Skip to main content
Log in

Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma

  • Leading Article
  • Published:
American Journal of Clinical Dermatology Aims and scope Submit manuscript

Abstract

Many mammalian viruses have properties that can be commandeered for the treatment of cancer. These characteristics include preferential infection and replication in tumor cells, the initiation of tumor cell lysis, and the induction of innate and adaptive anti-tumor immunity. Furthermore, viruses can be genetically engineered to reduce pathogenicity and increase immunogenicity resulting in minimally toxic therapeutic agents. Talimogene laherparepvec (T-VEC; Imlygic™), is a genetically modified herpes simplex virus, type 1, and is the first oncolytic virus therapy to be approved for the treatment of advanced melanoma by the US FDA. T-VEC is attenuated by the deletion of the herpes neurovirulence viral genes and enhanced for immunogenicity by the deletion of the viral ICP47 gene. Immunogenicity is further supported by expression of the human granulocyte–macrophage colony-stimulating factor (GM-CSF) gene, which helps promote the priming of T cell responses. T-VEC demonstrated significant improvement in durable response rate, objective response rate, and progression-free survival in a randomized phase III clinical trial for patients with advanced melanoma. This review will discuss the optimal selection of patients for such treatment and describe how therapy is optimally delivered. We will also discuss future directions for oncolytic virus immunotherapy, which will likely include combination T-VEC clinical trials, expansion of T-VEC to other types of non-melanoma skin cancers, and renewed efforts at oncolytic virus drug development with other viruses.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Brunssen A, et al. Impact of skin cancer screening and secondary prevention campaigns on skin cancer incidence and mortality: a systematic review. J Am Acad Dermatol. 2016. doi: 10.1016/j.jaad.2016.07.045.

  2. Feng Z, Zhang Z, Wu XC. Lifetime risks of cutaneous melanoma by histological subtype and race/ethnicity in the United States. J La State Med Soc. 2013;165(4):201–8.

    PubMed  Google Scholar 

  3. Coit DG, et al. NCCN Guidelines Insights: Melanoma, Version 3.2016. J Natl Compr Canc Netw. 2016;14(8):945–58.

    PubMed  Google Scholar 

  4. Radny P, et al. Phase II trial of intralesional therapy with interleukin-2 in soft-tissue melanoma metastases. Br J Cancer. 2003;89(9):1620–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lau PK, Ascierto PA, McArthur G. Melanoma: the intersection of molecular targeted therapy and immune checkpoint inhibition. Curr Opin Immunol. 2016;39:30–8.

    Article  CAS  PubMed  Google Scholar 

  6. Redmond KL, et al. Overcoming Resistance to Targeted Therapies in Cancer. Semin Oncol. 2015;42(6):896–908.

    Article  CAS  PubMed  Google Scholar 

  7. Ott PA, Hodi FS, Robert C. CTLA-4 and PD-1/PD-L1 blockade: new immunotherapeutic modalities with durable clinical benefit in melanoma patients. Clin Cancer Res. 2013;19(19):5300–9.

    Article  CAS  PubMed  Google Scholar 

  8. Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Robert C, et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372(26):2521–32.

    Article  CAS  PubMed  Google Scholar 

  10. Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–30.

    Article  CAS  PubMed  Google Scholar 

  11. Larkin J, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373(1):23–34.

    Article  PubMed  Google Scholar 

  12. Kluger HM, et al. Characterization of PD-L1 expression and associated T-cell infiltrates in metastatic melanoma samples from variable anatomic sites. Clin Cancer Res. 2015;21(13):3052–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Taube JM, et al. Differential expression of immune-regulatory genes associated with PD-L1 display in melanoma: implications for PD-1 pathway blockade. Clin Cancer Res. 2015;21(17):3969–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Inoue H, et al. Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma. Oncoimmunology. 2016;5(9):e1204507.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Gajewski TF, et al. Molecular profiling to identify relevant immune resistance mechanisms in the tumor microenvironment. Curr Opin Immunol. 2011;23(2):286–92.

    Article  CAS  PubMed  Google Scholar 

  16. Zaretsky JM, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):819–29.

    Article  CAS  PubMed  Google Scholar 

  17. Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015;14(9):642–62.

    Article  CAS  PubMed  Google Scholar 

  18. Douglas JT, et al. Efficient oncolysis by a replicating adenovirus (ad) in vivo is critically dependent on tumor expression of primary ad receptors. Cancer Res. 2001;61(3):813–7.

