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.

  • Perspective
  • Published:

Recent advances in the construction of antibody–drug conjugates

A Corrigendum to this article was published on 19 February 2016

This article has been updated

Abstract

Antibody–drug conjugates (ADCs) comprise antibodies covalently attached to highly potent drugs using a variety of conjugation technologies. As therapeutics, they combine the exquisite specificity of antibodies, enabling discrimination between healthy and diseased tissue, with the cell-killing ability of cytotoxic drugs. This powerful and exciting class of targeted therapy has shown considerable promise in the treatment of various cancers with two US Food and Drug Administration approved ADCs currently on the market (Adcetris and Kadcyla) and approximately 40 currently undergoing clinical evaluation. However, most of these ADCs exist as heterogeneous mixtures, which can result in a narrow therapeutic window and have major pharmacokinetic implications. In order for ADCs to deliver their full potential, sophisticated site-specific conjugation technologies to connect the drug to the antibody are vital. This Perspective discusses the strategies currently used for the site-specific construction of ADCs and appraises their merits and disadvantages.

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: General structure of an immunoglobulin G1 (IgG1) highlighting key components.
Figure 2: General scheme highlighting typical methods for the construction of ADCs based on engineered antibodies.
Figure 3: General scheme highlighting typical methods for the construction of ADCs based on native antibodies.

Similar content being viewed by others

Change history

  • 25 January 2016

    In the originally published version of this Perspective article, reference 42 contained an incorrect article number. The reference should have read 'Maruani, A. et al. A plug-and-play approach to antibody-based therapeutics via a chemoselective dual click strategy. Nature Commun. 6, 6645 (2015).' This has been corrected in all online versions.

References

  1. Ehrlich, P. Address in pathology, ON CHEMIOTHERAPY: Delivered before the seventeenth international congress of medicine. Brit. Med. J. 2, 353–359 (1913).

    Article  CAS  Google Scholar 

  2. Mathé, G., Loc, T. B. & Bernard, J. Effet sur la leucémie 1210 de la souris d'une combinaison par diazotation d'A-méthoptérine et de γ-globulines de hamsters porteurs de cette leucémie par hétérogreffe. C.R. Hebd. Séances Acad. Sci. 246, 1626–1628 (1958).

    PubMed  Google Scholar 

  3. Ghose, T. & Nigam, S. P. Antibody as carrier of chlorambucil. Cancer 29, 1398–1400 (1972).

    Article  CAS  Google Scholar 

  4. Rowland, G. F., O'Neill, G. J. & Davies, D. A. L. Suppression of tumour growth in mice by a drug–antibody conjugate using a novel approach to linkage. Nature 255, 487–488 (1975).

    Article  CAS  Google Scholar 

  5. Ford, C. H. J. et al. Localisation and toxicity study of a vindesine-anti-CEA conjugate in patients with advanced cancer. Brit. J. Cancer 47, 35–42 (1983).

    Article  CAS  Google Scholar 

  6. Trail, P. A. et al. Cure of xenografted human carcinomas by Br96–doxorubicin immunoconjugates. Science 261, 212–215 (1993).

    Article  CAS  Google Scholar 

  7. Pietersz, G. A. & Krauer, K. Antibody-targeted drugs for the therapy of cancer. J. Drug Target. 2, 183–215 (1994).

    Article  CAS  Google Scholar 

  8. Linenberger, M. L. et al. Multidrug-resistance phenotype and clinical responses to gemtuzumab ozogamicin. Blood 98, 988–994 (2001).

    Article  CAS  Google Scholar 

  9. Younes, A. et al. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N. Engl. J. Med. 363, 1812–1821 (2010).

    Article  CAS  Google Scholar 

  10. Senter, P. D. & Sievers, E. L. The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nature Biotechnol. 30, 631–637 (2012).

    Article  CAS  Google Scholar 

  11. Verma, S. et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N. Engl. J. Med. 367, 1783–1791 (2012).

    Article  CAS  Google Scholar 

  12. LoRusso, P. M., Weiss, D., Guardino, E., Girish, S. & Sliwkowski, M. X. Trastuzumab emtansine: a unique antibody-drug conjugate in development for human epidermal growth factor receptor 2-positive cancer. Clin. Cancer Res. 17, 6437–6447 (2011).

    Article  CAS  Google Scholar 

  13. Mullard, A. Maturing antibody–drug conjugate pipeline hits 30. Nature Rev. Drug Discov. 12, 329–332 (2013).

    Article  CAS  Google Scholar 

  14. Hamblett, K. J. et al. Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate. Clin. Cancer Res. 10, 7063–7070 (2004).

