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.

  • Letter
  • Published:

A coiled-coil masking domain for selective activation of therapeutic antibodies

Abstract

The use of monoclonal antibodies in cancer therapy is limited by their cross-reactivity to healthy tissue. Tumor targeting has been improved by generating masked antibodies that are selectively activated in the tumor microenvironment, but each such antibody necessitates a custom design. Here, we present a generalizable approach for masking the binding domains of antibodies with a heterodimeric coiled-coil domain that sterically occludes the complementarity-determining regions. On exposure to tumor-associated proteases, such as matrix metalloproteinases 2 and 9, the coiled-coil peptides are cleaved and antigen binding is restored. We test multiple coiled-coil formats and show that the optimized masking domain is broadly applicable to antibodies of interest. Our approach prevents anti-CD3-associated cytokine release in mice and substantially improves circulation half-life by protecting the antibody from an antigen sink. When applied to antibody–drug conjugates, our masked antibodies are preferentially unmasked at the tumor site and have increased anti-tumor efficacy compared with unmasked antibodies in mouse models of cancer.

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

Fig. 1: Steric blockade of antibody CDRs as a modular antibody masking system.
Fig. 2: Masking and reactivation of CC2B-masked antibodies.
Fig. 3: In vivo assessment of masked antibodies and ADCs.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available within the paper and its Supplementary Information files.

References

  1. Bugelski, P. J. & Martin, P. L. Concordance of preclinical and clinical pharmacology and toxicology of therapeutic monoclonal antibodies and fusion proteins: cell surface targets. Brit. J. Pharmacol. 166, 823–846 (2012).

    Article  CAS  Google Scholar 

  2. Hansel, T. T., Kropshofer, H., Singer, T., Mitchell, J. A. & George, A. J. The safety and side effects of monoclonal antibodies. Nat. Rev. Drug Discov. 9, 325–338 (2010).

    Article  CAS  Google Scholar 

  3. Polu, K. R. & Lowman, H. B. Probody therapeutics for targeting antibodies to diseased tissue. Expert Opin. Biol. Ther. 14, 1049–1053 (2014).

    Article  CAS  Google Scholar 

  4. Desnoyers, L. R. et al. Tumor-specific activation of an EGFR-targeting probody enhances therapeutic index. Sci. Transl. Med. 5, 207ra144 (2013).

    Article  Google Scholar 

  5. Donaldson, J., Kari, C., Fragoso, R., Rodeck, U. & Williams, J. C. Design and development of masked therapeutic antibodies to limit off-target effects: application to anti-EGFR antibodies. Cancer Biol. Ther. 8, 2147–2152 (2009).

    Article  CAS  Google Scholar 

  6. Thomas, J. & Daugherty, P. Proligands with protease-regulated binding activity identified from cell-displayed prodomain libraries. Protein Sci. 18, 2052–2059 (2009).

    Article  Google Scholar 

  7. Erster, O. et al. Site-specific targeting of antibody activity in vivo mediated by disease-associated proteases. J. Control. Release 161, 804–812 (2012).

    Article  CAS  Google Scholar 

  8. Yang, Y. et al. Preclinical studies of a pro-antibody-drug conjugate designed to selectively target EGFR-overexpressing tumors with improved therapeutic efficacy. mAbs 8, 405–413 (2015).

    Article  Google Scholar 

  9. Yang, Y. et al. Generation and characterization of a target-selectively activated antibody against epidermal growth factor receptor with enhanced anti-tumor potency. mAbs 7, 440–450 (2015).

    Article  CAS  Google Scholar 

  10. Sandersjoo, L., Jonsson, A. & Lofblom, J. A new prodrug form of Affibody molecules (pro-Affibody) is selectively activated by cancer-associated proteases. Cell. Mol. Life Sci. 72, 1405–1415 (2015).

