Generic placeholder image

Current Cancer Therapy Reviews

Editor-in-Chief

ISSN (Print): 1573-3947
ISSN (Online): 1875-6301

Review Article

The Important Role of Oncolytic Viruses in Common Cancer Treatments

Author(s): Amir Mohamadi, Gilles Pagès and Mohammad S. Hashemzadeh*

Volume 16, Issue 4, 2020

Page: [292 - 305] Pages: 14

DOI: 10.2174/1573394716666200211120906

Price: $65

Abstract

Oncolytic viruses (OV) are considered as promising tools in cancer treatment. In addition to direct cytolysis, the stimulation of both innate and adaptive immune responses is the most important mechanism in oncolytic virotherapy that finally leads to the long-standing tumor retardations in the advanced melanoma clinical trials. The OVs have become a worthy method in cancer treatment, due to their several biological advantages including (1) the selective replication in cancer cells without affecting normal cells; (2) the lack of resistance to the treatment; (3) cancer stem cell targeting; (4) the ability to be spread; and (5) the immune response induction against the tumors. Numerous types of viruses; for example, Herpes simplex viruses, Adenoviruses, Reoviruses, Poliovirus, and Newcastle disease virus have been studied as a possible cancer treatment strategy. Although some viruses have a natural orientation or tropism to cancer cells, several others need attenuation and genetic manipulation to increase the safety and tumor-specific replication activity. Two important mechanisms are involved in OV antitumor responses, which include the tumor cell death due to virus replication, and also induction of immunogenic cell death as a result of the immune system responses against the tumor cells. Furthermore, the high efficiency of OV on antitumor immune response stimulation can finally lead to a significant tumor shrinkage.

Keywords: Oncolytic virus, virotherapy, vaccinia virus, reovirus, adenovirus, vaccinia virus, measles virus, poliovirus, herpes simplex virus, cancer treatment.

