Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2827 word(s) 2827 2021-06-23 05:44:22 |
2 format correct Meta information modification 2827 2021-06-30 04:19:19 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lundstrom, K. Viral Vector-Based Melanoma Gene Therapy. Encyclopedia. Available online: https://encyclopedia.pub/entry/11428 (accessed on 29 March 2024).
Lundstrom K. Viral Vector-Based Melanoma Gene Therapy. Encyclopedia. Available at: https://encyclopedia.pub/entry/11428. Accessed March 29, 2024.
Lundstrom, Kenneth. "Viral Vector-Based Melanoma Gene Therapy" Encyclopedia, https://encyclopedia.pub/entry/11428 (accessed March 29, 2024).
Lundstrom, K. (2021, June 29). Viral Vector-Based Melanoma Gene Therapy. In Encyclopedia. https://encyclopedia.pub/entry/11428
Lundstrom, Kenneth. "Viral Vector-Based Melanoma Gene Therapy." Encyclopedia. Web. 29 June, 2021.
Viral Vector-Based Melanoma Gene Therapy
Edit

Gene therapy applications of oncolytic viruses represent an attractive alternative for cancer treatment. A broad range of oncolytic viruses, including adenoviruses, adeno-associated viruses, alphaviruses, herpes simplex viruses, retroviruses, lentiviruses, rhabdoviruses, reoviruses, measles virus, Newcastle disease virus, picornaviruses and poxviruses, have been used in diverse preclinical and clinical studies for the treatment of various diseases, including colon, head-and-neck, prostate and breast cancer as well as squamous cell carcinoma and glioma. The majority of studies have focused on immunotherapy and several drugs based on viral vectors have been approved. However, gene therapy for malignant melanoma based on viral vectors has not been utilized to its full potential yet.

melanoma cancer vector delivery gene therapy immunotherapy clinical trials

1. Introduction

Melanoma, or malignant melanoma, represents a cancer type that develops in melanocytes known as pigment-containing cells [1]. Since the beginning of the 21st century, melanoma has remained one of the most fatal malignancies. Most patients, when diagnosed early, are treated by local surgical excision following sentinel lymph node biopsy [1]. The incidence varies by country, skin phenotype and sun exposure. It mostly affects young and middle-aged female populations (below the age of 50 years), but more males are affected from the age of 55 onwards [1]. In men the incidence of melanoma is three times higher than in women by the age of 75. Ultraviolet (UV) light is known to be the main cause of malignant melanoma. A history of sunburn in childhood or adolescence has been suggested to be directly associated with the development of melanoma. Other risk factors include the number of melanocytic nevi, family history and genetic background. It has also been confirmed that patients with a previous history of melanoma are more prone to develop multiple primary melanomas [1]. In contrast, other environmental factors like alcohol or tobacco consumption have not been associated with melanoma development [2].
Skin melanoma has generally been classified according to the origin of the sun exposure, the degree of cumulative UV exposure, age at the time of diagnosis, types of oncogenic drivers and the mutational load [2]. It is known that B-Raf proto-oncogene (BRAF), neurofibromin 1 (NF1) and NRAS mutations, together with a high mutational load related to UV exposure, are the main genetic drivers [3]. Cases of periodic sun exposure are usually associated with BRAFV600E and a lower mutational load [2]. It is important to mention that each melanoma subtype may evolve from different precursor lesions, which can involve different gene mutations as well as different transformational stages [3].
Current medical treatments include various methods. Most patients with recently diagnosed melanoma have early-stage disease and can be treated by surgical excision, which is curative in the majority of cases. Some treatment methods involve lymph node biopsies in addition to standard surgical excision. Unfortunately, 10% of all melanoma cases are diagnosed at an advanced/late stage and are already metastatic, including visceral and brain metastases [2][3][4][5]. These patients have a poor prognosis and the probability of treatment success is lower. For patients with advanced stage disease, revolutionary therapy agents including RAF (Rapidly Accelerated Fibrosarcoma) and MEK (Mitogen-activated Protein Kinase) kinase inhibitors as well as immune checkpoint inhibitors like anti-CTLA4 and anti-PD1 have been approved, in 2011 and 2016, respectively [5][6][7][8]. Anti-PD1 and anti-CTLA4 antibodies (nivolumab, pembrolizumab and ipilimumab) as well as BRAF and MEK inhibitors (vemurafenib and trametinib) have shown promising results in clinical trials [9][10][11][12][13][14][15][16]. Today, the presence of the BRAFV600E mutation is verified in clinical settings, since it determines the correct treatment strategy. Mutations in the NRAS, NF1, CKIT, CDKN2A and PTEN genes have not been included in clinical practice yet.
Immunotherapy and kinase inhibitors are known as backbones for second-line systemic chemotherapy [17]. In the past, chemotherapy represented the treatment option for advanced melanoma. Although attempts to improve patient responses by combination therapy failed, it is still used for palliative treatment of progressed melanomas [18]. Dacarbazine, an alkylating agent, was approved by the FDA in 1974 for standard chemotherapy treatment of metastatic melanoma [19]. Despite moderate results, dacarbazine has been used as the sole standard of care, recently (in clinical trials) in combination with other chemotherapies and immunotherapies (ClinicalTrials.gov) [20]. Temozolomide (TMZ), an active metabolite of dacarbazine, has been applied in advanced melanoma [20].
Electrochemotherapy (ECT) combines two cytotoxic drugs (cisplatin and bleomycin) with high-intensity electric pulses, which enhances the delivery of the drug into cells [21][22]. This approach has been used for the treatment of cutaneous and subcutaneous melanoma nodules [23]. The overall response was 85%, and no major negative adverse events were reported [21]. Photodynamic therapy (PDT) is a light-based therapy. It represents a promising adjuvant treatment and can be used as a palliative method of choice for patients with stage III/IV cutaneous metastatic melanomas [24]. PDT is considered a minimally invasive procedure that requires a photosensitizer. Absorption is superior in metabolically active tissues [24]. The method applies non-toxic compounds, which create reactive oxygen species (ROS) when combined with oxygen [25]. ROS does irreversible damage to tumor cells and tumor-associated blood vessels, and contributes to the activation of various antitumor, immune and inflammatory responses [25][26][27][28]. Although PDT can be applied to both non-malignant and malignant diseases, several reports show that PDT alone has only limited efficacy in melanoma [22]. To improve PDT results, certain protective mechanisms, like pigmentation and oxidative stress resistance, need to be overcome [29][30]. The combination of PDT and dacarbazine chemotherapy displayed resistance reduction in pigmented and unpigmented metastatic melanomas [31]. On the other hand, combination of PDT and immunotherapy may increase the effect of eradication of the initial tumor and decrease in melanoma recurrence [29].
In the context of oncolytic viruses, their selective replication triggers tumor cell death and vector spread into neighboring cells, providing an interesting approach for cancer therapy [32]. Immunization studies in experimental animal models have employed a wide range of viral vectors based on adenoviruses, alphaviruses, herpes simplex viruses, coxsackie viruses and vaccinia viruses, targeting cancer types like glioblastoma, colon, cervix, and lung cancer as well as melanoma [33][34]. The first virus-based melanoma drug was approved by the FDA in 2015 [35].

