Cancer Res Treat.  2020 Jan;52(1):309-319. 10.4143/crt.2019.161.

Characterization of Oncolytic Vaccinia Virus Harboring the Human IFNB1 and CES2 Transgenes

Affiliations
  • 1Department of Pharmacology and Medical Research Center (MRC), Pusan National University School of Medicine, Yangsan, Korea
  • 2Department of Pharmacy and Pusan Cancer Research Center, Pusan National University, Busan, Korea
  • 3Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Korea
  • 4School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China

Abstract

Purpose
The purpose of this study was to assess characteristics of SJ-815, a novel oncolytic vaccinia virus lacking a functional thymidine kinase-encoding TK gene, and instead, having two human transgenes: the IFNB1 that encodes interferon β1, and the CES2 that encodes carboxylesterase 2, which metabolizes the prodrug, irinotecan, into cytotoxic SN-38.
Materials and Methods
Viral replication and dissemination of SJ-815 were measured by plaque assay and comet assay, respectively, and compared to the backbone of SJ-815, a modified Western Reserve virus named WI. Tumor cytotoxicity of SJ-815 (or mSJ-815, which has the murine IFNB1 transgene for mouse cancers) was evaluated using human and mouse cancer cells. Antitumor effects of SJ-815, with/without irinotecan, were evaluated using a human pancreatic cancer-bearing mouse model and a syngeneic melanoma-bearing mouse model. The SN-38/ irinotecan ratios in mouse melanoma tissue 4 days post irinotecan treatment were compared between groups with and without SJ-815 intravenous injection.
Results
SJ-815 demonstrated significantly lower viral replication and dissemination, but considerably stronger in vitro tumor cytotoxicity than WI. The combination use of SJ-815 plus irinotecan generated substantial tumor regression in the human pancreatic cancer model, and significantly prolonged survival in the melanoma model (hazard ratio, 0.11; 95% confidence interval, 0.02 to 0.50; p=0.013). The tumor SN-38/irinotecan ratios were over 3-fold higher in the group with SJ-815 than those without (p < 0.001).
Conclusion
SJ-815 demonstrates distinct characteristics gained from the inserted IFNB1 and CES2 transgenes. The potent antitumor effects of SJ-815, particularly when combined with irinotecan, against multiple solid tumors make SJ-815 an attractive candidate for further preclinical and clinical studies.

Keyword

Oncolytic viruses; Vaccinia virus; Carboxylesterase 2; Interferon β1; Irinotecan

Figure

  • Fig. 1. The activity of human proteins interferon β1 (IFN-β1) and carboxylesterase 2 (CES2) expressed by three final selected plaques of SJ-815 was demonstrated using HEK-Blue IFN-α/β cell assay (A), and p-nitrophenyl acetate assay (B).

  • Fig. 2. Viral replication of SJ-815 and other viruses in human (A, B) and mouse (C) cancer cell lines. Replication of Western Reserve (WR), modified Western Reserve (WI) (backbone), and SJ-815 in HeLa and HeLa S3 cells, with infection virus dose of 3 pfu/cell, at 2, 24, 48, and 72 hours (A); and in HeLa cells, with infection virus dose of 0.001, 0.01, 0.1, and 1 pfu/cell, at 24 hours (B). **p < 0.01, ***p < 0.001; WI (backbone) or SJ-815 vs. WR; the p-values were obtained using paired t tests. (C) Replication of WI (backbone), SJ-815, and mSJ-815 in MC-38 and B16-F10 mouse cancer cells, with infection virus dose of 0.1 pfu/cell at 72 hours (left), or 1 pfu/cell at 24 hours (right). *p < 0.05, **p < 0.01, ***p < 0.001; WI (backbone) or SJ-815 vs. mSJ-815; the p-values were obtained using paired t tests.

  • Fig. 3. Comet assays conducted on U-2 OS cells after infection with Western Reserve (WR) virus (A), modified Western Reserve (WI) (backbone) virus (B), or SJ-815 (C) for 2 hours first and then culture with Dulbecco's modified Eagle medium containing 2.5% fetal bovine serum for 72 hours; or after infection with WI (backbone) virus (D) for 2 hours first, and then culture with 1,000 U interferon β1 (IFN-β1), SJ-815 supernatant, or WR supernatant for 72 hours; or after infection with SJ-815 (E) for 2 hours first, and then culture with WR supernatant for 72 hours. The virus supernatants were collected after the U-2 OS cells were infected with the viruses: WR, WI (backbone), or SJ-815 for 16 hours, and filtered three times and stored at 4°C until use.

  • Fig. 4. (A) Comparison of tumor cytotoxicity (EC50) of SJ-815 and modified Western Reserve (WI) (backbone) in 12 human cancer cell lines. Each data was presented as mean±standard deviation of two EC50, which were calculated from two sets of dose-response data. (B) Tumor cytotoxicity (EC50) of mSJ-815 in 5 mouse cancer cell lines. Each data was presented as 95% confidence interval was EC50, which was calculated from only one set of dose-response data. *p < 0.05, **p < 0.01, ***p < 0.001; SJ-815 vs. WI (backbone); the p-values were obtained using paired t tests.

