The Open Virology Journal




(Discontinued)

ISSN: 1874-3579 ― Volume 15, 2021
RESEARCH ARTICLE

Targeting Melanoma with Cancer-Killing Viruses



Tiantian Zhang, Yogesh R. Suryawanshi, Helene M. Woyczesczyk, Karim Essani*
Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, U.S.A

Abstract

Melanoma is the deadliest skin cancer with ever-increasing incidence. Despite the development in diagnostics and therapies, metastatic melanoma is still associated with significant morbidity and mortality. Oncolytic viruses (OVs) represent a class of novel therapeutic agents for cancer by possessing two closely related properties for tumor reduction: virus-induced lysis of tumor cells and induction of host anti-tumor immune responses. A variety of viruses, either in “natural” or in genetically modified forms, have exhibited a remarkable therapeutic efficacy in regressing melanoma in experimental and/or clinical studies. This review provides a comprehensive summary of the molecular and cellular mechanisms of action of these viruses, which involve manipulating and targeting the abnormalities of melanoma, and can be categorized as enhancing viral tropism, targeting the tumor microenvironment and increasing the innate and adaptive antitumor responses. Additionally, this review describes the “biomarkers” and deregulated pathways of melanoma that are responsible for melanoma initiation, progression and metastasis. Advances in understanding these abnormalities of melanoma have resulted in effective targeted and immuno-therapies, and could potentially be applied for engineering OVs with enhanced oncolytic activity in future.

Keywords: Melanoma, Biomarkers, Oncolytic virotherapy, Tumor targeting, Tumor microenvironment, Immune enhancement, Immunotherapy, Combinational therapy.


Article Information


Identifiers and Pagination:

Year: 2017
Volume: 11
First Page: 28
Last Page: 47
Publisher Id: TOVJ-11-28
DOI: 10.2174/1874357901711010028

Article History:

Received Date: 30/09/2016
Revision Received Date: 05/01/2017
Acceptance Date: 17/01/2017
Electronic publication date: 31/03/2017
Collection year: 2017

© 2017 Zhang et al.

open-access license: This is an open access article distributed under the terms of the Creative Commons Attribution 4.0 International Public License (CC-BY 4.0), a copy of which is available at: https://creativecommons.org/licenses/by/4.0/legalcode. This license permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


* Address correspondence to this author at the Laboratory of Virology, Department of Biological Sciences, Western Michigan University, 3441 wood hall, Kalamazoo, MI 49008-5410, U.S.A; Tel: +269-387-2661; Fax: +269-387-5609; E-mail: karim.essani@wmich.edu





INTRODUCTION

Although melanoma accounts for only 3-5% of all skin cancers, it is responsible for nearly 80% of all skin-cancer related deaths [1Tsao H, Atkins MB, Sober AJ. Management of cutaneous melanoma. N Engl J Med 2004; 351(10): 998-1012.
[http://dx.doi.org/10.1056/NEJMra041245] [PMID: 15342808]
-3Johnson TM, Yahanda AM, Chang AE, Fader DJ, Sondak VK. Advances in melanoma therapy. J Am Acad Dermatol 1998; 38(5 Pt 1): 731-41.
[http://dx.doi.org/10.1016/S0190-9622(98)70202-6] [PMID: 9591818]
]. Like most other types of cancer, melanoma arises from a combination of genetic and epigenetic abnormalities. The genetic mutations such as over-activation of BRAF and Ras, which are found in respectively around 50% and 20% of metastatic melanomas, favor melanoma cell over-proliferation and tumor progression [4Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res 2015; 3(3): 288-95.
[http://dx.doi.org/10.1158/2326-6066.CIR-14-0207] [PMID: 25736262]
]. In addition, the innate and adaptive immune suppression further contributes to melanoma survival and metastasis, such as the loss of the expression of tumor specific antigens and tumor associated antigens (TAAs), the lack of co-stimulatory signals for T cell activation and the inefficient presentation of antigen to antigen presenting cells (APCs). Conventional cytotoxic therapies are not always effective for melanoma [5Karagiannis P, Fittall M, Karagiannis SN. Evaluating biomarkers in melanoma. Front Oncol 2015; 4: 383.
[PMID: 25667918]
]. For example, the application of dacarbazine or vermurafenib resulted in less than 20% response rate in patients with malignant melanoma [6Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
]. Although interleukin (IL)-2 seems beneficial to a selected group of patients, it is associated with significant toxicity [7Viale DL, Cafferata EG, Gould D, et al. Therapeutic improvement of a stroma-targeted CRAd by incorporating motives responsive to the melanoma microenvironment. J Invest Dermatol 2013; 133(11): 2576-84.
[http://dx.doi.org/10.1038/jid.2013.191] [PMID: 23604101]
].

Oncolytic viruses (OVs) emerge as a promising approach for melanoma therapy by possessing two closely-related properties. These include OVs’ ability to preferentially target and lyse the tumor cells and their capability to enhance antitumor immune responses [8Chan WM, Rahman MM, McFadden G. Oncolytic myxoma virus: the path to clinic. Vaccine 2013; 31(39): 4252-8.
[http://dx.doi.org/10.1016/j.vaccine.2013.05.056] [PMID: 23726825]
]. While the antiviral mechanism exists in normal cells, the malignantly activated or abnormally regulated pathways in tumor cells often favor OVs’ infection and replication, therefore generating natural tumor selectivity for some viruses. For example, melanoma cells over-expressing Ras and harboring defective interferon (IFN)-signaling pathways are effectively infected by reovirus and vesicular stomatitis virus (VSV) [9Steele L, Errington F, Prestwich R, et al. Pro-inflammatory cytokine/chemokine production by reovirus treated melanoma cells is PKR/NF-κB mediated and supports innate and adaptive anti-tumour immune priming. Mol Cancer 2011; 10: 20.
[http://dx.doi.org/10.1186/1476-4598-10-20] [PMID: 21338484]
]. In addition, the abnormal characteristics of tumors can be manipulated for generating or increasing virus oncoselectivity. For example, the promoters of tyrosinase or survivin genes that are over-expressed in melanoma, have been incorporated into virus genomes and used for the oncospecific targeting of engineered OVs [10Banerjee NS, Rivera AA, Wang M, et al. Analyses of melanoma-targeted oncolytic adenoviruses with tyrosinase enhancer/promoter-driven E1A, E4, or both in submerged cells and organotypic cultures. Mol Cancer Ther 2004; 3(4): 437-49.
[PMID: 15078987]
, 11Tamm I, Wang Y, Sausville E, et al. IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res 1998; 58(23): 5315-20.
[PMID: 9850056]
]. Further, OVs have been engineered to express immunostimulatory or immunomodulatory genes to increase their immumoreactivity and antiproliferative efficacy. Genes of cytokines such as granulocyte monocyte colony-stimulating factor (GM-CSF), IL-2, IL-12, IL-18 and IFN-γ have been incorporated into OVs [12Edukulla R, Woller N, Mundt B, et al. Antitumoral immune response by recruitment and expansion of dendritic cells in tumors infected with telomerase-dependent oncolytic viruses. Cancer Res 2009; 69(4): 1448-58.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-1160] [PMID: 19190348]
, 13Stephenson KB, Barra NG, Davies E, Ashkar AA, Lichty BD. Expressing human interleukin-15 from oncolytic vesicular stomatitis virus improves survival in a murine metastatic colon adenocarcinoma model through the enhancement of anti-tumor immunity. Cancer Gene Ther 2012; 19(4): 238-46.
[http://dx.doi.org/10.1038/cgt.2011.81] [PMID: 22158521]
]. Among these cytokines, GM-CSF has been the most widely engineered into OVs to recruit immune cells, enhancing the OVs’ clinical benefit. Talimogene laherparepvec (T-vec) and JX-594, the two most advanced OVs approved for treating melanoma and head and neck cancer respectively, are modified herpes simplex virus (HSV) and vaccinia virus (VV) both expressing human GM-CSF [14Lichty BD, Breitbach CJ, Stojdl DF, Bell JC. Going viral with cancer immunotherapy. Nat Rev Cancer 2014; 14(8): 559-67.
[http://dx.doi.org/10.1038/nrc3770] [PMID: 24990523]
]. By causing tumor lysis and immune cells recruitment, OVs armed with immunostimulatory genes exhibit higher efficacy in regressing tumors than immunostimulatory gene products alone. For example, the objective response rate was 26% including the complete response in 11% of patients with treatment of T-vec; while an objective response rate of 6% with 1% complete response rate was observed with treatment of GM-CSF [15Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J Clin Oncol 2015; 33(25): 2780-8.
[http://dx.doi.org/10.1200/JCO.2014.58.3377] [PMID: 26014293]
, 16Collichio FA, Amatruda T, Senzer NN, et al. OPTiM: A randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma. J Clin Oncol 2013; 31(18_suppl): LBA9008.
[PMID: 28136063]
].

Here, we provide a review about the oncolytic virotherapy focused on melanoma treatment, emphasizing the successful strategies used to attain increased tumor specificity, oncolysis and immunoreactivity. This review also describes the promising strategies with challenges, which lead the way for future modifications and usage of OVs, and is divided into three sections. The first section reviews biomarkers including the established, prognostic and immunotherapeutic biomarkers. The biomarkers are not only applicable for diagnostic purposes, but also significantly useful in OV genetic modifications for targeting melanoma cells. The second section reviews the successful strategies of engineering OVs for melanoma regression, which can be broadly divided into four categories: (1) modification of virus tropism to the tumor cells by transductional and transcriptional targeting or cancer signaling pathway targeting; (2) triggering vascular-disrupting effects and modifying the extracellular matrix (ECM) components for virus spread; (3) viral enhancement of innate and adaptive antitumor immune responses; and (4) combinational therapies. The subsequent section focuses on the prospective useful strategies along with the challenges for optimizing the clinical efficiency of OVs.

1. BIOMARKERS OF MELANOMA

In recent years, the development of large-scale sequencing approaches has uncovered the complex molecular and cellular abnormalities leading to the tumorigenesis of melanoma. These discoveries have shown that melanoma is a genetically heterogeneous disorder with many distinctive molecular and cellular defects, which strongly suggest the individualization of diagnosis and treatment. American Joint Committee on Cancer (AJCC) melanoma clarifies TNM staging (T: primary tumor; N: regional lymph nodes; M: metastasis) and provides well-established hallmarks for melanoma progression such as tumor thickness, ulceration, mitotic rate and lactate dehydrogenase (LDH) [17Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 2009; 27(36): 6199-206.
[http://dx.doi.org/10.1200/JCO.2009.23.4799] [PMID: 19917835]
]. Although these criteria correlate with the prognostic information, they are still unable to identify the prognosis of specific individuals and sometimes provide unreliable results. For example, some patients with thick tumors have excellent prognosis, and those with thin tumors result in poor outcome [18Griewank KG. Biomarkers in melanoma. Scand J Clin Lab Invest Suppl 2016; 245: S104-12.
[http://dx.doi.org/10.1080/00365513.2016.1210336] [PMID: 27467728]
]. Therefore, addition of the measurable diagnostic indicators of melanoma, referred as biomarkers, would be likely to determine more accurately the prognosis. Also, biomarkers are able to determine which kind of treatment the patients would benefit from. Therefore, detection of these biomarkers would indicate the therapies needed for better outcomes. For example, the detection of BRAF mutation and administration of BRAF V600E kinase inhibitors such as dabrafenib significantly improve the clinical situation of the patients [6Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
, 19Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380(9839): 358-65.
[http://dx.doi.org/10.1016/S0140-6736(12)60868-X] [PMID: 22735384]
]. In this section, we aim to briefly review the established biomarkers and biomarkers of interest in the last few years. These biomarkers not only identify high-risk melanoma patients, but also provide potential therapeutic targets for individualized treatment.

1.1. Established Biomarkers

1.1.1. Thickness Measurement

Alexander Breslow [20Breslow A. Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Ann Surg 1970; 172(5): 902-8.
[http://dx.doi.org/10.1097/00000658-197011000-00017] [PMID: 5477666]
] first reported the importance of using thickness and cross-sectional areas in reflecting the tumor burden. The prognostic significance of thickness measurement was also confirmed in the latest AJCC melanoma staging criteria. While 10-year survival rate is approximately 92% in patients with ≤ 1.00 mm thick melanoma, it drops to 50% in those with > 4mm thickness [17Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 2009; 27(36): 6199-206.
[http://dx.doi.org/10.1200/JCO.2009.23.4799] [PMID: 19917835]
].

1.1.2. Melanoma Ulceration

The presence of tumor ulceration has been shown to adversely affect the disease outcome [17Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 2009; 27(36): 6199-206.
[http://dx.doi.org/10.1200/JCO.2009.23.4799] [PMID: 19917835]
]. The ulceration caused by tumor eroding epidermis modifies the local environment and favors the expansion of the tumor burden [21Gogas H, Eggermont AM, Hauschild A, et al. Biomarkers in melanoma. Ann Oncol 2009; 20(Suppl. 6): vi8-vi13.
[http://dx.doi.org/10.1093/annonc/mdp251] [PMID: 19617299]
]. Recent studies have shown that treatment of IFN-α significantly increase the survival rate in the patients with ulcerated melanoma in comparison to those with non-ulcerated melanoma, suggesting that ulceration might lead to decreased production of endogenous IFN [22Eggermont AM, Suciu S, Testori A, Patel P, Spatz A. Ulceration of primary melanoma and responsiveness to adjuvant interferon therapy: analysis of the adjuvant trials EORTC18952 and EORTC18991 in 2,644 patients. J Clin Oncol 2009; 27(15S): 9007-7. PubMed PMID: WOS:000276606606023., 23Eggermont AM, Suciu S, Testori A, et al. Long-term results of the randomized phase III trial EORTC 18991 of adjuvant therapy with pegylated interferon alfa-2b versus observation in resected stage III melanoma. J Clin Oncol 2012; 30(31): 3810-8.
[http://dx.doi.org/10.1200/JCO.2011.41.3799] [PMID: 23008300]
].

1.1.3. Mitotic Activity

DNA replication involves the activation of replication origin firing (ROF) genes and securin which separates sister chromatids [24Winnepenninckx V, van den Oord JJ. Gene expression profiling and clinical outcome in melanoma: in search of novel prognostic factors. Expert Rev Anticancer Ther 2007; 7(11): 1611-31.
[http://dx.doi.org/10.1586/14737140.7.11.1611] [PMID: 18020928]
]. Melanoma in patients with poor prognosis is often characterized with over-activation of ROF and expression of securin gene [24Winnepenninckx V, van den Oord JJ. Gene expression profiling and clinical outcome in melanoma: in search of novel prognostic factors. Expert Rev Anticancer Ther 2007; 7(11): 1611-31.
[http://dx.doi.org/10.1586/14737140.7.11.1611] [PMID: 18020928]
, 25Winnepenninckx V, Lazar V, Michiels S, et al. Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst 2006; 98(7): 472-82.
[http://dx.doi.org/10.1093/jnci/djj103] [PMID: 16595783]
]. Increased mitotic rate represents higher metabolic activity of tumor cells and therefore the increased tumor proliferation and metastasis.

1.2. Prognostic Markers

Among serological biomarkers, LDH remains to date the strongest prognostic markers in melanoma [26Solassol J, Du-Thanh A, Maudelonde T, Guillot B. Serum proteomic profiling reveals potential biomarkers for cutaneous malignant melanoma. Int J Biol Markers 2011; 26(2): 82-7.
[http://dx.doi.org/10.5301/JBM.2011.8344] [PMID: 21607923]
]. LDH is less sensitive in the early stage. However, it is indicative of the tumor growth and malignancy progression [27Hofmann MA, Gussmann F, Fritsche A, et al. Diagnostic value of melanoma inhibitory activity serum marker in the follow-up of patients with stage I or II cutaneous melanoma. Melanoma Res 2009; 19(1): 17-23.
[http://dx.doi.org/10.1097/CMR.0b013e32831bc78c] [PMID: 19104451]
, 28Hofmann MA, Schicke B, Fritsch A, et al. Impact of lymph node metastases on serum level of melanoma inhibitory activity in stage III melanoma patients. J Dermatol 2011; 38(9): 880-6.
[PMID: 21658116]
]. It is still noteworthy that LDH, which occurs also in infection, inflammation and hemolysis, might lead to false positive readouts. S100 serves as diagnostic and prognostic marker for melanoma, as its expression is much higher in the malignant melanoma than in the normal tissues [29Davey GE, Murmann P, Hoechli M, Tanaka T, Heizmann CW. Calcium-dependent translocation of S100A11 requires tubulin filaments. Biochim Biophys Acta 2000; 1498(2-3): 220-32.
[http://dx.doi.org/10.1016/S0167-4889(00)00098-7] [PMID: 11108965]
, 30Hsieh HL, Schäfer BW, Cox JA, Heizmann CW. S100A13 and S100A6 exhibit distinct translocation pathways in endothelial cells. J Cell Sci 2002; 115(Pt 15): 3149-58.
[PMID: 12118070]
]. It has been found that elevated level of S100, in particular S100B, is related to the poor prognosis and low survival rate [31Oberholzer PA, Urosevic M, Steinert HC, Dummer R. Baseline staging of melanoma with unknown primary site: the value of serum s100 protein and positron emission tomography. Dermatology (Basel) 2008; 217(4): 351-5.
[http://dx.doi.org/10.1159/000155878] [PMID: 18799883]
, 32Beyeler M, Waldispuhl S, Strobel K, Joller-Jemelka HI, Burg G, Dummer R. Detection of melanoma relapse: first comparative analysis on imaging techniques versus S100 protein. Dermatology (Basel) 2006; 213(3): 187-91.
[http://dx.doi.org/10.1159/000095034] [PMID: 17033166]
]. Vascular endothelial growth factor (VEGF) and melanoma-inhibitory activity (MIA) protein are involved in tumor-associated angiogenesis and melanoma cell interaction with the ECM respectively. Studies have shown that elevated levels of VEGF and MIA are associated with more advanced disease and poor survival rates [33Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased serum concentration of angiogenic factors in malignant melanoma patients correlates with tumor progression and survival. J Clin Oncol 2001; 19(2): 577-83.
[PMID: 11208853]
, 34Díaz-Lagares A, Alegre E, Arroyo A, et al. Evaluation of multiple serum markers in advanced melanoma. Tumour Biol 2011; 32(6): 1155-61.
[http://dx.doi.org/10.1007/s13277-011-0218-x] [PMID: 21858537]
]. Other serum biomarkers for melanoma include tyrosinase [35Visús C, Andres R, Mayordomo JI, et al. Prognostic role of circulating melanoma cells detected by reverse transcriptase-polymerase chain reaction for tyrosinase mRNA in patients with melanoma. Melanoma Res 2007; 17(2): 83-9.
[http://dx.doi.org/10.1097/CMR.0b013e3280a60878] [PMID: 17496783]
], osteopontin [36McAllister SS, Gifford AM, Greiner AL, et al. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell 2008; 133(6): 994-1005.
[http://dx.doi.org/10.1016/j.cell.2008.04.045] [PMID: 18555776]
], IL-8 [37Sanmamed MF, Carranza-Rua O, Alfaro C, et al. Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins. Clin Cancer Res 2014; 20(22): 5697-707.
[http://dx.doi.org/10.1158/1078-0432.CCR-13-3203] [PMID: 25224278]
] etc.