2. Viral Vectors for Melanoma Treatment

As previously described, a broad range of oncolytic viruses have been evaluated for cancer gene therapy [36]. The specific targeting and killing of tumor cells and the simultaneous stimulation of the immune system have made oncolytic viruses attractive delivery vehicles [37][38][39]. This dual action promotes tumor regression as well as the induction of immune responses through innate and adaptive components. On the other hand, naturally occurring, ubiquitous, non-enveloped dsRNA viruses have shown generally mild infection in humans, and specific replication and cytopathogenicity in transformed cells, which possess active Ras signaling pathways [40][41]. Their specificity for Ras transformed cells and their relatively non-pathogenic nature in humans make them attractive anticancer therapy candidates [40][41]. This approach may lead to the recognition and removal of systemic disease and the prevention of tumor return [42].

2.1. Melanoma Treatment Using Herpes Simplex Virus Type 1

The prototype drug for virotherapy is an attenuated herpes simplex virus type 1 (HSV-1), which is engineered to express the human granulocyte–macrophage colony-stimulating factor (GM–CSF) [35]. The approved drug known as talimogene laherparepvec (TVEC) has the trade name Imlygic®. TVEC showed two mechanisms of action, one being the oncolytic effect of infecting and killing tumor cells at the local injection site, and the other being the immunotherapeutic effect through induction of local and systemic immune responses [43].
TVEC replicates in tumor cells, which results in lysis and release of soluble tumor-associated antigens and viral pathogens. Migration and maturation of dendritic cells is induced by local GM-CSF expression, leading to ingestion of dissolvable tumor antigens and apoptotic tumor cells. The dendritic cells are transported to the nearest lymph nodes, where antigens initiate a systemic immune response, specifically in CD4+ and CD8+ helper and cytotoxic T-cells. However, the response rate in metastases is lower than in injected tumors, which reflects insufficient effector T-cell expansion. The other reason could be the lack of efficacy at distant sites. To overcome this limitation, combination therapy with TVEC and immune checkpoint blockers might provide better results [35].
Generally, local lytic TVEC infection in tumor cells leads to the release of various proteins, such as interferons, chemokines, danger-associated molecular pattern (DAMP), and pathogen-associated molecular pattern (PAMP). These can provide more favorable surroundings for stimulation of anti-tumor immune responses [44]. Cancer cell lysis discharges tumor-associated neoantigens for processing by dendritic cells, which are activated by TVEC-encoded GM–CSF. This could lead to stimulation of anti-tumor CD8+ T-cell responses against unrecognized antigens and this effect has been clinically demonstrated [44].

2.2. Melanoma Treatment Using Retroviruses/Lentiviruses

Retroviruses and lentiviruses are ssRNA, which can provide long-term transgene expression by integration into the host genome. They have frequently been used as gene therapy vectors for indications such as glioma [45][46], and breast [47], gastric [48], liver [49], pancreatic [50], and hematologic [51] cancers. One limitation of using retroviruses such as Moloney murine leukemia virus (MoMLV) for gene therapy is the requirement of cell division for transduction and integration [52]. In contrast, lentiviruses are capable of transduction of both dividing and non-dividing cells.

