Biology:Oncolytic herpes virus

From HandWiki
Oncolytic herpes virus
Herpes simplex virus TEM B82-0474 lores.jpg
TEM micrograph of a herpes simplex virus.
Virus classification e
(unranked): Virus
Realm: Duplodnaviria
Kingdom: Heunggongvirae
Phylum: Peploviricota
Class: Herviviricetes
Order: Herpesvirales
Family: Orthoherpesviridae
Genus: Simplexvirus
Species:
Human alphaherpesvirus 1

Many variants of herpes simplex virus have been considered for viral therapy of cancer; the early development of these was thoroughly reviewed in the journal Cancer Gene Therapy in 2002.[1] This page describes (in the order of development) the most notable variants—those tested in clinical trials: G207, HSV1716, NV1020 and Talimogene laherparepvec (previously Oncovex-GMCSF). These attenuated versions are constructed by deleting viral genes required for infecting or replicating inside normal cells but not cancer cells, such as ICP34.5, ICP6/UL39, and ICP47.

Electron Micrograph of herpes virus

HSV1716

HSV1716 is a first generation oncolytic virus developed by the Glasgow Institute of Virology, and subsequently by Virttu Biologics (formerly Crusade Laboratories, a spin-out from The Institute of Virology), to selectively destroy cancer cells. The virus has the trade name SEPREHVIR. It is based on the herpes simplex virus (HSV-1). The HSV1716 strain has a deletion of the gene ICP34.5. ICP34.5 is a neurovirulence gene (enabling the virus to replicate in neurons of the brain and spinal cord). Deletion of this gene provides the property of tumor-selective replication to the virus (i.e. largely prevents replication in normal cells, while still allowing replication in tumor cells), although it also reduces replication in tumor cells as compared to wild type HSV.[2][3]

A vital part of the normal mechanism of HSV-1, the ICP34.5 protein has been proposed to condition post-mitotic cells for viral replication. With no ICP34.5 gene, the HSV-1716 variant is unable to overcome normal defences of healthy differentiated cells (mediated by PKR) to replicate efficiently. However, tumour cells have much weaker PKR-linked defences, which may be the reason why HSV1716 effectively kills a wide range of tumour cell lines in tissue culture.

An HSV1716 variant, HSV1716NTR is an oncolytic virus generated by inserting the enzyme NTR into the virus HSV1716 as a GDEPT strategy.[4] In-vivo, administration of the prodrug CB1954 to athymic mice bearing either A431 or A2780 tumour xenografts, 48 hours after intra-tumoral injection of HSV1790, resulted in a marked reduction in tumour volumes and significantly improved survival compared to administration of virus alone. A similar approach has been taken with a variant of HSV1716 that expresses the noradrenaline transporter to deliver radioactive iodine into individual infected cancer cells, by tagging a protein that cancer cells transport. The nor-adrenaline transporter specifically transports a compound containing radioactive iodine across the cell membrane, using genes from the virus. The only cells in the body that receive a significant radiation dose are those infected and their immediate neighbours.[5]

Clinical trials

G207

G207 was constructed as a second-generation vector from HSV-1 laboratory strain F, with ICP34.5 deleted and the ICP6 gene inactivated by insertion of the E. coli LacZ gene.[12]

Two phase I clinical trials in glioma were completed.[13][14][15] The results of the first trial were published simultaneously with the first trial of HSV1716 in 2000, with commentators praising the demonstration of safety of these viruses when injected into brain tumours but also expressing disappointment that viral replication could not be demonstrated due to the difficulty of taking biopsies from brain tumours.[16]

NV1020

NV1020 is an oncolytic herpes virus initially developed by Medigene Inc. and licensed for development by Catherex Inc. in 2010.[17] NV1020 has a deletion of just one copy of the ICP34.5 gene and ICP6 is intact.[1] A direct comparison of NV1020 and G207 in a mouse model of peritoneal cancer showed that NV1020 is more effective at lower doses.[18]

Clinical trials

A Phase I/II study completed in 2008 evaluating NV1020 for treatment of metastatic colorectal cancer in the liver.[19] The study assessed tumour response by CT scan and FDG-PET scans, showing 67% of patients had an initial increase in tumour size then followed by a decrease in 64% of patients.[20][18]

Talimogene laherparepvec

Main page: Biology:Talimogene laherparepvec

Talimogene laherparepvec is the USAN name for the oncolytic virus also known as 'OncoVEX GM-CSF'. It was developed by BioVex Inc. (Woburn, MA, USA & Oxford, UK) until BioVex was purchased by Amgen in January 2011.[21]

It is a second-generation herpes simplex virus based on the JS1 strain and expressing the immune stimulatory factor GM-CSF.[22][23] Like other oncolytic versions of HSV it has a deletion of the gene encoding ICP34.5, which provides tumor selectivity.[24] It also has a deletion of the gene encoding ICP47, a protein that inhibits antigen presentation,[25] and an insertion of a gene encoding GM-CSF, an immune stimulatory cytokine.[2][3] Deletion of the gene encoding ICP47 also puts the US11 gene (a late gene) under control of the immediate early ICP47 promoter. The earlier and greater expression of US11 (also involved in overcoming PKR-mediated responses) largely overcomes the reduction in replication in tumor cells of ICP34.5-deleted HSV as compared to wild type virus, but without reducing tumor selectivity.