    CAS  PubMed  Google Scholar 

  19. Goldufsky J, et al. Oncolytic virus therapy for cancer. Oncolytic Virother. 2013;2:31–46.

    PubMed  PubMed Central  Google Scholar 

  20. Andtbacka RH, Collichio F, Amatruda T, Senzer N, Chesney J, Delman K, et al. Final planned overall survival (OS) from OPTiM, a randomized phase III trial of talimogene laherparepvec (T-VEC) versus GM-CSF for the treatment of unresected stage IIIB/C/IV melanoma. J Immunother Cancer, 2014(suppl3):P263.

  21. Kaufman HL, et al. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17(3):718–30.

    Article  PubMed  Google Scholar 

  22. Huang B, et al. Synergistic anti-tumor effects between oncolytic vaccinia virus and paclitaxel are mediated by the IFN response and HMGB1. Gene Ther. 2011;18(2):164–72.

    Article  CAS  PubMed  Google Scholar 

  23. Miyamoto S, et al. Coxsackievirus B3 is an oncolytic virus with immunostimulatory properties that is active against lung adenocarcinoma. Cancer Res. 2012;72(10):2609–21.

    Article  CAS  PubMed  Google Scholar 

  24. Liikanen I, et al. Serum HMGB1 is a predictive and prognostic biomarker for oncolytic immunotherapy. Oncoimmunology. 2015;4(3):e989771.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Koks CA, et al. Newcastle disease virotherapy induces long-term survival and tumor-specific immune memory in orthotopic glioma through the induction of immunogenic cell death. Int J Cancer. 2015;136(5):E313–25.

    Article  CAS  PubMed  Google Scholar 

  26. Takasu A, et al. Immunogenic cell death by oncolytic herpes simplex virus type 1 in squamous cell carcinoma cells. Cancer Gene Ther. 2016;23(4):107–13.

    Article  CAS  PubMed  Google Scholar 

  27. West EE, et al. PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells. J Clin Invest. 2013;123(6):2604–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10.

    Article  PubMed  Google Scholar 

  29. Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol. 2012;30(7):658–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Xu F, et al. Seroprevalence and coinfection with herpes simplex virus type 1 and type 2 in the United States, 1988-1994. J Infect Dis. 2002;185(8):1019–24.

    Article  PubMed  Google Scholar 

  31. Peters C, Rabkin SD. Designing Herpes Viruses as Oncolytics. Mol Ther Oncolyt. 2015;2. doi:10.1038/mto.2015.10.

  32. Coen DM, et al. Thymidine kinase-negative herpes simplex virus mutants establish latency in mouse trigeminal ganglia but do not reactivate. Proc Natl Acad Sci USA. 1989;86(12):4736–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Martuza RL, et al. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science. 1991;252(5007):854–6.

    Article  CAS  PubMed  Google Scholar 

  34. Goldstein DJ, Weller SK. Herpes simplex virus type 1-induced ribonucleotide reductase activity is dispensable for virus growth and DNA synthesis: isolation and characterization of an ICP6 lacZ insertion mutant. J Virol. 1988;62(1):196–205.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. He B, Gross M, Roizman B. The gamma(1)34.5 protein of herpes simplex virus 1 complexes with protein phosphatase 1alpha to dephosphorylate the alpha subunit of the eukaryotic translation initiation factor 2 and preclude the shutoff of protein synthesis by double-stranded RNA-activated protein kinase. Proc Natl Acad Sci USA. 1997;94(3):843–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chou J, et al. Mapping of herpes simplex virus-1 neurovirulence to gamma 134.5, a gene nonessential for growth in culture. Science. 1990;250(4985):1262–6.

    Article  CAS  PubMed  Google Scholar 

  37. Mineta T, et al. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med. 1995;1(9):938–43.

    Article  CAS  PubMed  Google Scholar 

  38. MacLean AR, et al. Herpes simplex virus type 1 deletion variants 1714 and 1716 pinpoint neurovirulence-related sequences in Glasgow strain 17+ between immediate early gene 1 and the ‘a’ sequence. J Gen Virol. 1991;72(Pt 3):631–9.

    Article  CAS  PubMed  Google Scholar 

  39. McKie EA, et al. Selective in vitro replication of herpes simplex virus type 1 (HSV-1) ICP34.5 null mutants in primary human CNS tumours–evaluation of a potentially effective clinical therapy. Br J Cancer. 1996;74(5):745–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kesari S, et al. Therapy of experimental human brain tumors using a neuroattenuated herpes simplex virus mutant. Lab Invest. 1995;73(5):636–48.