In addition to the serum biomarkers, molecules over-expressed in the tumor tissues also function as an effective indicator of the melanoma progression and metastasis. For example, matrix metalloproteinases (MMP), which proteolytically degrade ECM proteins, have been shown to promote melanoma cell migration and tumor progression. The MMP-1/3 positive melanoma was shown to correlate with higher malignancy and decreased overall survival [38Nikkola J, Vihinen P, Vlaykova T, Hahka-Kemppinen M, Kähäri VM, Pyrhönen S. High expression levels of collagenase-1 and stromelysin-1 correlate with shorter disease-free survival in human metastatic melanoma. Int J Cancer 2002; 97(4): 432-8.
[http://dx.doi.org/10.1002/ijc.1636] [PMID: 11802203]
]. Cell adhesion molecules (CAMs) such as the carcinoembryonic antigen-related cell adhesion molecule-1 and melanoma-associated antigen (MAA), which play an essential role in cell adhesion, show abnormal expression in melanoma and participate in melanoma migration and metastasis [39Sapoznik S, Ortenberg R, Schachter J, Markel G. CEACAM1 in malignant melanoma: a diagnostic and therapeutic target. Curr Top Med Chem 2012; 12(1): 3-10.
[http://dx.doi.org/10.2174/156802612798919259] [PMID: 22196267]
, 40Vergilis IJ, Szarek M, Ferrone S, Reynolds SR. Presence and prognostic significance of melanoma-associated antigens CYT-MAA and HMW-MAA in serum of patients with melanoma. J Invest Dermatol 2005; 125(3): 526-31.
[http://dx.doi.org/10.1111/j.0022-202X.2005.23798.x] [PMID: 16117794]
]. Other tissue-specific biomarkers such as cyclooxygenase-2 (Cox-2) and galectin-3 have also been shown to associate with melanoma tumor size and malignancy and overall patient survival.

1.3. Genetic Biomarkers

About half of the melanomas harbor the mutation of BRAF gene (BRAFV600E, and possibly BRAFV600K), which results in constant activation of BRAF kinase [6Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
]. This likely induces the promotion of Ras-Raf-MEK-ERK signaling pathway and therefore the proliferation of melanoma. Like BRAF mutation, N-Ras gene mutation is also considered as the “driver mutation” and found in approximately 15-20% metastatic melanoma patients [4Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res 2015; 3(3): 288-95.
[http://dx.doi.org/10.1158/2326-6066.CIR-14-0207] [PMID: 25736262]
]. BRAF and N-Ras mutations have been shown to link to the metastasis and poor outcome [41Mann GJ, Pupo GM, Campain AE, et al. BRAF mutation, NRAS mutation, and the absence of an immune-related expressed gene profile predict poor outcome in patients with stage III melanoma. J Invest Dermatol 2013; 133(2): 509-17.
[http://dx.doi.org/10.1038/jid.2012.283] [PMID: 22931913]
]. With lower frequencies, KIT mutations have been observed in mucosal and acral melanomas and melanomas with sun-damaged skin [42Kong Y, Si L, Zhu Y, et al. Large-scale analysis of KIT aberrations in Chinese patients with melanoma. Clin Cancer Res 2011; 17(7): 1684-91.
[http://dx.doi.org/10.1158/1078-0432.CCR-10-2346] [PMID: 21325067]
]. Cutaneous melanomas are characterized with mutations of BRAF, N-Ras, MEK, NF1 and KIT, while most ocular melanomas harbor the mutations of GNAQ and GNA11 [43Hodis E, Watson IR, Kryukov GV, et al. A landscape of driver mutations in melanoma. Cell 2012; 150(2): 251-63.
[http://dx.doi.org/10.1016/j.cell.2012.06.024] [PMID: 22817889]
]. In order to discriminate Spitz nevi and Spitzoid melanomas, biomarkers such as CDKN2A, RREB1, MYC and CCND1 have been utilized [44Gerami P, Li G, Pouryazdanparast P, et al. A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms. Am J Surg Pathol 2012; 36(6): 808-17.
[http://dx.doi.org/10.1097/PAS.0b013e31824b1efd] [PMID: 22588064]
].

1.4. Biomarkers For Immunotherapy

Cytotoxic T-lymphocyte antigen-4 (CTLA-4), a protein receptor found on the surface of some T cells, functions as an immune checkpoint. By binding to the B7 on the APCs, it transmits an inhibitory signal to T cell activation, which confers vulnerability to the tumor invasion [45Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol 2009; 27: 591-619.
[http://dx.doi.org/10.1146/annurev.immunol.021908.132706] [PMID: 19132916]
]. Ipilimumab (FDA approved for melanoma therapy in 2011), the human monoclonal immunoglobulin G1 against CTLA-4, has been shown to expand anti-tumor T cell activity and inhibit immune tolerance, therefore significantly enhancing the melanoma patients overall survival rate [46Lee B, Mukhi N, Liu D. Current management and novel agents for malignant melanoma. J Hematol Oncol 2012; 5: 3.
[http://dx.doi.org/10.1186/1756-8722-5-3] [PMID: 22333219]
, 47Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364(26): 2517-26.
[http://dx.doi.org/10.1056/NEJMoa1104621] [PMID: 21639810]
]. Another anti CTLA-4 monoclonal antibody is tremelimumab, which is still undergoing human trials [48Poust J. Targeting metastatic melanoma. Am J Health Syst Pharm 2008; 65(24)(Suppl. 9): S9-S15.
[http://dx.doi.org/10.2146/ajhp080461] [PMID: 19052265]
].

Programmed death receptor-1 (PD-1) is a protein expressed on the cell surface of T cells. By binding to the ligands PD-L1 and PD-L2 expressed on tumor and stromal cells, it transmits the signal in down-regulating T cell activation and promoting self-tolerance [49Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 2012; 24(2): 207-12.
[http://dx.doi.org/10.1016/j.coi.2011.12.009] [PMID: 22236695]
]. As PD-1 is also expressed on B cells and macrophages, it is likely more potent compared to CTLA-4 in negatively inhibiting immune responses [50Agata Y, Kawasaki A, Nishimura H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 1996; 8(5): 765-72.
[http://dx.doi.org/10.1093/intimm/8.5.765] [PMID: 8671665]
]. PD-1 inhibitors, which block PD-1 and activate immune responses, have achieved varying success in different cancers including melanoma. Nivolumab, lambrolizumab and pembrolizumab, which are anti-PD-1 antibodies, have been approved by FDA for melanoma therapy [51Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013; 369(2): 134-44.
[http://dx.doi.org/10.1056/NEJMoa1305133] [PMID: 23724846]
, 52Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369(2): 122-33.
[http://dx.doi.org/10.1056/NEJMoa1302369] [PMID: 23724867]
].

2. MELANOMA THERAPIES AND EMERGENCE OF ONCOLYTIC VIROTHERAPY

The traditional targeted therapies for melanoma include BRAF inhibitors (vemurafenib and dabrafenib), MEK inhibitors (trametinib and cobimetinib), tyrosine kinase inhibitors (imatinib) and angiogenesis inhibitors (aflibercept and bevacizumab) [6Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
, 19Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380(9839): 358-65.
[http://dx.doi.org/10.1016/S0140-6736(12)60868-X] [PMID: 22735384]
, 53Gonzalez D, Fearfield L, Nathan P, et al. BRAF mutation testing algorithm for vemurafenib treatment in melanoma: recommendations from an expert panel. Br J Dermatol 2013; 168(4): 700-7.
[http://dx.doi.org/10.1111/bjd.12248] [PMID: 23360189]
-60Tarhini AA, Frankel P, Margolin KA, et al. Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin Cancer Res 2011; 17(20): 6574-81.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-1463] [PMID: 21880788]
]. The melanoma immunotherapies include the cytokine therapies consisting of IFN-α and IL-2 [61Kirkwood JM, Ernstoff MS. Interferons in the treatment of human cancer. J Clin Oncol 1984; 2(4): 336-52.
[PMID: 6323641]
-63Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999; 17(7): 2105-16.
[PMID: 10561265]
] and inhibitors to CTLA-4 (ipilimumab) and PD-1 (nivolumab, lambrolizumab and pembrolizumab) [47Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364(26): 2517-26.
[http://dx.doi.org/10.1056/NEJMoa1104621] [PMID: 21639810]
, 51Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013; 369(2): 134-44.
[http://dx.doi.org/10.1056/NEJMoa1305133] [PMID: 23724846]
, 52Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369(2): 122-33.
[http://dx.doi.org/10.1056/NEJMoa1302369] [PMID: 23724867]
].

OVs, with the initial clinical testing of the concept in 1950s [64Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther 2007; 15(4): 651-9.
[http://dx.doi.org/10.1038/sj.mt.6300108] [PMID: 17299401]
], have achieved fast development over the past decade. With the ability of self-replicating in tumor cells and carrying additional genes for immune-stimulatory products, OVs have shown tremendous potential at being better anticancer therapeutics than the conventional drugs. It is encouraging that T-vec, a modified HSV strain expressing human GM-CSF, has been approved by FDA for melanoma therapy in 2015 [65Kee D, McArthur G. Immunotherapy of melanoma. Eur J Surg Oncol 2016; S0748-7983(16)30684-9.
[PMID: 27514721]
]. The other most advanced oncolytic virotherapy used clinically is the JX-594, which has been approved in China for head and neck cancer therapy [66Garber K. China approves worlds first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst 2006; 98(5): 298-300.
[http://dx.doi.org/10.1093/jnci/djj111] [PMID: 16507823]
]. It is the oncolytic VV engineered by addition of GM-CSF gene and deletion of viral thymidine kinase gene. The expression of GM-CSF induces the recruitment and activation of APCs and therefore the tumor-specific T cell responses. Other viruses which are undergoing clinical tests for melanoma therapy include reovirus and coxsackievirus [9Steele L, Errington F, Prestwich R, et al. Pro-inflammatory cytokine/chemokine production by reovirus treated melanoma cells is PKR/NF-κB mediated and supports innate and adaptive anti-tumour immune priming. Mol Cancer 2011; 10: 20.
[http://dx.doi.org/10.1186/1476-4598-10-20] [PMID: 21338484]
, 67Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 2015; 14(9): 642-62.
[http://dx.doi.org/10.1038/nrd4663] [PMID: 26323545]
].

2.1. Transductional Targeting

One of the major challenges of engineering OVs is that the natural tropism of viruses does not match the therapeutic need. For example, the administration of adenovirus systemically, among which the majority is sequestered by hepatic macrophages and hepatocyte transduction, results in adenovirus-mediated liver toxicity [68Glasgow JN, Everts M, Curiel DT. Transductional targeting of adenovirus vectors for gene therapy. Cancer Gene Ther 2006; 13(9): 830-44.
[http://dx.doi.org/10.1038/sj.cgt.7700928] [PMID: 16439993]
]. Therefore, approaches that retarget the viruses are needed to avoid the toxic side effects and facilitate the efficient infection to the targeted tumor cells. By modifying the viral coat proteins, which is referred to as transductional targeting, the viruses are engineered to infect the tumor cells that they do not infect naturally [69Barker SD, Dmitriev IP, Nettelbeck DM, et al. Combined transcriptional and transductional targeting improves the specificity and efficacy of adenoviral gene delivery to ovarian carcinoma. Gene Ther 2003; 10(14): 1198-204.
[http://dx.doi.org/10.1038/sj.gt.3301974] [PMID: 12833129]
]. For example, the modification of hypervariable loop 5 in the capsid protein hexon in adenovirus prevents infection of hepatocytes and expand the anticancer therapeutic window [70Shashkova EV, May SM, Doronin K, Barry MA. Expanded anticancer therapeutic window of hexon-modified oncolytic adenovirus. Mol Ther 2009; 17(12): 2121-30.
[http://dx.doi.org/10.1038/mt.2009.217] [PMID: 19755961]
].

Pseudotyping, which is the first approach of transductional targeting, is to replace viral attachment proteins of one virus with the tropism determinant ligands of other viruses whose tropism is against the targeted tumor cells. Exciting results have been observed by using oncoretroviral vector pseudotyped with a modified chimeric Sindbis virus envelop glycoproteins and conjugated with antibody specific for P-glycoprotein, for targeting metastatic melanoma [71Morizono K, Xie Y, Ringpis GE, et al. Lentiviral vector retargeting to P-glycoprotein on metastatic melanoma through intravenous injection. Nat Med 2005; 11(3): 346-52.
[http://dx.doi.org/10.1038/nm1192] [PMID: 15711560]
].

While pseudotyping is limited by the viral attachment proteins and the receptors exclusively present on the target cells, the other approach of transductional targeting by using adaptors, the molecules with one end binding the viral attachment protein and the other binding the target cells’ receptors, possesses more flexibility without changing the structure of viral vectors [72Waehler R, Russell SJ, Curiel DT. Engineering targeted viral vectors for gene therapy. Nat Rev Genet 2007; 8(8): 573-87.
[http://dx.doi.org/10.1038/nrg2141] [PMID: 17607305]
]. For example, the fusing of ectodomain of coxsackie virus and adenovirus receptor (CAR) and a single chain antibody against carcinoembryonic antigen allowed the adenovirus to target on tumors and ablated the liver tropism [73Li HJ, Everts M, Pereboeva L, et al. Adenovirus tumor targeting and hepatic untargeting by a coxsackie/adenovirus receptor ectodomain anti-carcinoembryonic antigen bispecific adapter. Cancer Res 2007; 67(11): 5354-61.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-4679] [PMID: 17545616]
]. Avidin, Biotin and monoclonal antibodies are also used in adaptor systems. For example, a biotin-acceptor peptide has been incorporated into the adenovirus fiber capsid protein and biotinylated and coupled to an avidin-binding ligand [74Parrott MB, Adams KE, Mercier GT, Mok H, Campos SK, Barry MA. Metabolically biotinylated adenovirus for cell targeting, ligand screening, and vector purification. Mol Ther 2003; 8(4): 688-700.
[http://dx.doi.org/10.1016/S1525-0016(03)00213-2] [PMID: 14529842]
]. MMP and MAA, as the biomarkers of melanoma, play a critical role in melanoma progression and metastasis. Efficient targeting of retrovirus to melanoma cells has been done by fusing a single chain antibody recognizing the high molecular weight MAA (HMWMAA) followed by a proline-rich linker and a MMP-2 cleavage site, to the amino terminus of murine leukemia virus (MLV-A) envelop glycoprotein [75Martín F, Chowdhury S, Neil S, Phillipps N, Collins MK. Envelope-targeted retrovirus vectors transduce melanoma xenografts but not spleen or liver. Mol Ther 2002; 5(3): 269-74.
[http://dx.doi.org/10.1006/mthe.2002.0550] [PMID: 11863416]
, 76Martin F, Neil S, Kupsch J, Maurice M, Cosset F, Collins M. Retrovirus targeting by tropism restriction to melanoma cells. J Virol 1999; 73(8): 6923-9.
[PMID: 10400790]
]. Following the attachment with HMWMAA, MMP cleaves the MMP-2 cleavage site and allows the infection of MLV. Another example combining the avidin/biotin and monoclonal antibody is by first biotinylating VV to which the avidin is subsequently added and then coating the avidin bridge with biotinylated antibodies. The antibodies are against the cell surface makers of target cells [77Purow B, Staveley-OCarroll K. Targeting of vaccinia virus using biotin-avidin viral coating and biotinylated antibodies. J Surg Res 2005; 123(1): 49-54.
[http://dx.doi.org/10.1016/j.jss.2004.04.022] [PMID: 15652950]
].

2.2. Transcriptional Targeting

The transcriptional regions consisting of promoters and enhancers regulate the kinetics and levels of mRNA production, and activators recruit RNA polymerase II for initiating the transcription of mRNA. Cancer-specific promoters can be activated in the presence of a subset of activators existing in cancerous cells but not in normal cells [78Wu L, Johnson M, Sato M. Transcriptionally targeted gene therapy to detect and treat cancer. Trends Mol Med 2003; 9(10): 421-9.
[http://dx.doi.org/10.1016/j.molmed.2003.08.005] [PMID: 14557054]
]. Therefore, transcriptional targeting in which part of the viral genome is placed under the control of cancer-specific promoter serves as a feasible way for generating tumor selectivity.