2.2.1. Preclinical Studies with Retroviruses/Lentiviruses

Although retroviruses have demonstrated potential for treating chronic diseases such as severe combined immunodeficiency (SCID) in children [53], fewer studies have been conducted for cancer. For instance, recombinant retrovirus vectors expressing GM–CSF and IL-4 showed high-level expression in cultured primary glioma cells, which lasted for 14 days and could therefore present an attractive approach for immunotherapy [45]. However, in recent years, lentiviral vectors have replaced conventional retroviruses in gene therapy. For instance, a lentivirus carrying the EGFP reporter gene provided long-term expression in DU145 and PC3 human prostate cell lines and in vivo in pre-established and orthotopic tumors [54]. In the context of melanoma, a lentiviral vector expressing the VP22-CD/5-FC suicide gene system demonstrated superior antitumor activity in a murine uveal melanoma model [55]. In another study, a lentivirus vector expressing RNAi sequences targeting the MAT2B gene, the regulatory subunit of methionine adenosyltransferase resulted in suppressed growth, colony formation and induced apoptosis in A375 and Mel-RM malignant melanoma cell lines, and affected tumor growth in a xenograft model in vivo [56]. Moreover, antisense non-coding mitochondrial RNA (ASncmtRNAs) was downregulated by a lentivirus vector expressing short hairpin RNA (shRNA), which induced apoptosis in murine B16F10 and human A375 melanoma cell lines, significantly reduced B16F10 tumor growth in vivo, and reduced the number of lung metastases in a tail vein assay [57].

2.2.2. Clinical Trials of Retroviruses/Lentiviruses for Melanoma Treatment

Related to lentivirus-based clinical trials, 30 children and adults with relapsed acute lymphoblastic leukemia (ALL) were treated with a lentiviral vector-based chimeric antigen receptor T (CAR-T), targeting CD19 (CTL019), which resulted in sustained remission with a 6-month event-free survival rate of 67% and an overall survival rate of 78% [58]. The treatment of relapsed and refractory ALL was efficient, with a high remission rate lasting for up to 24 months. In preparation for lentivirus-based clinical trials, monocyte-derived conventional dendritic cells (ConvDCs) were transduced using a tricistronic lentivirus vector, expressing GM–CSF, IL-4 and the melanoma antigen tyrosine-related protein 2 (TRP2), to overcome the difficulties in manufacturing and potency of ConvDCs [59]. The feasibility of this approach was demonstrated with monocytes from five advanced melanoma patients indicating that a simpler GMP-compliant method for manufacturing individualized DC vaccines with a higher specificity against melanoma is possible. In another approach, to improve ex vivo manufacturing of engineered T cells, isolated human CD8+ T cells from healthy donors were transduced with a lentivirus vector expressing the gp100-specific tumor antigen-specific T cell receptor (TCR) in the presence of a novel chemical lentiviral transduction enhancer (Lentiboost) [60]. It was demonstrated that antigen-specific secretion of tumor necrosis factor (TNF) and interferon-γ (IFN-γ) occurred in the transduced cells and significant cytotoxicity was detected in the antigen-positive tumor cells, showing the potential of lentivirus-based cancer immunotherapy.
The success of CAR-T based lentivirus therapy for hematological cancers such as ALL has also triggered treatment of solid tumors [61]. However, with tumors, the transition might be limited by therapeutic barriers such as CAR-T cell expansion, persistence, trafficking, and fate. In the context of melanoma, the first results from CAR-T cell therapy could not reproduce the findings from the treatment of hematological diseases [62]. Issues to be addressed include the lack of migration of CAR-T cells from blood vessels to the tumor site, as well as the immunosuppressive tumor microenvironment within solid tumors, before this technology can be successfully applied for melanoma treatment.

2.3. Melanoma Treatment Using Reoviruses

The nonenveloped dsRNA Reovirus Serotype 3-Dearing Strain known as Reolysin has been shown to replicate in specifically transformed cells possessing an activated RAS signaling pathway, which makes it a potential candidate for anticancer therapy [63][64][65]. The inhibition of dsRNA-activated protein kinase (PKR) in Ras-activated cells inhibits autophosphorylation of PKR, permitting viral translation and oncolysis in tumor cells [66][43]. As Ras pathway activation occurs in approximately 60% of metastatic melanoma patients, it provides a great opportunity for Reolysin testing in malignancy treatment [67][68].

2.3.1. Preclinical Studies with Reoviruses

It has been observed that some tumor cells and spontaneously transformed cell lines show favorable sensitivity toward reoviruses [69]. Other studies have revealed that intratumoral injection of reoviruses leads to regression of v-erbB-transformed NIH 3T3 or human U87 glioblastoma tumors in 80% of SCID mice. Moreover, multiple reovirus injections resulted in total tumor regression in 65% of immune-competent C3H mice [70]. The oncolytic capacity of reoviruses allows their application as single drugs targeting various types of cancers. It was confirmed that reoviruses can kill six human breast cancer cell lines (SK-BR-3, KPL4, MDA-MB-453, CRL1500, MCFT and MDA-MB-231) expressing HER-2, but not the breast cancer cell line Hs578Bst, which did not show HER-2 expression [71]. Favorable anticancer effects have also been demonstrated in immunocompetent mouse models in the presence of cyclosporine A or anti-CD4/anti-SD8 antibodies as immunosuppressant agents [72]. This shows the potential for reoviruses to be implemented for the treatment of a wide range of cancer types.

