8,363
Views
220
CrossRef citations to date
0
Altmetric
Editorial

First oncolytic virus approved for melanoma immunotherapy

, &
Article: e1115641 | Received 30 Oct 2015, Accepted 31 Oct 2015, Published online: 17 Feb 2016

ABSTRACT

On 2015, October 27th, the US Food and Drug Administration (FDA) has officially approved talimogene laherparepvec (T-VEC, also known as OncoVEXGM-CSF) for use in melanoma patients with injectable but non-resectable lesions in the skin and lymph nodes. T-VEC (which is commercialized by Amgen, Inc. under the name of Imlygic®) becomes therefore the first oncolytic virus approved for cancer therapy in the US.

The term “oncolytic virus” refers to a non-pathogenic viral strain that kills malignant cells while virtually sparing their non-transformed counterparts.Citation1,2 A limited amount of viruses are naturally oncolytic as they display an elevated oncotropism (i.e., they selectively infect neoplastic cells) and mediate a robust cytopathic effect.Citation1,2 However, several obstacles prevent the use of most naturally occurring viruses for oncolytic virotherapy, including an excessive pathogenic potential, a sub-optimal oncotropism, an elevated susceptibility to neutralization by innate or adaptive immune effectors, and a limited capacity to trigger tumor-targeting immune responses.Citation3,4 Genetic engineering has been intensively employed throughout the past decade to circumvent these issues and generate oncolytic viral strains with superior clinical activity.Citation1,2

T-VEC is one of these genetically engineered strains. T-VEC derives from a primary isolate of human herpes simplex virus 1 (HSV-1) known as JS1 (ECACC Accession Number 01010209) that per se demonstrated enhanced oncolytic activity as compared to other clinical HSV-1 isolates and laboratory strains.Citation5,6 T-VEC was obtained in several consecutive rounds of genetic engineering. First, JS1 was attenuated by functionally deleting both copies of RL1 (encoding the neurovirulence factor ICP34.5), as well as US12 (encoding the neurovirulence factor ICP47).Citation5,6 Of note, the deletion of US12 converted the late gene US11 into an immediate-early gene driven by the US12 promoter.Citation5,6 Altogether, these genetic modifications provide T-VEC with the capacity to establish a productive infection in malignant cells (but not in normal cells), most likely because rapidly-dividing cells express a protein that, together with US11 expressed early after infection, can functionally substitute for ICP34.5.Citation7,8 Finally, one cassette encoding human granulocyte macrophage colony-stimulating factor (GM-CSF) under the control of the cytomegalovirus immediate-early promoter was inserted into each of the non-functional RL1 loci.Citation5,6 The expression of GM-CSF in the context of viral oncolysis favors the recruitment and activation of antigen-presenting cells,Citation9,10 hence endowing T-VEC with the ability to promote the initiation of a tumor-targeting immune response.Citation11-13 Importantly, T-VEC remains susceptible to anti-HSV-1 therapeutics such as acyclovir,Citation14,15 offering an additional safety control over viral replication and spread.

Results from early Phase I and II trials testing intratumoral T-VEC in patients with various tumors (including melanoma) were very encouraging,Citation16-18 prompting Amgen, Inc. initiate various Phase III clinical studies, including OPTiM (OncoVEXGM-CSF Pivotal Trial in Melanoma, NCT00769704).Citation19 In this context, 436 subjects with Stage IIIb-IV melanoma bearing injectable but not surgically resectable tumors were randomly assigned (at a 2:1 ratio) to receive intralesional T-VEC or subcutaneous GM-CSF. The primary end point of the study was durable response rate (DRR, defined as the rate of objective responses lasting continuously for at least 6 mo), per independent assessment. Secondary end points included overall response rate (ORR) and overall survival (OS).Citation20

As early as on 2013, March 19th, Amgen Inc. released preliminary data from the OPTiM study, indicating that patients in the T-VEC arm exhibited a significantly higher DDR than subjects receiving GM-CSF only.Citation21 This announcement generated considerable enthusiasm, and earlier this year (on 2015, April 29th), the US FDA emitted the first formal recommendation supporting the approval of T-VEC for the treatment of melanoma.Citation22-24 However, it was not until October 27th that the US FDA officially licensed T-VEC for use in melanoma patients with injectable but non-resectable lesions in the skin and lymph nodes (source http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm469571.htm).