Tissue targeted virotherapy has been investigated in a variety of cancers including melanoma. Promoters of genes over-expressed in melanoma have been chosen and utilized, among which most are the above mentioned melanoma biomarkers’ promoters such as Cox-2, C-X-C chemokine receptor type 4 (CXCR-4), tyrosinase and survivin promoters [10Banerjee NS, Rivera AA, Wang M, et al. Analyses of melanoma-targeted oncolytic adenoviruses with tyrosinase enhancer/promoter-driven E1A, E4, or both in submerged cells and organotypic cultures. Mol Cancer Ther 2004; 3(4): 437-49.
[PMID: 15078987]
, 78Wu L, Johnson M, Sato M. Transcriptionally targeted gene therapy to detect and treat cancer. Trends Mol Med 2003; 9(10): 421-9.
[http://dx.doi.org/10.1016/j.molmed.2003.08.005] [PMID: 14557054]
-80Zhu ZB, Makhija SK, Lu B, et al. Transcriptional targeting of adenoviral vector through the CXCR4 tumor-specific promoter. Gene Ther 2004; 11(7): 645-8.
[http://dx.doi.org/10.1038/sj.gt.3302089] [PMID: 15029227]
]. Cox-2 is closely associated with the progression and metastasis of melanoma, whereas benign nevi and tissues are Cox-2 negative [81Denkert C, Köbel M, Berger S, et al. Expression of cyclooxygenase 2 in human malignant melanoma. Cancer Res 2001; 61(1): 303-8.
[PMID: 11196178]
]. CXCR-4 over-expressed on human melanoma cells has also been shown to play a role in melanoma progression [82Robledo MM, Bartolome RA, Longo N, et al. Expression of functional chemokine receptors CXCR3 and CXCR4 on human melanoma cells. J Biol Chem 2001; 276(48): 45098-105.
[http://dx.doi.org/10.1074/jbc.M106912200] [PMID: 11571298]
]. Incorporation of Cox-2 and CXCR-4 promoters in adenovirus genome has been shown to significantly enhance the virus-mediated gene expression in the malignant melanoma instead of non-malignant primary melanocytes [79Lu B, Makhija SK, Nettelbeck DM, et al. Evaluation of tumor-specific promoter activities in melanoma. Gene Ther 2005; 12(4): 330-8.
[http://dx.doi.org/10.1038/sj.gt.3302385] [PMID: 15696177]
, 80Zhu ZB, Makhija SK, Lu B, et al. Transcriptional targeting of adenoviral vector through the CXCR4 tumor-specific promoter. Gene Ther 2004; 11(7): 645-8.
[http://dx.doi.org/10.1038/sj.gt.3302089] [PMID: 15029227]
]. Tyrosinase, which plays an essential role in melanin synthesis and melanocyte differentiation, has been used as a melanoma biomarker for disease detection purposes [83Smith B, Selby P, Southgate J, Pittman K, Bradley C, Blair GE. Detection of melanoma cells in peripheral blood by means of reverse transcriptase and polymerase chain reaction. Lancet 1991; 338(8777): 1227-9.
[http://dx.doi.org/10.1016/0140-6736(91)92100-G] [PMID: 1719320]
]. It has been demonstrated that incorporation of tyrosinase promoter to drive the expression of the viral genes of adenovirus leads to an obvious melanoma-selective viral cytotoxicity [84Nettelbeck DM, Rivera AA, Balagué C, Alemany R, Curiel DT. Novel oncolytic adenoviruses targeted to melanoma: specific viral replication and cytolysis by expression of E1A mutants from the tyrosinase enhancer/promoter. Cancer Res 2002; 62(16): 4663-70.
[PMID: 12183423]
]. Survivin is expressed on a variety of tumors but undetectable in terminally differentiated tissues. It is associated with tumor progression and resistance to chemo- and radio-therapies, which is likely due to its ability of modulating apoptosis pathways and inhibiting apoptotic enzymes such as caspase-3 and caspase-7 [11Tamm I, Wang Y, Sausville E, et al. IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res 1998; 58(23): 5315-20.
[PMID: 9850056]
, 85Asanuma K, Kobayashi D, Furuya D, Tsuji N, Yagihashi A, Watanabe N. A role for survivin in radioresistance of pancreatic cancer cells. Jpn J Cancer Res 2002; 93(9): 1057-62.
[http://dx.doi.org/10.1111/j.1349-7006.2002.tb02483.x] [PMID: 12359060]
]. Lu et al. have reported that compared with Cox-2 and CXCR-4 promoters, the activity of survivin promoter is higher in melanoma cells and lower in normal tissues such as livers and epithelial melanocytes [79Lu B, Makhija SK, Nettelbeck DM, et al. Evaluation of tumor-specific promoter activities in melanoma. Gene Ther 2005; 12(4): 330-8.
[http://dx.doi.org/10.1038/sj.gt.3302385] [PMID: 15696177]
]. A continued search for a better cancer-specificity is warranted, as higher expression in tumor cells and lower expression in normal tissues for transgenes in viral vectors is always desirable.

2.3. Targeting Cancer Signaling Pathways

Mutations in ras gene, which lead to the constant activation of Ras and therefore cell over-proliferation and tumorigenesis, have been reported in 30% of all cancers [86Marcato P, Shmulevitz M, Lee PW. Connecting reovirus oncolysis and Ras signaling. Cell Cycle 2005; 4(4): 556-9.
[http://dx.doi.org/10.4161/cc.4.4.1600] [PMID: 15753655]
]. The incidence of ras mutations in melanomas is around 20% and the majority is N-ras mutations [87Bos JL. ras oncogenes in human cancer: a review. Cancer Res 1989; 49(17): 4682-9.
[PMID: 2547513]
]. It has been demonstrated that reovirus and VSV, in unaltered form, replicate preferentially and effectively in the Ras-mutated cancer cells and therefore serve as natural OVs [88Errington F, White CL, Twigger KR, et al. Inflammatory tumour cell killing by oncolytic reovirus for the treatment of melanoma. Gene Ther 2008; 15(18): 1257-70.
[http://dx.doi.org/10.1038/gt.2008.58] [PMID: 18401435]
, 89Noser JA, Mael AA, Sakuma R, et al. The RAS/Raf1/MEK/ERK signaling pathway facilitates VSV-mediated oncolysis: implication for the defective interferon response in cancer cells. Mol Ther 2007; 15(8): 1531-6.
[http://dx.doi.org/10.1038/sj.mt.6300193] [PMID: 17505473]
]. It has been shown that reovirus efficiently infects and kills melanoma cells and regresses the xenografted melanoma tumors. In addition, Ras signaling pathway has been shown to be involved in reovirus-induced apoptosis in human melanoma cells [9Steele L, Errington F, Prestwich R, et al. Pro-inflammatory cytokine/chemokine production by reovirus treated melanoma cells is PKR/NF-κB mediated and supports innate and adaptive anti-tumour immune priming. Mol Cancer 2011; 10: 20.
[http://dx.doi.org/10.1186/1476-4598-10-20] [PMID: 21338484]
]. Similarly, Noser et al. [89Noser JA, Mael AA, Sakuma R, et al. The RAS/Raf1/MEK/ERK signaling pathway facilitates VSV-mediated oncolysis: implication for the defective interferon response in cancer cells. Mol Ther 2007; 15(8): 1531-6.
[http://dx.doi.org/10.1038/sj.mt.6300193] [PMID: 17505473]
] have reported that Ras-overexpressing cancer cells are effectively killed by VSV and the oncolytic properties of VSV are dependent on the activation of Ras/Raf/MEK/ERK signaling pathway. It is noteworthy that the activation of Ras pathway is usually coupled with the down-regulation of IFN-induced antiviral responses such as the induction of protein kinase PKR, which further favors the virus replication in cancer cells.

Dysfunction of IFN signaling pathway has been reported in different cancers such as melanoma and breast cancer. Impaired IFN pathway aids tumor progression and immune escape, as IFN signaling is critical for the proper activation of key immune cells such as T, B and natural killer (NK) cells [90Critchley-Thorne RJ, Simons DL, Yan N, et al. Impaired interferon signaling is a common immune defect in human cancer. Proc Natl Acad Sci USA 2009; 106(22): 9010-5.
[http://dx.doi.org/10.1073/pnas.0901329106] [PMID: 19451644]
]. It has been reported that several melanomas are deficient in the key components of IFN signaling pathway such as STAT1 and STAT2 [91Wong LH, Krauer KG, Hatzinisiriou I, et al. Interferon-resistant human melanoma cells are deficient in ISGF3 components, STAT1, STAT2, and p48-ISGF3gamma. J Biol Chem 1997; 272(45): 28779-85.
[http://dx.doi.org/10.1074/jbc.272.45.28779] [PMID: 9353349]
]. One example of enhancing the viral oncoselectivity via targeting the impaired IFN pathway in tumors involves in the nonstructural protein NS1 of influenza virus which has been demonstrated to block the function of PKR induced by IFN [92Hatada E, Saito S, Fukuda R. Mutant influenza viruses with a defective NS1 protein cannot block the activation of PKR in infected cells. J Virol 1999; 73(3): 2425-33.
[PMID: 9971827]
]. Influenza virus with NS1 deleted has been shown to selectively infect and lyse the melanoma cells that are PKR-deficient and regress the melanoma tumors in vivo, while failing to replicate in normal cells with intact PKR-mediated antiviral responses [93Bergmann M, Romirer I, Sachet M, et al. A genetically engineered influenza A virus with ras-dependent oncolytic properties. Cancer Res 2001; 61(22): 8188-93.
[PMID: 11719449]
]. It is worthwhile to further investigate the application of viruses with the IFN or PKR viral inhibitor deletion for melanoma virotherapies, as IFN/PKR inhibitors are expressed by a variety of viruses such as VV and retrovirus [93Bergmann M, Romirer I, Sachet M, et al. A genetically engineered influenza A virus with ras-dependent oncolytic properties. Cancer Res 2001; 61(22): 8188-93.
[PMID: 11719449]
].

Deficiency in apoptosis is a critical contributor to the therapeutic resistance of melanoma. Two pathways, the intrinsic and extrinsic pathways lead to the apoptosis. While Bcl-2 family proteins such as Bax and Bak and the induction of apoptosome and caspases are involved in the intrinsic pathway, the extrinsic pathway features the death ligands of tumor necrosis factor (TNF)-α protein family including TNF-α, Fas ligand (CD95L) and TNF-related apoptosis-inducing ligand (TRAIL) [94Eberle J, Fecker LF, Forschner T, Ulrich C, Röwert-Huber J, Stockfleth E. Apoptosis pathways as promising targets for skin cancer therapy. Br J Dermatol 2007; 156(Suppl. 3): 18-24.
[http://dx.doi.org/10.1111/j.1365-2133.2007.07855.x] [PMID: 17488402]
]. The application of tyrosinase promoter controlled CD95L has been shown to selectively induce apoptosis in melanoma cells while sparing the non-melanoma cells [95Fecker LF, Geilen CC, Hossini AM, et al. Selective induction of apoptosis in melanoma cells by tyrosinase promoter-controlled CD95 ligand overexpression. J Invest Dermatol 2005; 124(1): 221-8.
[http://dx.doi.org/10.1111/j.0022-202X.2004.23572.x] [PMID: 15654977]
]. Viruses such as Newcastle disease virus (NDV) have been demonstrated to possess the ability of naturally inducing apoptosis in various cancer cells [96Ravindra PV, Tiwari AK, Sharma B, Chauhan RS. Newcastle disease virus as an oncolytic agent. Indian J Med Res 2009; 130(5): 507-13.
[PMID: 20090097]
]. A wide array of viral proteins which have been shown to induce apoptosis include: NS3 protein of bovine viral diarrhea virus, viral protein R of human immunodeficiency virus, hemagglutinin-neuraminidase of NDV and EIA of adenovirus [96Ravindra PV, Tiwari AK, Sharma B, Chauhan RS. Newcastle disease virus as an oncolytic agent. Indian J Med Res 2009; 130(5): 507-13.
[PMID: 20090097]
-99Uttenthal A, Stadejek T, Nylin B. Genetic diversity of bovine viral diarrhoea viruses (BVDV) in Denmark during a 10-year eradication period. APMIS 2005; 113(7-8): 536-41.
[http://dx.doi.org/10.1111/j.1600-0463.2005.apm_227.x] [PMID: 16086824]
]. It is appropriate to incorporate these apoptosis-inducing proteins into the OVs for expression. In addition, the over-expression of anti-apoptotic proteins is a critical factor for melanoma’s resistance to the chemotherapy. Therefore, it is feasible to increase the viral oncolysis by co-administrating with inhibitors of the anti-apoptotic proteins, such as BCL-2 inhibitors. It has been reported that the co-treatment of BCL-2 inhibitor improves the viral oncolysis of VSV in the tumors which are resistant to the treatment of virus alone [100Tumilasci VF, Olière S, Nguyên TL, Shamy A, Bell J, Hiscott J. Targeting the apoptotic pathway with BCL-2 inhibitors sensitizes primary chronic lymphocytic leukemia cells to vesicular stomatitis virus-induced oncolysis. J Virol 2008; 82(17): 8487-99.
[http://dx.doi.org/10.1128/JVI.00851-08] [PMID: 18579592]
].

2.4. Targeting Tumor Micro-Environment

The components in the environment where tumors exist, including the blood vessels, fibroblasts, immune cells and elements of ECM, are collectively termed as “tumor microenvironment”. The interaction between tumor cells and tumor microenvironment is critical in tumor initiation, progression and invasiveness. Also, tumor microenvironment affects the delivery and spread of the therapies such as OVs in tumors. Therefore, in this section, we will review the strategies used for increasing the efficacy of OVs and combating the melanoma progression via targeting the melanoma tumor microenvironment.

2.4.1. Targeting Tumor Vasculature

The formation of neovasculature is essential for the supply of nutrients and oxygen and melanoma cell dissemination. Therefore, the angiogenesis and lymphangiogenesis are critical for melanoma progression and metastasis [101Villanueva J, Herlyn M. Melanoma and the tumor microenvironment. Curr Oncol Rep 2008; 10(5): 439-46.
[http://dx.doi.org/10.1007/s11912-008-0067-y] [PMID: 18706274]
]. Angiogenesis of melanoma is stimulated by the overproduction of growth factors such as VEGF, fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and transforming growth factor (TGF). The rapidly increased vessel density and compression of the melanoma cells lead to the hypoxic and acidic environment throughout the tumor, which in turn induces the enhanced production of VEGF, FGF and TGF [102Wojton J, Kaur B. Impact of tumor microenvironment on oncolytic viral therapy. Cytokine Growth Factor Rev 2010; 21(2-3): 127-34.
[http://dx.doi.org/10.1016/j.cytogfr.2010.02.014] [PMID: 20399700]
]. In this regard, drugs inhibiting angiogenesis such as anti-VEGF antibody (bevacizumab and aflibercept) have been designed and utilized.

Targeting tumor angiogenesis has been used as an effective strategy for attaining increased tumor regression of OVs. It has been reported that the mutant VV engineered to express GLAF-1 which is the single-chain antibody targeting human VEGF, exhibited significantly increased tumor regression in human tumor xenografts compared with the parental virus [103Frentzen A, Yu YA, Chen N, et al. Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy. Proc Natl Acad Sci USA 2009; 106(31): 12915-20.
[http://dx.doi.org/10.1073/pnas.0900660106] [PMID: 19617539]
]. The HSV armed with transgene of platelet factor 4 which is an antiangiogenic agent has been shown to be more efficacious in inhibiting the tumor growth than the parental HSV [104Liu TC, Zhang T, Fukuhara H, et al. Oncolytic HSV armed with platelet factor 4, an antiangiogenic agent, shows enhanced efficacy. Mol Ther 2006; 14(6): 789-97.
[http://dx.doi.org/10.1016/j.ymthe.2006.07.011] [PMID: 17045531]
]. In addition, the incorporation of VEGF-targeted short hairpin RNA in the adenovirus results in greater tumor inhibition [105Yoo JY, Kim JH, Kwon YG, et al. VEGF-specific short hairpin RNA-expressing oncolytic adenovirus elicits potent inhibition of angiogenesis and tumor growth. Mol Ther 2007; 15(2): 295-302.
[http://dx.doi.org/10.1038/sj.mt.6300023] [PMID: 17235307]
]. Cytokines such as IL-12 and IL-18 have also been shown to reduce tumor angiogenesis [106Coughlin CM, Salhany KE, Wysocka M, et al. Interleukin-12 and interleukin-18 synergistically induce murine tumor regression which involves inhibition of angiogenesis. J Clin Invest 1998; 101(6): 1441-52.
[http://dx.doi.org/10.1172/JCI1555] [PMID: 9502787]
]. Adenovirus expressing IL-18 has exhibited greater apoptosis-induction and antitumor effect in xenografted human melanoma through inhibiting angiogenesis [107Zheng JN, Pei DS, Mao LJ, et al. Oncolytic adenovirus expressing interleukin-18 induces significant antitumor effects against melanoma in mice through inhibition of angiogenesis. Cancer Gene Ther 2010; 17(1): 28-36.
[http://dx.doi.org/10.1038/cgt.2009.38] [PMID: 19498459]
].

2.4.2. Targeting ECM

Effective oncolysis requires the virus entry and the subsequent replication and spread in the tumor environment. Therefore, a variety of strategies have been utilized to modify the physical aspects of tumor microenvironment to attain the optimal virus dispersal and replication.

Consistent with this idea, transgenes of enzymes degrading ECM components such as collagenase, hyaluronidase and MMP have been incorporated into OV modifications. The treatment of collagenase prior to the injection of adenovirus has been shown to enhance the virus spread [108Kuriyama N, Kuriyama H, Julin CM, Lamborn K, Israel MA. Pretreatment with protease is a useful experimental strategy for enhancing adenovirus-mediated cancer gene therapy. Hum Gene Ther 2000; 11(16): 2219-30.
[http://dx.doi.org/10.1089/104303400750035744] [PMID: 11084679]
]. Similarly, the expression of hyaluronidase by the mutant adenovirus resulted in improved spread of the virus within the tumor and increased antitumor efficacy [109Guedan S, Rojas JJ, Gros A, Mercade E, Cascallo M, Alemany R. Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth. Mol Ther 2010; 18(7): 1275-83.
[http://dx.doi.org/10.1038/mt.2010.79] [PMID: 20442708]
]. MMPs, in particular, MMP-1 and MMP-8 involve in degrading the triple-helical collagen in ECM. As melanoma biomarkers, MMPs correlate closely with melanoma metastasis and decreased overall survival rate [38Nikkola J, Vihinen P, Vlaykova T, Hahka-Kemppinen M, Kähäri VM, Pyrhönen S. High expression levels of collagenase-1 and stromelysin-1 correlate with shorter disease-free survival in human metastatic melanoma. Int J Cancer 2002; 97(4): 432-8.
[http://dx.doi.org/10.1002/ijc.1636] [PMID: 11802203]
]. Intratumoral injection of mutant adenovirus constructed to express MMP-8 led to significantly reduced collagen expression and increased tumor eradication [110Cheng J, Sauthoff H, Huang Y, et al. Human matrix metalloproteinase-8 gene delivery increases the oncolytic activity of a replicating adenovirus. Mol Ther 2007; 15(11): 1982-90.
[http://dx.doi.org/10.1038/sj.mt.6300264] [PMID: 17653103]
]. In addition, engineered OVs expressing relaxin and decorin, which are known as the ECM modulating proteins, have yielded enhanced virus penetration, persistence and spread compared to the control virus in melanoma [102Wojton J, Kaur B. Impact of tumor microenvironment on oncolytic viral therapy. Cytokine Growth Factor Rev 2010; 21(2-3): 127-34.
[http://dx.doi.org/10.1016/j.cytogfr.2010.02.014] [PMID: 20399700]
, 111Choi JW, Lee YS, Yun CO, Kim SW. Polymeric oncolytic adenovirus for cancer gene therapy. J Control Release 2015; 219: 181-91.
[http://dx.doi.org/10.1016/j.jconrel.2015.10.009] [PMID: 26453806]
, 112De Silva N, Atkins H, Kirn DH, Bell JC, Breitbach CJ. Double trouble for tumours: exploiting the tumour microenvironment to enhance anticancer effect of oncolytic viruses. Cytokine Growth Factor Rev 2010; 21(2-3): 135-41.
[http://dx.doi.org/10.1016/j.cytogfr.2010.02.007] [PMID: 20338801]
].

2.5. Targeting Immune Response Enhancement

Oncolytic virotherapy may represent a unique combination of viral tumor lysis and the activation of innate and adaptive immune responses. Targeted infection of tumor cells causes the release of TAAs and localized inflammation, which subsequently induces the tumor recognition of host immune system [113Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012; 30(7): 658-70.
[http://dx.doi.org/10.1038/nbt.2287] [PMID: 22781695]
]. Therefore, in this section, we will review the strategies for enhancing immune recognition of cancer specific neo-antigens and antitumor immune responses.