Graphical Abstract
[1]
Zamarin D, Holmgaard RB, Ricca J, et al. Intratumoral modulation of the inducible co-stimulator ICOS by recombinant oncolytic virus promotes systemic anti-tumour immunity. Nat Commun 2017; 8: 14340.
[http://dx.doi.org/10.1038/ncomms14340] [PMID: 28194010]
[2]
Pol J, Kroemer G, Galluzzi L. First oncolytic virus approved for melanoma immunotherapy. OncoImmunology 2015; 5(1)e1115641
[http://dx.doi.org/10.1080/2162402X.2015.1115641] [PMID: 26942095]
[3]
Brown MC, Dobrikova EY, Dobrikov MI, et al. Oncolytic polio virotherapy of cancer. Cancer 2014; 120(21): 3277-86.
[http://dx.doi.org/10.1002/cncr.28862] [PMID: 24939611]
[4]
Conry RM, Westbrook B, McKee S, Norwood TG. Talimogene laherparepvec: First in class oncolytic virotherapy. Hum Vaccin Immunother 2018; 14(4): 839-46.
[http://dx.doi.org/10.1080/21645515.2017.1412896] [PMID: 29420123]
[5]
Fernandes MS, Gomes EM, Butcher LD, et al. Growth inhibition of human multiple myeloma cells by an oncolytic adenovirus carrying the CD40 ligand transgene. Clin Cancer Res 2009; 15(15): 4847-56.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-0451] [PMID: 19622582]
[6]
Oliva S, Gambella M, Boccadoro M, Bringhen S. Systemic virotherapy for multiple myeloma. Expert Opin Biol Ther 2017; 17(11): 1375-87.
[PMID: 28796556]
[7]
Bartee E. Potential of oncolytic viruses in the treatment of multiple myeloma. Oncolytic Virother 2018; 7: 1-12.
[http://dx.doi.org/10.2147/OV.S136644] [PMID: 29503813]
[8]
Calton CM, Kelly KR, Anwer F, Carew JS, Nawrocki ST. Oncolytic viruses for multiple myeloma therapy. Cancers (Basel) 2018; 10(6)E198
[http://dx.doi.org/10.3390/cancers10060198] [PMID: 29903988]
[9]
Hummel HD, Kuntz G, Russell SJ, et al. Genetically engineered attenuated measles virus specifically infects and kills primary multiple myeloma cells. J Gen Virol 2009; 90(Pt 3): 693-701.
[http://dx.doi.org/10.1099/vir.0.007302-0] [PMID: 19218216]
[10]
Li W, Turaga RC, Li X, et al. Overexpression of Smac by an armed vesicular stomatitis virus overcomes tumor resistance. Mol Ther Oncolytics 2019; 14: 188-95.
[http://dx.doi.org/10.1016/j.omto.2019.05.006]
[11]
Leveille S, Samuel S, Goulet ML, Hiscott J. Enhancing VSV oncolytic activity with an improved cytosine deaminase suicide gene strategy. Cancer Gene Ther 2011; 18(6): 435-43.
[http://dx.doi.org/10.1038/cgt.2011.14] [PMID: 21394109]
[12]
Stewart JH IV, Ahmed M, Northrup SA, Willingham M, Lyles DS. Vesicular stomatitis virus as a treatment for colorectal cancer. Cancer Gene Ther 2011; 18(12): 837-49.
[http://dx.doi.org/10.1038/cgt.2011.49] [PMID: 21886191]
[13]
Lee AJ, Kim S-G. Selective purging of human multiple myeloma cells from peripheral blood mononuclear cells: A preliminary study. J Blood Med 2019; 10: 105-9.
[http://dx.doi.org/10.2147/JBM.S193467] [PMID: 31043801]
[14]
Kelly KR, Espitia CM, Zhao W, et al. Oncolytic reovirus sensitizes multiple myeloma cells to anti-PD-L1 therapy. Leukemia 2018; 32(1): 230-3.
[http://dx.doi.org/10.1038/leu.2017.272] [PMID: 28832023]
[15]
Lei W, Wang S, Yang C, et al. Combined expression of miR-34a and Smac mediated by oncolytic vaccinia virus synergistically promote anti-tumor effects in Multiple Myeloma. Sci Rep 2016; 6: 32174.
[http://dx.doi.org/10.1038/srep32174] [PMID: 27552933]
[16]
Rahman MM, Bagdassarian E, Ali MAM, McFadden G. Identification of host DEAD-box RNA helicases that regulate cellular tropism of oncolytic Myxoma virus in human cancer cells. Sci Rep 2017; 7(1): 15710.