Clinical trials

Including phase III : See Talimogene laherparepvec

See also

References

  1. 1.0 1.1 "Oncolytic herpes simplex virus vectors for cancer virotherapy". Cancer Gene Therapy 9 (12): 967–78. December 2002. doi:10.1038/sj.cgt.7700537. PMID 12522436. 
  2. 2.0 2.1 "A herpesvirus genetic element which affects translation in the absence of the viral GADD34 function". The EMBO Journal 15 (17): 4759–66. September 1996. doi:10.1002/j.1460-2075.1996.tb00853.x. PMID 8887567. 
  3. 3.0 3.1 "ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties". Gene Therapy 10 (4): 292–303. February 2003. doi:10.1038/sj.gt.3301885. PMID 12595888. 
  4. "Antitumor activity of a selectively replication competent herpes simplex virus (HSV) with enzyme prodrug therapy". Anticancer Research 29 (6): 2159–66. June 2009. PMID 19528476. http://ar.iiarjournals.org/cgi/pmidlookup?view=long&pmid=19528476. 
  5. "In vivo evaluation of a cancer therapy strategy combining HSV1716-mediated oncolysis with gene transfer and targeted radiotherapy". Journal of Nuclear Medicine 53 (4): 647–54. April 2012. doi:10.2967/jnumed.111.090886. PMID 22414636. 
  6. "HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival". Gene Therapy 11 (22): 1648–58. November 2004. doi:10.1038/sj.gt.3302289. PMID 15334111. 
  7. "The potential for efficacy of the modified (ICP 34.5(-)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study". Gene Therapy 9 (6): 398–406. March 2002. doi:10.1038/sj.gt.3301664. PMID 11960316. 
  8. "Potential for efficacy of the oncolytic Herpes simplex virus 1716 in patients with oral squamous cell carcinoma". Head & Neck 30 (8): 1045–51. August 2008. doi:10.1002/hed.20840. PMID 18615711. 
  9. "Intralesional injection of herpes simplex virus 1716 in metastatic melanoma". Lancet 357 (9255): 525–6. February 2001. doi:10.1016/S0140-6736(00)04048-4. PMID 11229673. 
  10. Clinical trial number NCT01721018 for "Intrapleural Administration of HSV1716 to Treat Patients With Malignant Pleural Mesothelioma" at ClinicalTrials.gov
  11. Clinical trial number NCT00931931 for "HSV1716 in Patients With Non-Central Nervous System (Non-CNS) Solid Tumors" at ClinicalTrials.gov
  12. "Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas". Nature Medicine 1 (9): 938–43. September 1995. doi:10.1038/nm0995-938. PMID 7585221. 
  13. "Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial". Gene Therapy 7 (10): 867–74. May 2000. doi:10.1038/sj.gt.3301205. PMID 10845725. 
  14. Clinical trial number NCT00028158 for "Safety and Effectiveness Study of G207, a Tumor-Killing Virus, in Patients With Recurrent Brain Cancer" at ClinicalTrials.gov
  15. Clinical trial number NCT00157703 for "G207 Followed by Radiation Therapy in Malignant Glioma" at ClinicalTrials.gov
  16. "A tale of two trials: selectively replicating herpesviruses for brain tumors". Gene Therapy 7 (10): 815–6. May 2000. doi:10.1038/sj.gt.3301213. PMID 10845717. 
  17. "MediGene AG divests Oncolytic Herpes Simplex Viruses (oHSV) program to Catherex, Inc" (Press release). MediGene AG. April 13, 2010. Archived from the original on October 29, 2013. Retrieved May 7, 2013.
  18. 18.0 18.1 "Phase I/II study of oncolytic herpes simplex virus NV1020 in patients with extensively pretreated refractory colorectal cancer metastatic to the liver". Human Gene Therapy 21 (9): 1119–28. September 2010. doi:10.1089/hum.2010.020. PMID 20486770. 
  19. Clinical trial number NCT00149396 for "Safety and Efficacy of a Genetically Engineered Herpes Simplex Virus NV1020 to Treat Colorectal Cancer Metastatic to Liver" at ClinicalTrials.gov
  20. "Response to intra-arterial oncolytic virotherapy with the herpes virus NV1020 evaluated by [18F]fluorodeoxyglucose positron emission tomography and computed tomography". Human Gene Therapy 23 (1): 91–7. January 2012. doi:10.1089/hum.2011.141. PMID 21895536. 
  21. "Amgen, Form 8-K, Current Report, Filing Date Jan 26, 2012". secdatabase.com. http://pdf.secdatabase.com/2009/0001193125-12-024302.pdf. 
  22. "OncoVEXGM-CSF RAC Submission". NIH Genetic Modification Clinical Research Information System (GeMCRIS®). http://www.gemcris.od.nih.gov/Abstracts/787_s.pdf. 
  23. "A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor". Clinical Cancer Research 12 (22): 6737–47. November 2006. doi:10.1158/1078-0432.CCR-06-0759. PMID 17121894. 
  24. "Preventing vaccinia virus class-I epitopes presentation by HSV-ICP47 enhances the immunogenicity of a TAP-independent cancer vaccine epitope". International Journal of Cancer 131 (5): E659-69. September 2012. doi:10.1002/ijc.27362. PMID 22116674. 
  25. "The GM-CSF/IL-3/IL-5 cytokine receptor family: from ligand recognition to initiation of signaling". Immunological Reviews 250 (1): 277–302. November 2012. doi:10.1111/j.1600-065X.2012.01164.x. PMID 23046136. 

External links

Wikidata ☰ Q16992171 entry