2.5.1. Arming OVs with TAAs

TAAs are the antigenic substances produced in tumor cells that trigger the immune responses towards the tumor. Certain tumor antigens, such as tyrosinase and melanoma–associated antigen (MAGE) in melanoma, are therefore used as the biomarkers for diagnostic test or the development of cancer therapies [114Hareuveni M, Gautier C, Kieny MP, Wreschner D, Chambon P, Lathe R. Vaccination against tumor cells expressing breast cancer epithelial tumor antigen. Proc Natl Acad Sci USA 1990; 87(23): 9498-502.
[http://dx.doi.org/10.1073/pnas.87.23.9498] [PMID: 2251291]
].

The combination of virus-mediated tumor destruction and enhanced immune recognition of TAAs has been studied and exhibited encouraging results. Use of VSV as the vector expressing TAAs from the cDNA library of melanoma successfully induced IL-17 recall response of tumor specific CD4+ T cells and eradicated melanoma tumors [115Pulido J, Kottke T, Thompson J, et al. Using virally expressed melanoma cDNA libraries to identify tumor-associated antigens that cure melanoma. Nat Biotechnol 2012; 30(4): 337-43.
[http://dx.doi.org/10.1038/nbt.2157] [PMID: 22426030]
]. Also, VSV expressing epitope library of human prostate cancer induced rejection of established prostate tumors [116Kottke T, Errington F, Pulido J, et al. Broad antigenic coverage induced by vaccination with virus-based cDNA libraries cures established tumors. Nat Med 2011; 17(7): 854-9.
[http://dx.doi.org/10.1038/nm.2390] [PMID: 21685898]
]. These results warrant further studies by using OVs expressing cDNA of multiple selective TAAs for activating a wide repertoire of APC and T cell responses.

2.5.2. Arming OVs with immuno-stimulatory or immune-modulatory genes

Tumors have been evolving to escape the antitumor immune response and develop the therapy-resistance. The infection and lysis of tumor cells by OVs triggers the localized release of inflammatory cytokines which subsequently attract the immune cells for tumor elimination [115Pulido J, Kottke T, Thompson J, et al. Using virally expressed melanoma cDNA libraries to identify tumor-associated antigens that cure melanoma. Nat Biotechnol 2012; 30(4): 337-43.
[http://dx.doi.org/10.1038/nbt.2157] [PMID: 22426030]
]. However, it is questionable if the cytokines are sufficient for eliciting tumor-reduction immune responses. Therefore, it would be desirable to have OVs expressing cytokines effective in inducing antitumor immune responses.

The two most advanced OVs, T-vec and JX-594 are respectively the modified HSV and VV, both expressing human GM-CSF which recruits dendritic cells and NK cells and induces tumor-specific cytotoxic T lymphocytes responses [117Deng L, Fan J, Guo M, Huang B. Oncolytic and immunologic cancer therapy with GM-CSF-armed vaccinia virus of Tian Tan strain Guang9. Cancer Lett 2016; 372(2): 251-7.
[http://dx.doi.org/10.1016/j.canlet.2016.01.025] [PMID: 26803055]
]. T-vec shows significant improvement of durable response rate and overall survival rate of patients with advance melanoma [15Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J Clin Oncol 2015; 33(25): 2780-8.
[http://dx.doi.org/10.1200/JCO.2014.58.3377] [PMID: 26014293]
].

The induction of anti-tumor T cell responses requires two signals: (1) the interaction between the major histocompatibility complex protein presenting tumor antigens and T cell receptors, and (2) the costimulatory molecules on APCs or tumor cells [118Todo T, Martuza RL, Dallman MJ, Rabkin SD. In situ expression of soluble B71 in the context of oncolytic herpes simplex virus induces potent antitumor immunity. Cancer Res 2001; 61(1): 153-61.
[PMID: 11196154]
]. The costimulatory signals come from the binding of B7 on activated APCs to CD28 or CTLA-4 on T cells and the binding of CD40 on APCs to CD40L on T cells. While interaction of B7 and CD28 promotes T cell activation and survival, the binding of B7 and CTLA-4 transmits inhibitory signal to the activated T cells. The binding of CD40L and CD40 triggers T lymphocyte expansion and increases IL-12 release which is critical for the engagement of CTL for the antitumor immune response [119Loskog AS, Fransson ME, Totterman TT. AdCD40L gene therapy counteracts T regulatory cells and cures aggressive tumors in an orthotopic bladder cancer model. Clin Cancer Res 2005; 11(24 Pt 1): 8816-21.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-1817] [PMID: 16361570]
, 120Mackey MF, Gunn JR, Maliszewsky C, Kikutani H, Noelle RJ, Barth RJ Jr. Dendritic cells require maturation via CD40 to generate protective antitumor immunity. J Immunol 1998; 161(5): 2094-8.
[PMID: 9725199]
]. Adenovirus, VSV and HSV engineered to express soluble B7 or CD40L exhibited more effective melanoma regression and significantly enhanced CD4+ and CD8+ T-cell infiltration [121Galivo F, Diaz RM, Thanarajasingam U, et al. Interference of CD40L-mediated tumor immunotherapy by oncolytic vesicular stomatitis virus. Hum Gene Ther 2010; 21(4): 439-50.
[http://dx.doi.org/10.1089/hum.2009.143] [PMID: 19922169]
-123Lee YS, Kim JH, Choi KJ, et al. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B71 in an immunocompetent murine model. Clin Cancer Res 2006; 12(19): 5859-68.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-0935] [PMID: 17020994]
]. Similarly, potent antitumor activity along with increased T cell activity have been observed with the administration of adenovirus and VSV coding a fully monoclonal antibody specific for CTLA-4 [124Gao Y, Whitaker-Dowling P, Griffin JA, Barmada MA, Bergman I. Recombinant vesicular stomatitis virus targeted to Her2/neu combined with anti-CTLA4 antibody eliminates implanted mammary tumors. Cancer Gene Ther 2009; 16(1): 44-52.
[http://dx.doi.org/10.1038/cgt.2008.55] [PMID: 18654610]
, 125Dias JD, Hemminki O, Diaconu I, et al. Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4. Gene Ther 2012; 19(10): 988-98.
[http://dx.doi.org/10.1038/gt.2011.176] [PMID: 22071969]
]. Binding of PD-L1 or PD-L2 on tumor cells to PD-1 on T cells inhibits antitumor T cell response. Combination of Anti-PD-1 blockade with virotherapy has been shown to improve the virus’ antitumor activity and result in improved therapeutic outcome [126Engeland CE, Grossardt C, Veinalde R, et al. CTLA-4 and PD-L1 checkpoint blockade enhances oncolytic measles virus therapy. Mol Ther 2014; 22(11): 1949-59.
[http://dx.doi.org/10.1038/mt.2014.160] [PMID: 25156126]
].

A variety of cytokines have been incorporated into OVs (Table 1) to activate various lymphocytes and attain enhanced antitumor immune response. For example, IL-2, the T cell growth factor, not only promotes T cell activation and proliferation, but also activates NK cells and increases NK cells’ lysing ability. IL-2 has been engineered into viruses such as NDV and HSV, intended to stimulate the activation of T and NK cells [127Vigil A, Park MS, Martinez O, et al. Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res 2007; 67(17): 8285-92.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-1025] [PMID: 17804743]
-129Carew JF, Kooby DA, Halterman MW, Kim SH, Federoff HJ, Fong Y. A novel approach to cancer therapy using an oncolytic herpes virus to package amplicons containing cytokine genes. Mol Ther 2001; 4(3): 250-6.
[http://dx.doi.org/10.1006/mthe.2001.0448] [PMID: 11545616]
]. Other cytokines engineered into OVs for promoting the antitumor activity through enhancing immune cell activation include IL-12, IL-15, IL-18, CCL-3 and IFN-γ etc [12Edukulla R, Woller N, Mundt B, et al. Antitumoral immune response by recruitment and expansion of dendritic cells in tumors infected with telomerase-dependent oncolytic viruses. Cancer Res 2009; 69(4): 1448-58.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-1160] [PMID: 19190348]
, 13Stephenson KB, Barra NG, Davies E, Ashkar AA, Lichty BD. Expressing human interleukin-15 from oncolytic vesicular stomatitis virus improves survival in a murine metastatic colon adenocarcinoma model through the enhancement of anti-tumor immunity. Cancer Gene Ther 2012; 19(4): 238-46.
[http://dx.doi.org/10.1038/cgt.2011.81] [PMID: 22158521]
, 123Lee YS, Kim JH, Choi KJ, et al. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B71 in an immunocompetent murine model. Clin Cancer Res 2006; 12(19): 5859-68.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-0935] [PMID: 17020994]
, 130Choi IK, Lee JS, Zhang SN, et al. Oncolytic adenovirus co-expressing IL-12 and IL-18 improves tumor-specific immunity via differentiation of T cells expressing IL-12Rβ2 or IL-18Rα. Gene Ther 2011; 18(9): 898-909.
[http://dx.doi.org/10.1038/gt.2011.37] [PMID: 21451575]
-134Su C, Peng L, Sham J, et al. Immune gene-viral therapy with triplex efficacy mediated by oncolytic adenovirus carrying an interferon-gamma gene yields efficient antitumor activity in immunodeficient and immunocompetent mice. Mol Ther 2006; 13(5): 918-27.
[http://dx.doi.org/10.1016/j.ymthe.2005.12.011] [PMID: 16497559]
]. The expression of these cytokines results in overall increase of therapeutic effect, despite the effect that some cytokines such as GM-CSF and IFN-γ cause reduced virus replication and early clearance.

Table 1
Melanoma virotherapy.


2.6. Combination of OVs and Other Therapies

In the past decade, there has been a rapid development of viruses with oncolytic activity as the clinical or preclinical anticancer therapeutics. More and more studies have shown that rather than using OVs as tumor therapy alone, it would be likely to realize OVs’ full therapeutic potential by combining with other treatment strategies such as radiotherapy, chemotherapy or immunotherapy.

2.6.1. Oncolysis with Radiotherapy

The combination of OVs and radiotherapy has resulted in synergistic therapeutic effect, which significantly inhibits the tumor growth relative to either of the individual therapies [135Dilley J, Reddy S, Ko D, et al. Oncolytic adenovirus CG7870 in combination with radiation demonstrates synergistic enhancements of antitumor efficacy without loss of specificity. Cancer Gene Ther 2005; 12(8): 715-22.
[http://dx.doi.org/10.1038/sj.cgt.7700835] [PMID: 15832172]
]. It has been reported that radiation exposure significantly increased the virus replication in a variety of cancer cell lines in vitro and vivo [136Adusumilli PS, Chan MK, Hezel M, et al. Radiation-induced cellular DNA damage repair response enhances viral gene therapy efficacy in the treatment of malignant pleural mesothelioma. Ann Surg Oncol 2007; 14(1): 258-69.
[http://dx.doi.org/10.1245/s10434-006-9127-4] [PMID: 17080237]
-140Kim SH, Wong RJ, Kooby DA, et al. Combination of mutated herpes simplex virus type 1 (G207 virus) with radiation for the treatment of squamous cell carcinoma of the head and neck. Eur J Cancer 2005; 41(2): 313-22.
[http://dx.doi.org/10.1016/j.ejca.2004.10.018] [PMID: 15661558]
].

The prospective synergistic efficacy by combining OVs and radiotherapy in melanoma is due to four aspects. First, OVs and radiation individually shows therapeutic efficacy in melanoma treatment. Second, radiation may reduce the high pressure of tumor cells which serves as an obstacle for virus replication and spread in vivo. Third, Ras mutation is one of the driver mutations for melanoma and is associated with radio-resistance [141Gupta AK, Cerniglia GJ, Mick R, et al. Radiation sensitization of human cancer cells in vivo by inhibiting the activity of PI3K using LY294002. Int J Radiat Oncol Biol Phys 2003; 56(3): 846-53.
[http://dx.doi.org/10.1016/S0360-3016(03)00214-1] [PMID: 12788194]
-143McKenna WG, Muschel RJ, Gupta AK, Hahn SM, Bernhard EJ. The RAS signal transduction pathway and its role in radiation sensitivity. Oncogene 2003; 22(37): 5866-75.
[http://dx.doi.org/10.1038/sj.onc.1206699] [PMID: 12947393]
]. However, reovirus, VSV and HSV have been demonstrated to preferentially target the Ras mutated melanoma cells and induce apoptosis [88Errington F, White CL, Twigger KR, et al. Inflammatory tumour cell killing by oncolytic reovirus for the treatment of melanoma. Gene Ther 2008; 15(18): 1257-70.
[http://dx.doi.org/10.1038/gt.2008.58] [PMID: 18401435]
, 89Noser JA, Mael AA, Sakuma R, et al. The RAS/Raf1/MEK/ERK signaling pathway facilitates VSV-mediated oncolysis: implication for the defective interferon response in cancer cells. Mol Ther 2007; 15(8): 1531-6.
[http://dx.doi.org/10.1038/sj.mt.6300193] [PMID: 17505473]
]. Therefore, OVs which kill the radiation-resistant melanoma cells will exert a complementary therapeutic effect to radiotherapy. Fourth, infection of the melanoma cells induces the release of localized inflammatory cytokines such as TNF-α and TRAIL, which have been shown to sensitize the cells to the effects of radiation [144Chinnaiyan AM, Prasad U, Shankar S, et al. Combined effect of tumor necrosis factor-related apoptosis-inducing ligand and ionizing radiation in breast cancer therapy. Proc Natl Acad Sci USA 2000; 97(4): 1754-9.
[http://dx.doi.org/10.1073/pnas.030545097] [PMID: 10677530]
-148Mundt AJ, Vijayakumar S, Nemunaitis J, et al. A Phase I trial of TNFerade biologic in patients with soft tissue sarcoma in the extremities. Clin Cancer Res 2004; 10(17): 5747-53.
[http://dx.doi.org/10.1158/1078-0432.CCR-04-0296] [PMID: 15355902]
]. The combination of reovirus and radiation has shown to increase the tumor growth delay of the melanoma xenografts in the treated animals, and significantly enhance the overall survival rate compared to the treatment with either of the individual therapies [149Twigger K, Vidal L, White CL, et al. Enhanced in vitro and in vivo cytotoxicity of combined reovirus and radiotherapy. Clin Cancer Res 2008; 14(3): 912-23.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-1400] [PMID: 18245555]
].

2.6.2. Oncolysis with chemotherapy

Chemotherapies can be generally divided into several categories: (1) alkylating agents that bind covalently to DNA via their alkyl group and exert antitumor effect, such as cyclophosphamide (CPA), decarbazine, cisplatin and carboplatin; (2) anti-metabolites that inhibit DNA and RNA synthesis, such as fluorouracil; (3) anti-microtubule agents that interfere with the microtubule function and inhibit cell division, such as paclitaxel and docetaxel; (4) topoisomerase inhibitors that prevent DNA replication and translation, such as novobiocin and aclarubicin; and (5) cytotoxic antibiotics that interrupt cell division, such as mitomycin C and doxorubicin [150Ottolino-Perry K, Diallo JS, Lichty BD, Bell JC, McCart JA. Intelligent design: combination therapy with oncolytic viruses. Mol Ther 2010; 18(2): 251-63.
[http://dx.doi.org/10.1038/mt.2009.283] [PMID: 20029399]
]. With the consideration that chemotherapy and OVs work with different mechanisms via different pathways, it is probable that the combination of these two therapies would exert synergistic therapeutic effectiveness.

The preclinical study of combinational therapy on malignant melanomas has demonstrated the difference between a variety of chemotherapies in enhancing the viral oncolysis. Pandha et al. [151Pandha HS, Heinemann L, Simpson GR, et al. Synergistic effects of oncolytic reovirus and cisplatin chemotherapy in murine malignant melanoma. Clin Cancer Res 2009; 15(19): 6158-66.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-0796] [PMID: 19773377]
] have reported that reovirus, in combination with cisplatin or paclitaxel, resulted in more pronounced tumor-killing effect in malignant melanoma than with other chemotherapeutic agents such as dacarbazine, gemcitabine and carboplatin, despite the fact that synergistic effect has been seen when reovirus was combined with all these chemotherapeutic agents. The combinational therapy of reovirus and cisplatin resulted in more remarkable retardation of tumor growth, in comparison to using cisplatin or reovirus alone. In addition, the intravenous administrations of combinational therapy of reovirus with paclitaxel, carboplatin, dacarbazine and gemcitabine are undergoing phase I or phase II trials [152Galanis E, Markovic SN, Suman VJ, et al. Phase II trial of intravenous administration of Reolysin(®) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma. Mol Ther 2012; 20(10): 1998-2003.
[http://dx.doi.org/10.1038/mt.2012.146] [PMID: 22871663]
].

It is critical to understand the interplay between the virus and the chemotherapies of choice which likely modify the humoral and adaptive immune responses. For example, CPA has been reported to ablate the neutralizing antibodies against the virus intratumorally administered, and it is necessary to maintain sufficient levels of neutralizing antibodies to prevent the virus to cause toxicities in systemic organs [153Qiao J, Wang H, Kottke T, et al. Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus. Clin Cancer Res 2008; 14(1): 259-69.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-1510] [PMID: 18172278]
]. Therefore, it is critical to analyze the toxicity, carefully make chemotherapy dose decisions and supervise the real-time levels of neutralizing antibodies against virus.

2.6.3. Oncolysis with Immunotherapy

The current immunotherapy for melanoma include cytokine agents (IL-2 and IFN-α) and antibodies against CTLA-4 and PD-1. Targeting CTLA-4 and PD-1/PD-L1 pathways via using anti-CTLA-4 and anti-PD-1/anti-PD-L1 has achieved more efficacies in regressing melanoma, as compared to other cancers such as renal cell carcinoma [154Quezada SA, Peggs KS. Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer. Br J Cancer 2013; 108(8): 1560-5.
[http://dx.doi.org/10.1038/bjc.2013.117] [PMID: 23511566]
]. Combining two therapeutic agents with different mechanisms of action is highly desirable, due to the potentially enhanced efficacy than that of single drug alone. While in some instances the combinations lead to promising effectiveness, some do not. For example, a phase II trial in metastatic melanoma patients has shown that compared with ipilimumab (CTLA-4 blockade) monotherapy, the combination of ipilimumab and GM-CSF resulted in enhanced overall survival rate and tumor reduction with less toxicity [155Lee SJ, McDermott DF, Rao UN, et al. Multicenter, randomized phase II trial of GM-CSF (GM) plus ipilimumab (Ipi) versus Ipi alone in metastatic melanoma: E1608. J Clin Oncol 2013; 31(18_suppl): CRA9007.
[PMID: 28136075]
]. However, a phase I study of cotreatment of ipilimumab and vemurafenib demonstrated remarkable increase of the liver toxicity than that of ipilimumab single treatment, which ultimately caused the early termination of the clinical trial of this combination [156Ribas A, Hodi FS, Callahan M, Konto C, Wolchok J. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med 2013; 368(14): 1365-6.
[http://dx.doi.org/10.1056/NEJMc1302338] [PMID: 23550685]
].