[http://dx.doi.org/10.1038/s41598-017-15941-1] [PMID: 29146961]
[17]
Bartee MY, Dunlap KM, Bartee E. Myxoma virus induces ligand independent extrinsic apoptosis in human myeloma cells. Clin Lymphoma Myeloma Leuk 2016; 16(4): 203-12.
[http://dx.doi.org/10.1016/j.clml.2015.12.005] [PMID: 26803534]
[18]
Eissa IR, Bustos-Villalobos I, Ichinose T, et al. The current status and future prospects of oncolytic viruses in clinical trials against melanoma, glioma, pancreatic, and breast cancers. Cancers (Basel) 2018; 10(10)E356
[http://dx.doi.org/10.3390/cancers10100356] [PMID: 30261620]
[19]
Martikainen M, Essand M. Virus-based immunotherapy of glioblastoma. Cancers (Basel) 2019; 11(2)E186
[http://dx.doi.org/10.3390/cancers11020186] [PMID: 30764570]
[20]
Muscolini M, Castiello L, Palermo E, et al. SIRT1 modulates the sensitivity of prostate cancer cells to VSV oncolysis. J Virol 2019; 93(15)e00626-19
[http://dx.doi.org/10.1128/JVI.00626-19]
[21]
Foreman PM, Friedman GK, Cassady KA, Markert JM. Oncolytic Virotherapy for the treatment of malignant glioma. Neurotherapeutics 2017; 14(2): 333-44.
[http://dx.doi.org/10.1007/s13311-017-0516-0] [PMID: 28265902]
[22]
Varela-Guruceaga M, Tejada-Solís S, García-Moure M, et al. Oncolytic viruses as therapeutic tools for pediatric brain tumors. Cancers (Basel) 2018; 10(7)E226
[http://dx.doi.org/10.3390/cancers10070226] [PMID: 29987215]
[23]
Lun X, Yang W, Alain T, et al. Myxoma virus is a novel oncolytic virus with significant antitumor activity against experimental human gliomas. Cancer Res 2005; 65(21): 9982-90.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-1201] [PMID: 16267023]
[24]
Barrett JW, McFadden G. Myxoma virus mutants for cancer treatment. US Patent US9980994B22018.
[25]
Eissa IR, Naoe Y, Bustos-Villalobos I, et al. Genomic signature of the natural oncolytic herpes simplex virus HF10 and its therapeutic role in preclinical and clinical trials. Front Oncol 2017; 7: 149.
[http://dx.doi.org/10.3389/fonc.2017.00149] [PMID: 28770166]
[26]
Faghfuri E, Faramarzi MA, Nikfar S, Abdollahi M. Nivolumab and pembrolizumab as immune-modulating monoclonal antibodies tar-geting the PD-1 receptor to treat melanoma. Expert Rev Anticancer Ther 2015; 15(9): 981-93.
[http://dx.doi.org/10.1586/14737140.2015.1074862] [PMID: 26313415]
[27]
Streby KA, Geller JI, Currier MA, et al. Intratumoral injection of HSV1716, an oncolytic herpes virus, is safe and shows evidence of immune response and viral replication in young cancer patients. Clin Cancer Res 2017; 23(14): 3566-74.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-2900] [PMID: 28495911]
[28]
Aghi M, Visted T, Depinho RA, Chiocca EA. Oncolytic herpes virus with defective ICP6 specifically replicates in quiescent cells with homozygous genetic mutations in p16. Oncogene 2008; 27(30): 4249-54.
[http://dx.doi.org/10.1038/onc.2008.53] [PMID: 18345032]
[29]
Friedman GK, Nan L, Haas MC, et al. gamma(1)34.5-deleted HSV-1-expressing human cytomegalovirus IRS1 gene kills human glioblastoma cells as efficiently as wild-type HSV-1 in normoxia or hypoxia. Gene Ther 2015; 22(4): 348-55.
[http://dx.doi.org/10.1038/gt.2014.107] [PMID: 25427614]
[30]
Hashemzadeh M, Ayoubi S, Naimipoor F, et al. Detection of cytomegalovirus in human cells. IJBPAS 2015; 4: 7.
[31]
Kim JW, Chang AL, Kane JR, Young JS, Qiao J, Lesniak MS. Gene therapy and virotherapy of gliomas. Prog Neurol Surg 2018; 32: 112-23.