  • Fig. 5. Changes in tumor volume from day 0 to day 41 (sacrifice day) in human pancreatic cancer (MIA PaCa-2)-bearing mice, treated with phosphate-buffered saline (PBS), SJ-815 (1×106 pfu; intratumoral; day 7 and 14), irinotecan (25 mg/kg; intravenous; day 3, 10, and 17), or SJ-815 (1×106 pfu; intratumoral; day 7 and 14)+irinotecan (25 mg/kg; intravenous; day 3, 10, and 17). Statistical analyses were conducted to compare tumor volume on the sacrifice day. **p < 0.01, SJ-815 vs. SJ-815+irinotecan, ***p < 0.001, PBS vs. SJ-815+irinotecan; the p-values were obtained using t tests. Irinotecan was not compared because one mouse died before the sacrifice day.

  • Fig. 6. Percent survival (A) and body weight change (B) of B16-F10 melanoma-bearing mice, treated with phosphate-buffered saline (PBS), mSJ-815 (3×107 pfu; intratumoral; day 7 and 14), irinotecan (25 mg/kg; intravenous; day 3, 10, and 17), or mSJ-815 (3×107 pfu; intratumoral; day 7 and 14)+irinotecan (25 mg/kg; intravenous; day 3, 10, and 17). The dashed lines indicate the normal range of body weight change. The p-value was obtained using a log-rank test.

  • Fig. 7. Ratio of SN-38/irinotecan in tumor tissue, obtained on day 7 from B16-F10 melanoma-bearing mice, treated with phosphate-buffered saline, irinotecan (25 mg/kg; intravenous; day 3), modified Western Reserve (WI; 1×107 pfu; intravenous; day 0)+irinotecan (25 mg/kg; intravenous; day 3), or SJ-815 (1×107 pfu; intravenous; day 0) +irinotecan (25 mg/kg; intravenous; day 3). The p-value was obtained using an ANOVA test.