    CAS  PubMed  Google Scholar 

  41. Randazzo BP, et al. Treatment of experimental intracranial murine melanoma with a neuroattenuated herpes simplex virus 1 mutant. Virology. 1995;211(1):94–101.

    Article  CAS  PubMed  Google Scholar 

  42. McKie EA, et al. Histopathological responses in the CNS following inoculation with a non-neurovirulent mutant (1716) of herpes simplex virus type 1 (HSV 1): relevance for gene and cancer therapy. Neuropathol Appl Neurobiol. 1998;24(5):367–72.

    Article  CAS  PubMed  Google Scholar 

  43. Rampling R, et al. Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther. 2000;7(10):859–66.

    Article  CAS  PubMed  Google Scholar 

  44. MacKie RM, Stewart B, Brown SM. Intralesional injection of herpes simplex virus 1716 in metastatic melanoma. Lancet. 2001;357(9255):525–6.

    Article  CAS  PubMed  Google Scholar 

  45. Liu BL, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003;10(4):292–303.

    Article  CAS  PubMed  Google Scholar 

  46. Cassady KA, Gross M, Roizman B. The herpes simplex virus US11 protein effectively compensates for the gamma1(34.5) gene if present before activation of protein kinase R by precluding its phosphorylation and that of the alpha subunit of eukaryotic translation initiation factor 2. J Virol. 1998;72(11):8620–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Farassati F, Yang AD, Lee PW. Oncogenes in Ras signalling pathway dictate host-cell permissiveness to herpes simplex virus 1. Nat Cell Biol. 2001;3(8):745–50.

    Article  CAS  PubMed  Google Scholar 

  48. Pan W, et al. Utilizing ras signaling pathway to direct selective replication of herpes simplex virus-1. PLoS One. 2009;4(8):e6514.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Lim F, et al. Biosafety of gene therapy vectors derived from herpes simplex virus type 1. Curr Gene Ther. 2013;13(6):478–91.

    Article  CAS  PubMed  Google Scholar 

  50. Atherton MJ, Lichty BD. Evolution of oncolytic viruses: novel strategies for cancer treatment. Immunotherapy. 2013;5(11):1191–206.

    Article  CAS  PubMed  Google Scholar 

  51. Hu JC, et al. A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin Cancer Res. 2006;12(22):6737–47.

    Article  CAS  PubMed  Google Scholar 

  52. Shen Y, Nemunaitis J. Herpes simplex virus 1 (HSV-1) for cancer treatment. Cancer Gene Ther. 2006;13(11):975–92.

    Article  CAS  PubMed  Google Scholar 

  53. Coffin RS, et al. Gene delivery to the central and peripheral nervous systems of mice using HSV1 ICP34.5 deletion mutant vectors. Gene Ther. 1996;3(10):886–91.

    CAS  PubMed  Google Scholar 

  54. Hu JC, et al. A novel HSV-1 virus, JS1/34.5-/47-, purges contaminating breast cancer cells from bone marrow. Clin Cancer Res. 2006;12(22):6853–62.

    Article  CAS  PubMed  Google Scholar 

  55. Andtbacka RHI, Kaufaman HL, Collichio F, Amatruda T, Senzer N, Chesney J, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;2014.58:3377.

  56. Shen BH, Hermiston TW. Effect of hypoxia on Ad5 infection, transgene expression and replication. Gene Ther. 2005;12(11):902–10.

    Article  CAS  PubMed  Google Scholar 

  57. Shen BH, Bauzon M, Hermiston TW. The effect of hypoxia on the uptake, replication and lytic potential of group B adenovirus type 3 (Ad3) and type 11p (Ad11p). Gene Ther. 2006;13(12):986–90.

    Article  CAS  PubMed  Google Scholar 

  58. Mok W, Boucher Y, Jain RK. Matrix metalloproteinases-1 and -8 improve the distribution and efficacy of an oncolytic virus. Cancer Res. 2007;67(22):10664–8.

    Article  CAS  PubMed  Google Scholar 

  59. Nguyen A, Ho L, Wan Y. Chemotherapy and oncolytic virotherapy: advanced tactics in the war against cancer. Front Oncol. 2014;4:145.

    PubMed  PubMed Central  Google Scholar 

  60. Tesfay MZ, et al. PEGylation of vesicular stomatitis virus extends virus persistence in blood circulation of passively immunized mice. J Virol. 2013;87(7):3752–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Morrison J, et al. Virotherapy of ovarian cancer with polymer-cloaked adenovirus retargeted to the epidermal growth factor receptor. Mol Ther. 2008;16(2):244–51.