2.3.2. Clinical Trials of Reoviruses in Melanoma Treatment

Reovirus-based monotherapy has been conducted for solid tumors including soft-tissue sarcomas, melanoma, breast cancer and head and neck cancer [73]. One study included 18 patients and was mainly designed to verify the safety and tolerability of reovirus intralesional administration [74]. Monitoring was done over a period of six weeks. The toxic effects were measured according to the criteria of the National Cancer Institute Clinical Trials Group. The tumor responses were measured using the Response Evaluation Criteria in Solid Tumors. After the trial period of six weeks, one patient showed complete response (CR), two demonstrated partial responses (PRs), four patients had stable disease (SD) and ten showed progressive disease (PD) [74]. However, the results indicated that intralesional reovirus monotherapy was safe, well tolerated and did not reach dose-limiting toxicity. Local administration of reovirus in phase I/II malignant melanoma studies was as well tolerated as monotherapy [75][76]. Intravenous reovirus monotherapy was applied in a malignant melanoma phase II trial in 21 patients receiving a 3 × 1010 50% tissue-culture infective dose (TCID50) once every 60 min on days 1–5 every four weeks [77]. Clinical benefits as CR or PR were monitored for eight weeks. One patient demonstrated extensive tumor necrosis (75%–90%) in two metastatic lesions after two treatment cycles, whereas no other patient met the criteria for CR or PR [78]. Moreover, reovirus was detected in two out of 13 biopsies containing melanoma metastases. The findings from the phase II trial further support the positive clinical outcome obtained from phase I studies [79][80]. Furthermore, two out of 13 patients showed productive reoviral replication in melanoma metastases. Unfortunately, the trial could not progress as initially planned, as the clinical objective of having two or more patients reaching CR or PR was not achieved. According to these results, the phase II trial did not support the application of reovirus as a monotherapy for metastatic melanoma, but rather as part of a combination therapy with other therapeutic or chemotherapeutic agents [74].

2.4. Current Stage of Melanoma Treatment Using Coxsackievirus CVA21

CVA21, a member of the Picornaviridae family, is a nonenveloped ssRNA enterovirus enclosed in an icosahedral capsid. Two major subgroups, A and B, have been characterized in murine models [81]. Subgroup A contains 23 serotypes, with their main impact on skeletal muscles, while subgroup B contains six serotypes affecting a broad range of tissue types [79]. Their clinical significance in humans reflects their responsibility for mild upper respiratory tract infections spread by aerosol transmission [82]. The oncolytic CVA21 is commercially available as CAVATAK™ based on the wild-type Kuykendall strain [83]. Modelling of the attachment mechanisms and cell internalization has indicated that other group A serotypes may have similar oncolytic potential [84]. The CVA21 infection is characterized by attachment to the intracellular adhesion molecule-1 (ICAM-1), the primary receptor for attachment, and to the decay-accelerating factor (DAF), the secondary receptor for attachment [85]. ICAM-1 is a viral receptor common for the Picornaviridae family. Although DAF is expressed on almost all cells, its primary role is regulation of complement responses [85]. Since the attachment of CVA21 to DAF is not sufficient for host cell infection, DAF is considered to act as a membrane receptor, which accumulates the virus at the cell surface and optimizes viral entry via ICAM-1 [86]. The discovery of CVA21 and its lysis of cancer cells was mainly achieved through research conducted on ICAM-1 and DAF receptors, and comparisons between various cancer cell lines and non-malignant tissues. It has also been shown for melanoma as well as multiple myeloma, malignant glioma, breast, colon, endometrial and pancreatic cancer cell lines [87][88].

References

  1. Rastrelli, M.; Tropea, S.; Rossi, C.R.; Alaibac, M. Melanoma: Epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo 2014, 28, 1005–1011.
  2. Nakamura, Y. The role and necessity of sentinel lymph node biopsy for invasive melanoma. Front. Med. (Lausanne) 2019, 6.
  3. Ali, Z.; Yousaf, N.; Larkin, J. Melanoma epidemiology, biology and prognosis. EJC Suppl. 2013, 11, 81–91.
  4. Candido, S.; Rapisarda, V.; Marconi, A.; Malaponte, G.; Bevelacqua, V.; Gangemi, P.; Scalisi, A.; Mccubrey, J.A.; Maestro, R.; Spandidos, D.A.; et al. Analysis of the B-RAFV600E mutation in cutaneous melanoma patients with occupational sun exposure. Oncol. Rep. 2014, 31, 1079–1082.
  5. Bastian, B.C. The molecular pathology of melanoma: An integrated taxonomy of melanocytic neoplasia. Annu. Rev. Pathol. 2014, 9, 239–271.
  6. Curtin, J.A.; Patel, H.N.; Cho, K.-H.; LeBoit, P.E. Distinct sets of genetic alterations in melanoma. N. Engl. J. Med. 2005, 353, 2135–2147.
  7. Shain, A.H.; Bastian, B.C. From melanocytes to melanomas. Nat. Rev. Cancer 2016, 16, 345–358.
  8. Rossi, E.; Pagliara, M.M.; Orteschi, D.; Dosa, T.; Sammarco, M.G.; Caputo, C.G.; Petrone, G.; Rindi, G.; Zollino, M.; Blasi, M.A.; et al. Pembrolizumab as first-line treatment for metastatic uveal melanoma. Cancer Immunol. Immunother. 2019, 68, 1179–1185.
  9. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723.
  10. Hauschild, A.; Grob, J.-J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H.; Kaempgen, E.; et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012, 380, 358–365.
  11. McArthur, G.A.; Chapman, P.B.; Robert, C.; Larkin, J.; Haanen, J.B.; Dummer, R.; Ribas, A.; Hogg, D.; Hamid, O.; Ascierto, P.A.; et al. Safety and efficacy of vemurafenib in BRAFV600E and BRAFV600K mutation-positive melanoma (BRIM-3): Extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014, 15, 323–332.
  12. Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 2015, 372, 30–39.
  13. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; De Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.-J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451.
  14. Ribas, A.; Puzanov, I.; Dummer, R.; Schadendorf, D.; Hamid, O.; Robert, C.; Hodi, F.S.; Schachter, J.; Pavlick, A.C.; Lewis, K.D.; et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (Keynote-002): A randomised, controlled, phase 2 trial. Lancet Oncol. 2015, 16, 908–918.
  15. Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H.; Lao, C.D.; et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015, 16, 375–384.
  16. Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 2015, 372, 320–330.
  17. Malas, S.; Harrasser, M.; Lacy, K.E.; Karagiannis, S.N. Antibody therapies for melanoma: New and emerging opportunities to activate immunity (Review). Oncol. Rep. 2014, 32, 875–886.
  18. Wilson, M.A.; Schuchter, L.M. Chemotherapy for melanoma. In Melanoma; Kaufman, H.L., Mehnert, J.M., Eds.; Cancer Treatment and Research; Springer International Publishing: Cham, Switzerland, 2016; pp. 209–229. ISBN 978-3-319-22539-5.
  19. Kim, C.; Lee, C.W.; Kovacic, L.; Shah, A.; Klasa, R.; Savage, K.J. Long-term survival in patients with metastatic melanoma treated with DTIC or temozolomide. Oncologist 2010, 15, 765–771.
  20. Home—ClinicalTrials.gov. Available online: (accessed on 27 December 2019).
  21. Testori, A.; Ribero, S.; Bataille, V. Diagnosis and treatment of in-transit melanoma metastases. Eur. J. Surg. Oncol. 2017, 43, 544–560.
  22. Miklavčič, D.; Serša, G.; Brecelj, E.; Gehl, J.; Soden, D.; Bianchi, G.; Ruggieri, P.; Rossi, C.R.; Campana, L.G.; Jarm, T. Electrochemotherapy: Technological advancements for efficient electroporation-based treatment of internal tumors. Med. Biol. Eng. Comput. 2012, 50, 1213–1225.
  23. Matthiessen, L.W.; Chalmers, R.L.; Sainsbury, D.C.G.; Veeramani, S.; Kessell, G.; Humphreys, A.C.; Bond, J.E.; Muir, T.; Gehl, J. Management of cutaneous metastases using electrochemotherapy. Acta Oncol. 2011, 50, 621–629.
  24. Austin, E.; Mamalis, A.; Ho, D.; Jagdeo, J. Laser and light-based therapy for cutaneous and soft-tissue metastases of malignant melanoma: A systematic review. Arch. Dermatol. Res. 2017, 309, 229–242.