Reference

References

1. Kim MK, Breitbach CJ, Moon A, Heo J, Lee YK, Cho M, et al. Oncolytic and immunotherapeutic vaccinia induces antibody-mediated complement-dependent cancer cell lysis in humans. Sci Transl Med. 2013; 5:185ra63.
Article
2. Shen Y, Nemunaitis J. Fighting cancer with vaccinia virus: teaching new tricks to an old dog. Mol Ther. 2005; 11:180–95.
Article
3. Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol. 2008; 9:533–42.
Article
4. McCart JA, Ward JM, Lee J, Hu Y, Alexander HR, Libutti SK, et al. Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes. Cancer Res. 2001; 61:8751–7.
5. Topolcan O, Holubec L Jr. The role of thymidine kinase in cancer diseases. Expert Opin Med Diagn. 2008; 2:129–41.
Article
6. Qin XQ, Tao N, Dergay A, Moy P, Fawell S, Davis A, et al. Interferon-beta gene therapy inhibits tumor formation and causes regression of established tumors in immune-deficient mice. Proc Natl Acad Sci U S A. 1998; 95:14411–6.
7. Hervas-Stubbs S, Perez-Gracia JL, Rouzaut A, Sanmamed MF, Le Bon A, Melero I. Direct effects of type I interferons on cells of the immune system. Clin Cancer Res. 2011; 17:2619–27.
Article
8. Spaapen RM, Leung MY, Fuertes MB, Kline JP, Zhang L, Zheng Y, et al. Therapeutic activity of high-dose intratumoral IFN-β requires direct effect on the tumor vasculature. J Immunol. 2014; 193:4254–60.
Article
9. Kirn DH, Wang Y, Le Boeuf F, Bell J, Thorne SH. Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus. PLoS Med. 2007; 4:e353.
Article
10. Robertson J, Barr R, Shulman LN, Forte GB, Magrini N. Essential medicines for cancer: WHO recommendations and national priorities. Bull World Health Organ. 2016; 94:735–42.
Article
11. Chang JS, Chiu YF, Yu JC, Chen LT, Ch'ang HJ. The role of consolidation chemoradiotherapy in locally advanced pancreatic cancer receiving chemotherapy: an updated systematic review and meta-analysis. Cancer Res Treat. 2018; 50:562–74.
Article
12. Cecchin E, Corona G, Masier S, Biason P, Cattarossi G, Frustaci S, et al. Carboxylesterase isoform 2 mRNA expression in peripheral blood mononuclear cells is a predictive marker of the irinotecan to SN38 activation step in colorectal cancer patients. Clin Cancer Res. 2005; 11(19 Pt 1):6901–7.
Article
13. Laizure SC, Herring V, Hu Z, Witbrodt K, Parker RB. The role of human carboxylesterases in drug metabolism: have we overlooked their importance? Pharmacotherapy. 2013; 33:210–22.
Article
14. Capello M, Lee M, Wang H, Babel I, Katz MH, Fleming JB, et al. Carboxylesterase 2 as a determinant of response to irinotecan and neoadjuvant FOLFIRINOX therapy in pancreatic ductal adenocarcinoma. J Natl Cancer Inst. 2015; 107:djv132.
Article
15. Wadkins RM, Hyatt JL, Yoon KJ, Morton CL, Lee RE, Damodaran K, et al. Discovery of novel selective inhibitors of human intestinal carboxylesterase for the amelioration of irinotecan-induced diarrhea: synthesis, quantitative structure-activity relationship analysis, and biological activity. Mol Pharmacol. 2004; 65:1336–43.
Article
16. Smith GL, Vanderplasschen A, Law M. The formation and function of extracellular enveloped vaccinia virus. J Gen Virol. 2002; 83(Pt 12):2915–31.
Article
17. Bekisz J, Baron S, Balinsky C, Morrow A, Zoon KC. Antiproliferative properties of type I and type II interferon. Pharmaceuticals (Basel). 2010; 3:994–1015.
Article
18. Blasco R, Sisler JR, Moss B. Dissociation of progeny vaccinia virus from the cell membrane is regulated by a viral envelope glycoprotein: effect of a point mutation in the lectin homology domain of the A34R gene. J Virol. 1993; 67:3319–25.
Article
19. Sampath P, Li J, Hou W, Chen H, Bartlett DL, Thorne SH. Crosstalk between immune cell and oncolytic vaccinia therapy enhances tumor trafficking and antitumor effects. Mol Ther. 2013; 21:620–8.
Article
20. Khan S, Ahmad A, Guo W, Wang YF, Abu-Qare A, Ahmad I. A simple and sensitive LC/MS/MS assay for 7-ethyl-10-hydroxycamptothecin (SN-38) in mouse plasma and tissues: application to pharmacokinetic study of liposome entrapped SN-38 (LE-SN38). J Pharm Biomed Anal. 2005; 37:135–42.
Article
21. Vanderplasschen A, Hollinshead M, Smith GL. Antibodies against vaccinia virus do not neutralize extracellular enveloped virus but prevent virus release from infected cells and comet formation. J Gen Virol. 1997; 78(Pt 8):2041–8.
Article
22. Potts KG, Irwin CR, Favis NA, Pink DB, Vincent KM, Lewis JD, et al. Deletion of F4L (ribonucleotide reductase) in vaccinia virus produces a selective oncolytic virus and promotes antitumor immunity with superior safety in bladder cancer models. EMBO Mol Med. 2017; 9:638–54.
23. Leib DA, Machalek MA, Williams BR, Silverman RH, Virgin HW. Specific phenotypic restoration of an attenuated virus by knockout of a host resistance gene. Proc Natl Acad Sci U S A. 2000; 97:6097–101.
Article
24. Vaha-Koskela M, Hinkkanen A. Tumor restrictions to oncolytic virus. Biomedicines. 2014; 2:163–94.
Article
25. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, et al. Host type I IFN signals are required for anti-tumor CD8+ T cell responses through CD8α+ dendritic cells. J Exp Med. 2011; 208:2005–16.
Article
26. Yang X, Zhang X, Fu ML, Weichselbaum RR, Gajewski TF, Guo Y, et al. Targeting the tumor microenvironment with interferon-β bridges innate and adaptive immune responses. Cancer Cell. 2014; 25:37–48.
Article
27. Gujar S, Pol JG, Kroemer G. Heating it up: oncolytic viruses make tumors 'hot' and suitable for checkpoint blockade immunotherapies. Oncoimmunology. 2018; 7:e1442169.
Article
28. Patel MR, Jacobson BA, Ji Y, Drees J, Tang S, Xiong K, et al. Vesicular stomatitis virus expressing interferon-β is oncolytic and promotes antitumor immune responses in a syngeneic murine model of non-small cell lung cancer. Oncotarget. 2015; 6:33165–77.
Article
29. Hsieh YT, Lin HP, Chen BM, Huang PT, Roffler SR. Effect of cellular location of human carboxylesterase 2 on CPT-11 hydrolysis and anticancer activity. PLoS One. 2015; 10:e0141088.
Article
30. Chu C, Abbara C, Tandia M, Polrot M, Gonin P, Farinotti R, et al. Cetuximab increases concentrations of irinotecan and of its active metabolite SN-38 in plasma and tumour of human colorectal carcinoma-bearing mice. Fundam Clin Pharmacol. 2014; 28:652–60.
Article
31. Sharkey RM, McBride WJ, Cardillo TM, Govindan SV, Wang Y, Rossi EA, et al. Enhanced delivery of SN-38 to human tumor xenografts with an anti-Trop-2-SN-38 antibody conjugate (Sacituzumab Govitecan). Clin Cancer Res. 2015; 21:5131–8.
Article
32. Adkins CE, Nounou MI, Hye T, Mohammad AS, Terrell-Hall T, Mohan NK, et al. NKTR-102 Efficacy versus irinotecan in a mouse model of brain metastases of breast cancer. BMC Cancer. 2015; 15:685.
Article
33. Cho E, Ryu EJ, Jiang F, Jeon UB, Cho M, Kim CH, et al. Preclinical safety evaluation of hepatic arterial infusion of oncolytic poxvirus. Drug Des Devel Ther. 2018; 12:2467–74.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr