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.

Tracking the expression of therapeutic protein targets in rare cells by antibody-mediated nanoparticle labelling and magnetic sorting

Abstract

Molecular-level features of tumours can be tracked using single-cell analyses of circulating tumour cells (CTCs). However, single-cell measurements of protein expression for rare CTCs are hampered by the presence of a large number of non-target cells. Here, we show that antibody-mediated labelling of intracellular proteins in the nucleus, mitochondria and cytoplasm of human cells with magnetic nanoparticles enables analysis of target proteins at the single-cell level by sorting the cells according to their nanoparticle content in a microfluidic device with cell-capture zones sandwiched between arrays of magnets. We used the magnetic labelling and cell-sorting approach to track the expression of therapeutic protein targets in CTCs isolated from blood samples of mice with orthotopic prostate xenografts and from patients with metastatic castration-resistant prostate cancer. We also show that mutated proteins that are drug targets or markers of therapeutic response can be directly identified in CTCs, analysed at the single-cell level and used to predict how mice with drug-susceptible and drug-resistant pancreatic tumour xenografts respond to therapy.

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

Fig. 1: The single-cell intracellular protein analysis approach.
Fig. 2: Intracellular protein analysis and the sensitivity of the approach.
Fig. 3: Analysis of clinically relevant intracellular proteins.
Fig. 4: Analysis of c-Myc and vimentin in xenografts and clinical samples.
Fig. 5: Analysis of mutated proteins relevant for therapeutic selection.

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. The raw and analysed datasets generated during the study are too large to be publicly shared, but are available for research purposes from the corresponding authors on reasonable request.

References

  1. Alix-Panabieres, C. & Pantel, K. Challenges in circulating tumour cell research. Nat. Rev. Cancer 14, 623–631 (2014).

    CAS  PubMed  Google Scholar 

  2. Nagrath, S. et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 450, 1235–1239 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Yoon, H. J. et al. Sensitive capture of circulating tumour cells by functionalized graphene oxide nanosheets. Nat. Nanotechnol. 8, 735–741 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhao, W. et al. Bioinspired multivalent DNA network for capture and release of cells. Proc. Natl Acad. Sci. USA 109, 19626–19631 (2012).

    CAS  PubMed  Google Scholar 

  5. Stott, S. L. et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc. Natl Acad. Sci. USA 107, 18392–18397 (2010).

    CAS  PubMed  Google Scholar 

  6. Ozkumur, E. et al. Inertial focusing for tumor antigen-dependent and -independent sorting of rare circulating tumor cells. Sci. Transl. Med. 5, 179ra147 (2013).

    Google Scholar 

  7. Lee, A. et al. All-in-one centrifugal microfluidic device for size-selective circulating tumor cell isolation with high purity. Anal. Chem. 86, 11349–11356 (2014).

    CAS  PubMed  Google Scholar 

  8. Kim, T. H. et al. FAST: size-selective, clog-free isolation of rare cancer cells from whole blood at a liquid–liquid interface. Anal. Chem. 89, 1155–1162 (2017).

    CAS  PubMed  Google Scholar 

  9. Adams, A. A. et al. Highly efficient circulating tumor cell isolation from whole blood and label-free enumeration using polymer-based microfluidics with an integrated conductivity sensor. J. Am. Chem. Soc. 130, 8633–8641 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhang, Y., Zhou, L. & Qin, L. High-throughput 3D cell invasion chip enables accurate cancer metastatic assays. J. Am. Chem. Soc. 136, 15257–15262 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, Y., Zhang, W. & Qin, L. Mesenchymal-mode migration assay and antimetastatic drug screening with high-throughput microfluidic channel networks. Angew. Chem. Int. Ed. 53, 2344–2348 (2014).

    CAS  Google Scholar 

  12. Toriello, N. M. et al. Integrated microfluidic bioprocessor for single-cell gene expression analysis. Proc. Natl Acad. Sci. USA 105, 20173–20178 (2008).

    CAS  PubMed  Google Scholar 

  13. Reyes, E. E. et al. Quantitative characterization of androgen receptor protein expression and cellular localization in circulating tumor cells from patients with metastatic castration-resistant prostate cancer. J. Transl. Med. 12, 313 (2014).