[http://dx.doi.org/10.1159/000469685] [PMID: 29990979]
[32]
Friedman GK, Langford CP, Coleman JM, et al. Engineered herpes simplex viruses efficiently infect and kill CD133+ human glioma xenograft cells that express CD111. J Neurooncol 2009; 95(2): 199-209.
[http://dx.doi.org/10.1007/s11060-009-9926-0] [PMID: 19521665]
[33]
Friedman GK, Bernstock JD, Chen D, et al. Enhanced sensitivity of patient-derived pediatric high-grade brain tumor xenografts to on-colytic HSV-1 virotherapy correlates with Nectin-1 expression. Sci Rep 2018; 8(1): 13930.
[http://dx.doi.org/10.1038/s41598-018-32353-x] [PMID: 30224769]
[34]
Markert JM, Liechty PG, Wang W, et al. Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol 2009; 17(1): 199-207.
[http://dx.doi.org/10.1038/mt.2008.228]
[35]
Markert JM, Razdan SN, Kuo HC, et al. A phase 1 trial of oncolytic HSV-1, G207, given in combination with radiation for recurrent GBM demonstrates safety and radiographic responses. Mol Ther 2014; 22(5): 1048-55.
[36]
Lang FF, Conrad C, Gomez-Manzano C, et al. Phase I study of DNX-2401 (Delta-24-RGD) oncolytic adenovirus: Replication and immunotherapeutic effects in recurrent malignant glioma. J Clin Oncol 2018; 36(14): 1419-27.
[http://dx.doi.org/10.1200/JCO.2017.75.8219] [PMID: 29432077]
[37]
Martínez-Vélez N, Garcia-Moure M, Marigil M, et al. The oncolytic virus Delta-24-RGD elicits an antitumor effect in pediatric glioma and DIPG mouse models 2019; 10(1)2235
[http://dx.doi.org/10.1038/s41467-019-10043-0]
[38]
Niemann J, Kühnel F. Oncolytic viruses: Adenoviruses. Virus Genes 2017; 53(5): 700-6.
[http://dx.doi.org/10.1007/s11262-017-1488-1] [PMID: 28702840]
[39]
Gil-Hoyos R, Miguel-Camacho J, Alemany R. Oncolytic adenovirus characterization: Activity and immune responses. Methods Mol Biol 2014; 1089: 117-32.
[http://dx.doi.org/10.1007/978-1-62703-679-5_9] [PMID: 24132482]
[40]
Kemp V, Hoeben RC, van den Wollenberg DJ. Exploring reovirus plasticity for improving its use as oncolytic virus. Viruses 2015; 8(1)E4
[http://dx.doi.org/10.3390/v8010004] [PMID: 26712782]
[41]
Ou W, Marino MP, Suzuki A, et al. Specific targeting of human interleukin (IL)-13 receptor α2-positive cells with lentiviral vectors displaying IL-13. Hum Gene Ther Methods 2012; 23(2): 137-47.
[http://dx.doi.org/10.1089/hgtb.2012.054] [PMID: 22612657]
[42]
Gao J, Zheng Q, Xin N, Wang W, Zhao C. CD155, an onco-immunologic molecule in human tumors. Cancer Sci 2017; 108(10): 1934-8.
[http://dx.doi.org/10.1111/cas.13324] [PMID: 28730595]
[43]
Jammal MP, Michelin MA, Nomelini RS, Murta EFC. Recombinant poliovirus for cancer immunotherapy. Ann Transl Med 2018; 6(18): 368.
[http://dx.doi.org/10.21037/atm.2018.07.19] [PMID: 30370295]
[44]
Tayeb S, Zakay-Rones Z, Panet A. Therapeutic potential of oncolytic Newcastle disease virus: a critical review. Oncolytic Virother 2015; 4: 49-62.
[PMID: 27512670]
[45]
Raihan J, Ahmad U, Yong YK, Eshak Z, Othman F, Ideris A. Regression of solid breast tumours in mice by Newcastle disease virus is associated with production of apoptosis related-cytokines. BMC Cancer 2019; 19(1): 315.
[http://dx.doi.org/10.1186/s12885-019-5516-5] [PMID: 30947706]
[46]
Shafaati M, Hashemzadeh M, Dorostkar R. Cloning of fusion protein gene of Newcastle disease virus into a baculovirus derived bacmid shuttle vector, in order to express it in insect cell line. J Arak Univ Med Sci (AMUJ) 2015; 18: 80-9.
[47]