    Article  CAS  Google Scholar 

  15. Stan, A. C., Radu, D. L., Casares, S., Bona, C. A. & Brumeanu, T. D. Antineoplastic efficacy of doxorubicin enzymatically assembled on galactose residues of a monoclonal antibody specific for the carcinoembryonic antigen. Cancer Res. 59, 115–121 (1999).

    CAS  PubMed  Google Scholar 

  16. Strop, P. et al. Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem. Biol. 20, 161–167 (2013).

    Article  CAS  Google Scholar 

  17. Lyons, A. et al. Site-specific attachment to recombinant antibodies via introduced surface cysteine residues. Protein Eng. 3, 703–708 (1990).

    Article  CAS  Google Scholar 

  18. Junutula, J. R. et al. Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nature Biotechnol. 26, 925–932 (2008).

    Article  CAS  Google Scholar 

  19. Sunbul, M. & Yin, J. Site specific protein labeling by enzymatic posttranslational modification. Org. Biomol. Chem. 7, 3361–3371 (2009).

    Article  CAS  Google Scholar 

  20. Young, T. S., Ahmad, I., Yin, J. A. & Schultz, P. G. An enhanced system for unnatural amino acid mutagenesis in E. coli. J. Mol. Biol. 395, 361–374 (2010).

    Article  CAS  Google Scholar 

  21. Rabuka, D., Rush, J. S., deHart, G. W., Wu, P. & Bertozzi, C. R. Site-specific chemical protein conjugation using genetically encoded aldehyde tags. Nature Protoc. 7, 1052–1067 (2012).

  22. Axup, J. Y. et al. Synthesis of site-specific antibody-drug conjugates using unnatural amino acids. Proc. Natl Acad. Sci. USA 109, 16101–16106 (2012).

    Article  CAS  Google Scholar 

  23. Junutula, J. R. et al. Rapid identification of reactive cysteine residues for site-specific labeling of antibody-Fabs. J. Immunol. Methods 332, 41–52 (2008).

    Article  CAS  Google Scholar 

  24. Woo, H. J., Lotz, M. M., Jung, J. U. & Mercurio, A. M. Carbohydrate-binding protein 35 (Mac-2), a laminin-binding lectin, forms functional dimers using cysteine 186. J. Biol. Chem. 266, 18419–18422 (1991).

    CAS  PubMed  Google Scholar 

  25. Wootton, S. K. & Yoo, D. Homo-oligomerization of the porcine reproductive and respiratory syndrome virus nucleocapsid protein and the role of disulfide linkages. J. Virol. 77, 4546–4557 (2003).

    Article  CAS  Google Scholar 

  26. Greenberg, C. S., Birckbichler, P. J. & Rice, R. H. Transglutaminases: multifunctional cross-linking enzymes that stabilize tissues. FASEB J. 5, 3071–3077 (1991).

    Article  CAS  Google Scholar 

  27. Kanaji, T. et al. Primary structure of microbial transglutaminase from Streptoverticillium sp. strain s-8112. J. Biol. Chem. 268, 11565–11572 (1993).

    CAS  PubMed  Google Scholar 

  28. Kashiwagi, T. et al. Crystal structure of microbial transglutaminase from Streptoverticillium mobaraense. J. Biol. Chem. 277, 44252–44260 (2002).

    Article  CAS  Google Scholar 

  29. Jeger, S. et al. Site-specific and stoichiometric modification of antibodies by bacterial transglutaminase. Angew. Chem. Int. Ed. 49, 9995–9997 (2010).

    Article  CAS  Google Scholar 

  30. Wu, P. et al. Site-specific chemical modification of recombinant proteins produced in mammalian cells by using the genetically encoded aldehyde tag. Proc. Natl Acad. Sci. USA 106, 3000–3005 (2009).

    Article  CAS  Google Scholar 

  31. Drake, P. M. et al. Aldehyde tag coupled with HIPS chemistry enables the production of ADCs conjugated site-specifically to different antibody regions with distinct in vivo efficacy and PK outcomes. Bioconjugate Chem. 25, 1331–1341 (2014).

    Article  CAS  Google Scholar 

  32. Zimmerman, E. S. et al. Production of site-specific antibody-drug conjugates using optimized non-natural amino acids in a cell-free expression system. Bioconjugate Chem. 25, 351–361 (2014).

    Article  CAS  Google Scholar 

  33. Sapra, P. et al. Long-term tumor regression induced by an antibody-drug conjugate that targets 5T4, an oncofetal antigen expressed on tumor-initiating cells. Mol. Cancer Ther. 12, 38–47 (2013).

