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:

Oncolytic adenovirus encoding LIGHT (TNFSF14) inhibits tumor growth via activating anti-tumor immune responses in 4T1 mouse mammary tumor model in immune competent syngeneic mice

Abstract

LIGHT, also known as tumor-necrosis factor (TNF) superfamily member 14 (TNFSF14), is predominantly expressed on activated immune cells and some tumor cells. LIGHT is a pivotal regulator both for recruiting and activating immune cells in the tumor lesions. In this study, we armed human telomerase reverse transcriptase (TERT) promoter controlled oncolytic adenovirus with LIGHT to generate rAd.Light. rAd.Light effectively transduced both human and mouse breast tumor cell lines in vitro, and expressed LIGHT protein on the surface of tumor cells. Both rAd.Null, and rAd.Light could replicate in human breast cancer cells, and produced cytotoxicity to human and mouse mammary tumor cells. rAd.Light induced apoptosis resulting in tumor cell death. Using a subcutaneous model of 4T1 cells in BALB/c mice, rAd.Light was delivered intratumorally to evaluate the anti-tumor responses. Both rAd.Light and rAd.Null significantly inhibited the tumor growth, but rAd.Light produced much stronger anti-tumor effects. Histopathological analysis showed the infiltration of T lymphocytes in the tumor tissues. rAd.Light also induced stronger cellular apoptosis than rAd.Null in the tumors. Interestingly, on day 15, compared to rAd.Null, there was a significant reduction of Tregs following rAd.Light treatment. rAd.Light significantly increased Th1 cytokine interleukin (IL)-2 expression, and reduced Th2 cytokines expression, such as transforming growth factor β (TGF-β) and IL-10 in the tumors. These results suggest rAd.Light induced activation of anti-tumor immune responses. In conclusion, rAd.Light produced anti-tumor effect in a subcutaneous model of breast cancer via inducing tumor apoptosis and evoking strong anti-tumor immune responses. Therefore, rAd.Light has great promise to be developed as an effective therapeutic approach for the treatment of breast 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: Construction of oncolytic adenovirus rAd.Light, viral replication, viral-induced cytotoxicity, and viral-mediated LIGHT expression in breast cancer cell lines.
Fig. 2: Oncolytic adenovirus expressing LIGHT, rAd.Light produced anti-tumor responses in subcutaneous model of mouse breast cancer.
Fig. 3: rAd.Light promotes tumor infiltration of CD3+ T cells, produces LIGHT protein, promotes infiltration of T lymphocytes and regulates Th1/Th2 cytokines balance in tumor microenvironment.
Fig. 4: Intratumoral delivery of rAd.Light up-regulates CD8+ T lymphocytes, promoting CD4+ T memory cells and reducing regulatory T cells in peripheral blood.

Similar content being viewed by others

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J Clin. 2018;68:394–424.

    Google Scholar 

  2. Soysal SD, Tzankov A, Muenst SE. Role of the tumor microenvironment in breast cancer. Pathobiology. 2015;82:142–52.

    CAS  PubMed  Google Scholar 

  3. Duan H. Novel therapeutic strategies for solid tumor based on body’s intrinsic antitumor immune system. Cell Physiol Biochem. 2018;47:441–57.

    CAS  PubMed  Google Scholar 

  4. Parker KH, Beury DW, Ostrand-Rosenberg S. Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv cancer Res. 2015;128:95–139.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Truffi M, Mazzucchelli S, Bonizzi A, Sorrentino L, Allevi R, Vanna R, et al. Nano-strategies to target breast cancer-associated fibroblasts: rearranging the tumor microenvironment to achieve antitumor efficacy. Int J Mol Sci. 2019;20:E1263. https://doi.org/10.3390/ijms20061263.

    Article  CAS  PubMed  Google Scholar 

  6. Chen C, Bai L, Cao F, Wang S, He H, Song M, et al. Targeting LIN28B reprograms tumor glucose metabolism and acidic microenvironment to suppress cancer stemness and metastasis. Oncogene. 2019;38:4527–39.

    CAS  PubMed  Google Scholar 

  7. Amerizadeh F, Bahrami A, Khazaei M, Hesari A, Rezayi M, Talebian S, et al. Current status and future prospects of transforming growth factor-beta as a potential prognostic and therapeutic target in the treatment of breast cancer. J Cell Biochem. 2019. https://doi.org/10.1002/jcb.27831. [Epub ahead of print].

  8. Zhai Y, Guo R, Hsu TL, Yu GL, Ni J, Kwon BS, et al. LIGHT, a novel ligand for lymphotoxin beta receptor and TR2/HVEM induces apoptosis and suppresses in vivo tumor formation via gene transfer. J Clin Investig. 1998;102:1142–51.

    CAS  PubMed  Google Scholar 

  9. Pasero C, Barbarat B, Just-Landi S, Bernard A, Aurran-Schleinitz T, Rey J, et al. A role for HVEM, but not lymphotoxin-beta receptor, in LIGHT-induced tumor cell death and chemokine production. Eur J Immunol. 2009;39:2502–14.

    CAS  PubMed  Google Scholar 

  10. Maker AV, Ito H, Mo Q, Weisenberg E, Qin LX, Turcotte S, et al. Genetic evidence that intratumoral T-cell proliferation and activation are associated with recurrence and survival in patients with resected colorectal liver metastases. Cancer Immunol Res. 2015;3:380–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Maker AV. Precise identification of immunotherapeutic targets for solid malignancies using clues within the tumor microenvironment-evidence to turn on the LIGHT. Oncoimmunology. 2016;5:e1069937.

    PubMed  Google Scholar 

  12. Treps L. EnLIGHTenment of tumor vessel normalization and immunotherapy in glioblastoma. J Pathol. 2018;246:3–6.

    CAS  PubMed  Google Scholar 

  13. Yan L, Da Silva DM, Verma B, Gray A, Brand HE, Skeate JG, et al. Forced LIGHT expression in prostate tumors overcomes Treg mediated immunosuppression and synergizes with a prostate tumor therapeutic vaccine by recruiting effector T lymphocytes. Prostate. 2015;75:280–91.

    CAS  PubMed  Google Scholar 

  14. Johansson-Percival A, Li ZJ, Lakhiani DD, He B, Wang X, Hamzah J, et al. Intratumoral LIGHT restores pericyte contractile properties and vessel integrity. Cell Rep. 2015;13:2687–98.

    CAS  PubMed  Google Scholar 

  15. Johansson-Percival A, He B, Li ZJ, Kjellen A, Russell K, Li J, et al. De novo induction of intratumoral lymphoid structures and vessel normalization enhances immunotherapy in resistant tumors. Nat Immunol. 2017;18:1207–17.

    CAS  PubMed  Google Scholar 

  16. Pearl TM, Markert JM, Cassady KA, Ghonime MG. Oncolytic virus-based cytokine expression to improve immune activity in brain and solid tumors. Mol Ther oncolytics. 2019;13:14–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Yang Y, Xu W, Peng D, Wang H, Zhang X, Xiao F, et al. An oncolytic adenovirus targeting transforming growth factor beta inhibits protumorigenic signals and produces immune activation: a novel approach to enhance anti-PD-1 and anti-CTLA-4 therapy. Human Gene Ther. 2019;30:1117–32.

    CAS  Google Scholar 

  18. Zhao H, Wang H, Kong F, Xu W, Wang T, Xiao F, et al. Oncolytic adenovirus rAd.DCN inhibits breast tumor growth and lung metastasis in an immune-competent orthotopic xenograft model. Hum Gene Ther. 2019;30:197–210.

