DOI QR코드

DOI QR Code

Treatment of Human Thyroid Carcinoma Cells with the G47delta Oncolytic Herpes Simplex Virus

  • Wang, Jia-Ni (Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University) ;
  • Xu, Li-Hua (Department of Oncology and Hematology, The First Affiliated Hospital of Guangzhou Medical University) ;
  • Zeng, Wei-Gen (Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University) ;
  • Hu, Pan (Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University) ;
  • Rabkin, Samuel D. (Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School) ;
  • Liu, Ren-Rin (Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University)
  • Published : 2015.03.04

Abstract

Background: Thyroid carcinoma is the most common malignancy of the endocrine organs. Although the majority of thyroid cancer patients experience positive outcomes, anaplastic thyroid carcinoma is considered one of the most aggressive malignancies. Current therapeutic regimens do not confer a significant survival benefit, and new therapies are urgently needed. Oncolytic herpes simplex virus (oHSV) may represent a promising therapy for cancer. In the present study, we investigated the therapeutic effects of a third-generation HSV vector, $G47{\Delta}$, on various human thyroid carcinoma cell lines in vitro. Two subcutaneous (s.c.) models of anaplastic thyroid carcinoma were also established to evaluate the in vivo anti-tumor efficacy of $G47{\Delta}$. Materials and Methods: The human thyroid carcinoma cell line ARO, FRO, WRO, and KAT-5, were infected with $G47{\Delta}$ at different multiplicities of infection (MOIs) in vitro. The survival rates of infected cells were calculated each day. Two s.c. tumor models were established using ARO and FRO cells in Balb/c nude mice, which were intratumorally (i.t.) treated with either $G47{\Delta}$ or mock. Tumor volumes and mouse survival times were documented. Results: $G47{\Delta}$ was highly cytotoxic to different types of thyroid carcinomas. For ARO, FRO, and KAT-5, greater than 30% and 80% of cells were killed at MOI=0.01 and MOI=0.1, respectively on day 5. WRO cells displayed modest sensitivity to $G47{\Delta}$, with only 21% and 38% of cells killed. In the s.c. tumor model, both of the anaplastic thyroid carcinoma cell lines (ARO and FRO) were highly sensitive to $G47{\Delta}$; $G47{\Delta}$ significantly inhibited tumor growth and prolonged the survival of mice bearing s.c. ARO and FRO tumors. Conclusions: The oHSV $G47{\Delta}$ can effectively kill different types of human thyroid carcinomas in vitro. $G47{\Delta}$ significantly inhibited growth of anaplastic thyroid carcinoma in vivo and prolonged animal survival. Therefore, $G47{\Delta}$ may hold great promise for thyroid cancer patients.

