Skip to main content

Advertisement

Log in

Oriented conjugation of antibodies against the epithelial cell adhesion molecule on fluorescently doped silica nanoparticles for flow-cytometric determination and in vivo imaging of EpCAM, a biomarker for colorectal cancer

  • Original Paper
  • Published:
Microchimica Acta Aims and scope Submit manuscript

Abstract

The authors report on the conjugation of monoclonal antibodies against the biomarker epithelial cell adhesion molecule (EpCAM) to silica nanoparticles doped with the dye Cy5 (Cy5-SiNPs). Conjugation was performed on the Cy5-SiNPs that were previously coated with a layer of protein G which serves as a linker controlling the orientation of the antibody. The conjugation method takes advantage of site specific interactions between the protein G and constant domains (Fc) of the antibody. The method warrants the antibody binding sites (Fab) to be faced outwards such that the conjugates maintain their affinity for binding the analyte (EpCAM). In vitro analysis by confocal fluorescence imaging and flow cytometry using analytical wavelengths comparable with the excitation and emission wavelength of Cy5-SiNPs at 643 and 662 nm, respectively. The result demonstrated the oriented conjugate to specifically bind to target cells (HT-29) with a sensitivity that is 12 times higher than that of conjugates prepared by conventional EDC coupling. In vivo fluorescence imaging of mice bearing the HT-29 tumor highlighted time-dependent accumulation of the oriented conjugates at the tumor site. As indicated by biodistribution studies hepatic excretion of the oriented probes occurs, however tumor fluorescence still remains for up to 14 days post injection. This research demonstrates that the oriented conjugates derived herein can improve target cell detection sensitivity and can be successfully applied in tumor imaging, which should drive further development of new classes of effective fluorescence contrast agents for cancer diagnostics.

Cy5 dye-doped silica nanoparticles were conjugated to antibodies specific for the epithelial cell adhesion molecule. The nanoparticles were previously coated with protein G to control the orientation of the antibody. This warrants enhanced sensitivity for in vitro analysis and also enables in vivo imaging.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. van Dam GM, Themelis G, Crane LMA et al (2011) Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: first in-human results. Nat Med 17:1315–1319

    Article  Google Scholar 

  2. Sturm MB, Joshi BP, Lu S et al (2013) Targeted imaging of esophageal neoplasia with a fluorescently labeled peptide: first-in-human results. Sci Transl Med 5:184ra61

    Article  Google Scholar 

  3. Yang L, Sajja HK, Cao Z et al (2014) uPAR-targeted optical imaging contrasts as theranostic agents for tumor margin detection. Theranostics 4:106–118

    Article  CAS  Google Scholar 

  4. Hellebust A, Richards-Kortum R (2012) Advances in molecular imaging: targeted optical contrast agents for cancer diagnostics. Nanomedicine 7:429–445

    Article  CAS  Google Scholar 

  5. Chi C, Du Y, Ye J et al (2014) Intraoperative imaging-guided cancer surgery: from current fluorescence molecular imaging methods to future multi-modality imaging technology. Theranostics 4:1072–1084

    Article  Google Scholar 

  6. Burns AA, Vider J, Ow H et al (2009) Fluorescent silica nanoparticles with efficient urinary excretion for nanomedicine. Nano Lett 9:442–448

    Article  CAS  Google Scholar 

  7. Hong X, Wang Z, Yang J et al (2012) Silylated BODIPY dyes and their use in dye-encapsulated silica nanoparticles with switchable emitting wavelengths for cellular imaging. Analyst 137:4140–4149

    Article  CAS  Google Scholar 

  8. Bae SW, Tan W, Hong J-I (2012) Fluorescent dye-doped silica nanoparticles: new tools for bioapplications. Chem Commun 48:2270–2282

    Article  CAS  Google Scholar 

  9. Tiernan JP, Ingram N, Perry SL et al (2015) CEA-targeted nanoparticles allow specific in vivo fluorescent imaging of colorectal cancer models. Nanomedicine 19:1–9

    Google Scholar 

  10. Santra S, Zhang P, Wang K, Tapec R, Tan W (2001) Conjugation of biomolecules with luminophore-doped silica nanoparticles for photostable biomarkers. Anal Chem 73:4988–4993

    Article  CAS  Google Scholar 

  11. Tao L, Zhang K, Sun Y, Jin B, Zhang Z, Yang K (2012) Anti-epithelial cell adhesion molecule monoclonal antibody conjugated fluorescent nanoparticle biosensor for sensitive detection of colon cancer cells. Biosens Bioelectron 35:186–192

    Article  CAS  Google Scholar 

  12. Xing Y, Chaudry Q, Shen C et al (2007) Bioconjugated quantum dots for multiplexed and quantitative immunohistochemistry. Nat Protoc 2:1152–1165

    Article  CAS  Google Scholar 

  13. Liu Y, Yu J (2016) Oriented immobilization of proteins on solid supports for use in biosensors and biochips: a review. Microchim Acta 183:1–19

    Article  CAS  Google Scholar 

  14. Jha RK, Gaiotto T, Bradbury ARM, Strauss CEM (2014) An improved protein G with higher affinity for human/rabbit IgG Fc domains exploiting a computationally designed polar network. Protein Eng Des Sel 27:127–134

    Article  CAS  Google Scholar 

  15. Bailey LJ, Sheehy KM, Hoey RJ, Schaefer ZP, Ura M, Kossiakoff AA (2014) Applocations for an engineered protein-G varient with a pH controllable affinity to antibody fragments. J Immunol Methods 415:24–30

    Article  CAS  Google Scholar 

  16. Kato K, Lian L, Barsukov IL et al (1995) Model for the complex between protein G and an antibody Fc fragment in solution. Protein Eng 3:79–85

    CAS  Google Scholar 

  17. Bae YM, Oh B-K, Lee W, Lee WH, Choi J-W (2005) Study on orientation of immunoglobulin G on protein G layer. Biosens Bioelectron 21:103–110

    Article  CAS  Google Scholar 

  18. Schnell U, Cirulli V, Giepmans BNG (2013) EpCAM: structure and function in health and disease. Biochim Biophys 1828:1989–2001

    Article  CAS  Google Scholar 

  19. van der Gun BTF, Melchers LJ, Ruiters MHJ, de Leij LFMH, McLaughlin PMJ, Rots MG (2010) EpCAM in carcinogenesis: the good, the bad or the ugly. Carcinogenesis 31:1913–1921

    Article  Google Scholar 

  20. Spizzo G, Fong D, Wurm M et al (2011) EpCAM expression in primary tumour tissues and metastases: an immunohistochemical analysis. J Clin Pathol 64:415–420

    Article  Google Scholar 

  21. Kausaite-Minkstimiene A, Ramanaviciene A, Kirlyte J, Ramanavicius A (2010) Comparative study of random and oriented antibody immobilization techniques on the binding capacity of immunosensor. Anal Chem 82:6401–6408

    Article  CAS  Google Scholar 

  22. Stöber W, Fink A, Bohn E (1968) Controlled growth of monodisperse silica spheres in the micron size range. J Colloid Interface Sci 26:62–69

    Article  Google Scholar 

  23. Bamrungsap S, Apiwat C, Chantima W, Dharakul T, Wiriyachaiporn N (2013) Rapid and sensitive lateral flow immunoassay for influenza antigen using fluorescently-doped silica nanoparticles. Microchim Acta 181:223–230

    Article  Google Scholar 

  24. Bailey LJ, Sheehy KM, Hoey RJ, Schaefer ZP, Ura M, Kossiakoff AA (2014) Applications for an engineered protein-G variant with a pH controllable affinity to antibody fragments. J Immunol Methods 415:24–30

    Article  CAS  Google Scholar 

  25. Xing Y, Chaudry Q, Shen C et al (2008) Bioconjugated quantum dots for multiplexed and quantitative immunohistochemistry. Nat Protoc 5:1152–1165

    Google Scholar 

  26. Park K-S, Tae J, Choi B et al (2010) Characterization, in vitro cytotoxicity assessment, and in vivo visualization of multimodal, RITC-labeled, silica-coated magnetic nanoparticles for labeling human cord blood-derived mesenchymal stem cells. Nanomedicine 6:263–276

    Article  CAS  Google Scholar 

  27. Koo H, Huh MS, Sun I-C et al (2011) In vivo targeted delivery of nanoparticles for theranosis. Acc Chem Res 44:1018–1028

    Article  CAS  Google Scholar 

  28. Lu J, Liong M, Li Z, Zink JI, Tamanoi F (2010) Biocompatibility, Biodistribution, and drug-delivery Effiency of Mesoporous silica nanoparticles for cancer therapy in animals. Small 6:1794–1805

    Article  CAS  Google Scholar 

  29. Thurber GM, Weissleder R (2011) Quantitating antibody uptake in vivo: conditional dependence on antigen expression levels. Mol Imaging Biol 13:623–632

    Article  Google Scholar 

  30. Rosenholm JM, Mamaeva V, Sahlgren C, Linden M (2012) Nanoparticles in targeted cancer therapy: mesoporous silica nanoparticles entering preclinical development stage. Nanomedicine 7:111–120

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Suwussa Bamrungsap.

Ethics declarations

The authors declare that they have no competing interests.

Electronic supplementary material

ESM 1

(DOCX 762 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Treerattrakoon, K., Chanthima, W., Apiwat, C. et al. Oriented conjugation of antibodies against the epithelial cell adhesion molecule on fluorescently doped silica nanoparticles for flow-cytometric determination and in vivo imaging of EpCAM, a biomarker for colorectal cancer. Microchim Acta 184, 1941–1950 (2017). https://doi.org/10.1007/s00604-017-2211-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00604-017-2211-6

Keywords

Navigation