    PubMed  PubMed Central  Google Scholar 

  14. Ciaccio, M. F., Wagner, J. P., Chuu, C. P., Lauffenburger, D. A. & Jones, R. B. Systems analysis of EGF receptor signaling dynamics with microwestern arrays. Nat. Methods 7, 148–155 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Rimm, D. L. What brown cannot do for you. Nat. Biotechnol. 24, 914–916 (2006).

    CAS  PubMed  Google Scholar 

  16. Liu, Y. et al. Modulation of fluorescent protein chromophores to detect protein aggregation with turn-on fluorescence. J. Am. Chem. Soc. 140, 7381–7384 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Pollock, S. B. et al. Highly multiplexed and quantitative cell-surface protein profiling using genetically barcoded antibodies. Proc. Natl Acad. Sci. USA 115, 2836–2841 (2018).

    CAS  PubMed  Google Scholar 

  18. Engelen, W., Meijer, L. H., Somers, B., de Greef, T. F. & Merkx, M. Antibody-controlled actuation of DNA-based molecular circuits. Nat. Commun. 8, 14473 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Fan, R. et al. Integrated barcode chips for rapid, multiplexed analysis of proteins in microliter quantities of blood. Nat. Biotechnol. 26, 1373–1378 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Nam, J. M., Thaxton, C. S. & Mirkin, C. A. Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins. Science 301, 1884–1886 (2003).

    CAS  PubMed  Google Scholar 

  21. Angelo, M. et al. Multiplexed ion beam imaging of human breast tumors. Nat. Med. 20, 436–442 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Rissin, D. M. et al. Single-molecule enzyme-linked immunosorbent assay detects serum proteins at subfemtomolar concentrations. Nat. Biotechnol. 28, 595–599 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. de la Rica, R. & Stevens, M. M. Plasmonic ELISA for the ultrasensitive detection of disease biomarkers with the naked eye. Nat. Nanotechnol. 7, 821–824 (2012).

    PubMed  Google Scholar 

  24. Xue, M. et al. Chemical methods for the simultaneous quantitation of metabolites and proteins from single cells. J. Am. Chem. Soc. 137, 4066–4069 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Gerdtsson, T. et al. Multiplex protein detection on circulating tumor cells from liquid biopsies using imaging mass cytometry. Converg. Sci. Phys. Oncol. 4, 015002 (2018).

    PubMed  PubMed Central  Google Scholar 

  26. Hughes, A. J. et al. Single-cell western blotting. Nat. Methods 11, 749–755 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Sinkala, E. et al. Profiling protein expression in circulating tumour cells using microfluidic western blotting. Nat. Commun. 8, 14622 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Poudineh, M. et al. Tracking the dynamics of circulating tumour cell phenotypes using nanoparticle-mediated magnetic ranking. Nat. Nanotechnol. 12, 274–281 (2017).

    CAS  PubMed  Google Scholar 

  29. Labib, M. et al. Single-cell mRNA cytometry via sequence-specific nanoparticle clustering and trapping. Nat. Chem. 10, 489–495 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Herold, S., Herkert, B. & Eilers, M. Facilitating replication under stress: an oncogenic function of MYC? Nat. Rev. Cancer 9, 441–444 (2009).

    CAS  PubMed  Google Scholar 

  31. Dang, C. V. MYC on the path to cancer. Cell 149, 22–35 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Beaulieu, M. E. et al. Intrinsic cell-penetrating activity propels Omomyc from proof of concept to viable anti-MYC therapy. Sci. Transl. Med. 11, eaar5012 (2019).

    PubMed  PubMed Central  Google Scholar 

  33. Burke, A. J., Ali, H., O’Connell, E., Sullivan, F. J. & Glynn, S. A. Sensitivity profiles of human prostate cancer cell lines to an 80 kinase inhibitor panel. Anticancer Res. 36, 633–641 (2016).

    CAS  PubMed  Google Scholar 

  34. Lakshman, M. et al. Dietary genistein inhibits metastasis of human prostate cancer in mice. Cancer Res. 68, 2024–2032 (2008).

    CAS  PubMed  Google Scholar 

  35. Thiery, J. P. Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2, 442–454 (2002).

    CAS  PubMed  Google Scholar 

  36. Lindsay, C. R. et al. Vimentin and Ki67 expression in circulating tumour cells derived from castrate-resistant prostate cancer. BMC Cancer 16, 168 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ray Chaudhuri, A. & Nussenzweig, A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 18, 610–621 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Schiewer, M. J. et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov. 2, 1134–1149 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kosaka, T. et al. The prognostic significance of OCT4 expression in patients with prostate cancer. Hum. Pathol. 51, 1–8 (2016).