Keywords

References

  1. Are C, Shaha AR (2006). Anaplastic thyroid carcinoma: biology, pathogenesis, prognostic factors, and treatment approaches. Ann Surg Oncol, 13, 453-64. https://doi.org/10.1245/ASO.2006.05.042
  2. Campadelli-Fiume G, De Giovanni C, Gatta V, et al (2011). Rethinking herpes simplex virus: the way to oncolytic agents. Rev Med Virol, 21, 213-26. https://doi.org/10.1002/rmv.691
  3. Goldstein DJ, Weller SK (1988). Factor (s) present in herpes simplex virus type 1-infected cells can compensate for the loss of the large subunit of the viral ribonucleotide reductase: characterization of an ICP6 deletion mutant. Virology, 166, 41-51. https://doi.org/10.1016/0042-6822(88)90144-4
  4. Huang YY, Yu Z, Lin SF, et al (2007). Nectin-1 is a marker of thyroid cancer sensitivity to herpes oncolytic therapy. J Clin Endocrinol Metab, 92, 1965-70. https://doi.org/10.1210/jc.2007-0040
  5. Lai ML, Faa G, Serra S, et al (2005). Rhabdoid tumor of the thyroid gland: a variant of anaplastic carcinoma. Arch Pathol Lab Med, 129, 55-7.
  6. Li J, Zeng W, Huang Y, et al (2012). Treatment of breast cancer stem cells with oncolytic herpes simplex virus. Cancer Gene Ther, 19, 707-14. https://doi.org/10.1038/cgt.2012.49
  7. Lin SF, Gao SP, Price DL, et al (2008). Synergy of a herpes oncolytic virus and paclitaxel for anaplastic thyroid cancer. Clin Cancer Res, 14, 1519-28. https://doi.org/10.1158/1078-0432.CCR-07-4628
  8. Martuza RL, Malick A, Markert JM, et al (1991). Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science, 252, 854-6. https://doi.org/10.1126/science.1851332
  9. Mohr I, Sternberg D, Ward S, et al (2001). A herpes simplex virus type 1 gamma34.5 second-site suppressor mutant that exhibits enhanced growth in cultured glioblastoma cells is severely attenuated in animals. J Virol, 75, 5189-96. https://doi.org/10.1128/JVI.75.11.5189-5196.2001
  10. Pacini F, Castagna MG, Cipri C, et al (2010). Medullary thyroid carcinoma. Clin Oncol (R Coll Radiol), 22, 475-85. https://doi.org/10.1016/j.clon.2010.05.002
  11. Reinblatt M, Pin RH, Fong Y (2007). Herpes viral oncolysis: a novel cancer therapy. J Am Coll Surg, 205, S69-75.
  12. Russell SJ, Peng KW, Bell JC (2012). Oncolytic virotherapy. Nat Biotechnol, 30, 658-70. https://doi.org/10.1038/nbt.2287
  13. Sherman SI, Wirth LJ, Droz JP, et al (2008). Motesanib diphosphate in progressive differentiated thyroid cancer. N Engl J Med, 359, 31-42. https://doi.org/10.1056/NEJMoa075853
  14. Sipos JA, Mazzaferri EL (2010). Thyroid cancer epidemiology and prognostic variables. Clin Oncol (R Coll Radiol), 22, 395-404. https://doi.org/10.1016/j.clon.2010.05.004
  15. Smallridge RC, Ain KB, Asa SL, et al (2012). American thyroid association guidelines for management of patients with anaplastic thyroid cancer. Thyroid, 22, 1104-39. https://doi.org/10.1089/thy.2012.0302
  16. Todo T, Martuza RL, Rabkin SD, et al (2001). Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc Natl Acad Sci USA, 98, 6396-401. https://doi.org/10.1073/pnas.101136398
  17. Vanderpump MP (2011). The epidemiology of thyroid disease. Br Med Bull, 99, 39-51. https://doi.org/10.1093/bmb/ldr030
  18. Varghese S, Rabkin SD (2002). Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther, 9, 967-78. https://doi.org/10.1038/sj.cgt.7700537
  19. Wang J, Hu P, Zeng M, et al (2011a). Oncolytic herpes simplex virus treatment of metastatic breast cancer. Int J Oncol, 40, 757-63.
  20. Wang JN, Hu P, Zeng MS, et al (2011b). Anti-tumor effect of oncolytic herpes simplex virus G47delta on human nasopharyngeal carcinoma. Chin J Cancer, 30, 831-41. https://doi.org/10.5732/cjc.011.10301
  21. Yu Z, Eisenberg DP, Singh B, et al (2004). Treatment of aggressive thyroid cancer with an oncolytic herpes virus. Int J Cancer, 112, 525-32. https://doi.org/10.1002/ijc.20421
  22. Zaman M, Fatima N, Sajjad Z, et al (2012). Threshold primary tumour sizes for nodal and distant metastases in papillary and follicular thyroid cancers. Asian Pac J Cancer Prev, 13, 2473-6. https://doi.org/10.7314/APJCP.2012.13.6.2473
  23. Zeng W, Hu P, Wu J, et al (2013a). The oncolytic herpes simplex virus vector G47 effectively targets breast cancer stem cells. Oncol Rep, 29, 1108-14.
  24. Zeng WG, Li JJ, Hu P, et al (2013b). An oncolytic herpes simplex virus vector, G47Delta, synergizes with paclitaxel in the treatment of breast cancer. Oncol Rep, 29, 2355-61.

Cited by

  1. MiR-99a Inhibits Cell Proliferation and Tumorigenesis through Targeting mTOR in Human Anaplastic Thyroid Cancer vol.16, pp.12, 2015, https://doi.org/10.7314/APJCP.2015.16.12.4937
  2. The oncolytic herpes simplex virus vector, G47Δ, effectively targets tamoxifen-resistant breast cancer cells vol.35, pp.3, 2016, https://doi.org/10.3892/or.2015.4539
  3. Oncolytic virus therapy: A new era of cancer treatment at dawn vol.107, pp.10, 2016, https://doi.org/10.1111/cas.13027
  4. Current status of clinical trials assessing oncolytic virus therapy for urological cancers vol.24, pp.5, 2017, https://doi.org/10.1111/iju.13325
  5. Oncolytic virotherapy for anaplastic and poorly differentiated thyroid cancer: a promise or a clinical reality? pp.2045-0877, 2018, https://doi.org/10.2217/ije-2017-0028