In regard of these findings, cotreatment of immunotherapy and OVs provides another appealing option for enhancing the therapeutic effect for melanoma treatment. Rajani et al. [157Rajani K, Parrish C, Kottke T, 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.
[http://dx.doi.org/10.1038/mt.2015.156] [PMID: 26310630]
] demonstrated that intravenous injection of anti-PD-1 and intertumoral injection of reovirus enhanced the survival of mice with melanoma xenografts compared to either therapy alone. Addition of anti-PD-1 not only reduced the T-reg activity and increased the cytotoxic T cell-mediated tumor reduction, but also significantly enhanced the NK cell activation mediated by the reovirus-infected tumor cells [157Rajani K, Parrish C, Kottke T, 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.
[http://dx.doi.org/10.1038/mt.2015.156] [PMID: 26310630]
]. Similarly, it has been shown that compared with combination with anti-TGF or anti-IL-10R, combination with anti-CTLA-4 strikingly improves the therapeutic effect of VSV-mediated tumor regression by eliciting CD4+ and CD8+ T cells response [124Gao Y, Whitaker-Dowling P, Griffin JA, Barmada MA, Bergman I. Recombinant vesicular stomatitis virus targeted to Her2/neu combined with anti-CTLA4 antibody eliminates implanted mammary tumors. Cancer Gene Ther 2009; 16(1): 44-52.
[http://dx.doi.org/10.1038/cgt.2008.55] [PMID: 18654610]
]. IL-2 has been known as the T cells growth factor and is able to enhance the proliferation of T cells and activate NK cells and macrophages. It has been shown that OVs expressing IL-2 successfully increased the regulatory CD4+ and cytotoxic CD8+ T cells proliferation and memory T cells generation, resulting in effective reduction of tumors [128Zhao H, Janke M, Fournier P, Schirrmacher V. Recombinant Newcastle disease virus expressing human interleukin-2 serves as a potential candidate for tumor therapy. Virus Res 2008; 136(1-2): 75-80.
[http://dx.doi.org/10.1016/j.virusres.2008.04.020] [PMID: 18538434]
, 158Bai F, Niu Z, Tian H, et al. Genetically engineered Newcastle disease virus expressing interleukin 2 is a potential drug candidate for cancer immunotherapy. Immunol Lett 2014; 159(1-2): 36-46.
[http://dx.doi.org/10.1016/j.imlet.2014.02.009] [PMID: 24613899]
]. In addition, Kottke et al. [159Kottke T, Galivo F, Wongthida P, et al. Treg depletion-enhanced IL-2 treatment facilitates therapy of established tumors using systemically delivered oncolytic virus. Mol Ther 2008; 16(7): 1217-26.
[http://dx.doi.org/10.1038/mt.2008.83] [PMID: 18431359]
] have shown that systemic IL-2 causes the endothelial cell injury (vascular leak syndrome), which facilitates the efflux of circulating VSV to locate into melanoma tumors and increases the number of NK cells.

FUTURE DIRECTIONS

A major part of the current standard therapeutic strategy for melanoma includes a range of targeted treatment entities based on specific gene mutations (BRAF inhibitors), pathways or enzymes (MEK inhibitors, tyrosine kinase inhibitors) and growth factors (VEGF inhibitors) [6Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
, 19Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380(9839): 358-65.
[http://dx.doi.org/10.1016/S0140-6736(12)60868-X] [PMID: 22735384]
, 53Gonzalez D, Fearfield L, Nathan P, et al. BRAF mutation testing algorithm for vemurafenib treatment in melanoma: recommendations from an expert panel. Br J Dermatol 2013; 168(4): 700-7.
[http://dx.doi.org/10.1111/bjd.12248] [PMID: 23360189]
-60Tarhini AA, Frankel P, Margolin KA, et al. Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin Cancer Res 2011; 17(20): 6574-81.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-1463] [PMID: 21880788]
]. Melanoma associated biomarkers like LDH, MIA, VEGF and MMP have shown a notable prognostic value where the level of these biomarkers has closely correlated with the advancement of the disease [33Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased serum concentration of angiogenic factors in malignant melanoma patients correlates with tumor progression and survival. J Clin Oncol 2001; 19(2): 577-83.
[PMID: 11208853]
, 34Díaz-Lagares A, Alegre E, Arroyo A, et al. Evaluation of multiple serum markers in advanced melanoma. Tumour Biol 2011; 32(6): 1155-61.
[http://dx.doi.org/10.1007/s13277-011-0218-x] [PMID: 21858537]
, 38Nikkola J, Vihinen P, Vlaykova T, Hahka-Kemppinen M, Kähäri VM, Pyrhönen S. High expression levels of collagenase-1 and stromelysin-1 correlate with shorter disease-free survival in human metastatic melanoma. Int J Cancer 2002; 97(4): 432-8.
[http://dx.doi.org/10.1002/ijc.1636] [PMID: 11802203]
]. In the context of FDA approved therapies targeting melanoma biomarkers such as BRAF, MEK and VEGF, it is safe to say that biomarkers can not only serve as prognostic tools but are also useful in developing new therapies targeting melanoma. Targeted therapies fail to provide expected outcomes in many patients due to the genetic variability of melanoma, as melanoma tumors can be genetically heterogeneous and may or may not harbor mutations in target genes of therapeutic interest. Efficacy of treatment strategy for melanoma increases significantly when multiple targeted therapies are used in combination versus use of a single therapeutic agent, indicating the high level of dependence of targeted melanoma therapies on the genetic profile of the cancer. While nearly 50% of melanoma patients usually respond to BRAF inhibitors, the number of non-responding patients decreased by 40% when BRAF inhibitors were used along with MEK inhibitors as a combination therapy [6Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
, 19Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380(9839): 358-65.
[http://dx.doi.org/10.1016/S0140-6736(12)60868-X] [PMID: 22735384]
, 55Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med 2012; 366(8): 707-14.
[http://dx.doi.org/10.1056/NEJMoa1112302] [PMID: 22356324]
, 160Larkin J, Del Vecchio M, Ascierto PA, et al. Vemurafenib in patients with BRAF(V600) mutated metastatic melanoma: an open-label, multicentre, safety study. Lancet Oncol 2014; 15(4): 436-44.
[http://dx.doi.org/10.1016/S1470-2045(14)70051-8] [PMID: 24582505]
]. In regard of this, immunotherapy (IFN-α and IL-2) and oncolytic virotherapy that are not heavily dependent on genetic heterogeneity of melanoma would circumvent limitations associated with targeted therapies that target specific gene mutations.

The OVs’ ability of causing tumor lysis, stimulating local inflammatory responses and serving as gene delivery vehicles, supports them as promising therapeutic agents for melanoma. While OVs are able to induce cell lysis and local inflammation that are not relying on genetic heterogeneity of melanoma, some viruses, in native forms, preferentially infect melanoma cells with aberrant genetic properties. Viruses such as reovirus, NDV and VSV have been shown to effectively kill the Ras-mutated tumor cells or lead to apoptosis, which make them as natural OVs for melanoma [88Errington F, White CL, Twigger KR, et al. Inflammatory tumour cell killing by oncolytic reovirus for the treatment of melanoma. Gene Ther 2008; 15(18): 1257-70.
[http://dx.doi.org/10.1038/gt.2008.58] [PMID: 18401435]
, 89Noser JA, Mael AA, Sakuma R, et al. The RAS/Raf1/MEK/ERK signaling pathway facilitates VSV-mediated oncolysis: implication for the defective interferon response in cancer cells. Mol Ther 2007; 15(8): 1531-6.
[http://dx.doi.org/10.1038/sj.mt.6300193] [PMID: 17505473]
, 96Ravindra PV, Tiwari AK, Sharma B, Chauhan RS. Newcastle disease virus as an oncolytic agent. Indian J Med Res 2009; 130(5): 507-13.
[PMID: 20090097]
]. In addition, for those viruses without natural oncoselectivity, modifications targeting them to the genetic abnormalities of melanoma, often represented as biomarkers, have shown significant effectiveness in enhancing their oncolytic activity. For example, promoters of melanoma biomarkers such as Cox-2, CXCR-4, tyrosinase and survivin have been incorporated into the viral genomes, which result in significantly enhanced oncospecificity [10Banerjee NS, Rivera AA, Wang M, et al. Analyses of melanoma-targeted oncolytic adenoviruses with tyrosinase enhancer/promoter-driven E1A, E4, or both in submerged cells and organotypic cultures. Mol Cancer Ther 2004; 3(4): 437-49.
[PMID: 15078987]
, 78Wu L, Johnson M, Sato M. Transcriptionally targeted gene therapy to detect and treat cancer. Trends Mol Med 2003; 9(10): 421-9.
[http://dx.doi.org/10.1016/j.molmed.2003.08.005] [PMID: 14557054]
-80Zhu ZB, Makhija SK, Lu B, et al. Transcriptional targeting of adenoviral vector through the CXCR4 tumor-specific promoter. Gene Ther 2004; 11(7): 645-8.
[http://dx.doi.org/10.1038/sj.gt.3302089] [PMID: 15029227]
]. A single chain antibody recognizing the HMWMAA, which is another melanoma biomarker, has been engineered into the virus for transductionally targeting melanoma cells [77Purow B, Staveley-OCarroll K. Targeting of vaccinia virus using biotin-avidin viral coating and biotinylated antibodies. J Surg Res 2005; 123(1): 49-54.
[http://dx.doi.org/10.1016/j.jss.2004.04.022] [PMID: 15652950]
]. Beyond the genetic and epigenetic alterations within the melanoma cells, the interaction between the melanoma cells and the microenvironment critically determines the transformation process of melanoma. Microenvironmental factors such as growth factors (VEGF, FGF, PDGF and TGF) and proteolytic enzymes (MMPs) involve in angiogenesis and tissue remodeling, highly promoting melanoma progression and metastasis. Modifications of OVs to target these microenvironmental factors have resulted in enhanced viral penetration and spread and melanoma regression [103Frentzen A, Yu YA, Chen N, et al. Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy. Proc Natl Acad Sci USA 2009; 106(31): 12915-20.
[http://dx.doi.org/10.1073/pnas.0900660106] [PMID: 19617539]
, 110Cheng J, Sauthoff H, Huang Y, et al. Human matrix metalloproteinase-8 gene delivery increases the oncolytic activity of a replicating adenovirus. Mol Ther 2007; 15(11): 1982-90.
[http://dx.doi.org/10.1038/sj.mt.6300264] [PMID: 17653103]
, 111Choi JW, Lee YS, Yun CO, Kim SW. Polymeric oncolytic adenovirus for cancer gene therapy. J Control Release 2015; 219: 181-91.
[http://dx.doi.org/10.1016/j.jconrel.2015.10.009] [PMID: 26453806]
].

OVs represent a class of novel agents as they can be engineered to harness the benefits of targeted therapy and immunotherapy, by inducing cancer cell death along with induction of a strong innate and adaptive immune response against cancer cells. The greatest success has been achieved with HSV (T-vec) and VV (JX-594) when developing OVs for cancer treatment. Some other viruses that are being studied in pre-clinical and clinical trials include HF10 (HSV), reovirus, NDV, VSV and coxsackie virus [115Pulido J, Kottke T, Thompson J, et al. Using virally expressed melanoma cDNA libraries to identify tumor-associated antigens that cure melanoma. Nat Biotechnol 2012; 30(4): 337-43.
[http://dx.doi.org/10.1038/nbt.2157] [PMID: 22426030]
, 127Vigil A, Park MS, Martinez O, et al. Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res 2007; 67(17): 8285-92.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-1025] [PMID: 17804743]
, 152Galanis E, Markovic SN, Suman VJ, et al. Phase II trial of intravenous administration of Reolysin(®) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma. Mol Ther 2012; 20(10): 1998-2003.
[http://dx.doi.org/10.1038/mt.2012.146] [PMID: 22871663]
, 161Watanabe D, Goshima F, Mori I, Tamada Y, Matsumoto Y, Nishiyama Y. Oncolytic virotherapy for malignant melanoma with herpes simplex virus type 1 mutant HF10. J Dermatol Sci 2008; 50(3): 185-96.
[http://dx.doi.org/10.1016/j.jdermsci.2007.12.001] [PMID: 18226503]
, 162Shafren D, Quah M, Wong Y, Andtbacka RH, Au G. Combination of a novel oncolytic immunotherapeutic agent, coxsackievirus a21 and pd-1 blockade significantly reduces tumor growth and improves survival in an immune competent mouse melanoma model. Ann Oncol 2014; •••: 25. [PMID: WOS:000346901000307].]. TAAs and immunostimulatory cytokines have been engineered into OVs, intended to attain enhanced immune recognition of tumor antigens and antitumor immune responses [115Pulido J, Kottke T, Thompson J, et al. Using virally expressed melanoma cDNA libraries to identify tumor-associated antigens that cure melanoma. Nat Biotechnol 2012; 30(4): 337-43.
[http://dx.doi.org/10.1038/nbt.2157] [PMID: 22426030]
]. Using the viruses as the platforms for expressing immunostimulatory cytokines such as GM-CSF is appealing as it is likely to increase the local concentration of the gene products and minimize the side effects due to the systemic exposure [163Simmons AD, Moskalenko M, Creson J, et al. Local secretion of anti-CTLA-4 enhances the therapeutic efficacy of a cancer immunotherapy with reduced evidence of systemic autoimmunity. Cancer Immunol Immunother 2008; 57(8): 1263-70.
[http://dx.doi.org/10.1007/s00262-008-0451-3] [PMID: 18236040]
]. While in some instances the expression of cytokines showed no inhibitive effect in virus replication, in some other cases the incorporation of cytokines such as IL-2, IFN and GM-CSF resulted in significantly decreased virus titer [126Engeland CE, Grossardt C, Veinalde R, et al. CTLA-4 and PD-L1 checkpoint blockade enhances oncolytic measles virus therapy. Mol Ther 2014; 22(11): 1949-59.
[http://dx.doi.org/10.1038/mt.2014.160] [PMID: 25156126]
, 128Zhao H, Janke M, Fournier P, Schirrmacher V. Recombinant Newcastle disease virus expressing human interleukin-2 serves as a potential candidate for tumor therapy. Virus Res 2008; 136(1-2): 75-80.
[http://dx.doi.org/10.1016/j.virusres.2008.04.020] [PMID: 18538434]
, 164Kirn DH, Wang Y, Le Boeuf F, Bell J, Thorne SH. Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus. PLoS Med 2007; 4(12): e353.
[http://dx.doi.org/10.1371/journal.pmed.0040353] [PMID: 18162040]
-166Lei N, Shen FB, Chang JH, et al. An oncolytic adenovirus expressing granulocyte macrophage colony-stimulating factor shows improved specificity and efficacy for treating human solid tumors. Cancer Gene Ther 2009; 16(1): 33-43.
[http://dx.doi.org/10.1038/cgt.2008.46] [PMID: 18670453]
]. To cope with this issue, strategies have been designed to fuse the cytokine transgene to an exogenous cytokine-expressing regulator, which regulates the expression of the cytokine at specific time [167Chen H, Sampath P, Hou W, Thorne SH. Regulating cytokine function enhances safety and activity of genetic cancer therapies. Mol Ther 2013; 21(1): 167-74.
[http://dx.doi.org/10.1038/mt.2012.225] [PMID: 23281444]
].

While some OVs are being studied for their efficacy as monotherapy, some are under investigation in combination with other therapies such as chemotherapy, radiotherapy, targeted therapy and more recently adoptive T cell therapy. T-vec is under investigation in several clinical studies where its efficacy is being assessed when combined with ipilimumab (CTLA4 inhibitor), and the results of a phase Ib/2 study of T-vec+ipilimumab suggest the enhanced synergistic efficacy with higher complete response and objective response rate [168Puzanov I, Milhem MM, Andtbacka RH, Minor DR, Hamid O, Li A, et al. Primary analysis of a phase 1b multicenter trial to evaluate safety and efficacy of talimogene laherparepvec (T-VEC) and ipilimumab (ipi) in previously untreated, unresected stage IIIB-IV melanoma. J Clin Oncol 2014; 32(15) [PMID: WOS:000358613204315].]. Similar synergistic effects have also been observed when OVs were combined with anti-PD-1 and IL-2 [157Rajani K, Parrish C, Kottke T, 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.
[http://dx.doi.org/10.1038/mt.2015.156] [PMID: 26310630]
]. Adoptive T cell therapy, with priming of T cells against tumor antigens followed by transfusion of the primed T cells in cancer patients, has shown consistent efficacy in clinical trials for melanoma [169Weber JS. At the bedside: adoptive cell therapy for melanoma-clinical development. J Leukoc Biol 2014; 95(6): 875-82.
[http://dx.doi.org/10.1189/jlb.0513293] [PMID: 24732024]
]. Adoptive T cell therapy is also being investigated as combination therapy along with OVs. It has been shown that oncolytic adenovirus expressing IFN-γ combined with adoptive T cell therapy resulted in overall objective response of 38.5% in melanoma patients [170Khammari A, Nguyen JM, Saint-Jean M, et al. Adoptive T cell therapy combined with intralesional administrations of TG1042 (adenovirus expressing interferon-γ) in metastatic melanoma patients. Cancer Immunol Immunother 2015; 64(7): 805-15.
[http://dx.doi.org/10.1007/s00262-015-1691-7] [PMID: 25846669]
].

Although combination therapies have shown significant improvement and encouraging results, careful attention is still paid to the clinical trial designs, timing and dosing regimens, biosafety and biodistribution. For example, when combining the OVs and immunotherapy such as anti-PD-1, the timing of immunotherapy administration would be critical to avoid augmenting the anti-viral immune response or any earlier clearance of the virus. Rajani et al. [157Rajani K, Parrish C, Kottke T, 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.
[http://dx.doi.org/10.1038/mt.2015.156] [PMID: 26310630]
] have demonstrated that administration of anti-PD-1 7 days after the intratumoral injection of virus is optimal to maximize the virus spread within the tumors. When combining OVs with chemotherapies such as CPA, CPA is able to eliminate the neutralizing antibodies against the virus and allow the virus to cause toxicities in systemic organs [153Qiao J, Wang H, Kottke T, et al. Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus. Clin Cancer Res 2008; 14(1): 259-69.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-1510] [PMID: 18172278]
]. Therefore, it is essential to critically supervise the toxicity and the real-time levels of neutralizing antibodies against the virus, and carefully make dose-escalation decisions. For the combinational therapy of OVs and immunotherapy, it has been proposed to additionally combine with the systemic treatment such as T-reg depletion, to exert even higher synergistic efficacy [125Dias JD, Hemminki O, Diaconu I, et al. Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4. Gene Ther 2012; 19(10): 988-98.
[http://dx.doi.org/10.1038/gt.2011.176] [PMID: 22071969]
]. However, one should be cautious to avoid autoimmune reactions when combining T cell enhancement with T-reg depletion, as immunotherapy such as anti-CTLA-4 treatment is already closely related to decreased number of myeloid derived suppressor cells [171Martens A, Wistuba-Hamprecht K, Geukes Foppen M, et al. Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22(12): 2908-18.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-2412] [PMID: 26787752]
].