This decision ensued the release of final data from the OPTiM trial, which confirmed the superiority of intralesional T-VEC over subcutaneous GM-CSF in terms of DRR (16.3% vs. 2.1%), ORR (26.4% vs. 5.7%) and median OS (23.3 mo vs. 18.9 mo).Citation20 Moreover, the administration of T-VEC was associated with a very favorable safety profile, the most common adverse events being fatigue, chills, and pyrexia. The only severe (Grade 3–4) toxicity occurring in more than 2% of patients receiving T-VEC was cellulitis, and no fatal treatment-related adverse events occurred.Citation20

Although H101 (a recombinant oncolytic adenovirus commercialized under the name of Oncorine®) is licensed by the China Food and Drug Administration (CFDA) for use together with chemotherapy in subjects with refractory head and neck carcinoma since November 2005,Citation25 the addition of oncolytic virotherapy to the clinical armamentarium represents a first-in-history in the US. Based on recent official recommendations emitted by the European Medicine Agency (EMA), T-VEC may soon enter the clinical practice in Europe, too (http://www.ema.europa.eu/ema/index.jsp?curl=pages/news_and_events/news/2015/10/news_detail_002421.jsp&mid=WC0b01ac058004d5c1). Now, it will be interesting to see if t T-VEC can be successfully employed for the treatment of other solid tumors, and to test whether the efficacy of T-VEC can be boosted by combining it with chemotherapy,Citation26,27 radiation therapy,Citation28,29 or alternative immunotherapeutic interventions,Citation30 like checkpoint blockers.Citation31,32 Finally, it will be important to elucidate the molecular and cellular circuitries explaining why T-VEC has been so successful as compared to several other GM-CSF-encoding oncolytic viruses, most of which are still in preclinical or early clinical development.Citation3,4 Perhaps, T-VEC is able to promote a particularly immunogenic form of cellular demiseCitation33-36 associated with Type I interferon signaling, which is also required for the full-blown efficacy of various chemotherapeutics.Citation37,38 Further investigation is required to formally address this intriguing possibility.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

Authors are supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt-Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