    Article  Google Scholar 

  11. Chen, I. et al. Selective antibody activation through protease-activated pro-antibodies that mask binding sites with inhibitory domains. Sci. Rep. 7, 1–12 (2017).

    Article  Google Scholar 

  12. Burkhard, P., Stetefeld, J. & Strelkov, S. V. Coiled coils: a highly versatile protein folding motif. Trends Cell Biol. 11, 82–88 (2001).

    Article  CAS  Google Scholar 

  13. Thomas, F., Boyle, A. L., Burton, A. J. & Woolfson, D. N. A set of de novo designed parallel heterodimeric coiled coils with quantified dissociation constants in the micromolar to sub-nanomolar regime. J. Am. Chem. Soc. 135, 5161–5166 (2013).

    Article  CAS  Google Scholar 

  14. Arndt, K., Pelletier, J., Müller, K., Plückthun, A. & Alber, T. Comparison of in vivo selection and rational design of heterodimeric coiled coils. Structure 10, 1235–1248 (2002).

    Article  CAS  Google Scholar 

  15. Schmidt, M. M. Engineering antibodies for improved targeting of solid tumors. PhD thesis, Massachusetts Institute of Technology, Ch. 5 (2010); http://hdl.handle.net/1721.1/61239

  16. McClain, D., Woods, H. & Oakley, M. Design and characterization of a heterodimeric coiled coil that forms exclusively with an antiparallel relative helix orientation. J. Am. Chem. Soc. 123, 3151–3152 (2001).

    Article  CAS  Google Scholar 

  17. Plückthun, A. & Pack, P. New protein engineering approaches to multivalent and bispecific antibody fragments. Immunotechnology 3, 83–105 (1997).

    Article  Google Scholar 

  18. Gerber, H. P. et al. Potent antitumor activity of the anti-CD19 auristatin antibody drug conjugate hBU12-vcMMAE against rituximab-sensitive and -resistant lymphomas. Blood 113, 4352–4361 (2009).

    Article  CAS  Google Scholar 

  19. Jiang, T. et al. Tumor imaging by means of proteolytic activation of cell-penetrating peptides. Proc. Natl Acad. Sci. USA 101, 17867–17872 (2004).

    Article  CAS  Google Scholar 

  20. Turk, B. E., Huang, L. L., Piro, E. T. & Cantley, L. C. Determination of protease cleavage site motifs using mixture-based oriented peptide libraries. Nat. Biotechnol. 19, 661–667 (2001).

    Article  CAS  Google Scholar 

  21. Shay, G., Lynch, C. C. & Fingleton, B. Moving targets: emerging roles for MMPs in cancer progression and metastasis. Matrix Biol. 44–46, 200–206 (2015).

    Article  Google Scholar 

  22. Kessenbrock, K., Plaks, V. & Werb, Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 141, 52–67 (2010).

    Article  CAS  Google Scholar 

  23. King, K. M. & Younes, A. Rituximab: review and clinical applications focusing on non-Hodgkin’s lymphoma. Expert Rev. Anticancer Ther. 1, 177–186 (2001).

    Article  CAS  Google Scholar 

  24. Maximiano, S., Magalhães, P., Guerreiro, M. P. & Morgado, M. Trastuzumab in the treatment of breast cancer. BioDrugs 30, 75–86 (2016).

    Article  CAS  Google Scholar 

  25. Ryan, M. C. et al. Integrin αVα6 is expressed on multiple solid tumors and is a potential therapeutic target for auristatin-based antibody-drug conjugates. Canc. Res. 72, abstr. 4630 (2012).

  26. Leo, O., Foo, M., Sachs, D. H., Samelson, L. E. & Bluestone, J. A. Identification of a monoclonal antibody specific for a murine T3 polypeptide. Proc. Natl Acad. Sci. USA 84, 1374–1378 (1987).

    Article  CAS  Google Scholar 

  27. Lin, J. & Sagert, J. in Innovations for Next-Generation Antibody-Drug Conjugates (ed. Damelin, M.) 281–298 (Springer Int. Pub., 2018).

