. USA 113, 5173–5178 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Saks, M.E. et al. An engineered Tetrahymena tRNAGln for in vivo incorporation of unnatural amino acids into proteins by nonsense suppression. J. Biol. Chem. 271, 23169–23175 (1996).

    Article  CAS  PubMed  Google Scholar 

  28. Liu, W., Brock, A., Chen, S., Chen, S. & Schultz, P.G. Genetic incorporation of unnatural amino acids into proteins in mammalian cells. Nat. Methods 4, 239–244 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Link, A.J., Mock, M.L. & Tirrell, D.A. Non-canonical amino acids in protein engineering. Curr. Opin. Biotechnol. 14, 603–609 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Rodriguez, E.A., Lester, H.A. & Dougherty, D.A. In vivo incorporation of multiple unnatural amino acids through nonsense and frameshift suppression. Proc. Natl. Acad. Sci. USA 103, 8650–8655 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Laughlin, S.T. & Bertozzi, C.R. Metabolic labeling of glycans with azido sugars and subsequent glycan-profiling and visualization via Staudinger ligation. Nat. Protoc. 2, 2930–2944 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Breidenbach, M.A. et al. Targeted metabolic labeling of yeast N-glycans with unnatural sugars. Proc. Natl. Acad. Sci. USA 107, 3988–3993 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Luchansky, S.J. et al. Constructing azide-labeled cell surfaces using polysaccharide biosynthetic pathways. Methods Enzymol. 362, 249–272 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Chang, P.V., Dube, D.H., Sletten, E.M. & Bertozzi, C.R. A strategy for the selective imaging of glycans using caged metabolic precursors. J. Am. Chem. Soc. 132, 9516–9518 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hao, J., Vann, W.F., Hinderlich, S. & Sundaramoorthy, M. Elimination of 2-keto-3-deoxy-D-glycero-D-galacto-nonulosonic acid 9-phosphate synthase activity from human N-acetylneuraminic acid 9-phosphate synthase by a single mutation. Biochem. J. 397, 195–201 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gunawan, J. et al. Structural and mechanistic analysis of sialic acid synthase NeuB from Neisseria meningitidis in complex with Mn2+, phosphoenolpyruvate, and N-acetylmannosaminitol. J. Biol. Chem. 280, 3555–3563 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Il'ichev, Y.V., Schwörer, M.A. & Wirz, J. Photochemical reaction mechanisms of 2-nitrobenzyl compounds: methyl ethers and caged ATP. J. Am. Chem. Soc. 126, 4581–4595 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Chauhan, S.S., Goldstein, L.J. & Gottesman, M.M. Expression of cathepsin L in human tumors. Cancer Res. 51, 1478–1481 (1991).

    CAS  PubMed  Google Scholar 

  39. Witt, O., Deubzer, H.E., Milde, T. & Oehme, I. HDAC family: what are the cancer relevant targets? Cancer Lett. 277, 8–21 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Ueki, N., Lee, S., Sampson, N.S. & Hayman, M.J. Selective cancer targeting with prodrugs activated by histone deacetylases and a tumour-associated protease. Nat. Commun. 4, 2735 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Bonfils, C. et al. Evaluation of the pharmacodynamic effects of MGCD0103 from preclinical models to human using a novel HDAC enzyme assay. Clin. Cancer Res. 14, 3441–3449 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wegener, D., Wirsching, F., Riester, D. & Schwienhorst, A. A fluorogenic histone deacetylase assay well suited for high-throughput activity screening. Chem. Biol. 10, 61–68 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Lodish, H. et al. in Molecular Cell Biology 4th edn. Ch. 20.2 (W.H. Freeman, 2000).

  44. Malinoff, H.L. & Wicha, M.S. Isolation of a cell surface receptor protein for laminin from murine fibrosarcoma cells. J. Cell Biol. 96, 1475–1479 (1983).

    Article  CAS  PubMed  Google Scholar 

  45. Dent, R. et al. Triple-negative breast cancer: clinical features and patterns of recurrence. Clin. Cancer Res. 13, 4429–4434 (2007).

