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:

CF33-hNIS-antiPDL1 virus primes pancreatic ductal adenocarcinoma for enhanced anti-PD-L1 therapy

Abstract

Immunotherapeutic strategies that combine oncolytic virus (OV) and immune checkpoint inhibitors have the potential to overcome treatment resistance in pancreatic ductal adenocarcinoma (PDAC), one of the least immunogenic solid tumors. Oncolytic viral chimera, CF33-hNIS-antiPDL1 genetically modified to express anti-human PD-L1 antibody and CF33-hNIS-Δ without the anti-PD-L1 gene, were used to investigate the immunogenic effects of OVs and virus-delivered anti-PD-L1 in PDAC in vitro. Western blot, flow cytometry, and immunofluorescence microscopy were used to evaluate the effects of CF33-hNIS-Δ and IFNγ on PD-L1 upregulation in AsPC-1 and BxPC-3 cells, and CF33-hNIS-antiPDL1 production of anti-PD-L1 and surface PD-L1 blockade of AsPC-1 and BxPC-3 with or without cocultured activated T cells. The cytosolic and cell surface levels of PD-L1 in PDAC cell lines varied; only BxPC-3 showed high cell surface expression. Treatment of these cells with CF33-hNIS-Δ and IFNγ significantly upregulated PD-L1 expression and translocation of PD-L1 from the cytosol onto the cell surface. Following coculture of activated T cells and BxPC-3 with CF33-hNIS-antiPDL1, the cell surface PD-L1 blockade on BxPC-3 cells by virus-delivered anti-PD-L1 antibody increased granzyme B release and prevented virus-induced decrease of perforin release from activated CD8+ T cells. Our results suggest that CF33-IOVs can prime immune checkpoint inhibition of PDAC and enhance antitumor immune killing.

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: PD-L1 is found mostly in the cytosol of human pancreatic ductal adenocarcinoma cells (PDACs).
Fig. 2: CF33-hNIS-Δ increases the translocation/expression of PD-L1 in human PDAC cell lines.
Fig. 3: IFNγ induces PD-L1 translocation/upregulation from the cytosol onto the surface of human PDAC cell lines.
Fig. 4: Expression of anti-PD-L1 antibody encoded by CF33-hNIS-antiPDL1 in PDACs over time.
Fig. 5: Anti-PD-L1 antibody encoded by the virus blocks CF33-hNIS-Δ-induced surface PD-L1/CD274 binding of PDACs.
Fig. 6: Virus-encoded anti-PD-L1 antibody blocks IFNγ-induced surface PD-L1 binding of PDACs.
Fig. 7: CF33-hNIS-antiPDL1 induces BxPC-3 cell killing and increases T cell granzyme B and perforin release in coculture with activated T cells in vitro.

Similar content being viewed by others

Data availability

All data relevant to the study are provided in the article or uploaded as supplementary information.

References

  1. Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. Lancet. 2020;395:2008–20.

    Article  CAS  Google Scholar 

  2. Tempero MA. NCCN guidelines updates: pancreatic cancer. J Natl Compr Canc Netw. 2019;17:603–5.

    CAS  PubMed  Google Scholar 

  3. Moore A, Donahue T. Pancreatic cancer. JAMA. 2019;322:1426.

    Article  Google Scholar 

  4. Huang L, Jansen L, Balavarca Y, Babaei M, van der Geest L, Lemmens V, et al. Stratified survival of resected and overall pancreatic cancer patients in Europe and the USA in the early twenty-first century: a large, international population-based study. BMC Med. 2018;16:125.

    Article  Google Scholar 

  5. Balsano R, Tommasi C, Garajova I. State of the art for metastatic pancreatic cancer treatment: where are we now? Anticancer Res. 2019;39:3405–12.

    Article  CAS  Google Scholar 

  6. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913–21.

    Article  CAS  Google Scholar 

  7. Sunami Y, Kleeff J. Immunotherapy of pancreatic cancer. Prog Mol Biol Transl Sci. 2019;164:189–216.

    Article  CAS  Google Scholar 

  8. Singh RR, O’Reilly EM. New treatment strategies for metastatic pancreatic ductal adenocarcinoma. Drugs. 2020;80:647–69.

    Article  Google Scholar 

  9. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175:313–26.

    Article  CAS  Google Scholar 

  10. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–61.

