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:

An oncolytic herpesvirus expressing E-cadherin improves survival in mouse models of glioblastoma

A Publisher Correction to this article was published on 03 January 2019

This article has been updated

Abstract

The efficacy of oncolytic herpes simplex virus (oHSV) is limited by rapid viral clearance by innate immune effector cells and poor intratumoral viral spread. We combine two approaches to overcome these barriers: inhibition of natural killer (NK) cells and enhancement of intratumoral viral spread. We engineered an oHSV to express CDH1, encoding E-cadherin, an adherent molecule and a ligand for KLRG1, an inhibitory receptor expressed on NK cells. In vitro, infection with this engineered virus, named OV-CDH1, induced high surface E-cadherin expression on infected glioblastoma (GBM) cells, which typically lack endogenous E-cadherin. Ectopically expressed E-cadherin enhanced the spread of OV-CDH1 by facilitating cell-to-cell infection and viral entry and reduced viral clearance by selectively protecting OV-CDH1-infected cells from KLRG1+ NK cell killing. In vivo, OV-CDH1 treatment substantially prolonged the survival in GBM-bearing mouse models, primarily because of improved viral spread rather than inhibition of NK cell activity. Thus, virus-induced overexpression of E-cadherin may be a generalizable strategy for improving cancer virotherapy.

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: OV-CDH1 infection reduces the cytotoxicity of human NK cells against OV-infected GBM cells.
Figure 2: Cell–cell fusion facilitates viral spread of OV-CDH1.
Figure 3: Cadherin interaction facilitates cell-to-cell infection by OV-CDH1.
Figure 4: E-cadherin may accelerate viral entry and virus production.
Figure 5: OV-CDH1 improves the efficacy of GBM virotherapy in vivo.
Figure 6: OV-CDH1 treatment leads to increased intracranial NK cell infiltration, enhanced viral spread, viral production and oncolysis.

Similar content being viewed by others

Change history

  • 19 December 2018

    In the HTML and PDF versions of this article initially published online, KLRG1 should have read KLRG1+ in the first instance of the phrase in the Figure 1e legend reading "For KLRG1 NK cells, uninfected vs. OV-Q1." In the HTML version, KLRG1 should have read KLRG1+ in the following locations: the first instance in the Figure 1b legend, the first instance in the Figure 1c legend, and the first instance in the Figure 1e legend. Finally, 1 × 105 in the Figure 5 legend should have read 1 × 105. The errors have been corrected in the print, PDF and HTML versions of this article.

References

  1. Markert, J.M. et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol. Ther. 17, 199–207 (2009).

    CAS  PubMed  Google Scholar 

  2. Kaufman, H.L. & Bines, S.D. OPTIM trial: a Phase III trial of an oncolytic herpes virus encoding GM-CSF for unresectable stage III or IV melanoma. Future Oncol. 6, 941–949 (2010).

    CAS  PubMed  Google Scholar 

  3. Hu, J.C. et al. A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin. Cancer Res. 12, 6737–6747 (2006).

    CAS  PubMed  Google Scholar 

  4. Nakamori, M. et al. Effective therapy of metastatic ovarian cancer with an oncolytic herpes simplex virus incorporating two membrane fusion mechanisms. Clin. Cancer Res. 9, 2727–2733 (2003).

    CAS  PubMed  Google Scholar 

  5. Pol, J., Kroemer, G. & Galluzzi, L. First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology 5, e1115641 (2015).

    PubMed  PubMed Central  Google Scholar 

  6. Ikeda, K. et al. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat. Med. 5, 881–887 (1999).

    CAS  PubMed  Google Scholar 

  7. Alvarez-Breckenridge, C.A. et al. NK cells impede glioblastoma virotherapy through NKp30 and NKp46 natural cytotoxicity receptors. Nat. Med. 18, 1827–1834 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Han, J. et al. TGFβ treatment enhances glioblastoma virotherapy by inhibiting the innate immune response. Cancer Res. 75, 5273–5282 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kolodkin-Gal, D. et al. Herpes simplex virus type 1 preferentially targets human colon carcinoma: role of extracellular matrix. J. Virol. 82, 999–1010 (2008).

    CAS  PubMed  Google Scholar 

  10. Fulci, G. et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc. Natl. Acad. Sci. USA 103, 12873–12878 (2006).

    CAS  PubMed  Google Scholar 

  11. Ayala-Breton, C., Suksanpaisan, L., Mader, E.K., Russell, S.J. & Peng, K.W. Amalgamating oncolytic viruses to enhance their safety, consolidate their killing mechanisms, and accelerate their spread. Mol. Ther. 21, 1930–1937 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Guedan, S. et al. Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth. Mol. Ther. 18, 1275–1283 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Simpson, G.R. et al. Combination of a fusogenic glycoprotein, prodrug activation, and oncolytic herpes simplex virus for enhanced local tumor control. Cancer Res. 66, 4835–4842 (2006).

    CAS  PubMed  Google Scholar 

  14. Ito, M. et al. Killer cell lectin-like receptor G1 binds three members of the classical cadherin family to inhibit NK cell cytotoxicity. J. Exp. Med. 203, 289–295 (2006).

