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.

  • Article
  • Published:

Priming of naive T cells inside tumors leads to eradication of established tumors

Abstract

The tumor barrier comprised of nonantigenic stromal cells may contribute to the failure of tumor rejection. The tumor-necrosis factor superfamily member LIGHT (also known as TNFSF-14) is a ligand of stromal cell–expressed lymphotoxin-β receptor and T cell–expressed herpes viral entry mediator (HVEM). Here we show that forced expression of LIGHT in the tumor environment induces a massive infiltration of naive T lymphocytes that correlates with an upregulation of both chemokine production and expression of adhesion molecules. Activation of these infiltrating T cells, possibly through HVEM, leads to the rejection of established, highly progressive tumors at local and distal sites. Our study indicates that targeting the tumor barrier may be an effective strategy for cancer 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: Growth kinetics of LIGHT-expressing Ag104Ld and parental tumors in C3B6F1 and B6-Rag1−/− mice.
Figure 2: Increased infiltration of CD8+ T cells in LIGHT-expressing Ag104Ld tumor tissues.
Figure 3: The modified extracellular domain of LIGHT is sufficient to costimulate purified T cell responses.
Figure 4: Increased LTβR-regulated chemokines and adhesion molecules in Ag104Ld-LIGHT tumors.
Figure 5: LIGHT-mediated Ag104Ld tumor environment recruits and activates naive 2C T cells leading to tumor rejection.
Figure 6: Injection of LIGHT-expressing Ag104Ld cells leads to eradication of established parental tumors and B16-OVA tumors.

Similar content being viewed by others

References

  1. Boon, T. & van der Bruggen, P. Human tumor antigens recognized by T lymphocytes. J. Exp. Med. 183, 725–729 (1996).

    Article  CAS  Google Scholar 

  2. Rosenberg, S.A. A new era for cancer immunotherapy based on the genes that encode cancer antigens. Immunity 10, 281–287 (1999).

    Article  CAS  Google Scholar 

  3. Groh, V., Wu, J., Yee, C. & Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419, 734–738 (2002).

    Article  CAS  Google Scholar 

  4. Berzofsky, J.A., Ahlers, J.D. & Belyakov, I.M. Strategies for designing and optimizing new generation vaccines. Nat. Rev. Immunol. 1, 209–219 (2001).

    Article  CAS  Google Scholar 

  5. Pardoll, D.M. Spinning molecular immunology into successful immunotherapy. Nat. Rev. Immunol. 2, 227–238 (2002).

    Article  CAS  Google Scholar 

  6. Chambers, C.A., Kuhns, M.S., Egen, J.G. & Allison, J.P. CTLA-4-mediated inhibition in regulation of T cell responses: mechanisms and manipulation in tumor immunotherapy. Annu. Rev. Immunol. 19, 565–594 (2001).

    Article  CAS  Google Scholar 

  7. Terabe, M. et al. NKT cell–mediated repression of tumor immunosurveillance by IL-13 and the IL-4R-STAT6 pathway. Nat. Immunol. 1, 515–520 (2000).

    Article  CAS  Google Scholar 

  8. Ochsenbein, A.F. et al. Immune surveillance against a solid tumor fails because of immunological ignorance. Proc. Natl. Acad. Sci. USA 96, 2233–2238 (1999).

    Article  CAS  Google Scholar 

  9. Singh, S., Ross, S.R., Acena, M., Rowley, D.A. & Schreiber, H. Stroma is critical for preventing or permitting immunological destruction of antigenic cancer cells. J. Exp. Med. 175, 139–146 (1992).

    Article  CAS  Google Scholar 

  10. Perdrizet, G.A. et al. Animals bearing malignant grafts reject normal grafts that express through gene transfer the same antigen. J. Exp. Med. 171, 1205–1220 (1990).

    Article  CAS  Google Scholar 

  11. Sarma, S. et al. Cytotoxic T lymphocytes to an unmutated tumor rejection antigen P1A: normal development but restrained effector function in vivo . J. Exp. Med. 189, 811–820 (1999).

    Article  CAS  Google Scholar 

  12. Wick, M. et al. Antigenic cancer cells grow progressively in immune hosts without evidence for T cell exhaustion or systemic anergy. J. Exp. Med. 186, 229–238 (1997).

    Article  CAS  Google Scholar 

  13. Hanson, H.L. et al. Eradication of established tumors by CD8+ T cell adoptive immunotherapy. Immunity 13, 265–276 (2000).