    Article  CAS  Google Scholar 

  11. Kyi C, Postow MA. Immune checkpoint inhibitor combinations in solid tumors: opportunities and challenges. Immunotherapy. 2016;8:821–37.

    Article  CAS  Google Scholar 

  12. Feng M, Xiong G, Cao Z, Yang G, Zheng S, Song X, et al. PD-1/PD-L1 and immunotherapy for pancreatic cancer. Cancer Lett. 2017;407:57–65.

    Article  CAS  Google Scholar 

  13. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65.

    Article  CAS  Google Scholar 

  14. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–7.

    Article  CAS  Google Scholar 

  15. Barrueto L, Caminero F, Cash L, Makris C, Lamichhane P, Deshmukh RR. Resistance to checkpoint inhibition in cancer immunotherapy. Transl Oncol. 2020;13:100738.

    Article  Google Scholar 

  16. Dougall WC, Kurtulus S, Smyth MJ, Anderson AC. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunol Rev. 2017;276:112–20.

    Article  CAS  Google Scholar 

  17. Dempke WCM, Fenchel K, Uciechowski P, Dale SP. Second- and third-generation drugs for immuno-oncology treatment—the more the better? Eur J Cancer. 2017;74:55–72.

    Article  CAS  Google Scholar 

  18. Zeng S, Pottler M, Lan B, Grutzmann R, Pilarsky C, Yang H. Chemoresistance in pancreatic cancer. Int J Mol Sci. 2019;20:4504.

    Article  CAS  Google Scholar 

  19. Balachandran VP, Beatty GL, Dougan SK. Broadening the impact of immunotherapy to pancreatic cancer: challenges and opportunities. Gastroenterology. 2019;156:2056–72.

    Article  CAS  Google Scholar 

  20. Sivanandam V, LaRocca CJ, Chen NG, Fong Y, Warner SG. Oncolytic viruses and immune checkpoint inhibition: the best of both worlds. Mol Ther Oncolytics. 2019;13:93–106.

    Article  CAS  Google Scholar 

  21. Vijayakumar G, McCroskery S, Palese P. Engineering Newcastle disease virus as an oncolytic vector for intratumoral delivery of immune checkpoint inhibitors and immunocytokines. J Virol. 2020;94:e01677–19.

    Article  CAS  Google Scholar 

  22. Liu Z, Ravindranathan R, Kalinski P, Guo ZS, Bartlett DL. Rational combination of oncolytic vaccinia virus and PD-L1 blockade works synergistically to enhance therapeutic efficacy. Nat Commun. 2017;8:14754.

    Article  CAS  Google Scholar 

  23. Woo Y, Zhang Z, Yang A, Chaurasiya S, Park AK, Lu J, et al. Novel chimeric immuno-oncolytic virus CF33-hNIS-antiPDL1 for the treatment of pancreatic cancer. J Am Coll Surg. 2020;230:709–17.

    Article  Google Scholar 

  24. Chaurasiya S, Yang A, Kang S, Lu J, Kim SI, Park AK, et al. Oncolytic poxvirus CF33-hNIS-DeltaF14.5 favorably modulates tumor immune microenvironment and works synergistically with anti-PD-L1 antibody in a triple-negative breast cancer model. Oncoimmunology. 2020;9:1729300.

    Article  Google Scholar 

  25. Warner SG, Kim SI, Chaurasiya S, O’Leary MP, Lu J, Sivanandam V, et al. A novel chimeric poxvirus encoding hNIS is tumor-tropic, imageable, and synergistic with radioiodine to sustain colon cancer regression. Mol Ther Oncolytics. 2019;13:82–92.

    Article  CAS  Google Scholar 

  26. Chaurasiya S, Chen NG, Lu J, Martin N, Shen Y, Kim SI, et al. A chimeric poxvirus with J2R (thymidine kinase) deletion shows safety and anti-tumor activity in lung cancer models. Cancer Gene Ther. 2020;27:125–35.

    Article  CAS  Google Scholar 

  27. Zhang Z, Shively JE. Generation of novel bone forming cells (monoosteophils) from the cathelicidin-derived peptide LL-37 treated monocytes. PLoS ONE. 2010;5:e13985.

    Article  Google Scholar 

  28. Zhang Z, Le K, La Placa D, Armstrong B, Miller MM, Shively JE. CXCR2 specific endocytosis of immunomodulatory peptide LL-37 in human monocytes and formation of LL-37 positive large vesicles in differentiated monoosteophils. Bone Rep. 2020;12:100237.

    Article  Google Scholar 

  29. Yang S, He P, Wang J, Schetter A, Tang W, Funamizu N, et al. A novel MIF signaling pathway drives the malignant character of pancreatic cancer by targeting NR3C2. Cancer Res. 2016;76:3838–50.

    Article  CAS  Google Scholar 

  30. Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An integrated TCGA Pan-Cancer Clinical Data Resource to drive high-quality survival outcome analytics. Cell. 2018;173:400–416.e11.

    Article  CAS  Google Scholar 

  31. Levy DE, Marie IJ, Durbin JE. Induction and function of type I and III interferon in response to viral infection. Curr Opin Virol. 2011;1:476–86.

    Article  CAS  Google Scholar 

  32. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.

    Article  CAS  Google Scholar 

  33. Cullen SP, Brunet M, Martin SJ. Granzymes in cancer and immunity. Cell Death Differ. 2010;17:616–23.

    Article  CAS  Google Scholar 

  34. Brennan AJ, Chia J, Trapani JA, Voskoboinik I. Perforin deficiency and susceptibility to cancer. Cell Death Differ. 2010;17:607–15.

    Article  CAS  Google Scholar 

  35. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Disco. 2019;18:197–218.

    Article  CAS  Google Scholar 

  36. Atkins MB, Larkin J. Immunotherapy combined or sequenced with targeted therapy in the treatment of solid tumors: current perspectives. J Natl Cancer Inst. 2016;108:djv414.

    Article  Google Scholar 

  37. Russell L, Peng KW, Russell SJ, Diaz RM. Oncolytic viruses: priming time for cancer immunotherapy. BioDrugs. 2019;33:485–501.

    Article  CAS  Google Scholar 

  38. Engeland CE, Grossardt C, Veinalde R, Bossow S, Lutz D, Kaufmann JK, et al. CTLA-4 and PD-L1 checkpoint blockade enhances oncolytic measles virus therapy. Mol Ther. 2014;22:1949–59.

    Article  CAS  Google Scholar 

  39. Puzanov I, Milhem MM, Minor D, Hamid O, Li A, Chen L, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol. 2016;34:2619–26.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors are grateful to Imugene Limited for providing approval to use their licensed oncolytic virus CF33 in the studies reported in this paper. Research reported in this publication included work performed in the Pathology Core, Flow Cytometry Core, Light Microscopy Core, and Integrative Genomics and Bioinformatics core facilities supported by the National Cancer Institute of the National Institutes of Health under grant number P30CA033572. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The authors would also like to thank Dr Jinhui Wang in Integrative Genomics Core, Lucy Brown in Flow Cytometry core, and Dr Brian Armstrong in the Light Microscopy Core of City of Hope for supporting the work.

The authors would like to thank Byungwook Kim, Martha Magallanes, Seonah Kang, and Dr Maria Hahn in our laboratory and Dr Chunyan Zhang in Department of Immuno-Oncology for technical support, and Supriya Deshpande, PhD, for assistance with manuscript editing.

SGW and SC are supported through the generosity of Natalie and David Roberts. These authors wish to thank them for their philanthropy. The authors would also like to thank Samuel Kuo and Grace Liu of Samson Holding, Ltd. for their generosity.

Funding

Department of Defense E01 Award W81XWH-19-1-0225 (CA180425).

Author information

Authors and Affiliations

Authors

Contributions

ZZ, YW, and YF conceived the experiments. ZZ, AY, SC, AKP, JL, and S-IK performed the experiments, researched, and interpreted data. ZZ, AY, Y-CY, ZL, and YW analyzed data. YW, SGW, HH, DVH, and YF secured funding and interpreted the data. ZZ, YW, AY, and YF wrote the manuscript. All authors edited and approved the final manuscript.

Corresponding author

Correspondence to Yanghee Woo.

Ethics declarations

Conflict of interest

The authors declare no competing interests.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Z., Yang, A., Chaurasiya, S. et al. CF33-hNIS-antiPDL1 virus primes pancreatic ductal adenocarcinoma for enhanced anti-PD-L1 therapy. Cancer Gene Ther 29, 722–733 (2022). https://doi.org/10.1038/s41417-021-00350-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-021-00350-4

This article is cited by

Search

Quick links