    Article  CAS  PubMed  Google Scholar 

  62. O’Riordan CR, et al. PEGylation of adenovirus with retention of infectivity and protection from neutralizing antibody in vitro and in vivo. Hum Gene Ther. 1999;10(8):1349–58.

    Article  PubMed  Google Scholar 

  63. Fulci G, et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc Natl Acad Sci USA. 2006;103(34):12873–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wakimoto H, et al. The complement response against an oncolytic virus is species-specific in its activation pathways. Mol Ther. 2002;5(3):275–82.

    Article  CAS  PubMed  Google Scholar 

  65. Ikeda K, et al. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med. 1999;5(8):881–7.

    Article  CAS  PubMed  Google Scholar 

  66. Evgin L, et al. Complement inhibition prevents oncolytic vaccinia virus neutralization in immune humans and cynomolgus macaques. Mol Ther. 2015;23(6):1066–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Diallo JS, et al. A high-throughput pharmacoviral approach identifies novel oncolytic virus sensitizers. Mol Ther. 2010;18(6):1123–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Berghauser Pont LM, et al. In vitro screening of clinical drugs identifies sensitizers of oncolytic viral therapy in glioblastoma stem-like cells. Gene Ther. 2015;22(12):947–59.

    Article  CAS  PubMed  Google Scholar 

  69. Ilkow CS, et al. Reciprocal cellular cross-talk within the tumor microenvironment promotes oncolytic virus activity. Nat Med. 2015;21(5):530–6.

    Article  CAS  PubMed  Google Scholar 

  70. Puzanov I, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol. 2016;34(22):2619–26.

    Article  CAS  PubMed  Google Scholar 

  71. Rehman H, et al. Into the clinic: talimogene laherparepvec (T-VEC), a first-in-class intratumoral oncolytic viral therapy. J Immunother Cancer. 2016;4:53.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Rajani K, et al. Combination Therapy With Reovirus and Anti-PD-1 Blockade Controls Tumor Growth Through Innate and Adaptive Immune Responses. Mol Ther. 2016;24(1):166–74.

    Article  CAS  PubMed  Google Scholar 

  73. Ilett, E., et al., Prime-boost using separate oncolytic viruses in combination with checkpoint blockade improves anti-tumor therapy. Gene Ther. 2016. doi:10.1038/gt.2016.70.

  74. Hart SL. Multifunctional nanocomplexes for gene transfer and gene therapy. Cell Biol Toxicol. 2010;26(1):69–81.

    Article  CAS  PubMed  Google Scholar 

  75. Mader EK, et al. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res. 2009;15(23):7246–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Muthana M, et al. Use of macrophages to target therapeutic adenovirus to human prostate tumors. Cancer Res. 2011;71(5):1805–15.

    Article  CAS  PubMed  Google Scholar 

  77. Debinski W, Tatter SB. Convection-enhanced delivery to achieve widespread distribution of viral vectors: predicting clinical implementation. Curr Opin Mol Ther. 2010;12(6):647–53.

    CAS  PubMed  Google Scholar 

  78. Senzer NN, 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):5763–71.

    Article  CAS  PubMed  Google Scholar 

  79. Aris M, Barrio MM. Combining immunotherapy with oncogene-targeted therapy: a new road for melanoma treatment. Front Immunol. 2015;6:46.

    PubMed  PubMed Central  Google Scholar 

  80. Stanley NF. Reovirus—a ubiquitous orphan. Med J Aust. 1961;48(2):815–8.

    PubMed  Google Scholar 

  81. Coffey MC, et al. Reovirus therapy of tumors with activated Ras pathway. Science. 1998;282(5392):1332–4.

    Article  CAS  PubMed  Google Scholar 

  82. Villalona-Calero MA, et al. Oncolytic reovirus in combination with chemotherapy in metastatic or recurrent non-small cell lung cancer patients with KRAS-activated tumors. Cancer. 2016;122(6):875–83.