  28. Doronina, S. O. et al. Enhanced activity of monomethylauristatin F through monoclonal antibody delivery: effects of linker technology on efficacy and toxicity. Bioconjug. Chem. 17, 114–124 (2006).

    Article  CAS  Google Scholar 

  29. Stanková, J., Hoskin, D. W. & Roder, J. C. Murine anti-CD3 monoclonal antibody induces potent cytolytic activity in both T and NK cell populations. Cell. Immunol. 121, 13–29 (1989).

    Article  Google Scholar 

  30. Alegre, M. et al. Hypothermia and hypoglycemia induced by anti-CD3 monoclonal antibody in mice: role of tumor necrosis factor. Eur. J. Immunol. 20, 707–710 (1990).

    Article  CAS  Google Scholar 

  31. Ferran, C. et al. Cytokine-related syndrome following injection of anti-CD3 monoclonal antibody: further evidence for transient in vivo T cell activation. Eur. J. Immunol. 20, 509–515 (1990).

    Article  CAS  Google Scholar 

  32. Huang, X. et al. Inactivation of the integrin B6 subunit gene reveals a role of epithelial integrins in regulating inflammation in the lungs and skin. J. Cell Biol. 133, 921–928 (1996).

    Article  CAS  Google Scholar 

  33. Lyon, R. P. et al. Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33, 733–735 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank our Seattle Genetics colleagues L. Benoit, D. Meyer and J. Mitchell for help with antibody–drug conjugation and C. Yu for help with internalization experiments.

Author information

Authors and Affiliations

Authors

Contributions

V.H.T., M.R.L. and P.D.S. participated in writing, reviewing and editing of the manuscript. V.H.T. and M.R.L. participated in the planning, initiation, data generation and analysis of biological experiments. V.H.T., X.Z. and M.R.L. performed cell-based assays. V.H.T. performed zymography experiments. S.W.W. and M.M.D performed biophysical characterization. J.H.C. conducted analysis of pharmacokinetics experiments. R.C.Y., W.Z., J.K.S. and I.J.S. performed in vivo anti-tumor activity and pharmacodynamics experiments. M.C.R., R.P.L. and P.D.S. provided project oversight and review.

Corresponding author

Correspondence to Matthew R. Levengood.

Ethics declarations

Competing interests

All of the authors were employees and shareholders of Seattle Genetics at the time of these studies.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Representative size-exclusion chromatographs and expression titers for select antibodies.

Analytical size-exclusion chromatography (SEC) data was collected as described in the Materials and Methods.

Supplementary Figure 2 Protease cleavage of coiled-coil masked antibodies.

a) Representative characterization of coiled-coil masked antibodies and MMP activation by LC-MS. Shown is the deconvoluted spectra of CC2B-IPVSLRSG-rituximab heavy and light chains (top) as well as MMP-2-cleaved CC-IPVSLRSG-rituximab heavy and light chains (bottom) following reduced reverse-phase LC-MS. b) Representative data for the cleavage of CC3-IPVSLRSG-hBU12 with a panel of human (h) and mouse (m) MMPs. Activated MMP (6 Enzyme Units, EU, amount of enzyme to process 1 μg substrate/minute) was incubated with antibody (10 μg) at for 20 min at 37 °C. Reactions were stopped by addition of 25 mM EDTA and samples were analyzed using the method described in (A). The percent cleaved heavy or light chain was calculated using peak intensities from deconvoluted mass spectra. Two independent experiments were performed with similar results.

Supplementary Figure 3 Kinetics of antibody binding.

Kinetics of binding for hBU12 and CC2B-hBU12 were assessed via biolayer interferometry using recombinant human CD19. Biotinylated antigen was immobilized on a streptavidin-coated biosensor and antibodies were then assessed for association and dissociation over time. Kinetic parameters were determined using the Octet analysis software using a 1:1 global fit binding model. This experiment was repeated in two independent experiments with similar results.

