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.

  • Technical Report
  • Published:

Monoclonal TCR-redirected tumor cell killing

Abstract

T cell immunity can potentially eradicate malignant cells and lead to clinical remission in a minority of patients with cancer. In the majority of these individuals, however, there is a failure of the specific T cell receptor (TCR)–mediated immune recognition and activation process. Here we describe the engineering and characterization of new reagents termed immune-mobilizing monoclonal TCRs against cancer (ImmTACs). Four such ImmTACs, each comprising a distinct tumor-associated epitope-specific monoclonal TCR with picomolar affinity fused to a humanized cluster of differentiation 3 (CD3)-specific single-chain antibody fragment (scFv), effectively redirected T cells to kill cancer cells expressing extremely low surface epitope densities. Furthermore, these reagents potently suppressed tumor growth in vivo. Thus, ImmTACs overcome immune tolerance to cancer and represent a new approach to tumor immunotherapy.

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: The biological activity and biophysical characteristics of ImmTAC molecules with different specificities.
Figure 2: Efficient activation of multiple CD8+ T cell effector functions by ImmTAC-gp100.
Figure 3: Redirected lysis of tumor cells and peptide-pulsed targets by CD8+ T cells in the presence of ImmTAC molecules.
Figure 4: Visualization of the redirected lysis of Mel642 melanoma cells by PBMCs or CD8+ T cells in the presence of ImmTAC-gp100.
Figure 5: In vivo efficacy of ImmTAC molecules in NOD-SCID and Beige-SCID xenograft models.

Similar content being viewed by others

References

  1. Köhler, G. & Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256, 495–497 (1975).

    Article  Google Scholar 

  2. Isaacs, J.D. et al. Humanised monoclonal antibody therapy for rheumatoid arthritis. Lancet 340, 748–752 (1992).

    Article  CAS  Google Scholar 

  3. Maynard, J. & Georgiou, G. Antibody engineering. Annu. Rev. Biomed. Eng. 2, 339–376 (2000).

    Article  CAS  Google Scholar 

  4. Kufer, P., Lutterbuse, R. & Baeuerle, P.A. A revival of bispecific antibodies. Trends Biotechnol. 22, 238–244 (2004).

    Article  CAS  Google Scholar 

  5. Clynes, R.A., Towers, T.L., Presta, L.G. & Ravetch, J.V. Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets. Nat. Med. 6, 443–446 (2000).

    Article  CAS  Google Scholar 

  6. Cartron, G. et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcγRIIIa gene. Blood 99, 754–758 (2002).

    Article  CAS  Google Scholar 

  7. von Mehren, M., Adams, G.P. & Weiner, L.M. Monoclonal antibody therapy for cancer. Annu. Rev. Med. 54, 343–369 (2003).

    Article  CAS  Google Scholar 

  8. Withoff, S., Helfrich, W., de Leij, L.F. & Molema, G. Bi-specific antibody therapy for the treatment of cancer. Curr. Opin. Mol. Ther. 3, 53–62 (2001).

    CAS  PubMed  Google Scholar 

  9. Bargou, R. et al. Tumor regression in cancer patients by very low doses of a T cell–engaging antibody. Science 321, 974–977 (2008).

    Article  CAS  Google Scholar 

  10. Baeuerle, P.A., Kufer, P. & Bargou, R. BiTE: teaching antibodies to engage T-cells for cancer therapy. Curr. Opin. Mol. Ther. 11, 22–30 (2009).

    CAS  PubMed  Google Scholar 

  11. Kiewe, P. et al. Phase I trial of the trifunctional anti-HER2 x anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clin. Cancer Res. 12, 3085–3091 (2006).

    Article  CAS  Google Scholar 

  12. Sebastian, M. et al. Treatment of malignant pleural effusion with the trifunctional antibody catumaxomab (Removab) (anti-EpCAM x anti-CD3): results of a phase 1/2 study. J. Immunother. 32, 195–202 (2009).

    Article  CAS  Google Scholar 

  13. Cole, D.K. et al. Human TCR-binding affinity is governed by MHC class restriction. J. Immunol. 178, 5727–5734 (2007).

    Article  CAS  Google Scholar 

  14. Boulter, J.M. et al. Stable, soluble T-cell receptor molecules for crystallization and therapeutics. Protein Eng. 16, 707–711 (2003).