    Article  CAS  Google Scholar 

  14. Dudley, M.E. et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298, 850–854 (2002).

    Article  CAS  Google Scholar 

  15. Rosenberg, S.A. Progress in the development of immunotherapy for the treatment of patients with cancer. J. Intern. Med. 250, 462–475 (2001).

    Article  CAS  Google Scholar 

  16. Lee, K.H. et al. Increased vaccine-specific T cell frequency after peptide-based vaccination correlates with increased susceptibility to in vitro stimulation but does not lead to tumor regression. J. Immunol. 163, 6292–6300 (1999).

    CAS  PubMed  Google Scholar 

  17. Mauri, D.N. et al. LIGHT, a new member of the TNF superfamily, and lymphotoxin α are ligands for herpesvirus entry mediator. Immunity 8, 21–30 (1998).

    Article  CAS  Google Scholar 

  18. Ngo, V.N. et al. Lymphotoxin α/β and tumor necrosis factor are required for stromal cell expression of homing chemokines in B and T cell areas of the spleen. J. Exp. Med. 189, 403–412 (1999).

    Article  CAS  Google Scholar 

  19. Wang, J. et al. The complementation of lymphotoxin deficiency with LIGHT, a newly discovered TNF family member, for the restoration of secondary lymphoid structure and function. Eur. J. Immunol. 32, 1969–1979 (2002).

    Article  CAS  Google Scholar 

  20. Ueno, T. et al. Role for CCR7 ligands in the emigration of newly generated T lymphocytes from the neonatal thymus. Immunity 16, 205–218 (2002).

    Article  CAS  Google Scholar 

  21. Gunn, M.D. et al. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J. Exp. Med. 189, 451–460 (1999).

    Article  CAS  Google Scholar 

  22. Tamada, K. et al. Modulation of T-cell-mediated immunity in tumor and graft-versus-host disease models through the LIGHT co-stimulatory pathway. Nat. Med. 6, 283–289 (2000).

    Article  CAS  Google Scholar 

  23. Dranoff, G. et al. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc. Natl. Acad. Sci. USA 90, 3539–3543 (1993).

    Article  CAS  Google Scholar 

  24. Lenschow, D.J., Walunas, T.L. & Bluestone, J.A. CD28/B7 system of T cell costimulation. Annu. Rev. Immunol. 14, 233–258 (1996).

    Article  CAS  Google Scholar 

  25. Chen, L. et al. Tumor immunogenicity determines the effect of B7 costimulation on T cell-mediated tumor immunity. J. Exp. Med. 179, 523–532 (1994).

    Article  CAS  Google Scholar 

  26. Montgomery, R.I., Warner, M.S., Lum, B.J. & Spear, P.G. Herpes simplex virus-1 entry into cells mediated by a novel member of the TNF/NGF receptor family. Cell 87, 427–436 (1996).

    Article  CAS  Google Scholar 

  27. Kwon, B.S. et al. A newly identified member of the tumor necrosis factor receptor superfamily with a wide tissue distribution and involvement in lymphocyte activation. J. Biol. Chem. 272, 14272–14276 (1997).

    Article  CAS  Google Scholar 

  28. Harrop, J.A. et al. Antibodies to TR2 (herpesvirus entry mediator), a new member of the TNF receptor superfamily, block T cell proliferation, expression of activation markers, and production of cytokines. J. Immunol. 161, 1786–1794 (1998).

    CAS  PubMed  Google Scholar 

  29. Fu, Y.X. & Chaplin, D.D. Development and maturation of secondary lymphoid tissues. Annu. Rev. Immunol. 17, 399–433 (1999).

    Article  CAS  Google Scholar 

  30. Cyster, J.G. Chemokines and cell migration in secondary lymphoid organs. Science 286, 2098–2102 (1999).

    Article  CAS  Google Scholar 

  31. Farber, J.M. Mig and IP-10: CXC chemokines that target lymphocytes. J. Leukoc. Biol. 61, 246–257 (1997).

    Article  CAS  Google Scholar 

  32. Kang, H.S. et al. Signaling via LTβR on the lamina propria stromal cells of the gut is required for IgA production. Nat. Immunol. 3, 576–582 (2002).

    Article  CAS  Google Scholar 

  33. Yu, P., Spiotto, M.T., Lee, Y., Schreiber, H. & Fu, Y.X. Complementary role of CD4+ T cells and secondary lymphoid tissues for cross-presentation of tumor antigen to CD8+ T cells. J. Exp. Med. 197, 985–995 (2003).