Supplementary Figure 4 Binding of partially masked antibodies.

Half-masked hBU12 antibodies were assessed for binding on CD19(+) Raji cells. Antibodies were expressed containing an MMP-cleavable linker on either the heavy or light chain and a non-cleavable linker on the opposite chain. a) Characterization of LC cleavable CC2B-hBU12 constructs by reverse phase LC-MS. Shown is the deconvoluted mass spectra of LC-CC2B-hBU12 heavy and light chain (top) as well as MMP-2-cleaved heavy and light chain (bottom). The additional peak (*, 55922 Da) on the heavy chain is due to complexation of mercury from the activation buffer for MMP-2. Following incubation with MMP-2, the light chain containing the IPVSLRSG sequence was cleaved whereas the heavy chain containing the LALGPG sequence was not. Similar results were observed for HC CC2B-hBU12 except the heavy chain was cleaved whereas the light chain was not (data not shown). b) The half-masked antibodies were assessed for binding via saturation analysis on human CD19-expressing Raji cells using flow cytometry. Data represent the mean of duplicate cell samples. Half-cleaved antibodies restored binding to within approximately 10-fold of the parent antibody whereas fully cleavage restores binding to the same level as un-masked hBU12. For S4B, two independent experiments were conducted with similar results.

Supplementary Figure 5 Proliferation of mouse T cells in the presence of either masked or unmasked anti-mouse CD3 antibodies.

Mouse T-cells were labeled with carboxyfluorescein succinimidyl ester (CFSE) and incubated with either CC2B-masked or unmasked anti-mouse CD3 antibodies at 500 ng/mL along with anti-CD28 stimulation at 37 °C. T-cell activation was monitored after 4 days of incubation using flow cytometry. A leftward shift of fluorescence indicates that CFSE has been diluted as cells have proliferated. Robust proliferation was observed for the anti-mouse CD3 antibody whereas no proliferation occurred for the masked antibody. Two independent experiments were conducted with similar results.

Supplementary Figure 6 In situ zymography of frozen Ramos xenograft tumor slices.

Protease activity was assessed by incubating Alexa Fluor 647-labeled hBU12 or CC3-IPVSLRSG-hBU12 antibodies (1 µg/mL) with fresh frozen Ramos tumor slices (5 µm). Proteolysis of the mask results in binding of the antibody, as detected using an Olympus fluorescence microscope. The addition of protease inhibitors resulted in reduced binding of CC3-IPVSLRSG-hBU12, whereas hBU12 binding was not impacted. Images were taken at 20x magnification and the scale bar indicates a 50 μm distance. Two independent experiments were conducted with similar results.

Supplementary Figure 7 Binding of masked and unmasked h15H3-gluc-MMAE(8) ADCs.

Binding was assessed by saturating binding on αVβ6-transfected human embryonic kidney (HEK) cells. Data represent the mean of duplicate cell samples. For the cleaved antibody, the CC2B mask was removed via recombinant MMP-2, which was confirmed by reverse-phase LC-MS analysis prior to the binding study. Two independent experiments were performed with similar results.

Supplementary Figure 8 Cytotoxicity of masked and unmasked h15H3-gluc-MMAE(8) ADCs.

ADCs were incubated with BxPC3 cells for 96 h at 37 °C. Cytotoxicity was assessed using CellTiter-Glo (Promega) and luminescence was measured on an EnVision plate reader (Perkin Elmer). Data represent the mean ± s.d. of quadruplicate cell samples. Two independent experiments were performed with similar results.

Supplementary Figure 9 Efficacy of anti-αVβ6 h15H3-gluc-MMAE(8) ADCs in Detroit-562 xenograft.