    Article  CAS  Google Scholar 

  15. Li, Y. et al. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nat. Biotechnol. 23, 349–354 (2005).

    Article  CAS  Google Scholar 

  16. Holler, P.D. et al. In vitro evolution of a T cell receptor with high affinity for peptide/MHC. Proc. Natl. Acad. Sci. USA 97, 5387–5392 (2000).

    Article  CAS  Google Scholar 

  17. Chlewicki, L.K., Holler, P.D., Monti, B.C., Clutter, M.R. & Kranz, D.M. High-affinity, peptide-specific T cell receptors can be generated by mutations in CDR1, CDR2 or CDR3. J. Mol. Biol. 346, 223–239 (2005).

    Article  CAS  Google Scholar 

  18. Adema, G.J. et al. Melanocyte lineage-specific antigens recognized by monoclonal antibodies NKI-beteb, HMB-50, and HMB-45 are encoded by a single cDNA. Am. J. Pathol. 143, 1579–1585 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Gaugler, B. et al. Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J. Exp. Med. 179, 921–930 (1994).

    Article  CAS  Google Scholar 

  20. Coulie, P.G. et al. A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J. Exp. Med. 180, 35–42 (1994).

    Article  CAS  Google Scholar 

  21. Kawakami, Y. et al. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc. Natl. Acad. Sci. USA 91, 3515–3519 (1994).

    Article  CAS  Google Scholar 

  22. Chen, Y.T. et al. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc. Natl. Acad. Sci. USA 94, 1914–1918 (1997).

    Article  CAS  Google Scholar 

  23. Cox, A.L. et al. Identification of a peptide recognized by five melanoma-specific human cytotoxic T cell lines. Science 264, 716–719 (1994).

    Article  CAS  Google Scholar 

  24. Celis, E. et al. Induction of anti-tumor cytotoxic T lymphocytes in normal humans using primary cultures and synthetic peptide epitopes. Proc. Natl. Acad. Sci. USA 91, 2105–2109 (1994).

    Article  CAS  Google Scholar 

  25. Kawakami, Y. et al. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2–restricted tumor infiltrating lymphocytes. J. Exp. Med. 180, 347–352 (1994).

    Article  CAS  Google Scholar 

  26. Jäger, E. et al. Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2–binding peptide epitopes. J. Exp. Med. 187, 265–270 (1998).

    Article  Google Scholar 

  27. Purbhoo, M.A. et al. Quantifying and imaging NY-ESO-1/LAGE-1–derived epitopes on tumor cells using high affinity T cell receptors. J. Immunol. 176, 7308–7316 (2006).

    Article  CAS  Google Scholar 

  28. Chattopadhyay, P.K. et al. The cytolytic enzymes granyzme A, granzyme B, and perforin: expression patterns, cell distribution, and their relationship to cell maturity and bright CD57 expression. J. Leukoc. Biol. 85, 88–97 (2009).

    Article  CAS  Google Scholar 

  29. Schietinger, A., Philip, M., Liu, R.B., Schreiber, K. & Schreiber, H. Bystander killing of cancer requires the cooperation of CD4+ and CD8+ T cells during the effector phase. J. Exp. Med. 207, 2469–2477 (2010).

    Article  CAS  Google Scholar 

  30. Arstila, T.P. et al. A direct estimate of the human αβ T cell receptor diversity. Science 286, 958–961 (1999).

    Article  CAS  Google Scholar 

  31. Purbhoo, M.A., Irvine, D.J., Huppa, J.B. & Davis, M.M. T cell killing does not require the formation of a stable mature immunological synapse. Nat. Immunol. 5, 524–530 (2004).

    Article  CAS  Google Scholar 

  32. Price, D.A., Klenerman, P., Booth, B.L., Phillips, R.E. & Sewell, A.K. Cytotoxic T lymphocytes, chemokines and antiviral immunity. Immunol. Today 20, 212–216 (1999).

    Article  CAS  Google Scholar 

  33. Quezada, S.A. et al. Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J. Exp. Med. 207, 637–650 (2010).

    Article  CAS  Google Scholar 

  34. Rosenberg, S.A. & Dudley, M.E. Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr. Opin. Immunol. 21, 233–240 (2009).