Although it is reasonable to expect sufficient efficacy exerted by a one-shot curative agent, the immune “barriers” such as virus neutralization and sequestration make it more realistic to attain the optimal therapeutic effect with serial OVs for priming the antitumor immune responses. Strategies have been developed to prime the antitumor responses with one viral vector and boost with another, in order to minimize the anti-viral immune responses [172Irvine KR, Chamberlain RS, Shulman EP, Surman DR, Rosenberg SA, Restifo NP. Enhancing efficacy of recombinant anticancer vaccines with prime/boost regimens that use two different vectors. J Natl Cancer Inst 1997; 89(21): 1595-601.
[http://dx.doi.org/10.1093/jnci/89.21.1595] [PMID: 9362157]
, 173Marshall JL, Hoyer RJ, Toomey MA, et al. Phase I study in advanced cancer patients of a diversified prime-and-boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avipox virus to elicit anti-carcinoembryonic antigen immune responses. J Clin Oncol 2000; 18(23): 3964-73.
[PMID: 11099326]
].

CONCLUSION

In summary, this review describes OVs and their mechanisms in melanoma tumor regression. We also discuss different combinational therapies, which overcome the limitations associated with each therapeutic agent such as chemo-, radio- and targeted- therapies. Nonetheless, the quest for identifying new melanoma biomarkers that can channelize efforts for developing newer targeted therapies for melanoma should be continued.

ABBREVIATIONS

AJCC  = American Joint Committee on Cancer
APC  = antigen presenting cell
CAM  = cell adhesion molecule
CAR  = coxsackie virus and adenovirus receptor
CD  = cluster of differentiation
Cox  = cyclooxygenase
CPA  = cyclophosphamide
CTLA  = cytotoxic T-lymphocyte antigen
CXCR-4  = C-X-C chemokine receptor type 4
ECM  = extracellular matrix
FGF  = fibroblast growth factor
GM-CSF  = granulocyte monocyte colony-stimulating factor
HMWMAA  = high molecular weight melanoma-associated antigen
HSV  = herpes simplex virus
IFN  = interferon
IL  = interleukin
LDH  = lactate dehydrogenase
MAA/MAGE  = melanoma-associated antigen
MIA  = melanoma-inhibitory activity
MLV  = murine leukemia virus
MMP  = matrix metalloproteinase
NK  = natural killer
NDV  = Newcastle disease virus
OV  = oncolytic virus
PD  = programmed death receptor
PDGF  = platelet-derived growth factor
PK  = protein kinase
ROF  = replication origin firing
T-vec  = talimogene laherparepvec
TAA  = tumor associated antigen
TGF  = transforming growth factor
TNF  = tumor necrosis factor
TRAIL  = tumor necrosis factor-related apoptosis-inducing ligand
VEGF  = vascular endothelial growth factor
VSV  = vesicular stomatitis virus
VV  = vaccinia virus

CONFLICT OF INTEREST

The authors confirm that this article content has no conflict of interest.

ACKNOWLEDGMENTS

We are grateful to Drs. Bruce Bejcek and Rob Eversole for editorial comments.