    CAS  PubMed  Google Scholar 

  19. Liu Z, Yang Y, Zhang X, Wang H, Xu W, Xiao F, et al. An oncolytic adenovirus encoding decorin and granulocyte macrophage colony stimulating factor inhibits tumor growth in a colorectal tumor model by targeting pro-tumorigenic signals and via immune activation. Hum Gene Ther. 2017;28:667–80.

    CAS  PubMed  Google Scholar 

  20. Yang Y, Xu W, Neill T, Hu Z, Wang CH, Xiao X, et al. Systemic delivery of an oncolytic adenovirus expressing decorin for the treatment of breast cancer bone metastases. Hum Gene Ther. 2015;26:813–25.

    PubMed  PubMed Central  Google Scholar 

  21. Hu ZB, Wu CT, Wang H, Zhang QW, Wang L, Wang RL, et al. A simplified system for generating oncolytic adenovirus vector carrying one or two transgenes. Cancer Gene Ther. 2008;15:173–82.

    CAS  PubMed  Google Scholar 

  22. Qiao G, Qin J, Kunda N, Calata JF, Mahmud DL, Gann P, et al. LIGHT elevation enhances immune eradication of colon cancer metastases. Cancer Res. 2017;77:1880–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Yu P, Lee Y, Liu W, Chin RK, Wang J, Wang Y, et al. Priming of naive T cells inside tumors leads to eradication of established tumors. Nat Immunol. 2004;5:141–9.

    CAS  PubMed  Google Scholar 

  24. Hu Z, Gupta J, Zhang Z, Gerseny H, Berg A, Chen YJ, et al. Systemic delivery of oncolytic adenoviruses targeting transforming growth factor-beta inhibits established bone metastasis in a prostate cancer mouse model. Hum Gene Ther. 2012;23:871–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Hu Z, Gerseny H, Zhang Z, Chen YJ, Berg A, Stock S, et al. Oncolytic adenovirus expressing soluble TGFbeta receptor II-Fc-mediated inhibition of established bone metastases: a safe and effective systemic therapeutic approach for breast cancer. Mol Ther. 2011;19:1609–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Katayose D, Gudas J, Nguyen H, Srivastava S, Cowan KH, Seth P. Cytotoxic effects of adenovirus-mediated wild-type p53 protein expression in normal and tumor mammary epithelial cells. Clin Cancer Res. 1995;1:889–97.

    CAS  PubMed  Google Scholar 

  27. Ries SJ, Brandts CH, Chung AS, Biederer CH, Hann BC, Lipner EM, et al. Loss of p14ARF in tumor cells facilitates replication of the adenovirus mutant dl1520 (ONYX-015). Nat Med. 2000;6:1128–33.

    CAS  PubMed  Google Scholar 

  28. Andtbacka RH, Ross M, Puzanov I, Milhem M, Collichio F, Delman KA, et al. Patterns of clinical response with Talimogene Laherparepvec (T-VEC) in patients with melanoma treated in the OPTiM phase III clinical trial. Ann Surgical Oncol. 2016;23:4169–77.

    Google Scholar 

  29. Poh A. First oncolytic viral therapy for melanoma. Cancer Discov. 2016;6:6.

    PubMed  Google Scholar 

  30. Breitbach CJ, Moon A, Burke J, Hwang TH, Kirn DH. A phase 2, open-label, randomized study of Pexa-Vec (JX-594) administered by intratumoral injection in patients with unresectable primary hepatocellular carcinoma. Methods Mol Biol. 2015;1317:343–57.

    PubMed  Google Scholar 

  31. Nishio N, Dotti G. Oncolytic virus expressing RANTES and IL-15 enhances function of CAR-modified T cells in solid tumors. Oncoimmunology. 2015;4:e988098.

    PubMed  PubMed Central  Google Scholar 

  32. Roy DM, Walsh LA. Candidate prognostic markers in breast cancer: focus on extracellular proteases and their inhibitors. Breast Cancer (Dove. Med Press). 2014;6:81–91.