    Article  CAS  Google Scholar 

  34. Wherry, E.J. et al. Lineage relationship and protective immunity of memory CD8+ T cell subsets. Nat. Immunol. 4, 225–234 (2003).

    Article  CAS  Google Scholar 

  35. Chen, L. Immunological ignorance of silent antigens as an explanation of tumor evasion. Immunol. Today 19, 27–30 (1998).

    Article  CAS  Google Scholar 

  36. Ochsenbein, A.F. et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 411, 1058–1064 (2001).

    Article  CAS  Google Scholar 

  37. Spiotto, M.T. et al. Increasing tumor antigen expression overcomes 'ignorance' to solid tumors via crosspresentation by bone marrow–derived stromal cells. Immunity 17, 737–747 (2002).

    Article  CAS  Google Scholar 

  38. Schrama, D. et al. Targeting of lymphotoxin-α to the tumor elicits an efficient immune response associated with induction of peripheral lymphoid-like tissue. Immunity 14, 111–121 (2001).

    Article  CAS  Google Scholar 

  39. Sharma, S. et al. Secondary lymphoid tissue chemokine mediates T cell–dependent antitumor responses in vivo. J. Immunol. 164, 4558–4563 (2000).

    Article  CAS  Google Scholar 

  40. Bai, X.F. et al. On the site and mode of antigen presentation for the initiation of clonal expansion of CD8 T cells specific for a natural tumor antigen. Cancer Res. 61, 6860–6867 (2001).

    CAS  PubMed  Google Scholar 

  41. Wang, J. et al. The regulation of T cell homeostasis and autoimmunity by T cell–derived LIGHT. J. Clin. Invest. 108, 1771–1780 (2001).

    Article  CAS  Google Scholar 

  42. Sharma, S. et al. SLC/CCL21-mediated antitumor responses require IFNγ, MIG/CXCL9 and IP-10/CXCL10. Mol. Cancer 2, 22 (2003).

    Article  Google Scholar 

  43. Zhai, Y. et al. LIGHT, a novel ligand for lymphotoxin β receptor and TR2/HVEM induces apoptosis and suppresses in vivo tumor formation via gene transfer. J. Clin. Invest. 102, 1142–1151 (1998).

    Article  CAS  Google Scholar 

  44. Harrop, J.A. et al. Herpesvirus entry mediator ligand (HVEM-L), a novel ligand for HVEM/TR2, stimulates proliferation of T cells and inhibits HT29 cell growth. J. Biol. Chem. 273, 27548–27556 (1998).

    Article  CAS  Google Scholar 

  45. Browning, J.L. et al. Signaling through the lymphotoxin β receptor induces the death of some adenocarcinoma tumor lines. J. Exp. Med. 183, 867–878 (1996).

    Article  CAS  Google Scholar 

  46. Ward, P.L., Koeppen, H., Hurteau, T. & Schreiber, H. Tumor antigens defined by cloned immunological probes are highly polymorphic and are not detected on autologous normal cells. J. Exp. Med. 170, 217–232 (1989).

    Article  CAS  Google Scholar 

  47. Yu, P. et al. B cells control the migration of a subset of dendritic cells into B cell follicles via CXC chemokine ligand 13 in a lymphotoxin-dependent fashion. J. Immunol. 168, 5117–5123 (2002).

    Article  CAS  Google Scholar 

  48. Wu, Q. et al. The requirement of membrane lymphotoxin for the presence of dendritic cells in lymphoid tissues. J. Exp. Med. 190, 629–638 (1999).

    Article  CAS  Google Scholar 

  49. Hathcock, K.S. T cell depletion by cytotoxic elimination. in Current Protocols in Immunology (Coligan, J.E. et al.) 3.4.1–3.4.3 (Wiley, New York, 1991).

    Google Scholar 

Download references

Acknowledgements

We thank L. Chen for advice and support, and M. Spiotto and J. Lo for their technical assistance. This work was supported by grants from the NIH (R01-HD37104, R01-DK58897 and P01-CA09296-01). P.Y. is a recipient of an NIH training grant (5T32DK07074).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yang-Xin Fu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yu, P., Lee, Y., Liu, W. et al. Priming of naive T cells inside tumors leads to eradication of established tumors. Nat Immunol 5, 141–149 (2004). https://doi.org/10.1038/ni1029

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni1029

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