    Article  CAS  Google Scholar 

  35. Benowitz, S. Rethinking cancer vaccine trials: would new measures of success make a difference? J. Natl. Cancer Inst. 100, 237–238 (2008).

    Article  Google Scholar 

  36. Johnson, L.A. et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 114, 535–546 (2009).

    Article  CAS  Google Scholar 

  37. Moysey, R., Vuidepot, A.L. & Boulter, J.M. Amplification and one-step expression cloning of human T cell receptor genes. Anal. Biochem. 326, 284–286 (2004).

    Article  CAS  Google Scholar 

  38. Dunn, S.M. et al. Directed evolution of human T cell receptor CDR2 residues by phage display dramatically enhances affinity for cognate peptide-MHC without increasing apparent cross-reactivity. Protein Sci. 15, 710–721 (2006).

    Article  CAS  Google Scholar 

  39. Price, D.A. et al. Avidity for antigen shapes clonal dominance in CD8+ T cell populations specific for persistent DNA viruses. J. Exp. Med. 202, 1349–1361 (2005).

    Article  CAS  Google Scholar 

  40. Bertozzi, C.C. et al. Multiple initial culture conditions enhance the establishment of cell lines from primary ovarian cancer specimens. In Vitro Cell. Dev. Biol. Anim. 42, 58–62 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank Sanofi Pasteur for funding affinity maturation of the gp100 and MAGE-A3 mTCRs; C. Yee (Fred Hutchinson Cancer Research Centre, Seattle, Washington, USA), P. Coulie (University of Louvain, Brussels, Belgium) and V. Cerundolo (Weatherall Institute of Molecular Medicine, University of Oxford, UK) for providing T cell clones; Southern Research and Cellvax for conducting mouse xenograft experiments; Southern Research for immunohistochemistry staining; R. Liu for assistance with mouse imaging and tumor measurements; K. Haines (Translational and Correlative Studies Laboratory, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA) for technical support; and A. Secreto, C. Keefer and G. Danet-Desnoyers (Stem Cell and Xenograft Core, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA) for assistance with the established tumor xenograft studies.

Author information

Authors and Affiliations

Authors

Contributions

N.L., N.E.H., P.E.M. and E.G. isolated wild-type mTCRs and carried out the phage display process under the supervision of Y.L.; A.V. and Y.L. were involved in ImmTAC construct optimization; T.M.M., J.G., A.V., E.E.B., N.J.P., N.M.L. and B.J.C. were involved in protein production and biochemical testing; G.B., K.J.A., A.L., N.J.H., K.L., S.J.P., J.V.H. and R.E.D. performed in vitro experiments under the supervision of D.D.W., R.A., D.H.S., A.K.S. and D.A.P. Large-scale production, stability testing, quality control and biochemical testing of ImmTACs was conducted by F.C.B., M.S., A.J., E.E.B., P.T.T. and S.M.D. under the supervision of Y.M. Mouse xenograft experiments were designed, coordinated and conducted by G.P., C.H.J., M.K. and D.D.W. Data analysis and interpretation were performed by D.H.S. and D.A.P.; D.H.S. and D.A.P. wrote the paper. B.K.J. conceived the idea and directed the project. All authors contributed to discussions.

Corresponding author

Correspondence to Bent K Jakobsen.

Ethics declarations

Competing interests

N.L., G.B., K.J.A., T.M.M., N.J.H., J.G., F.C.B., N.J.P., N.M.L., N.E.H., P.E.M., Y.L., B.J.C., M.S., E.E.B., P.T.T., S.J.P., R.E.D., J.V.H., S.M.D., R.A., A.J., Y.M., A.V., D.D.W., D.H.S. and B.K.J. are employed by ImmunoCore Ltd., and the reagents studied in this manuscript were developed by ImmunoCore Ltd.

Supplementary information

Supplementary Text and Figures

Supplementary Methods and Supplementary Table 1 (PDF 305 kb)

Supplementary Video 1a

Supplementary Video 1a (MOV 382 kb)

Supplementary Video 1b

Supplementary Video 1b (MOV 3096 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Liddy, N., Bossi, G., Adams, K. et al. Monoclonal TCR-redirected tumor cell killing. Nat Med 18, 980–987 (2012). https://doi.org/10.1038/nm.2764

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2764

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