    CAS  Google Scholar 

  33. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol. 2012;196:395–406.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Criscitiello C, Esposito A, Curigliano G. Tumor-stroma crosstalk: targeting stroma in breast cancer. Curr Opin Oncol. 2014;26:551–5.

    CAS  PubMed  Google Scholar 

  35. Nwabo Kamdje AH, Seke Etet PF, Vecchio L, Muller JM, Krampera M, Lukong KE. Signaling pathways in breast cancer: therapeutic targeting of the microenvironment. Cell Signal. 2014;26:2843–56.

    CAS  PubMed  Google Scholar 

  36. Ma HS, Poudel B, Torres ER, Sidhom JW, Robinson TM, Christmas B, et al. A CD40 agonist and PD-1 antagonist antibody reprogram the microenvironment of nonimmunogenic tumors to allow T-cell-mediated anticancer activity. Cancer Immunol Res. 2019;7:428–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Grither WR, Longmore GD. Inhibition of tumor-microenvironment interaction and tumor invasion by small-molecule allosteric inhibitor of DDR2 extracellular domain. Proc Natl Acad Sci USA. 2018;115:E7786–94.

    CAS  PubMed  Google Scholar 

  38. Allen M, Louise Jones J. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J Pathol. 2011;223:162–76.

    CAS  PubMed  Google Scholar 

  39. Bohling SD, Allison KH. Immunosuppressive regulatory T cells are associated with aggressive breast cancer phenotypes: a potential therapeutic target. Mod Pathol. 2008;21:1527–32.

    CAS  PubMed  Google Scholar 

  40. Ohara M, Yamaguchi Y, Matsuura K, Murakami S, Arihiro K, Okada M. Possible involvement of regulatory T cells in tumor onset and progression in primary breast cancer. Cancer Immunol, Immunotherapy. 2009;58:441–7.

    CAS  Google Scholar 

  41. Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM, et al. Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature. 2011;470:548–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Qin JZ, Upadhyay V, Prabhakar B, Maker AV. Shedding LIGHT (TNFSF14) on the tumor microenvironment of colorectal cancer liver metastases. J Transl Med. 2013;11:70.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Yu P, Fu YX. Targeting tumors with LIGHT to generate metastasis-clearing immunity. Cytokine Growth Factor Rev. 2008;19:285–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Tran TTP, Eichholz K, Amelio P, Moyer C, Nemerow GR, Perreau M, et al. Humoral immune response to adenovirus induce tolerogenic bystander dendritic cells that promote generation of regulatory T cells. PLoS Pathog. 2018;14:e1007127.

    PubMed  PubMed Central  Google Scholar 

  45. Holmes TD, Wilson EB, Black EV, Benest AV, Vaz C, Tan B, et al. Licensed human natural killer cells aid dendritic cell maturation via TNFSF14/LIGHT. Proc Natl Acad Sci USA. 2014;111:E5688–96.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by Nova Program of Beijing (Z171100001117118). Funding at NorthShore was provided by a Clinical and Translational Science Award (PS).

Author information

Authors and Affiliations

Authors

Contributions

HW and PS, conceived and designed the work; SD, YL, WX, CL, XD, and HZ, performed experiments and collected data; YY, BP and AM, analyze and interpret the results; HW and PS wrote the manuscript; PS, BP, and AM revised the manuscript and modified the language.

Corresponding authors

Correspondence to Prem Seth or Hua Wang.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dai, S., Lv, Y., Xu, W. et al. Oncolytic adenovirus encoding LIGHT (TNFSF14) inhibits tumor growth via activating anti-tumor immune responses in 4T1 mouse mammary tumor model in immune competent syngeneic mice. Cancer Gene Ther 27, 923–933 (2020). https://doi.org/10.1038/s41417-020-0173-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-020-0173-z

This article is cited by

Search

Quick links