    Article  CAS  PubMed  Google Scholar 

  83. Mahalingam D, et al. The combination of intravenous Reolysin and gemcitabine induces reovirus replication and endoplasmic reticular stress in a patient with KRAS-activated pancreatic cancer. BMC Cancer. 2015;15:513.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Kolb EA, et al. A phase I trial and viral clearance study of reovirus (Reolysin) in children with relapsed or refractory extra-cranial solid tumors: a Children’s Oncology Group Phase I Consortium report. Pediatr Blood Cancer. 2015;62(5):751–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Danthi P, et al. Reovirus receptors, cell entry, and proapoptotic signaling. Adv Exp Med Biol. 2013;790:42–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Strong JE, Lee PW. The v-erbB oncogene confers enhanced cellular susceptibility to reovirus infection. J Virol. 1996;70(1):612–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Strong JE, et al. The molecular basis of viral oncolysis: usurpation of the Ras signaling pathway by reovirus. EMBO J. 1998;17(12):3351–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Prestwich RJ, et al. Reciprocal human dendritic cell-natural killer cell interactions induce antitumor activity following tumor cell infection by oncolytic reovirus. J Immunol. 2009;183(7):4312–21.

    Article  CAS  PubMed  Google Scholar 

  89. Au GG, et al. Oncolysis of vascular malignant human melanoma tumors by Coxsackievirus A21. Int J Oncol. 2005;26(6):1471–6.

    CAS  PubMed  Google Scholar 

  90. Skelding KA, Barry RD, Shafren DR. Systemic targeting of metastatic human breast tumor xenografts by Coxsackievirus A21. Breast Cancer Res Treat. 2009;113(1):21–30.

    Article  PubMed  Google Scholar 

  91. Berry LJ, et al. Potent oncolytic activity of human enteroviruses against human prostate cancer. Prostate. 2008;68(6):577–87.

    Article  PubMed  Google Scholar 

  92. Andtbacka RH, et al. A phase II study of Coxsackievirus A21 (CVA21) oncolytic virus immunotherapy in patients with advanced melanoma, in ASCO. J Clin Oncol. 2015;9030.

  93. Parato KA, et al. The oncolytic poxvirus JX-594 selectively replicates in and destroys cancer cells driven by genetic pathways commonly activated in cancers. Mol Ther. 2012;20(4):749–58.

    Article  CAS  PubMed  Google Scholar 

  94. Park SH, et al. Phase 1b trial of biweekly intravenous pexa-vec (JX-594), an oncolytic and immunotherapeutic vaccinia virus in colorectal cancer. Mol Ther. 2015;23(9):1532–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Zamarin D, Postow MA. Immune checkpoint modulation: rational design of combination strategies. Pharmacol Ther. 2015;150:23–32.

    Article  CAS  PubMed  Google Scholar 

  96. Hu-Lieskovan S, et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci Transl Med. 2015;7(279):279ra41.

  97. al., A.R.e., A multicenter, open-label trial of talimogene laherparepvec (T-VEC) plus pembrolizumab vs pembrolizumab monotherapy in previously untreated, unresected, stage IIIB-IV melanoma. J Clin Oncol. 2015; 33 (suppl; abstr TPS9081).

  98. Andtbacka R, e.a., Phase 2, multicenter, randomized, open-label trial assessing efficacy and safety of talimogene laherparepvec (T-VEC) neoadjuvant treatment (tx) plus surgery vs surgery for resectable stage IIIB/C and IVM1a melanoma (MEL). J Clin Oncol 33, 2015 (suppl; abstr TPS9094).

  99. Zeng J, et al. Immune modulation and stereotactic radiation: improving local and abscopal responses. Biomed Res Int. 2013;2013:658126.

    PubMed  PubMed Central  Google Scholar 

  100. Milhem M. TVEC and Preop Radiation for Sarcoma. https://clinicaltrials.gov/ct2/show/NCT02453191. Accessed 11 Feb 2016.

  101. al., H.e., A Phase I, multicenter, open-label trial to evaluate the safety of talimogene laherparepvec (T-VEC) injected into liver tumors. J ImmunoTher Cancer. 2015;3(Suppl 2):P180.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Howard L. Kaufman.

Ethics declarations

Funding

This work was supported, in part, by grant UM1 CA 186716-01 from the National Cancer Institute to HLK.

Conflict of interest

Dr. Kaufman has served on advisory boards for Amgen, Celldex, EMD Serono, Merck, Prometheus, and Sanofi. He is a member of the Merck Speaker’s Bureau but does not received direct compensation for this activity. Mr. Bommareddy, Dr. Patel and Dr. Hossain have no conflicts to declare.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bommareddy, P.K., Patel, A., Hossain, S. et al. Talimogene Laherparepvec (T-VEC) and Other Oncolytic Viruses for the Treatment of Melanoma. Am J Clin Dermatol 18, 1–15 (2017). https://doi.org/10.1007/s40257-016-0238-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40257-016-0238-9

Keywords

Navigation