REFERENCES

[1] Tsao H, Atkins MB, Sober AJ. Management of cutaneous melanoma. N Engl J Med 2004; 351(10): 998-1012.
[http://dx.doi.org/10.1056/NEJMra041245] [PMID: 15342808]
[2] Kim BS, Seo H, Kim HJ, et al. Decursin from Angelica gigas nakai Inhibits B16F10 melanoma growth through induction of apoptosis. J Med Food 2015; 18(10): 1121-7.
[http://dx.doi.org/10.1089/jmf.2014.3397] [PMID: 26336081]
[3] Johnson TM, Yahanda AM, Chang AE, Fader DJ, Sondak VK. Advances in melanoma therapy. J Am Acad Dermatol 1998; 38(5 Pt 1): 731-41.
[http://dx.doi.org/10.1016/S0190-9622(98)70202-6] [PMID: 9591818]
[4] Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res 2015; 3(3): 288-95.
[http://dx.doi.org/10.1158/2326-6066.CIR-14-0207] [PMID: 25736262]
[5] Karagiannis P, Fittall M, Karagiannis SN. Evaluating biomarkers in melanoma. Front Oncol 2015; 4: 383.
[PMID: 25667918]
[6] Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364(26): 2507-16.
[http://dx.doi.org/10.1056/NEJMoa1103782] [PMID: 21639808]
[7] Viale DL, Cafferata EG, Gould D, et al. Therapeutic improvement of a stroma-targeted CRAd by incorporating motives responsive to the melanoma microenvironment. J Invest Dermatol 2013; 133(11): 2576-84.
[http://dx.doi.org/10.1038/jid.2013.191] [PMID: 23604101]
[8] Chan WM, Rahman MM, McFadden G. Oncolytic myxoma virus: the path to clinic. Vaccine 2013; 31(39): 4252-8.
[http://dx.doi.org/10.1016/j.vaccine.2013.05.056] [PMID: 23726825]
[9] Steele L, Errington F, Prestwich R, et al. Pro-inflammatory cytokine/chemokine production by reovirus treated melanoma cells is PKR/NF-κB mediated and supports innate and adaptive anti-tumour immune priming. Mol Cancer 2011; 10: 20.
[http://dx.doi.org/10.1186/1476-4598-10-20] [PMID: 21338484]
[10] Banerjee NS, Rivera AA, Wang M, et al. Analyses of melanoma-targeted oncolytic adenoviruses with tyrosinase enhancer/promoter-driven E1A, E4, or both in submerged cells and organotypic cultures. Mol Cancer Ther 2004; 3(4): 437-49.
[PMID: 15078987]
[11] Tamm I, Wang Y, Sausville E, et al. IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res 1998; 58(23): 5315-20.
[PMID: 9850056]
[12] Edukulla R, Woller N, Mundt B, et al. Antitumoral immune response by recruitment and expansion of dendritic cells in tumors infected with telomerase-dependent oncolytic viruses. Cancer Res 2009; 69(4): 1448-58.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-1160] [PMID: 19190348]
[13] Stephenson KB, Barra NG, Davies E, Ashkar AA, Lichty BD. Expressing human interleukin-15 from oncolytic vesicular stomatitis virus improves survival in a murine metastatic colon adenocarcinoma model through the enhancement of anti-tumor immunity. Cancer Gene Ther 2012; 19(4): 238-46.
[http://dx.doi.org/10.1038/cgt.2011.81] [PMID: 22158521]
[14] Lichty BD, Breitbach CJ, Stojdl DF, Bell JC. Going viral with cancer immunotherapy. Nat Rev Cancer 2014; 14(8): 559-67.
[http://dx.doi.org/10.1038/nrc3770] [PMID: 24990523]
[15] Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. J Clin Oncol 2015; 33(25): 2780-8.
[http://dx.doi.org/10.1200/JCO.2014.58.3377] [PMID: 26014293]
[16] Collichio FA, Amatruda T, Senzer NN, et al. OPTiM: A randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma. J Clin Oncol 2013; 31(18_suppl): LBA9008.
[PMID: 28136063]
[17] Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 2009; 27(36): 6199-206.
[http://dx.doi.org/10.1200/JCO.2009.23.4799] [PMID: 19917835]
[18] Griewank KG. Biomarkers in melanoma. Scand J Clin Lab Invest Suppl 2016; 245: S104-12.
[http://dx.doi.org/10.1080/00365513.2016.1210336] [PMID: 27467728]
[19] Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380(9839): 358-65.
[http://dx.doi.org/10.1016/S0140-6736(12)60868-X] [PMID: 22735384]
[20] Breslow A. Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Ann Surg 1970; 172(5): 902-8.
[http://dx.doi.org/10.1097/00000658-197011000-00017] [PMID: 5477666]
[21] Gogas H, Eggermont AM, Hauschild A, et al. Biomarkers in melanoma. Ann Oncol 2009; 20(Suppl. 6): vi8-vi13.
[http://dx.doi.org/10.1093/annonc/mdp251] [PMID: 19617299]
[22] Eggermont AM, Suciu S, Testori A, Patel P, Spatz A. Ulceration of primary melanoma and responsiveness to adjuvant interferon therapy: analysis of the adjuvant trials EORTC18952 and EORTC18991 in 2,644 patients. J Clin Oncol 2009; 27(15S): 9007-7. PubMed PMID: WOS:000276606606023.
[23] Eggermont AM, Suciu S, Testori A, et al. Long-term results of the randomized phase III trial EORTC 18991 of adjuvant therapy with pegylated interferon alfa-2b versus observation in resected stage III melanoma. J Clin Oncol 2012; 30(31): 3810-8.
[http://dx.doi.org/10.1200/JCO.2011.41.3799] [PMID: 23008300]
[24] Winnepenninckx V, van den Oord JJ. Gene expression profiling and clinical outcome in melanoma: in search of novel prognostic factors. Expert Rev Anticancer Ther 2007; 7(11): 1611-31.
[http://dx.doi.org/10.1586/14737140.7.11.1611] [PMID: 18020928]
[25] Winnepenninckx V, Lazar V, Michiels S, et al. Gene expression profiling of primary cutaneous melanoma and clinical outcome. J Natl Cancer Inst 2006; 98(7): 472-82.
[http://dx.doi.org/10.1093/jnci/djj103] [PMID: 16595783]
[26] Solassol J, Du-Thanh A, Maudelonde T, Guillot B. Serum proteomic profiling reveals potential biomarkers for cutaneous malignant melanoma. Int J Biol Markers 2011; 26(2): 82-7.
[http://dx.doi.org/10.5301/JBM.2011.8344] [PMID: 21607923]
[27] Hofmann MA, Gussmann F, Fritsche A, et al. Diagnostic value of melanoma inhibitory activity serum marker in the follow-up of patients with stage I or II cutaneous melanoma. Melanoma Res 2009; 19(1): 17-23.
[http://dx.doi.org/10.1097/CMR.0b013e32831bc78c] [PMID: 19104451]
[28] Hofmann MA, Schicke B, Fritsch A, et al. Impact of lymph node metastases on serum level of melanoma inhibitory activity in stage III melanoma patients. J Dermatol 2011; 38(9): 880-6.
[PMID: 21658116]
[29] Davey GE, Murmann P, Hoechli M, Tanaka T, Heizmann CW. Calcium-dependent translocation of S100A11 requires tubulin filaments. Biochim Biophys Acta 2000; 1498(2-3): 220-32.
[http://dx.doi.org/10.1016/S0167-4889(00)00098-7] [PMID: 11108965]
[30] Hsieh HL, Schäfer BW, Cox JA, Heizmann CW. S100A13 and S100A6 exhibit distinct translocation pathways in endothelial cells. J Cell Sci 2002; 115(Pt 15): 3149-58.
[PMID: 12118070]
[31] Oberholzer PA, Urosevic M, Steinert HC, Dummer R. Baseline staging of melanoma with unknown primary site: the value of serum s100 protein and positron emission tomography. Dermatology (Basel) 2008; 217(4): 351-5.
[http://dx.doi.org/10.1159/000155878] [PMID: 18799883]
[32] Beyeler M, Waldispuhl S, Strobel K, Joller-Jemelka HI, Burg G, Dummer R. Detection of melanoma relapse: first comparative analysis on imaging techniques versus S100 protein. Dermatology (Basel) 2006; 213(3): 187-91.
[http://dx.doi.org/10.1159/000095034] [PMID: 17033166]
[33] Ugurel S, Rappl G, Tilgen W, Reinhold U. Increased serum concentration of angiogenic factors in malignant melanoma patients correlates with tumor progression and survival. J Clin Oncol 2001; 19(2): 577-83.
[PMID: 11208853]
[34] Díaz-Lagares A, Alegre E, Arroyo A, et al. Evaluation of multiple serum markers in advanced melanoma. Tumour Biol 2011; 32(6): 1155-61.
[http://dx.doi.org/10.1007/s13277-011-0218-x] [PMID: 21858537]
[35] Visús C, Andres R, Mayordomo JI, et al. Prognostic role of circulating melanoma cells detected by reverse transcriptase-polymerase chain reaction for tyrosinase mRNA in patients with melanoma. Melanoma Res 2007; 17(2): 83-9.
[http://dx.doi.org/10.1097/CMR.0b013e3280a60878] [PMID: 17496783]
[36] McAllister SS, Gifford AM, Greiner AL, et al. Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell 2008; 133(6): 994-1005.
[http://dx.doi.org/10.1016/j.cell.2008.04.045] [PMID: 18555776]
[37] Sanmamed MF, Carranza-Rua O, Alfaro C, et al. Serum interleukin-8 reflects tumor burden and treatment response across malignancies of multiple tissue origins. Clin Cancer Res 2014; 20(22): 5697-707.
[http://dx.doi.org/10.1158/1078-0432.CCR-13-3203] [PMID: 25224278]
[38] Nikkola J, Vihinen P, Vlaykova T, Hahka-Kemppinen M, Kähäri VM, Pyrhönen S. High expression levels of collagenase-1 and stromelysin-1 correlate with shorter disease-free survival in human metastatic melanoma. Int J Cancer 2002; 97(4): 432-8.
[http://dx.doi.org/10.1002/ijc.1636] [PMID: 11802203]
[39] Sapoznik S, Ortenberg R, Schachter J, Markel G. CEACAM1 in malignant melanoma: a diagnostic and therapeutic target. Curr Top Med Chem 2012; 12(1): 3-10.
[http://dx.doi.org/10.2174/156802612798919259] [PMID: 22196267]
[40] Vergilis IJ, Szarek M, Ferrone S, Reynolds SR. Presence and prognostic significance of melanoma-associated antigens CYT-MAA and HMW-MAA in serum of patients with melanoma. J Invest Dermatol 2005; 125(3): 526-31.
[http://dx.doi.org/10.1111/j.0022-202X.2005.23798.x] [PMID: 16117794]
[41] Mann GJ, Pupo GM, Campain AE, et al. BRAF mutation, NRAS mutation, and the absence of an immune-related expressed gene profile predict poor outcome in patients with stage III melanoma. J Invest Dermatol 2013; 133(2): 509-17.
[http://dx.doi.org/10.1038/jid.2012.283] [PMID: 22931913]
[42] Kong Y, Si L, Zhu Y, et al. Large-scale analysis of KIT aberrations in Chinese patients with melanoma. Clin Cancer Res 2011; 17(7): 1684-91.
[http://dx.doi.org/10.1158/1078-0432.CCR-10-2346] [PMID: 21325067]
[43] Hodis E, Watson IR, Kryukov GV, et al. A landscape of driver mutations in melanoma. Cell 2012; 150(2): 251-63.
[http://dx.doi.org/10.1016/j.cell.2012.06.024] [PMID: 22817889]
[44] Gerami P, Li G, Pouryazdanparast P, et al. A highly specific and discriminatory FISH assay for distinguishing between benign and malignant melanocytic neoplasms. Am J Surg Pathol 2012; 36(6): 808-17.
[http://dx.doi.org/10.1097/PAS.0b013e31824b1efd] [PMID: 22588064]
[45] Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu Rev Immunol 2009; 27: 591-619.
[http://dx.doi.org/10.1146/annurev.immunol.021908.132706] [PMID: 19132916]
[46] Lee B, Mukhi N, Liu D. Current management and novel agents for malignant melanoma. J Hematol Oncol 2012; 5: 3.
[http://dx.doi.org/10.1186/1756-8722-5-3] [PMID: 22333219]
[47] Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364(26): 2517-26.
[http://dx.doi.org/10.1056/NEJMoa1104621] [PMID: 21639810]
[48] Poust J. Targeting metastatic melanoma. Am J Health Syst Pharm 2008; 65(24)(Suppl. 9): S9-S15.
[http://dx.doi.org/10.2146/ajhp080461] [PMID: 19052265]
[49] Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol 2012; 24(2): 207-12.
[http://dx.doi.org/10.1016/j.coi.2011.12.009] [PMID: 22236695]
[50] Agata Y, Kawasaki A, Nishimura H, et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 1996; 8(5): 765-72.
[http://dx.doi.org/10.1093/intimm/8.5.765] [PMID: 8671665]
[51] Hamid O, Robert C, Daud A, et al. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 2013; 369(2): 134-44.
[http://dx.doi.org/10.1056/NEJMoa1305133] [PMID: 23724846]
[52] Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013; 369(2): 122-33.
[http://dx.doi.org/10.1056/NEJMoa1302369] [PMID: 23724867]
[53] Gonzalez D, Fearfield L, Nathan P, et al. BRAF mutation testing algorithm for vemurafenib treatment in melanoma: recommendations from an expert panel. Br J Dermatol 2013; 168(4): 700-7.
[http://dx.doi.org/10.1111/bjd.12248] [PMID: 23360189]
[54] Halait H, Demartin K, Shah S, et al. Analytical performance of a real-time PCR-based assay for V600 mutations in the BRAF gene, used as the companion diagnostic test for the novel BRAF inhibitor vemurafenib in metastatic melanoma. Diagn Mol Pathol 2012; 21(1): 1-8.
[http://dx.doi.org/10.1097/PDM.0b013e31823b216f] [PMID: 22306669]
[55] Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med 2012; 366(8): 707-14.
[http://dx.doi.org/10.1056/NEJMoa1112302] [PMID: 22356324]
[56] Guilhot F. Indications for imatinib mesylate therapy and clinical management. Oncologist 2004; 9(3): 271-81.
[http://dx.doi.org/10.1634/theoncologist.9-3-271] [PMID: 15169982]
[57] Cho JH, Kim KM, Kwon M, Kim JH, Lee J. Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Invest New Drugs 2012; 30(5): 2008-14.
[http://dx.doi.org/10.1007/s10637-011-9763-9] [PMID: 22068222]
[58] Shah NP, Tran C, Lee FY, Chen P, Norris D, Sawyers CL. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science 2004; 305(5682): 399-401.
[http://dx.doi.org/10.1126/science.1099480] [PMID: 15256671]
[59] Kluger HM, Dudek AZ, McCann C, et al. A phase 2 trial of dasatinib in advanced melanoma. Cancer 2011; 117(10): 2202-8.
[http://dx.doi.org/10.1002/cncr.25766] [PMID: 21523734]
[60] Tarhini AA, Frankel P, Margolin KA, et al. Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin Cancer Res 2011; 17(20): 6574-81.
[http://dx.doi.org/10.1158/1078-0432.CCR-11-1463] [PMID: 21880788]
[61] Kirkwood JM, Ernstoff MS. Interferons in the treatment of human cancer. J Clin Oncol 1984; 2(4): 336-52.
[PMID: 6323641]
[62] Kirkwood JM, Ibrahim JG, Sosman JA, et al. High-dose interferon alfa-2b significantly prolongs relapse-free and overall survival compared with the GM2-KLH/QS-21 vaccine in patients with resected stage IIB-III melanoma: results of intergroup trial E1694/S9512/C509801. J Clin Oncol 2001; 19(9): 2370-80.
[PMID: 11331315]
[63] Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999; 17(7): 2105-16.
[PMID: 10561265]
[64] Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther 2007; 15(4): 651-9.
[http://dx.doi.org/10.1038/sj.mt.6300108] [PMID: 17299401]
[65] Kee D, McArthur G. Immunotherapy of melanoma. Eur J Surg Oncol 2016; S0748-7983(16)30684-9.
[PMID: 27514721]
[66] Garber K. China approves worlds first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst 2006; 98(5): 298-300.
[http://dx.doi.org/10.1093/jnci/djj111] [PMID: 16507823]
[67] Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 2015; 14(9): 642-62.
[http://dx.doi.org/10.1038/nrd4663] [PMID: 26323545]
[68] Glasgow JN, Everts M, Curiel DT. Transductional targeting of adenovirus vectors for gene therapy. Cancer Gene Ther 2006; 13(9): 830-44.
[http://dx.doi.org/10.1038/sj.cgt.7700928] [PMID: 16439993]
[69] Barker SD, Dmitriev IP, Nettelbeck DM, et al. Combined transcriptional and transductional targeting improves the specificity and efficacy of adenoviral gene delivery to ovarian carcinoma. Gene Ther 2003; 10(14): 1198-204.
[http://dx.doi.org/10.1038/sj.gt.3301974] [PMID: 12833129]
[70] Shashkova EV, May SM, Doronin K, Barry MA. Expanded anticancer therapeutic window of hexon-modified oncolytic adenovirus. Mol Ther 2009; 17(12): 2121-30.
[http://dx.doi.org/10.1038/mt.2009.217] [PMID: 19755961]
[71] Morizono K, Xie Y, Ringpis GE, et al. Lentiviral vector retargeting to P-glycoprotein on metastatic melanoma through intravenous injection. Nat Med 2005; 11(3): 346-52.
[http://dx.doi.org/10.1038/nm1192] [PMID: 15711560]
[72] Waehler R, Russell SJ, Curiel DT. Engineering targeted viral vectors for gene therapy. Nat Rev Genet 2007; 8(8): 573-87.
[http://dx.doi.org/10.1038/nrg2141] [PMID: 17607305]
[73] Li HJ, Everts M, Pereboeva L, et al. Adenovirus tumor targeting and hepatic untargeting by a coxsackie/adenovirus receptor ectodomain anti-carcinoembryonic antigen bispecific adapter. Cancer Res 2007; 67(11): 5354-61.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-4679] [PMID: 17545616]
[74] Parrott MB, Adams KE, Mercier GT, Mok H, Campos SK, Barry MA. Metabolically biotinylated adenovirus for cell targeting, ligand screening, and vector purification. Mol Ther 2003; 8(4): 688-700.
[http://dx.doi.org/10.1016/S1525-0016(03)00213-2] [PMID: 14529842]
[75] Martín F, Chowdhury S, Neil S, Phillipps N, Collins MK. Envelope-targeted retrovirus vectors transduce melanoma xenografts but not spleen or liver. Mol Ther 2002; 5(3): 269-74.
[http://dx.doi.org/10.1006/mthe.2002.0550] [PMID: 11863416]
[76] Martin F, Neil S, Kupsch J, Maurice M, Cosset F, Collins M. Retrovirus targeting by tropism restriction to melanoma cells. J Virol 1999; 73(8): 6923-9.
[PMID: 10400790]
[77] Purow B, Staveley-OCarroll K. Targeting of vaccinia virus using biotin-avidin viral coating and biotinylated antibodies. J Surg Res 2005; 123(1): 49-54.
[http://dx.doi.org/10.1016/j.jss.2004.04.022] [PMID: 15652950]
[78] Wu L, Johnson M, Sato M. Transcriptionally targeted gene therapy to detect and treat cancer. Trends Mol Med 2003; 9(10): 421-9.
[http://dx.doi.org/10.1016/j.molmed.2003.08.005] [PMID: 14557054]
[79] Lu B, Makhija SK, Nettelbeck DM, et al. Evaluation of tumor-specific promoter activities in melanoma. Gene Ther 2005; 12(4): 330-8.
[http://dx.doi.org/10.1038/sj.gt.3302385] [PMID: 15696177]
[80] Zhu ZB, Makhija SK, Lu B, et al. Transcriptional targeting of adenoviral vector through the CXCR4 tumor-specific promoter. Gene Ther 2004; 11(7): 645-8.
[http://dx.doi.org/10.1038/sj.gt.3302089] [PMID: 15029227]
[81] Denkert C, Köbel M, Berger S, et al. Expression of cyclooxygenase 2 in human malignant melanoma. Cancer Res 2001; 61(1): 303-8.
[PMID: 11196178]
[82] Robledo MM, Bartolome RA, Longo N, et al. Expression of functional chemokine receptors CXCR3 and CXCR4 on human melanoma cells. J Biol Chem 2001; 276(48): 45098-105.
[http://dx.doi.org/10.1074/jbc.M106912200] [PMID: 11571298]
[83] Smith B, Selby P, Southgate J, Pittman K, Bradley C, Blair GE. Detection of melanoma cells in peripheral blood by means of reverse transcriptase and polymerase chain reaction. Lancet 1991; 338(8777): 1227-9.
[http://dx.doi.org/10.1016/0140-6736(91)92100-G] [PMID: 1719320]
[84] Nettelbeck DM, Rivera AA, Balagué C, Alemany R, Curiel DT. Novel oncolytic adenoviruses targeted to melanoma: specific viral replication and cytolysis by expression of E1A mutants from the tyrosinase enhancer/promoter. Cancer Res 2002; 62(16): 4663-70.
[PMID: 12183423]
[85] Asanuma K, Kobayashi D, Furuya D, Tsuji N, Yagihashi A, Watanabe N. A role for survivin in radioresistance of pancreatic cancer cells. Jpn J Cancer Res 2002; 93(9): 1057-62.
[http://dx.doi.org/10.1111/j.1349-7006.2002.tb02483.x] [PMID: 12359060]
[86] Marcato P, Shmulevitz M, Lee PW. Connecting reovirus oncolysis and Ras signaling. Cell Cycle 2005; 4(4): 556-9.
[http://dx.doi.org/10.4161/cc.4.4.1600] [PMID: 15753655]
[87] Bos JL. ras oncogenes in human cancer: a review. Cancer Res 1989; 49(17): 4682-9.
[PMID: 2547513]
[88] Errington F, White CL, Twigger KR, et al. Inflammatory tumour cell killing by oncolytic reovirus for the treatment of melanoma. Gene Ther 2008; 15(18): 1257-70.
[http://dx.doi.org/10.1038/gt.2008.58] [PMID: 18401435]
[89] Noser JA, Mael AA, Sakuma R, et al. The RAS/Raf1/MEK/ERK signaling pathway facilitates VSV-mediated oncolysis: implication for the defective interferon response in cancer cells. Mol Ther 2007; 15(8): 1531-6.
[http://dx.doi.org/10.1038/sj.mt.6300193] [PMID: 17505473]
[90] Critchley-Thorne RJ, Simons DL, Yan N, et al. Impaired interferon signaling is a common immune defect in human cancer. Proc Natl Acad Sci USA 2009; 106(22): 9010-5.
[http://dx.doi.org/10.1073/pnas.0901329106] [PMID: 19451644]
[91] Wong LH, Krauer KG, Hatzinisiriou I, et al. Interferon-resistant human melanoma cells are deficient in ISGF3 components, STAT1, STAT2, and p48-ISGF3gamma. J Biol Chem 1997; 272(45): 28779-85.
[http://dx.doi.org/10.1074/jbc.272.45.28779] [PMID: 9353349]
[92] Hatada E, Saito S, Fukuda R. Mutant influenza viruses with a defective NS1 protein cannot block the activation of PKR in infected cells. J Virol 1999; 73(3): 2425-33.
[PMID: 9971827]
[93] Bergmann M, Romirer I, Sachet M, et al. A genetically engineered influenza A virus with ras-dependent oncolytic properties. Cancer Res 2001; 61(22): 8188-93.
[PMID: 11719449]
[94] Eberle J, Fecker LF, Forschner T, Ulrich C, Röwert-Huber J, Stockfleth E. Apoptosis pathways as promising targets for skin cancer therapy. Br J Dermatol 2007; 156(Suppl. 3): 18-24.
[http://dx.doi.org/10.1111/j.1365-2133.2007.07855.x] [PMID: 17488402]
[95] Fecker LF, Geilen CC, Hossini AM, et al. Selective induction of apoptosis in melanoma cells by tyrosinase promoter-controlled CD95 ligand overexpression. J Invest Dermatol 2005; 124(1): 221-8.
[http://dx.doi.org/10.1111/j.0022-202X.2004.23572.x] [PMID: 15654977]
[96] Ravindra PV, Tiwari AK, Sharma B, Chauhan RS. Newcastle disease virus as an oncolytic agent. Indian J Med Res 2009; 130(5): 507-13.
[PMID: 20090097]
[97] Zeng J, Fournier P, Schirrmacher V. Induction of interferon-alpha and tumor necrosis factor-related apoptosis-inducing ligand in human blood mononuclear cells by hemagglutinin-neuraminidase but not F protein of Newcastle disease virus. Virology 2002; 297(1): 19-30.
[http://dx.doi.org/10.1006/viro.2002.1413] [PMID: 12083832]
[98] Patel CA, Mukhtar M, Pomerantz RJ. Human immunodeficiency virus type 1 Vpr induces apoptosis in human neuronal cells. J Virol 2000; 74(20): 9717-26.
[http://dx.doi.org/10.1128/JVI.74.20.9717-9726.2000] [PMID: 11000244]
[99] Uttenthal A, Stadejek T, Nylin B. Genetic diversity of bovine viral diarrhoea viruses (BVDV) in Denmark during a 10-year eradication period. APMIS 2005; 113(7-8): 536-41.
[http://dx.doi.org/10.1111/j.1600-0463.2005.apm_227.x] [PMID: 16086824]
[100] Tumilasci VF, Olière S, Nguyên TL, Shamy A, Bell J, Hiscott J. Targeting the apoptotic pathway with BCL-2 inhibitors sensitizes primary chronic lymphocytic leukemia cells to vesicular stomatitis virus-induced oncolysis. J Virol 2008; 82(17): 8487-99.
[http://dx.doi.org/10.1128/JVI.00851-08] [PMID: 18579592]
[101] Villanueva J, Herlyn M. Melanoma and the tumor microenvironment. Curr Oncol Rep 2008; 10(5): 439-46.
[http://dx.doi.org/10.1007/s11912-008-0067-y] [PMID: 18706274]
[102] Wojton J, Kaur B. Impact of tumor microenvironment on oncolytic viral therapy. Cytokine Growth Factor Rev 2010; 21(2-3): 127-34.
[http://dx.doi.org/10.1016/j.cytogfr.2010.02.014] [PMID: 20399700]
[103] Frentzen A, Yu YA, Chen N, et al. Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy. Proc Natl Acad Sci USA 2009; 106(31): 12915-20.
[http://dx.doi.org/10.1073/pnas.0900660106] [PMID: 19617539]
[104] Liu TC, Zhang T, Fukuhara H, et al. Oncolytic HSV armed with platelet factor 4, an antiangiogenic agent, shows enhanced efficacy. Mol Ther 2006; 14(6): 789-97.
[http://dx.doi.org/10.1016/j.ymthe.2006.07.011] [PMID: 17045531]
[105] Yoo JY, Kim JH, Kwon YG, et al. VEGF-specific short hairpin RNA-expressing oncolytic adenovirus elicits potent inhibition of angiogenesis and tumor growth. Mol Ther 2007; 15(2): 295-302.
[http://dx.doi.org/10.1038/sj.mt.6300023] [PMID: 17235307]
[106] Coughlin CM, Salhany KE, Wysocka M, et al. Interleukin-12 and interleukin-18 synergistically induce murine tumor regression which involves inhibition of angiogenesis. J Clin Invest 1998; 101(6): 1441-52.
[http://dx.doi.org/10.1172/JCI1555] [PMID: 9502787]
[107] Zheng JN, Pei DS, Mao LJ, et al. Oncolytic adenovirus expressing interleukin-18 induces significant antitumor effects against melanoma in mice through inhibition of angiogenesis. Cancer Gene Ther 2010; 17(1): 28-36.
[http://dx.doi.org/10.1038/cgt.2009.38] [PMID: 19498459]
[108] Kuriyama N, Kuriyama H, Julin CM, Lamborn K, Israel MA. Pretreatment with protease is a useful experimental strategy for enhancing adenovirus-mediated cancer gene therapy. Hum Gene Ther 2000; 11(16): 2219-30.
[http://dx.doi.org/10.1089/104303400750035744] [PMID: 11084679]
[109] Guedan S, Rojas JJ, Gros A, Mercade E, Cascallo M, Alemany R. Hyaluronidase expression by an oncolytic adenovirus enhances its intratumoral spread and suppresses tumor growth. Mol Ther 2010; 18(7): 1275-83.
[http://dx.doi.org/10.1038/mt.2010.79] [PMID: 20442708]
[110] Cheng J, Sauthoff H, Huang Y, et al. Human matrix metalloproteinase-8 gene delivery increases the oncolytic activity of a replicating adenovirus. Mol Ther 2007; 15(11): 1982-90.
[http://dx.doi.org/10.1038/sj.mt.6300264] [PMID: 17653103]
[111] Choi JW, Lee YS, Yun CO, Kim SW. Polymeric oncolytic adenovirus for cancer gene therapy. J Control Release 2015; 219: 181-91.
[http://dx.doi.org/10.1016/j.jconrel.2015.10.009] [PMID: 26453806]
[112] De Silva N, Atkins H, Kirn DH, Bell JC, Breitbach CJ. Double trouble for tumours: exploiting the tumour microenvironment to enhance anticancer effect of oncolytic viruses. Cytokine Growth Factor Rev 2010; 21(2-3): 135-41.
[http://dx.doi.org/10.1016/j.cytogfr.2010.02.007] [PMID: 20338801]
[113] Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012; 30(7): 658-70.
[http://dx.doi.org/10.1038/nbt.2287] [PMID: 22781695]
[114] Hareuveni M, Gautier C, Kieny MP, Wreschner D, Chambon P, Lathe R. Vaccination against tumor cells expressing breast cancer epithelial tumor antigen. Proc Natl Acad Sci USA 1990; 87(23): 9498-502.
[http://dx.doi.org/10.1073/pnas.87.23.9498] [PMID: 2251291]
[115] Pulido J, Kottke T, Thompson J, et al. Using virally expressed melanoma cDNA libraries to identify tumor-associated antigens that cure melanoma. Nat Biotechnol 2012; 30(4): 337-43.
[http://dx.doi.org/10.1038/nbt.2157] [PMID: 22426030]
[116] Kottke T, Errington F, Pulido J, et al. Broad antigenic coverage induced by vaccination with virus-based cDNA libraries cures established tumors. Nat Med 2011; 17(7): 854-9.
[http://dx.doi.org/10.1038/nm.2390] [PMID: 21685898]
[117] Deng L, Fan J, Guo M, Huang B. Oncolytic and immunologic cancer therapy with GM-CSF-armed vaccinia virus of Tian Tan strain Guang9. Cancer Lett 2016; 372(2): 251-7.
[http://dx.doi.org/10.1016/j.canlet.2016.01.025] [PMID: 26803055]
[118] Todo T, Martuza RL, Dallman MJ, Rabkin SD. In situ expression of soluble B71 in the context of oncolytic herpes simplex virus induces potent antitumor immunity. Cancer Res 2001; 61(1): 153-61.
[PMID: 11196154]
[119] Loskog AS, Fransson ME, Totterman TT. AdCD40L gene therapy counteracts T regulatory cells and cures aggressive tumors in an orthotopic bladder cancer model. Clin Cancer Res 2005; 11(24 Pt 1): 8816-21.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-1817] [PMID: 16361570]
[120] Mackey MF, Gunn JR, Maliszewsky C, Kikutani H, Noelle RJ, Barth RJ Jr. Dendritic cells require maturation via CD40 to generate protective antitumor immunity. J Immunol 1998; 161(5): 2094-8.
[PMID: 9725199]
[121] Galivo F, Diaz RM, Thanarajasingam U, et al. Interference of CD40L-mediated tumor immunotherapy by oncolytic vesicular stomatitis virus. Hum Gene Ther 2010; 21(4): 439-50.
[http://dx.doi.org/10.1089/hum.2009.143] [PMID: 19922169]
[122] Lee TH, Cho YH, Lee JD, Yang WI, Shin JL, Lee MG. Enhanced antitumor effect of dendritic cell based immunotherapy after intratumoral injection of radionuclide Ho-166 against B16 melanoma. Immunol Lett 2006; 106(1): 19-26.
[http://dx.doi.org/10.1016/j.imlet.2006.03.007] [PMID: 16647143]
[123] Lee YS, Kim JH, Choi KJ, et al. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B71 in an immunocompetent murine model. Clin Cancer Res 2006; 12(19): 5859-68.
[http://dx.doi.org/10.1158/1078-0432.CCR-06-0935] [PMID: 17020994]
[124] Gao Y, Whitaker-Dowling P, Griffin JA, Barmada MA, Bergman I. Recombinant vesicular stomatitis virus targeted to Her2/neu combined with anti-CTLA4 antibody eliminates implanted mammary tumors. Cancer Gene Ther 2009; 16(1): 44-52.
[http://dx.doi.org/10.1038/cgt.2008.55] [PMID: 18654610]
[125] Dias JD, Hemminki O, Diaconu I, et al. Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4. Gene Ther 2012; 19(10): 988-98.
[http://dx.doi.org/10.1038/gt.2011.176] [PMID: 22071969]
[126] Engeland CE, Grossardt C, Veinalde R, et al. CTLA-4 and PD-L1 checkpoint blockade enhances oncolytic measles virus therapy. Mol Ther 2014; 22(11): 1949-59.
[http://dx.doi.org/10.1038/mt.2014.160] [PMID: 25156126]
[127] Vigil A, Park MS, Martinez O, et al. Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res 2007; 67(17): 8285-92.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-1025] [PMID: 17804743]
[128] Zhao H, Janke M, Fournier P, Schirrmacher V. Recombinant Newcastle disease virus expressing human interleukin-2 serves as a potential candidate for tumor therapy. Virus Res 2008; 136(1-2): 75-80.
[http://dx.doi.org/10.1016/j.virusres.2008.04.020] [PMID: 18538434]
[129] Carew JF, Kooby DA, Halterman MW, Kim SH, Federoff HJ, Fong Y. A novel approach to cancer therapy using an oncolytic herpes virus to package amplicons containing cytokine genes. Mol Ther 2001; 4(3): 250-6.
[http://dx.doi.org/10.1006/mthe.2001.0448] [PMID: 11545616]
[130] Choi IK, Lee JS, Zhang SN, et al. Oncolytic adenovirus co-expressing IL-12 and IL-18 improves tumor-specific immunity via differentiation of T cells expressing IL-12Rβ2 or IL-18Rα. Gene Ther 2011; 18(9): 898-909.
[http://dx.doi.org/10.1038/gt.2011.37] [PMID: 21451575]
[131] van Rikxoort M, Michaelis M, Wolschek M, et al. Oncolytic effects of a novel influenza A virus expressing interleukin-15 from the NS reading frame. PLoS One 2012; 7(5): e36506.
[http://dx.doi.org/10.1371/journal.pone.0036506] [PMID: 22563505]
[132] Fukuhara H, Ino Y, Kuroda T, Martuza RL, Todo T. Triple gene-deleted oncolytic herpes simplex virus vector double-armed with interleukin 18 and soluble B71 constructed by bacterial artificial chromosome-mediated system. Cancer Res 2005; 65(23): 10663-8.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-2534] [PMID: 16322208]
[133] Ino Y, Saeki Y, Fukuhara H, Todo T. Triple combination of oncolytic herpes simplex virus-1 vectors armed with interleukin-12, interleukin-18, or soluble B71 results in enhanced antitumor efficacy. Clin Cancer Res 2006; 12(2): 643-52.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-1494] [PMID: 16428511]
[134] Su C, Peng L, Sham J, et al. Immune gene-viral therapy with triplex efficacy mediated by oncolytic adenovirus carrying an interferon-gamma gene yields efficient antitumor activity in immunodeficient and immunocompetent mice. Mol Ther 2006; 13(5): 918-27.
[http://dx.doi.org/10.1016/j.ymthe.2005.12.011] [PMID: 16497559]
[135] Dilley J, Reddy S, Ko D, et al. Oncolytic adenovirus CG7870 in combination with radiation demonstrates synergistic enhancements of antitumor efficacy without loss of specificity. Cancer Gene Ther 2005; 12(8): 715-22.
[http://dx.doi.org/10.1038/sj.cgt.7700835] [PMID: 15832172]
[136] Adusumilli PS, Chan MK, Hezel M, et al. Radiation-induced cellular DNA damage repair response enhances viral gene therapy efficacy in the treatment of malignant pleural mesothelioma. Ann Surg Oncol 2007; 14(1): 258-69.
[http://dx.doi.org/10.1245/s10434-006-9127-4] [PMID: 17080237]
[137] Adusumilli PS, Stiles BM, Chan MK, et al. Radiation therapy potentiates effective oncolytic viral therapy in the treatment of lung cancer. Ann Thorac Surg 2005; 80(2): 409-16.
[http://dx.doi.org/10.1016/j.athoracsur.2005.01.048] [PMID: 16039175]
[138] Advani SJ, Sibley GS, Song PY, et al. Enhancement of replication of genetically engineered herpes simplex viruses by ionizing radiation: a new paradigm for destruction of therapeutically intractable tumors. Gene Ther 1998; 5(2): 160-5.
[http://dx.doi.org/10.1038/sj.gt.3300546] [PMID: 9578834]
[139] Jarnagin WR, Zager JS, Hezel M, et al. Treatment of cholangiocarcinoma with oncolytic herpes simplex virus combined with external beam radiation therapy. Cancer Gene Ther 2006; 13(3): 326-34.
[http://dx.doi.org/10.1038/sj.cgt.7700890] [PMID: 16138120]
[140] Kim SH, Wong RJ, Kooby DA, et al. Combination of mutated herpes simplex virus type 1 (G207 virus) with radiation for the treatment of squamous cell carcinoma of the head and neck. Eur J Cancer 2005; 41(2): 313-22.
[http://dx.doi.org/10.1016/j.ejca.2004.10.018] [PMID: 15661558]
[141] Gupta AK, Cerniglia GJ, Mick R, et al. Radiation sensitization of human cancer cells in vivo by inhibiting the activity of PI3K using LY294002. Int J Radiat Oncol Biol Phys 2003; 56(3): 846-53.
[http://dx.doi.org/10.1016/S0360-3016(03)00214-1] [PMID: 12788194]
[142] Gupta AK, McKenna WG, Weber CN, et al. Local recurrence in head and neck cancer: relationship to radiation resistance and signal transduction. Clin Cancer Res 2002; 8(3): 885-92.
[PMID: 11895923]
[143] McKenna WG, Muschel RJ, Gupta AK, Hahn SM, Bernhard EJ. The RAS signal transduction pathway and its role in radiation sensitivity. Oncogene 2003; 22(37): 5866-75.
[http://dx.doi.org/10.1038/sj.onc.1206699] [PMID: 12947393]
[144] Chinnaiyan AM, Prasad U, Shankar S, et al. Combined effect of tumor necrosis factor-related apoptosis-inducing ligand and ionizing radiation in breast cancer therapy. Proc Natl Acad Sci USA 2000; 97(4): 1754-9.
[http://dx.doi.org/10.1073/pnas.030545097] [PMID: 10677530]
[145] Farone AL, OBrien PC, Cox DC. Tumor necrosis factor-alpha induction by reovirus serotype 3. J Leukoc Biol 1993; 53(2): 133-7.
[PMID: 8383165]
[146] Weichselbaum RR, Hallahan DE, Beckett MA, et al. Gene therapy targeted by radiation preferentially radiosensitizes tumor cells. Cancer Res 1994; 54(16): 4266-9.
[PMID: 8044769]
[147] Mauceri HJ, Hanna NN, Wayne JD, Hallahan DE, Hellman S, Weichselbaum RR. Tumor necrosis factor alpha (TNF-alpha) gene therapy targeted by ionizing radiation selectively damages tumor vasculature. Cancer Res 1996; 56(19): 4311-4.
[PMID: 8813113]
[148] Mundt AJ, Vijayakumar S, Nemunaitis J, et al. A Phase I trial of TNFerade biologic in patients with soft tissue sarcoma in the extremities. Clin Cancer Res 2004; 10(17): 5747-53.
[http://dx.doi.org/10.1158/1078-0432.CCR-04-0296] [PMID: 15355902]
[149] Twigger K, Vidal L, White CL, et al. Enhanced in vitro and in vivo cytotoxicity of combined reovirus and radiotherapy. Clin Cancer Res 2008; 14(3): 912-23.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-1400] [PMID: 18245555]
[150] Ottolino-Perry K, Diallo JS, Lichty BD, Bell JC, McCart JA. Intelligent design: combination therapy with oncolytic viruses. Mol Ther 2010; 18(2): 251-63.
[http://dx.doi.org/10.1038/mt.2009.283] [PMID: 20029399]
[151] Pandha HS, Heinemann L, Simpson GR, et al. Synergistic effects of oncolytic reovirus and cisplatin chemotherapy in murine malignant melanoma. Clin Cancer Res 2009; 15(19): 6158-66.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-0796] [PMID: 19773377]
[152] Galanis E, Markovic SN, Suman VJ, et al. Phase II trial of intravenous administration of Reolysin(®) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma. Mol Ther 2012; 20(10): 1998-2003.
[http://dx.doi.org/10.1038/mt.2012.146] [PMID: 22871663]
[153] Qiao J, Wang H, Kottke T, et al. Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus. Clin Cancer Res 2008; 14(1): 259-69.
[http://dx.doi.org/10.1158/1078-0432.CCR-07-1510] [PMID: 18172278]
[154] Quezada SA, Peggs KS. Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer. Br J Cancer 2013; 108(8): 1560-5.
[http://dx.doi.org/10.1038/bjc.2013.117] [PMID: 23511566]
[155] Lee SJ, McDermott DF, Rao UN, et al. Multicenter, randomized phase II trial of GM-CSF (GM) plus ipilimumab (Ipi) versus Ipi alone in metastatic melanoma: E1608. J Clin Oncol 2013; 31(18_suppl): CRA9007.
[PMID: 28136075]
[156] Ribas A, Hodi FS, Callahan M, Konto C, Wolchok J. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med 2013; 368(14): 1365-6.
[http://dx.doi.org/10.1056/NEJMc1302338] [PMID: 23550685]
[157] Rajani K, Parrish C, Kottke T, 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.
[http://dx.doi.org/10.1038/mt.2015.156] [PMID: 26310630]
[158] Bai F, Niu Z, Tian H, et al. Genetically engineered Newcastle disease virus expressing interleukin 2 is a potential drug candidate for cancer immunotherapy. Immunol Lett 2014; 159(1-2): 36-46.
[http://dx.doi.org/10.1016/j.imlet.2014.02.009] [PMID: 24613899]
[159] Kottke T, Galivo F, Wongthida P, et al. Treg depletion-enhanced IL-2 treatment facilitates therapy of established tumors using systemically delivered oncolytic virus. Mol Ther 2008; 16(7): 1217-26.
[http://dx.doi.org/10.1038/mt.2008.83] [PMID: 18431359]
[160] Larkin J, Del Vecchio M, Ascierto PA, et al. Vemurafenib in patients with BRAF(V600) mutated metastatic melanoma: an open-label, multicentre, safety study. Lancet Oncol 2014; 15(4): 436-44.
[http://dx.doi.org/10.1016/S1470-2045(14)70051-8] [PMID: 24582505]
[161] Watanabe D, Goshima F, Mori I, Tamada Y, Matsumoto Y, Nishiyama Y. Oncolytic virotherapy for malignant melanoma with herpes simplex virus type 1 mutant HF10. J Dermatol Sci 2008; 50(3): 185-96.
[http://dx.doi.org/10.1016/j.jdermsci.2007.12.001] [PMID: 18226503]
[162] Shafren D, Quah M, Wong Y, Andtbacka RH, Au G. Combination of a novel oncolytic immunotherapeutic agent, coxsackievirus a21 and pd-1 blockade significantly reduces tumor growth and improves survival in an immune competent mouse melanoma model. Ann Oncol 2014; •••: 25. [PMID: WOS:000346901000307].
[163] Simmons AD, Moskalenko M, Creson J, et al. Local secretion of anti-CTLA-4 enhances the therapeutic efficacy of a cancer immunotherapy with reduced evidence of systemic autoimmunity. Cancer Immunol Immunother 2008; 57(8): 1263-70.
[http://dx.doi.org/10.1007/s00262-008-0451-3] [PMID: 18236040]
[164] Kirn DH, Wang Y, Le Boeuf F, Bell J, Thorne SH. Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus. PLoS Med 2007; 4(12): e353.
[http://dx.doi.org/10.1371/journal.pmed.0040353] [PMID: 18162040]
[165] Willmon CL, Saloura V, Fridlender ZG, et al. Expression of IFN-beta enhances both efficacy and safety of oncolytic vesicular stomatitis virus for therapy of mesothelioma. Cancer Res 2009; 69(19): 7713-20.
[http://dx.doi.org/10.1158/0008-5472.CAN-09-1013] [PMID: 19773437]
[166] Lei N, Shen FB, Chang JH, et al. An oncolytic adenovirus expressing granulocyte macrophage colony-stimulating factor shows improved specificity and efficacy for treating human solid tumors. Cancer Gene Ther 2009; 16(1): 33-43.
[http://dx.doi.org/10.1038/cgt.2008.46] [PMID: 18670453]
[167] Chen H, Sampath P, Hou W, Thorne SH. Regulating cytokine function enhances safety and activity of genetic cancer therapies. Mol Ther 2013; 21(1): 167-74.
[http://dx.doi.org/10.1038/mt.2012.225] [PMID: 23281444]
[168] Puzanov I, Milhem MM, Andtbacka RH, Minor DR, Hamid O, Li A, et al. Primary analysis of a phase 1b multicenter trial to evaluate safety and efficacy of talimogene laherparepvec (T-VEC) and ipilimumab (ipi) in previously untreated, unresected stage IIIB-IV melanoma. J Clin Oncol 2014; 32(15) [PMID: WOS:000358613204315].
[169] Weber JS. At the bedside: adoptive cell therapy for melanoma-clinical development. J Leukoc Biol 2014; 95(6): 875-82.
[http://dx.doi.org/10.1189/jlb.0513293] [PMID: 24732024]
[170] Khammari A, Nguyen JM, Saint-Jean M, et al. Adoptive T cell therapy combined with intralesional administrations of TG1042 (adenovirus expressing interferon-γ) in metastatic melanoma patients. Cancer Immunol Immunother 2015; 64(7): 805-15.
[http://dx.doi.org/10.1007/s00262-015-1691-7] [PMID: 25846669]
[171] Martens A, Wistuba-Hamprecht K, Geukes Foppen M, et al. Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22(12): 2908-18.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-2412] [PMID: 26787752]
[172] Irvine KR, Chamberlain RS, Shulman EP, Surman DR, Rosenberg SA, Restifo NP. Enhancing efficacy of recombinant anticancer vaccines with prime/boost regimens that use two different vectors. J Natl Cancer Inst 1997; 89(21): 1595-601.
[http://dx.doi.org/10.1093/jnci/89.21.1595] [PMID: 9362157]
[173] Marshall JL, Hoyer RJ, Toomey MA, et al. Phase I study in advanced cancer patients of a diversified prime-and-boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avipox virus to elicit anti-carcinoembryonic antigen immune responses. J Clin Oncol 2000; 18(23): 3964-73.
[PMID: 11099326]
Track Your Manuscript:


Endorsements



"Open access will revolutionize 21st century knowledge work and accelerate the diffusion of ideas and evidence that support just in time learning and the evolution of thinking in a number of disciplines."


Daniel Pesut
(Indiana University School of Nursing, USA)

"It is important that students and researchers from all over the world can have easy access to relevant, high-standard and timely scientific information. This is exactly what Open Access Journals provide and this is the reason why I support this endeavor."


Jacques Descotes
(Centre Antipoison-Centre de Pharmacovigilance, France)

"Publishing research articles is the key for future scientific progress. Open Access publishing is therefore of utmost importance for wider dissemination of information, and will help serving the best interest of the scientific community."


Patrice Talaga
(UCB S.A., Belgium)

"Open access journals are a novel concept in the medical literature. They offer accessible information to a wide variety of individuals, including physicians, medical students, clinical investigators, and the general public. They are an outstanding source of medical and scientific information."


Jeffrey M. Weinberg
(St. Luke's-Roosevelt Hospital Center, USA)

"Open access journals are extremely useful for graduate students, investigators and all other interested persons to read important scientific articles and subscribe scientific journals. Indeed, the research articles span a wide range of area and of high quality. This is specially a must for researchers belonging to institutions with limited library facility and funding to subscribe scientific journals."


Debomoy K. Lahiri
(Indiana University School of Medicine, USA)

"Open access journals represent a major break-through in publishing. They provide easy access to the latest research on a wide variety of issues. Relevant and timely articles are made available in a fraction of the time taken by more conventional publishers. Articles are of uniformly high quality and written by the world's leading authorities."


Robert Looney
(Naval Postgraduate School, USA)

"Open access journals have transformed the way scientific data is published and disseminated: particularly, whilst ensuring a high quality standard and transparency in the editorial process, they have increased the access to the scientific literature by those researchers that have limited library support or that are working on small budgets."


Richard Reithinger
(Westat, USA)

"Not only do open access journals greatly improve the access to high quality information for scientists in the developing world, it also provides extra exposure for our papers."


J. Ferwerda
(University of Oxford, UK)

"Open Access 'Chemistry' Journals allow the dissemination of knowledge at your finger tips without paying for the scientific content."


Sean L. Kitson
(Almac Sciences, Northern Ireland)

"In principle, all scientific journals should have open access, as should be science itself. Open access journals are very helpful for students, researchers and the general public including people from institutions which do not have library or cannot afford to subscribe scientific journals. The articles are high standard and cover a wide area."


Hubert Wolterbeek
(Delft University of Technology, The Netherlands)

"The widest possible diffusion of information is critical for the advancement of science. In this perspective, open access journals are instrumental in fostering researches and achievements."


Alessandro Laviano
(Sapienza - University of Rome, Italy)

"Open access journals are very useful for all scientists as they can have quick information in the different fields of science."


Philippe Hernigou
(Paris University, France)

"There are many scientists who can not afford the rather expensive subscriptions to scientific journals. Open access journals offer a good alternative for free access to good quality scientific information."


Fidel Toldrá
(Instituto de Agroquimica y Tecnologia de Alimentos, Spain)

"Open access journals have become a fundamental tool for students, researchers, patients and the general public. Many people from institutions which do not have library or cannot afford to subscribe scientific journals benefit of them on a daily basis. The articles are among the best and cover most scientific areas."


M. Bendandi
(University Clinic of Navarre, Spain)

"These journals provide researchers with a platform for rapid, open access scientific communication. The articles are of high quality and broad scope."


Peter Chiba
(University of Vienna, Austria)

"Open access journals are probably one of the most important contributions to promote and diffuse science worldwide."


Jaime Sampaio
(University of Trás-os-Montes e Alto Douro, Portugal)

"Open access journals make up a new and rather revolutionary way to scientific publication. This option opens several quite interesting possibilities to disseminate openly and freely new knowledge and even to facilitate interpersonal communication among scientists."


Eduardo A. Castro
(INIFTA, Argentina)

"Open access journals are freely available online throughout the world, for you to read, download, copy, distribute, and use. The articles published in the open access journals are high quality and cover a wide range of fields."


Kenji Hashimoto
(Chiba University, Japan)

"Open Access journals offer an innovative and efficient way of publication for academics and professionals in a wide range of disciplines. The papers published are of high quality after rigorous peer review and they are Indexed in: major international databases. I read Open Access journals to keep abreast of the recent development in my field of study."


Daniel Shek
(Chinese University of Hong Kong, Hong Kong)

"It is a modern trend for publishers to establish open access journals. Researchers, faculty members, and students will be greatly benefited by the new journals of Bentham Science Publishers Ltd. in this category."


Jih Ru Hwu
(National Central University, Taiwan)


Browse Contents




Webmaster Contact: info@benthamopen.net
Copyright © 2023 Bentham Open