    Article  CAS  Google Scholar 

  34. Tian, F. et al. A general approach to site-specific antibody drug conjugates. Proc. Natl Acad. Sci. USA 111, 1766–1771 (2014).

    Article  CAS  Google Scholar 

  35. Sun, M. M. et al. Reduction-alkylation strategies for the modification of specific monoclonal antibody disulfides. Bioconjugate Chem. 16, 1282–1290 (2005).

    Article  CAS  Google Scholar 

  36. Doronina, S. O. et al. Development of potent monoclonal antibody auristatin conjugates for cancer therapy. Nature Biotechnol. 21, 778–784 (2003).

    Article  CAS  Google Scholar 

  37. Beckley, N. S., Lazzareschi, K. P., Chih, H.-W., Sharma, V. K. & Flores, H. L. Investigation into temperature-induced aggregation of an antibody drug conjugate. Bioconjugate Chem. 24, 1674–1683 (2013).

    Article  CAS  Google Scholar 

  38. Shen, B. Q. et al. Conjugation site modulates the in vivo stability and therapeutic activity of antibody-drug conjugates. Nature Biotechnol. 30, 184–189 (2012).

    Article  CAS  Google Scholar 

  39. Lyon, R. P. et al. Self-hydrolyzing maleimides improve the stability and pharmacological properties of antibody-drug conjugates. Nature Biotechnol. 32, 1059–1062 (2014).

    Article  CAS  Google Scholar 

  40. Badescu, G. et al. Bridging disulfides for stable and defined antibody drug conjugates. Bioconjugate Chem. 25, 1124–1136 (2014).

    Article  CAS  Google Scholar 

  41. Nunes, J. P. M. et al. Functional native disulfide bridging enables delivery of a potent, stable and targeted antibody-drug conjugate (ADC). Chem. Commun. 51, 10624–10627 (2015).

    Article  CAS  Google Scholar 

  42. Maruani, A. et al. A plug-and-play approach to antibody-based therapeutics via a chemoselective dual click strategy. Nature Commun. 6, 6645 (2015).

    Article  CAS  Google Scholar 

  43. Maruani, A. et al. Site-selective multi-porphyrin attachment enables the formation of a next-generation antibody-based photodynamic therapeutic. Chem. Commun. 51, 15304–15307 (2015).

    Article  CAS  Google Scholar 

  44. Lee, M. T. W., Maruani, A., Baker, J., Caddick, S. & Chudasama, V. Next-generation disulfide stapling: Reduction and functional re-bridging all in one. Chem. Sci. 7, 799–802 (2016).

    Article  CAS  Google Scholar 

  45. Hinman, L. M. et al. Preparation and characterization of monoclonal antibody conjugates of the calicheamicins: a novel and potent family of antitumor antibiotics. Cancer Res. 53, 3336–3342 (1993).

    CAS  PubMed  Google Scholar 

  46. Hamann, P. R. et al. An anti-MUC1 antibody-calicheamicin conjugate for treatment of solid tumors. Choice of linker and overcoming drug resistance. Bioconjugate Chem. 16, 346–353 (2005).

    Article  CAS  Google Scholar 

  47. Wang, W. et al. Impact of methionine oxidation in human IgG1 Fc on serum half-life of monoclonal antibodies. Mol. Immunol. 48, 860–866 (2011).

    Article  CAS  Google Scholar 

  48. Zhou, Q. et al. Site-specific antibody-drug conjugation through glycoengineering. Bioconjugate Chem. 25, 510–520 (2014).

    Article  CAS  Google Scholar 

  49. Li, X., Fang, T. & Boons, G. J. Preparation of well-defined antibody-drug conjugates through glycan remodeling and strain-promoted azide-alkyne cycloadditions. Angew. Chem. Int. Ed. 53, 7179–7182 (2014).

    Article  CAS  Google Scholar 

  50. Jefferis, R. Glycosylation of recombinant antibody therapeutics. Biotechnol. Prog. 21, 11–16 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to UCL, UCLB, NIHR BRC, MRC, BBSRC and EPSRC for support of our work in this area.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Vijay Chudasama or Stephen Caddick.

Ethics declarations

Competing interests

V.C. and S.C. are co-founders and directors of the company ThioLogics.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chudasama, V., Maruani, A. & Caddick, S. Recent advances in the construction of antibody–drug conjugates. Nature Chem 8, 114–119 (2016). https://doi.org/10.1038/nchem.2415

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchem.2415

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research