    CAS  PubMed  Google Scholar 

  40. Salem, A. F., Whitaker-Menezes, D., Howell, A., Sotgia, F. & Lisanti, M. P. Mitochondrial biogenesis in epithelial cancer cells promotes breast cancer tumor growth and confers autophagy resistance. Cell Cycle 11, 4174–4180 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Gao, L. et al. Androgen receptor promotes ligand-independent prostate cancer progression through c-Myc upregulation. PLoS ONE 8, e63563 (2013).

    PubMed  PubMed Central  Google Scholar 

  42. Wu, M. et al. Proteome analysis of human androgen-independent prostate cancer cell lines: variable metastatic potentials correlated with vimentin expression. Proteomics 7, 1973–1983 (2007).

    CAS  PubMed  Google Scholar 

  43. Stratton, M. R., Campbell, P. J. & Futreal, P. A. The cancer genome. Nature 458, 719–724 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Tran, C. et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324, 787–790 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Scher, H. I. et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1–2 study. Lancet 375, 1437–1446 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Watson, P. A., Arora, V. K. & Sawyers, C. L. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat. Rev. Cancer 15, 701–711 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Sinkevicius, K. W. et al. Neurotrophin receptor TrkB promotes lung adenocarcinoma metastasis. Proc. Natl Acad. Sci. USA 111, 10299–10304 (2014).

    CAS  PubMed  Google Scholar 

  48. Chen, Y. & Chi, P. Basket trial of TRK inhibitors demonstrates efficacy in TRK fusion-positive cancers. J. Hematol. Oncol. 11, 78–78 (2018).

    PubMed  PubMed Central  Google Scholar 

  49. Wylie, A. A. et al. The allosteric inhibitor ABL001 enables dual targeting of BCR–ABL1. Nature 543, 733–737 (2017).

    CAS  PubMed  Google Scholar 

  50. Holderfield, M., Deuker, M. M., McCormick, F. & McMahon, M. Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat. Rev. Cancer 14, 455–467 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Hauschild, A. et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 380, 358–365 (2012).

    CAS  PubMed  Google Scholar 

  52. Rebbeck, T. R. et al. Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N. Engl. J. Med. 346, 1616–1622 (2002).

    PubMed  Google Scholar 

  53. Jeyasekharan, A. D. et al. A cancer-associated BRCA2 mutation reveals masked nuclear export signals controlling localization. Nat. Struct. Mol. Biol. 20, 1191–1198 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Eriksson, I., Wettermark, B. & Bergfeldt, K. Real-world use and outcomes of olaparib: a population-based cohort study. Target. Oncol. 13, 725–733 (2018).

    PubMed  PubMed Central  Google Scholar 

  55. Mateo, J. et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 373, 1697–1708 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Rowe, B. P. & Glazer, P. M. Emergence of rationally designed therapeutic strategies for breast cancer targeting DNA repair mechanisms. Breast Cancer Res. 12, 203 (2010).

    PubMed  PubMed Central  Google Scholar 

  57. Green, B. J. et al. Isolation of phenotypically distinct cancer cells using nanoparticle-mediated sorting. ACS Appl. Mater. Interfaces 9, 20435–20443 (2017).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Canadian Institutes of Health Research (grant no. FDN-148415), the Natural Sciences and Engineering Research Council of Canada (grant no. 2016-06090), the Province of Ontario though the Ministry of Research, Innovation and Science (grant no. RE05-009) and the National Cancer Institute of the National Institutes of Health (grant no. 1R33CA204574). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or the other funding agencies. We thank A. Joshua for providing the clinical specimens.

Author information

Authors and Affiliations

Authors

Contributions

M.L., S.O.K. and E.H.S. conceived and designed the experiments. M.L., Z.W., S.U.A., R.M.M., B.D. and B.G. performed the experiments and analysed the data. All authors discussed the results and contributed to the preparation and editing of the manuscript.

Corresponding author

Correspondence to Shana O. Kelley.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary note, figures and tables.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Labib, M., Wang, Z., Ahmed, S.U. et al. Tracking the expression of therapeutic protein targets in rare cells by antibody-mediated nanoparticle labelling and magnetic sorting. Nat Biomed Eng 5, 41–52 (2021). https://doi.org/10.1038/s41551-020-0590-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-020-0590-1

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