    PubMed  PubMed Central  Google Scholar 

  15. Li, Y. et al. Structure of natural killer cell receptor KLRG1 bound to E-cadherin reveals basis for MHC-independent missing self recognition. Immunity 31, 35–46 (2009).

    PubMed  PubMed Central  Google Scholar 

  16. Schwartzkopff, S. et al. Tumor-associated E-cadherin mutations affect binding to the killer cell lectin-like receptor G1 in humans. J. Immunol. 179, 1022–1029 (2007).

    CAS  PubMed  Google Scholar 

  17. Drees, F., Pokutta, S., Yamada, S., Nelson, W.J. & Weis, W.I. Alpha-catenin is a molecular switch that binds E-cadherin-beta-catenin and regulates actin-filament assembly. Cell 123, 903–915 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Tachibana, K. et al. Two cell adhesion molecules, nectin and cadherin, interact through their cytoplasmic domain-associated proteins. J. Cell Biol. 150, 1161–1176 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Geraghty, R.J., Krummenacher, C., Cohen, G.H., Eisenberg, R.J. & Spear, P.G. Entry of alphaherpesviruses mediated by poliovirus receptor-related protein 1 and poliovirus receptor. Science 280, 1618–1620 (1998).

    CAS  PubMed  Google Scholar 

  20. Stupp, R. et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10, 459–466 (2009).

    CAS  PubMed  Google Scholar 

  21. Terada, K., Wakimoto, H., Tyminski, E., Chiocca, E.A. & Saeki, Y. Development of a rapid method to generate multiple oncolytic HSV vectors and their in vivo evaluation using syngeneic mouse tumor models. Gene Ther. 13, 705–714 (2006).

    CAS  PubMed  Google Scholar 

  22. Mineta, T., Rabkin, S.D., Yazaki, T., Hunter, W.D. & Martuza, R.L. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat. Med. 1, 938–943 (1995).

    CAS  PubMed  Google Scholar 

  23. Gottardi, C.J., Wong, E. & Gumbiner, B.M. E-cadherin suppresses cellular transformation by inhibiting beta-catenin signaling in an adhesion-independent manner. J. Cell Biol. 153, 1049–1060 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Bolyard, C. et al. BAI1 orchestrates macrophage inflammatory response to HSV infection—implications for oncolytic viral therapy. Clin. Cancer Res. 23, 1809–1819 (2017).

    CAS  PubMed  Google Scholar 

  25. Kim, I.J., Chouljenko, V.N., Walker, J.D. & Kousoulas, K.G. Herpes simplex virus 1 glycoprotein M and the membrane-associated protein UL11 are required for virus-induced cell fusion and efficient virus entry. J. Virol. 87, 8029–8037 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Qian, X., Karpova, T., Sheppard, A.M., McNally, J. & Lowy, D.R. E-cadherin-mediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. EMBO J. 23, 1739–1748 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Labernadie, A. et al. A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat. Cell Biol. 19, 224–237 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Halford, W.P., Balliet, J.W. & Gebhardt, B.M. Re-evaluating natural resistance to herpes simplex virus type 1. J. Virol. 78, 10086–10095 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Cheema, T.A. et al. Multifaceted oncolytic virus therapy for glioblastoma in an immunocompetent cancer stem cell model. Proc. Natl. Acad. Sci. USA 110, 12006–12011 (2013).

    CAS  PubMed  Google Scholar 

  30. Lopez, C. Genetics of natural resistance to herpesvirus infections in mice. Nature 258, 152–153 (1975).

    CAS  PubMed  Google Scholar 

  31. Nakashima, H. et al. Toxicity and efficacy of a novel GADD34-expressing oncolytic HSV-1 for the treatment of experimental glioblastoma. Clin. Cancer Res. 24, 2574–2584 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Bridle, B.W. et al. HDAC inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity during tumor immunotherapy. Mol. Ther. 21, 887–894 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. MacTavish, H. et al. Enhancement of vaccinia virus based oncolysis with histone deacetylase inhibitors. PLoS One 5, e14462 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Altomonte, J. et al. Enhanced oncolytic potency of vesicular stomatitis virus through vector-mediated inhibition of NK and NKT cells. Cancer Gene Ther. 16, 266–278 (2009).

    CAS  PubMed  Google Scholar 

  35. Fehniger, T.A. et al. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: a potential new link between adaptive and innate immunity. Blood 101, 3052–3057 (2003).

    CAS  PubMed  Google Scholar 

  36. Nagler, A., Lanier, L.L., Cwirla, S. & Phillips, J.H. Comparative studies of human FcRIII-positive and negative natural killer cells. J. Immunol. 143, 3183–3191 (1989).

    CAS  PubMed  Google Scholar 

  37. Markert, J.M., Malick, A., Coen, D.M. & Martuza, R.L. Reduction and elimination of encephalitis in an experimental glioma therapy model with attenuated herpes simplex mutants that retain susceptibility to acyclovir. Neurosurgery 32, 597–603 (1993).