    Article  PubMed  Google Scholar 

  46. Foulkes, W.D., Smith, I.E. & Reis-Filho, J.S. Triple-negative breast cancer. N. Engl. J. Med. 363, 1938–1948 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Steeg, P.S. Tumor metastasis: mechanistic insights and clinical challenges. Nat. Med. 12, 895–904 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Yan, S., Sameni, M. & Sloane, B.F. Cathepsin B and human tumor progression. Biol. Chem. 379, 113–123 (1998).

    CAS  PubMed  Google Scholar 

  49. Deller, M.C. & Yvonne Jones, E. Cell surface receptors. Curr. Opin. Struct. Biol. 10, 213–219 (2000).

    Article  CAS  PubMed  Google Scholar 

  50. Lehmann, B.D. et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 121, 2750–2767 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dubowchik, G.M. et al. Cathepsin B-labile dipeptide linkers for lysosomal release of doxorubicin from internalizing immunoconjugates: model studies of enzymatic drug release and antigen-specific in vitro anticancer activity. Bioconjug. Chem. 13, 855–869 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Tang, L. et al. Investigating the optimal size of anticancer nanomedicine. Proc. Natl. Acad. Sci. USA 111, 15344–15349 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

J.C. acknowledges support from the United States National Institute of Health (Director's New Innovator Award 1DP2OD007246), which partially supported the in vivo part of the research, and National Science Foundation (DMR 1309525), which partially supported the chemical design and synthesis of the work. L.Y. acknowledges the support from the National Natural Science Foundation of China (51403145 and 51573123), the Ministry of Science and Technology of China (2016YFA0201200), the Collaborative Innovation Center of Suzhou Nano Science and Technology, and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). X.C. acknowledges the support from the National Natural Science Foundation of China (51528303). H.W. was supported via a Howard Hughes Medical Institute International Student Research Fellowship. K.C. and Q.Y. acknowledge Beckman Institute Graduate Fellowship support at the University of Illinois at Urbana–Champaign. K.C., Q.Y., and L.T. acknowledge support from the NIH National Cancer Institute Alliance for Nanotechnology in Cancer “Midwest Cancer Nanotechnology Training Center” Grant R25 CA154015A. R.W. acknowledges the support of a CSTAR/T32 Fellowship through the NIH T32 Tissue Microenvironment Training Program. We acknowledge R. Tong and V. Mirshafiee for their early work related to the design of this project. We also acknowledge H. Ying and Y. Zhang for their useful discussions.

Author information

Authors and Affiliations

Authors

Contributions

J.C. conceived the original concept of the two-step targeting strategy and supervised the entire project. J.C., H.W., Q.Y. and L.T. initiated this project. H.W. demonstrated the controlled labeling strategy using ether-caged Ac3ManAz derivatives and designed DCL-AAM under the supervision of J.C. H.W. performed the initial synthesis of DCL-AAM. R.W. and K.C. enabled the synthesis of high-purity DCL-AAM. H.W. performed the experiments of flow cytometry and confocal imaging with the help of Y.L. and J.Y. H.W., M.X., Y.S., X.Z. and E.J.C. designed and performed in vivo imaging studies under the supervision of J.C., I.T.D., L.W.D. and S.A.B. H.W., H.H., R.W., Z.W. and K.C. designed and performed tumor efficacy studies under the supervision of J.C., L.Y., X.C., S.L. and T.M.F. H.W., J.C., L.Y., S.L., T.M.F. and X.C. analyzed data. L.Y., X.C., T.M.F. and S.L. provided other expertise and critical feedback. H.W., J.C., L.Y., S.L. and T.M.F. wrote the paper with input from other authors.

Corresponding authors

Correspondence to Xuesi Chen, Lichen Yin or Jianjun Cheng.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results and Supplementary Figures 1–17. (PDF 7431 kb)

Supplementary Note

Synthetic Procedures. (PDF 697 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, H., Wang, R., Cai, K. et al. Selective in vivo metabolic cell-labeling-mediated cancer targeting. Nat Chem Biol 13, 415–424 (2017). https://doi.org/10.1038/nchembio.2297

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.2297

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