  25. Yin, R.; Wang, M.; Huang, Y.-Y.; Huang, H.-C.; Avci, P.; Chiang, L.Y.; Hamblin, M.R. Photodynamic therapy with decacationic [60]fullerene monoadducts: Effect of a light absorbing electron-donor antenna and micellar formulation. Nanomedicine 2014, 10, 795–808.
  26. Dougherty, T.J.; Gomer, C.J.; Henderson, B.W.; Jori, G.; Kessel, D.; Korbelik, M.; Moan, J.; Peng, Q. Photodynamic therapy. J. Natl. Cancer Inst. 1998, 90, 889–905.
  27. Brown, S.B.; Brown, E.A.; Walker, I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 2004, 5, 497–508.
  28. Longo, J.; Muehlmann, L.; Almeida-Santos, M.; Azevedo, R. Preventing metastasis by targeting lymphatic vessels with photodynamic therapy based on nanostructured photosensitizers. J. Nanomed. Nanotechnol. 2015, 6, 5.
  29. Baldea, I.; Filip, A.G. Photodynamic therapy in melanoma—An update. J. Physiol. Pharmacol. 2012, 63, 109–118.
  30. Huang, Y.-Y.; Vecchio, D.; Avci, P.; Yin, R.; Garcia-Diaz, M.; Hamblin, M.R. Melanoma resistance to photodynamic therapy: New insights. Biol. Chem. 2013, 394, 239–250.
  31. Biteghe, F.N.; Davids, L. A combination of photodynamic therapy and chemotherapy displays a differential cytotoxic effect on human metastatic melanoma cells. J. Photochem. Photobiol. B Biol. 2017, 166, 18–27.
  32. Lundstrom, K. Oncolytic alphaviruses in cancer immunotherapy. Vaccines (Basel) 2017, 5, 9.
  33. Stone, D. Novel viral vector systems for gene therapy. Viruses 2010, 2, 1002–1007.
  34. Brenner, M.K.; Rill, D.R.; Krance, R.A.; Ihle, J.N.; Moen, R.C.; Mirro, J.; Anderson, W.F. Gene-marking to trace origin of relapse after autologous bone-marrow transplantation. Lancet 1993, 341, 85–86.
  35. Conry, R.M.; Westbrook, B.; McKee, S.; Norwood, T.G. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum. Vaccin. Immunother. 2018, 14, 839–846.
  36. Lundstrom, K. Self-replicating RNA viruses for RNA therapeutics. Molecules 2018, 23, 3310.
  37. Roberts, M.S.; Groene, W.S.; Lorence, R.M.; Bamat, M.K. Naturally occurring viruses for the treatment of cancer. Discov. Med. 2009, 6, 217–222.
  38. Prestwich, R.J.; Harrington, K.J.; Pandha, H.S.; Vile, R.G.; Melcher, A.A.; Errington, F. Oncolytic viruses: A novel form of immunotherapy. Expert Rev. Anticancer Ther. 2008, 8, 1581–1588.
  39. Chiocca, E.; Rabkin, S. Oncolytic viruses and their application to cancer immunotherapy. Cancer Immunol. Res. 2014, 2, 295–300.
  40. Aurelian, L. Oncolytic viruses as immunotherapy: Progress and remaining challenges. Onco Targets Ther. 2016, 9, 2627–2637.
  41. Kelly, E.; Russell, S.J. History of oncolytic viruses: Genesis to genetic engineering. Mol. Ther. 2007, 15, 651–659.
  42. Jhawar, S.R.; Thandoni, A.; Bommareddy, P.K.; Hassan, S.; Kohlhapp, F.J.; Goyal, S.; Schenkel, J.M.; Silk, A.W.; Zloza, A. Oncolytic viruses—Natural and genetically engineered cancer immunotherapies. Front. Oncol. 2017, 7.
  43. Bommareddy, P.K.; Patel, A.; Hossain, S.; Kaufman, H.L. Talimogene laherparepvec (T-VEC) and other oncolytic viruses for the treatment of melanoma. Am. J. Clin. Dermatol. 2017, 18, 1–15.
  44. Coffin, R.S.; MacLean, A.R.; Latchman, D.S.; Brown, S.M. Gene delivery to the central and peripheral nervous systems of mice using HSV1 ICP34.5 deletion mutant vectors. Gene Ther. 1996, 3, 886–891.
  45. Huang, T.T.; Parab, S.; Burnett, R.; Diago, O.; Ostertag, D.; Hofman, F.M.; Espinoza, F.L.; Martin, B.; Ibañez, C.E.; Kasahara, N.; et al. Intravenous administration of retroviral replicating vector, Toca 511, demonstrates therapeutic efficacy in orthotopic immune-competent mouse glioma model. Hum. Gene Ther. 2015, 26, 82–93.
  46. Wakimoto, H.; Yoshida, Y.; Ayoyagi, M.; Hirakawa, K.; Hamada, H. Efficient retrovirus-mediated cytokine-gene transduction of primary-cultured human glioma cells for tumor vaccination therapy. Jpn. J. Cancer Res. 1997, 88, 296–305.
  47. Wang, Y.; Jin, T.; Dai, X.; Xu, I. Lentivirus-mediated knockdown of CEP55 suppressed cell proliferation of breast cancer cells. Biosc. Trends 2016, 10, 67–73.
  48. Li, R.; Yang, H.-Q.; Xi, H.-L.; Feng, S.; Qin, R.-H. Inhibition of CDH17 gene expression via RNA interference reduces proliferation and apoptosis of human MKN28 gastric cancer cells. Int. J. Oncol. 2017, 50, 15–22.
  49. Pan, Z.Y.; Yang, Y.; Pan, H.; Zhang, J.; Liu, H.; Yang, Y.; Haung, G.; Yin, L.; Huang, J.; Zhou, W.P. Lentivirus-mediated TDP52L2 depletion inhibits the proliferation of liver cancer cells in vitro. Int. J. Clin. Exp. Med. 2015, 8, 233–241.
