CC BY-NC-ND 4.0 · Journal of Clinical Interventional Radiology ISVIR 2017; 01(02): 096-099
DOI: 10.1055/s-0037-1602389
Review Article
Indian Society of Vascular and Interventional Radiology

Viral Oncolytic Therapy

Omar Zurkiya
1   Division of Interventional Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
,
Suvranu Ganguli
1   Division of Interventional Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
› Author Affiliations
Further Information

Publication History

Publication Date:
28 July 2017 (online)

Abstract

Viral oncolysis broadly refers to the use of modified viruses to infect and subsequently lyse tumor cells. This concept arises from the observation that viral replication is itself effective in destroying tumor cells. This effect is then amplified by reinfection of adjacent tumor cells by the progeny virion released from lysed tumor cells. Herpes simplex virus 1 (HSV-1) has been the primary focus of current efforts in viral oncolysis. It is a double-stranded DNA virus that is a ubiquitous pathogen transmitted by direct mucosal contact. HSV-1 possesses several features well suited to viral oncolytic therapy. It does not integrate into the cellular genome, has a large transgene capacity of up to 50 kb, and is already highly prevalent in the general population. In addition, effective antiherpetic agents are available to stop unwanted viral replication. HSV-1 mutants that preferentially replicate in neoplastic cells rather than normal cells have been characterized, and several variants of replication deficient HSV-1 mutants have been created and studied. They follow a common theme in that their replication is significantly attenuated in normal cells, while activated in cancer cells. Studies have been performed in various strains including those known as G207, NV1020, talimogene laherparepvec, and rRp450, and are reviewed here. Viral oncolysis is an exciting area of research with applications to tumors throughout the body. It holds promise as a new treatment for primary and metastatic liver cancer and may soon become a relevant therapy in interventional oncology.

 
  • References

  • 1 Chase M, Chung RY, Chiocca EA. An oncolytic viral mutant that delivers the CYP2B1 transgene and augments cyclophosphamide chemotherapy. Nat Biotechnol 1998; 16 (05) 444-448
  • 2 Geevarghese SK, Geller DA, de Haan HA. , et al. Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver. Hum Gene Ther 2010; 21 (09) 1119-1128
  • 3 Kuruppu D, Tanabe KK. Viral oncolysis by herpes simplex virus and other viruses. Cancer Biol Ther 2005; 4 (05) 524-531
  • 4 Braidwood L, Graham SV, Graham A, Conner J. Oncolytic herpes viruses, chemotherapeutics, and other cancer drugs. Oncolytic Virother 2013; 2: 57-74
  • 5 Goins WF, Huang S, Cohen JB, Glorioso JC. Engineering HSV-1 vectors for gene therapy. Methods Mol Biol 2014; 1144: 63-79
  • 6 Markert JM, Medlock MD, Rabkin SD. , et al. Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther 2000; 7 (10) 867-874
  • 7 Jackson JD, McMorris AM, Roth JC. , et al. Assessment of oncolytic HSV efficacy following increased entry-receptor expression in malignant peripheral nerve sheath tumor cell lines. Gene Ther 2014; 21 (11) 984-990
  • 8 Kemeny N, Brown K, Covey A. , et al. Phase I, open-label, dose-escalating study of a genetically engineered herpes simplex virus, NV1020, in subjects with metastatic colorectal carcinoma to the liver. Hum Gene Ther 2006; 17 (12) 1214-1224
  • 9 Senzer NN, Kaufman HL, Amatruda T. , et al. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol 2009; 27 (34) 5763-5771
  • 10 Pawlik TM, Nakamura H, Yoon SS. , et al. Oncolysis of diffuse hepatocellular carcinoma by intravascular administration of a replication-competent, genetically engineered herpesvirus. Cancer Res 2000; 60 (11) 2790-2795