References

  • Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012; 30:658–70; PMID:22781695; http://dx.doi.org/10.1038/nbt.2287
  • Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 2015; 14:642–62; PMID:26323545; http://dx.doi.org/10.1038/nrd4663
  • Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Cremer I, Erbs P, Limacher JM, Preville X, Zitvogel L et al. Trial watch: oncolytic viruses for cancer therapy. Oncoimmunology 2014; 3:e28694; PMID:25097804; http://dx.doi.org/10.4161/onci.28694
  • Vacchelli E, Eggermont A, Sautes-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: oncolytic viruses for cancer therapy. Oncoimmunology 2013; 2:e24612; PMID:23894720; http://dx.doi.org/10.4161/onci.24612
  • Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, McGrath Y, Thomas SK, Thornton M, Bullock P et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther 2003; 10:292–303; PMID:12595888; http://dx.doi.org/10.1038/sj.gt.3301885
  • Goins WF, Huang S, Cohen JB, Glorioso JC. Engineering HSV-1 vectors for gene therapy. Methods Mol Biol 2014; 1144:63–79; PMID:24671677; http://dx.doi.org/10.1007/978-1-4939-0428-0_5
  • 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; 72:8620–6; PMID:9765401
  • Cassady KA, Gross M, Roizman B. The second-site mutation in the herpes simplex virus recombinants lacking the gamma134.5 genes precludes shutoff of protein synthesis by blocking the phosphorylation of eIF-2alpha. J Virol 1998; 72:7005–11; PMID:9696792
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory cytokines. Oncoimmunology 2013; 2:e24850; PMID:24073369; http://dx.doi.org/10.4161/onci.24850
  • Vacchelli E, Aranda F, Obrist F, Eggermont A, Galon J, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory cytokines in cancer therapy. Oncoimmunology 2014; 3:e29030; PMID:25083328; http://dx.doi.org/10.4161/onci.29030
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013; 31:51–72; PMID:23157435; http://dx.doi.org/10.1146/annurev-immunol-032712-100008
  • Ranki T, Joensuu T, Jager E, Karbach J, Wahle C, Kairemo K, Alanko T, Partanen K, Turkki R, Linder N et al. Local treatment of a pleural mesothelioma tumor with ONCOS-102 induces a systemic antitumor CD8 T-cell response, prominent infiltration of CD8 lymphocytes and Th1 type polarization. Oncoimmunology 2014; 3:e958937; PMID:25941579; http://dx.doi.org/10.4161/21624011.2014.958937
  • Spel L, Boelens JJ, Nierkens S, Boes M. Antitumor immune responses mediated by dendritic cells: how signals derived from dying cancer cells drive antigen cross-presentation. Oncoimmunology 2013; 2:e26403; PMID:24482744; http://dx.doi.org/10.4161/onci.26403
  • De Clercq E, Field HJ. Antiviral prodrugs - the development of successful prodrug strategies for antiviral chemotherapy. Br J Pharmacol 2006; 147:1–11; PMID:16284630; http://dx.doi.org/10.1038/sj.bjp.0706446
  • Razonable RR. Antiviral drugs for viruses other than human immunodeficiency virus. Mayo Clin Proc 2011; 86:1009–26; PMID:21964179; http://dx.doi.org/10.4065/mcp.2011.0309
  • Hu JC, Coffin RS, Davis CJ, Graham NJ, Groves N, Guest PJ, Harrington KJ, James ND, Love CA, McNeish I 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; 12:6737–47; PMID:17121894; http://dx.doi.org/10.1158/1078-0432.CCR-06-0759
  • Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, Gonzalez R, Glaspy J, Whitman E, Harrington K 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; 27:5763–71; PMID:19884534; http://dx.doi.org/10.1200/JCO.2009.24.3675
  • Kaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. 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; 17:718–30; PMID:19915919; http://dx.doi.org/10.1245/s10434-009-0809-6
  • Kaufman HL, Bines SD. OPTIM trial: a Phase III trial of an oncolytic herpes virus encoding GM-CSF for unresectable stage III or IV melanoma. Future Oncol 2010; 6:941–9; PMID:20528232; http://dx.doi.org/10.2217/fon.10.66
  • Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol 2015; 33:2780–8; PMID:26014293; http://dx.doi.org/10.1200/JCO.2014.58.3377
  • Galluzzi L, Lugli E. Cancer immunotherapy turns viral. Oncoimmunology 2013; 2:e24802; PMID:23734338; http://dx.doi.org/10.4161/onci.24802
  • Dharmadhikari N, Mehnert JM, Kaufman HL. Oncolytic virus immunotherapy for melanoma. Curr Treat Options Oncol 2015; 16:326; PMID:25777572; http://dx.doi.org/10.1007/s11864-014-0326-0
  • Dolgin E. Oncolytic viruses get a boost with first FDA-approval recommendation. Nat Rev Drug Discov 2015; 14:369–71; PMID:26027526; http://dx.doi.org/10.1038/nrd4643
  • Killock D. Skin cancer: T-VEC oncolytic viral therapy shows promise in melanoma. Nat Rev Clin Oncol 2015; 12:438; PMID:26077044; http://dx.doi.org/10.1038/nrclinonc.2015.106
  • Liang M. Clinical development of oncolytic viruses in China. Curr Pharm Biotechnol 2012; 13:1852–7; PMID:21740357; http://dx.doi.org/10.2174/138920112800958760
  • Gujar SA, Clements D, Lee PW. Two is better than one: complementing oncolytic virotherapy with gemcitabine to potentiate antitumor immune responses. Oncoimmunology 2014; 3:e27622; PMID:24804161; http://dx.doi.org/10.4161/onci.27622
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74–88; PMID:23890065; http://dx.doi.org/10.1016/j.immuni.2013.06.014
  • Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014; 3:e28518; PMID:25071979; http://dx.doi.org/10.4161/onci.28518
  • Bloy N, Pol J, Manic G, Vitale I, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: radioimmunotherapy for oncological indications. Oncoimmunology 2014; 3:e954929; PMID:25941606; http://dx.doi.org/10.4161/21624011.2014.954929
  • Galluzzi L, Vacchelli E, Bravo-San Pedro JM, Buque A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P et al. Classification of current anticancer immunotherapies. Oncotarget 2014; 5:12472–508; PMID:25537519; http://dx.doi.org/10.18632/oncotarget.2998
  • Vacchelli E, Eggermont A, Galon J, Sautes-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: monoclonal antibodies in cancer therapy. Oncoimmunology 2013; 2:e22789; PMID:23482847; http://dx.doi.org/10.4161/onci.22789
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27297; PMID:24701370; http://dx.doi.org/10.4161/onci.27297
  • Galluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ 2015; 22:58–73; PMID:25236395; http://dx.doi.org/10.1038/cdd.2014.137
  • Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3:e955691; PMID:25941621; http://dx.doi.org/10.4161/21624011.2014.955691
  • Workenhe ST, Mossman KL. Rewiring cancer cell death to enhance oncolytic viro-immunotherapy. Oncoimmunology 2013; 2:e27138; PMID:24498567; http://dx.doi.org/10.4161/onci.27138
  • Jiang H, Fueyo J. Healing after death: antitumor immunity induced by oncolytic adenoviral therapy. Oncoimmunology 2014; 3:e947872; PMID:25954598; http://dx.doi.org/10.4161/21624011.2014.947872
  • Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Nat Rev Immunol 2015; 15:405–14; PMID:26027717; http://dx.doi.org/10.1038/nri3845
  • Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell 2015; 28:690–714; PMID:26678337; http://dx.doi.org/10.1016/j.ccell.2015.10.012

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.