Mice were administered a single 3 mg/kg IP dose once tumors reached 100 mm3 and tumor size was measured at various time points. Data represent the mean of n = 5 biologically independent animals. The experiment was performed once, but the h15H3-gluc-MMAE(8) ADC has been tested in at least one additional independent experiment and provided similar results.

Supplementary Figure 10 Efficacy of anti-αVβ6 h15H3-gluc-MMAE(8) ADCs in HPAF-II xenograft.

Mice were administered a single 3 mg/kg IP dose once tumors reached 100 mm3 and tumor size was measured at various time points. Data represent the mean of n = 5 biologically independent animals. The experiment was performed once, but the h15H3-gluc-MMAE(8) and isotype ADCs have been tested in at least one additional independent experiment and provided similar results.

Supplementary Figure 11 Cleavage of masked ADCs in vivo.

Western blot analysis of tumor, plasma, and liver samples from an HPAF-II xenograft tumor model. Mice bearing HPAF-II tumors (100 mm3) were dosed with 3 mg/kg of CC2B-h15H3-gluc-MMAE(8) ADCs bearing either cleavable (PLGLAG) or scrambled (LALGPG) cleavage sequences, or the h15H3-gluc-MMAE(8) parent ADC. At 4 days post-dose, mice were sacrificed and tissues were harvested. Antibody was purified from homogenized tissue samples using protein A capture and the presence of intact or cleaved antibody was assessed via western blot. The experiment was conducted once with n = 3 mice for CC2B-PLGLAG-h15H3, n = 2 mice for CC2B-LALGPG-h15H3, and n = 3 mice for h15H3.

Supplementary Figure 12 In situ zymography of frozen HPAF-II tumor slices.

Protease activity was assessed via incubation of anti-αVβ6 CC2B-h15H3 antibodies with HPAF-II tumor slices. Proteolysis of the mask results in antibody binding, which was detected using an AF594 labeled anti-human secondary antibody. The scrambled control antibody CC2B-LALGPG-h15H3 was less effectively cleaved than the cleavable antibody CC2B-IPVSLRSG-h15H3, as indicated by reduced staining of the tumor slice. Images were taken at a 10x magnification and the scale bar indicates a 100 μm distance. Two independent experiments were performed with similar results.

Supplementary Figure 13 Partial MMP cleavage and binding analysis of CC2B-PLGLAG-h15H3.

100 μg of antibody was incubated with 9 EU of human MMP2 at 37 °C for 0, 5, 10, 40, and 60 min. The reaction was stopped at the indicated times using 25 mM EDTA and samples were analyzed using reduced reverse-phase LC-MS. The percent cleaved antibody was calculated using mass spectrometry peak intensities and represents an average of heavy and light chain cleavage. Binding was assessed using human embryonic kidney cells (HEK) expressing αVβ6. Data represent the mean of duplicate cell samples. Two independent experiments were performed with similar results.

Supplementary Figure 14 Cleavage of non-binding antibodies in vivo.

Cleavage analysis of CC3-masked non-binding antibodies in the plasma of naïve BALB/c (left) or HT1080 xenograft-bearing nude mice (right). Naïve BALB/c mice were dosed with 3 mg/kg of a CC3-IPVSLRSG-masked IgG1 isotype control antibody (left) and HT1080 tumor-bearing nude mice dosed with 5 mg/kg of CC3-IPVSLRSG-IgG1 (right). At the times indicated, plasma samples were collected from mice, human antibodies was captured using protein A resin, and the presence of intact or cleaved antibody heavy chain was assessed via Western blot. Data represent the mean ± SD (n = 3 biologically independent animals). Two independent experiments were performed with similar results.

Supplementary information

Supplementary Information

Supplementary Figs. 1–14 and Supplementary Tables 1 and 2

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Trang, V.H., Zhang, X., Yumul, R.C. et al. A coiled-coil masking domain for selective activation of therapeutic antibodies. Nat Biotechnol 37, 761–765 (2019). https://doi.org/10.1038/s41587-019-0135-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0135-x

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