  50. Ravet, E.; Lulka, H.; Gross, F.; Casteilla, L.; Buscail, L.; Cordelier, P. Using lentiviral vectors for efficient pancreatic cancer gene therapy. Cancer Gene Ther. 2010, 17, 315–324.
  51. Tian, L.; Liu, J.; Xia, G.-H.; Chen, B.-A. RNAi-mediated knockdown of MCM7 gene on CML cells and its therapeutic potential for leukemia. Med. Oncol. 2017, 34, 21.
  52. Scherr, M.; Eder, M. Gene transfer into hematopoietic stem cells using lentiviral vectors. Curr. Gene Ther. 2002, 2, 45–55.
  53. Cavazzana-Calvo, M.; Hacein-Bey, S.; De Saint Basile, G.; Gross, F.; Yvon, E.; Nusbaum, P.; Selz, F.; Hue, C.; Certain, S.; Casanova, J.-L.; et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 2000, 288, 669–672.
  54. Bastide, C.; Maroc, N.; Bladou, F.; Hassoun, J.; Maitland, N.; Mannoni, P.; Bagnis, C. Expression of a model gene in prostate cancer cells lentivirally transduced in vitro and in vivo. Prostate Cancer Prostatic Dis. 2003, 6, 228–234.
  55. Liu, S.; Song, W.; Liu, F.; Zhang, J.; Zhu, S. Antitumor efficacy of VP22-CD/5-FC suicide gene system mediated by lentivirus in a murine uveal melanoma model. Exp. Eye Res. 2018, 172, 144–151.
  56. Lei, Y.; Zhang, B.; Zhang, Y.; Zhao, Y.; Sun, J.; Zhang, X.; Yang, S. Lentivirus-mediated downregulation of MAT2B inhibits cell proliferation and induces apoptosis in melanoma. Int. J. Oncol. 2016, 49, 981–990.
  57. Varas-Godoy, M.; Lladser, A.; Farfan, N.; Villota, C.; Villegas, J.; Tapia, J.C.; Burzio, L.O.; Burzio, V.A.; Valenzuela, P.D.T. In vivo knockdown of antisense non-coding mitochondrial RNAs by a lentiviral-encoded shRNA inhibits melanoma tumor growth and lung colonization. Pigment Cell Melanoma Res. 2018, 31, 64–72.
  58. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517.
  59. Sundarasetty, B.S.; Chan, L.; Darling, D.; Giunti, G.; Frazaneh, F.; Schemck, F.; Naundorf, S.; Kuehlcke, K.; Ruggiero, E.; Schmidt, M.; et al. Lentivirus-induced “Smart”dendritic cells: Pharmacodynamics and GMP-compliant production for immunotherapy against TRP2-positive melanoma. Gene Ther. 2015, 22, 707–720.
  60. Simon, B.; Harrer, D.C.; Thirion, C.; Schuler-Thurner, B.; Schuler, G.; Uslu, U. Enhancing lentiviral transduction to generate melanoma-specific human T cells for cancer immunotherapy. J. Immunol. Methods 2019, 472, 55–64.
  61. Milotou, A.N.; Papadopoulou, L.C. CAR T-cell therapy: A new era in cancer immunotherapy. Curr. Pharm. Biotechnol. 2018, 19, 5–18.
  62. Simon, B.; Uslu, U. CAR-T cell therapy in melanoma: A future success story? Exp. Dermatol. 2018, 27, 1315–1321.
  63. Bodkin, D.K.; Nibert, M.L.; Fields, B.N. Proteolytic digestion of reovirus in the intestinal lumens of neonatal mice. J. Virol. 1989, 63, 4676–4681.
  64. Strong, J.E.; Coffey, M.C.; Tang, D.; Sabinin, P.; Lee, P.W. The molecular basis of viral oncolysis: Usurpation of the Ras signaling pathway by reovirus. EMBO J. 1998, 17, 3351–3362.
  65. Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.-K.; Attar, N.; Sazegar, H.; et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977.
  66. Lundstrom, K. Viral vectors in gene therapy. Diseases 2018, 6, 42.
  67. Errington, F.; White, C.L.; Twigger, K.R.; Rose, A.; Scott, K.; Steele, L.; Ilett, L.J.; Prestwich, R.; Pandha, H.S.; Coffey, M.; et al. Inflammatory tumour cell killing by oncolytic reovirus for the treatment of melanoma. Gene Ther. 2008, 15, 1257.
  68. Hashiro, G.; Loh, P.C.; Yau, J.T. The preferential cytotoxicity of reovirus for certain transformed cell lines. Arch. Virol. 1977, 54, 307–315.
  69. Coffey, M.C.; Strong, J.E.; Forsyth, P.A.; Lee, P.W.K. Reovirus therapy of tumors with activated Ras pathway. Science 1998, 282, 1332–1334.
  70. Hata, Y.; Etoh, T.; Inomata, M.; Shiraishi, N.; Nishizono, A.; Kitano, S. Efficacy of oncolytic reovirus against human breast cancer cells. Oncol. Rep. 2008, 19, 1395–1398.
  71. Twigger, K.; Vidal, L.; White, C.L.; Bono, J.S.D.; Bhide, S.; Coffey, M.; Thompson, B.; Vile, R.G.; Heinemann, L.; Pandha, H.S.; et al. Enhanced in vitro and in vivo cytotoxicity of combined reovirus and radiotherapy. Clin. Cancer Res. 2008, 14, 912–923.