    CAS  PubMed  Google Scholar 

  38. Kambara, H., Okano, H., Chiocca, E.A. & Saeki, Y. An oncolytic HSV-1 mutant expressing ICP34.5 under control of a nestin promoter increases survival of animals even when symptomatic from a brain tumor. Cancer Res. 65, 2832–2839 (2005).

    CAS  PubMed  Google Scholar 

  39. Bolyard, C. et al. Doxorubicin synergizes with 34.5ENVE to enhance antitumor efficacy against metastatic ovarian cancer. Clin. Cancer Res. 20, 6479–6494 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Menotti, L. et al. Inhibition of human tumor growth in mice by an oncolytic herpes simplex virus designed to target solely HER-2-positive cells. Proc. Natl. Acad. Sci. USA 106, 9039–9044 (2009).

    CAS  PubMed  Google Scholar 

  41. Gatta, V., Petrovic, B. & Campadelli-Fiume, G. The engineering of a novel ligand in gH confers to HSV an expanded tropism independent of gD activation by its receptors. PLoS Pathog. 11, e1004907 (2015).

    PubMed  PubMed Central  Google Scholar 

  42. Uchida, H. et al. Effective treatment of an orthotopic xenograft model of human glioblastoma using an EGFR-retargeted oncolytic herpes simplex virus. Mol. Ther. 21, 561–569 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank A. Yilmaz, B. McNeil and V. Sellers for critical reading. This work was supported by grants from the NIH (NS106170, AI129582, J.Y.; CA185301, CA068458, CA210087, CA163205-5805, M.A.C.; CA163205-5806, E.A.C.), the Leukemia & Lymphoma Society (6503-17, 1364-19, J.Y.), the American Cancer Society (RSG-14-243-01-LIB, J.Y.) and the Gabrielle's Angel Cancer Research Foundation (87, J.Y.).

Author information

Authors and Affiliations

Authors

Contributions

B.X. performed experiments, designed research and wrote the manuscript; R.M., L.R., J.Y.Y. and J.H., H.C., P.Y. performed experiments; H.D. designed research; J.Z. analyzed the data; H.N. provided materials; E.A.C. and B.K. designed research and reviewed the manuscript. M.A.C. designed research, reviewed and edited the manuscript and acquired funding. J.Y. designed research, wrote the manuscript, acquired funding and supervised the study.

Corresponding author

Correspondence to Jianhua Yu.

Ethics declarations

Competing interests

A patent application on OV-CDH1 virus related to this work has been submitted by The Ohio State University on behalf of the inventors, J.Y., M.A.C. and B.X.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–19, Supplementary Table 1 (PDF 7693 kb)

Life Sciences Reporting Summary (PDF 240 kb)

Supplementary Notes

Supplementary Notes 1–4 (PDF 150 kb)

Supplementary Video 1

The plaque-forming process of OV-Q1-infected U251 cells. U251 cells were infected with OV-Q1 at a MOI of 0.005. At 2 hpi, infection media were replaced with fresh media. The video was recorded from 24 to 72 hpi using Zeiss fluorescence microscope (AXIO observer Z1). The time interval is 5 min. Green fluorescence (GFP) indicates virus-infected cells. This experiment was repeated 3 times with similar results. (AVI 30223 kb)

Supplementary Video 2

The plaque-forming process of OV-CDH1-infected U251 cells. U251 cells were infected with OV-CDH1 at a MOI of 0.005. At 2 hpi, infection media were replaced with fresh media. The video was recorded from 24 to 72 hpi using Zeiss fluorescence microscope (AXIO observer Z1). The time interval is 5 min. Green fluorescence (GFP) indicates virus-infected cells. This experiment was repeated 3 times with similar results. (AVI 44622 kb)

Supplementary Video 3

The plaque-forming process of OV-Q1-infected U251 cells after staining with CellTracker. U251 cells were infected with OV-Q1 at a MOI of 0.005. At 2 hpi, infection media were replaced with fresh media. At 24 hpi, cells were stained with Celltracker Deep Red. The video was recorded from 24 to 72 hpi using Zeiss fluorescence microscope (AXIO observer Z1). The time interval is 5 min. Green fluorescence (GFP) indicates virus-infected cells; red fluorescence indicates the cytoplasmic content of all the cells stained with CellTracker. This experiment was repeated 3 times with similar results. (AVI 26612 kb)

Supplementary Video 4

The plaque-forming process of OV-CDH1-infected U251 cells after staining with CellTracker. U251 cells were infected with OV-CDH1 at a MOI of 0.005. At 2 hpi, infection media were replaced with fresh media. At 24 hpi, cells were stained with Celltracker Deep Red. The video was recorded from 24 to 72 hpi using Zeiss fluorescence microscope (AXIO observer Z1). The time interval is 5 min. Green fluorescence (GFP) indicates virus-infected cells; red fluorescence indicates the cytoplasmic content of all the cells stained with CellTracker. This experiment was repeated 3 times with similar results. (AVI 40372 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, B., Ma, R., Russell, L. et al. An oncolytic herpesvirus expressing E-cadherin improves survival in mouse models of glioblastoma. Nat Biotechnol 37, 45–54 (2019). https://doi.org/10.1038/nbt.4302

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.4302

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