  72. Morris, D.G.; Feng, X.; DiFrancesco, L.M.; Fonseca, K.; Forsyth, P.A.; Paterson, A.H.; Coffey, M.C.; Thompson, B. REO-001: A phase I trial of percutaneous intralesional administration of reovirus type 3 dearing (Reolysin®) in patients with advanced solid tumors. Investig. New Drugs 2013, 31, 696–706.
  73. Clements, D.; Helson, E.; Gujar, S.A.; Lee, P.W. Reovirus in cancer therapy: An evidence-based review. Oncolytic Virother. 2014, 3, 69–82.
  74. Forsyth, P.; Roldán, G.; George, D.; Wallace, C.; Palmer, C.A.; Morris, D.; Cairncross, G.; Matthews, M.V.; Markert, J.; Gillespie, Y.; et al. A phase I trial of intratumoral administration of reovirus in patients with histologically confirmed recurrent malignant gliomas. Mol. Ther. 2008, 16, 627–632.
  75. Kicielinski, K.P.; Chiocca, E.A.; Yu, J.S.; Gill, G.M.; Coffey, M.; Markert, J.M. Phase 1 clinical trial of intratumoral reovirus infusion for the treatment of recurrent malignant gliomas in adults. Mol. Ther. 2014, 22, 1056–1062.
  76. Gollamudi, R.; Ghalib, M.H.; Desai, K.K.; Chaudhary, I.; Wong, B.; Einstein, M.; Coffey, M.; Gill, G.M.; Mettinger, K.; Mariadason, J.M.; et al. Intravenous administration of Reolysin®, a live replication competent RNA virus is safe in patients with advanced solid tumors. Investig. New Drugs 2010, 28, 641–649.
  77. Galanis, E.; Markovic, S.N.; Suman, V.J.; Nuovo, G.J.; Vile, R.G.; Kottke, T.J.; Nevala, W.K.; Thompson, M.A.; Lewis, J.E.; Rumilla, K.M.; et al. Phase II trial of intravenous administration of Reolysin® (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma. Mol. Ther. 2012, 20, 1998.
  78. Silk, A.W.; Kaufman, H.; Gabrail, N.; Mehnert, J.; Bryan, J.; Norrell, J.; Medina, D.; Bommareddy, P.; Shafren, D.; Grose, M.; et al. Abstract CT026: Phase 1b study of intratumoral Coxsackievirus A21 (CVA21) and systemic pembrolizumab in advanced melanoma patients: Interim results of the CAPRA clinical trial. Cancer Res. 2017, 77, CT026.
  79. Vidal, L.; Pandha, H.S.; Yap, T.A.; White, C.L.; Twigger, K.; Vile, R.G.; Melcher, A.; Coffey, M.; Harrington, K.J.; DeBono, J.S. A phase I study of intravenous oncolytic reovirus type 3 dearing in patients with advanced cancer. Clin. Cancer Res. 2008, 14, 7127–7137.
  80. Ali, S.; Kaur, J.; Patel, K.D. Intercellular cell adhesion molecule-1, vascular cell adhesion molecule-1, and regulated on activation normal T cell expressed and secreted are expressed by human breast carcinoma cells and support eosinophil adhesion and activation. Am. J. Pathol. 2000, 157, 313–321.
  81. Au, G.G.; Beagley, L.G.; Haley, E.S.; Barry, R.D.; Shafren, D.R. Oncolysis of malignant human melanoma tumors by Coxsackieviruses A13, A15 and A18. Virol. J. 2011, 8, 22.
  82. Newcombe, N.G.; Beagley, L.G.; Christiansen, D.; Loveland, B.E.; Johansson, E.S.; Beagley, K.W.; Barry, R.D.; Shafren, D.R. Novel role for decay-accelerating factor in Coxsackievirus A21-mediated cell infectivity. J. Virol. 2004, 78, 12677–12682.
  83. Annels, N.E.; Mansfield, D.; Arif, M.; Ballesteros-Merino, C.; Simpson, G.R.; Denyer, M.; Sandhu, S.S.; Melcher, A.A.; Harrington, K.J.; Davies, B.; et al. Phase I trial of an ICAM-1-targeted immunotherapeutic-Coxsackievirus A21 (CVA21) as an oncolytic agent against non muscle-invasive bladder cancer. Clin. Cancer Res. 2019, 25, 5818–5831.
  84. Shafren, D.R.; Dorahy, D.J.; Ingham, R.A.; Burns, G.F.; Barry, R.D. Coxsackievirus A21 binds to decay-accelerating factor but requires intercellular adhesion molecule 1 for cell entry. J. Virol. 1997, 71, 4736–4743.
  85. Johansson, E.S.; Xing, L.; Cheng, R.H.; Shafren, D.R. Enhanced cellular receptor usage by a bioselected variant of Coxsackievirus A21. J. Virol. 2004, 78, 12603–12612.
  86. Kageshita, T.; Ono, T. Clinical relevance of ICAM-1 expression in primary lesions and serum of patients with malignant melanoma. J. Dermatol. Sci. 1993, 6, 11.
  87. Komi, J.; Lassila, O. Toremifene increases the expression of intercellular adhesion molecule-1 (ICAM-1) on MCF-7 breast cancer cells and jurkat cells. Scand. J. Immunol. 2000, 51, 73–78.
  88. Au, G.G.; Lincz, L.F.; Enno, A.; Shafren, D.R. Oncolytic Coxsackievirus A21 as a novel therapy for multiple myeloma. Br. J. Haematol. 2007, 137, 133–141.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 433
Revisions: 2 times (View History)
Update Date: 30 Jun 2021
1000/1000