Wagner S, Csatary CM, Gosztonyi G, et al. Combined treatment of pediatric high-grade glioma with the oncolytic viral strain MTH-68/H and oral valproic acid. APMIS 2006; 114(10): 731-43.
[48]
Freeman AI, Zakay-Rones Z, Gomori JM, et al. Phase I/II trial of intravenous NDV-HUJ oncolytic virus in recurrent glioblastoma multiforme. Mol Ther 2006; 13(1): 221-8.
[49]
Payandeh Z, Yarahmadi M, Nariman-Saleh-Fam Z, et al. Immune therapy of melanoma: Overview of therapeutic vaccines. J Cell Physiol 2019. Epub ahead of print
[http://dx.doi.org/10.1002/jcp.28181] [PMID: 30706472]
[50]
Puzanov I, Milhem MM, Minor D, et al. Talimogene laherparepvec in combination with ipilimumab in previously untreated, unresectable stage IIIB-IV melanoma. J Clin Oncol 2016; 34(22): 2619-26.
[http://dx.doi.org/10.1200/JCO.2016.67.1529] [PMID: 27298410]
[51]
Chesney J, Puzanov I, Collichio F, et al. Randomized, open-label phase II study evaluating the efficacy and safety of talimogene la-herparepvec in combination with ipilimumab versus ipilimumab alone in patients with advanced, unresectable melanoma. J Clin Oncol 2018; 36(17): 1658-67.
[http://dx.doi.org/10.1200/JCO.2017.73.7379] [PMID: 28981385]
[52]
Ferris RL, Gross ND, Nemunaitis JJ, et al. Phase I trial of intratumoral therapy using HF10, an oncolytic HSV-1, demonstrates safety in HSV+/HSV- patients with refractory and superficial cancers 2014; 32(15)(_suppl.).6082
[53]
Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother (Hagerstown, Md: 1997) 2010; 33(8): 823-33.
[54]
Andtbacka RHI, Ross MI, Agarwala SS, et al. Preliminary results from phase II study of combination treatment with HF10, a replication-competent HSV-1 oncolytic virus, and ipilimumab in patients with stage IIIb, IIIc, or IV unresectable or metastatic melanoma. J Clin Oncol 2016; 34(15)(_suppl.).9543
[55]
Andtbacka RHI, Ross MI, Agarwala SS, et al. Preliminary results from phase II study of combination treatment with HF10, a replication-competent HSV-1 oncolytic virus, and ipilimumab in patients with stage IIIb, IIIc, or IV unresectable or metastatic melanoma. J Clin Oncol 2016; 34(15)(Suppl.): 9543.
[http://dx.doi.org/10.1200/JCO.2016.34.15_suppl.9543]
[56]
Zou L, Yi L, Song Y, et al. A cluster of coxsackievirus A21 associated acute respiratory illness: The evidence of efficient transmission of CVA21. Arch Virol 2017; 162(4): 1057-9.
[http://dx.doi.org/10.1007/s00705-016-3201-4] [PMID: 28025712]
[57]
Dharmadhikari N, Mehnert JM, Kaufman HL. Oncolytic virus immunotherapy for melanoma. Curr Treat Options Oncol 2015; 16(3): 326.
[http://dx.doi.org/10.1007/s11864-014-0326-0] [PMID: 25777572]
[58]
Deng L, Fan J, Ding Y, et al. Oncolytic cancer therapy with a vaccinia virus strain. Oncol Rep 2019; 41(1): 686-92.
[PMID: 30365140]
[59]
Passaro C, Alayo Q, De Laura I, et al. Arming an oncolytic herpes simplex virus type 1 with a single-chain fragment variable antibody against PD-1 for experimental glioblastoma therapy. Clin Cancer Res 2019; 25(1): 290-9.
[http://dx.doi.org/10.1158/1078-0432.CCR-18-2311] [PMID: 30279232]
[60]
Falasca M, Kim M, Casari I. Pancreatic cancer: Current research and future directions. Biochim Biophys Acta 2016; 1865(2): 123-32.
[PMID: 26794394]
[61]
Sahin IH, Askan G, Hu ZI, O’Reilly EM. Immunotherapy in pancreatic ductal adenocarcinoma: an emerging entity? Ann Oncol 2017; 28(12): 2950-61.
[http://dx.doi.org/10.1093/annonc/mdx503] [PMID: 28945842]
[62]
Wang Y, Camateros P, Cheung WY. A real-world comparison of FOLFIRINOX, Gemcitabine Plus nab-Paclitaxel, and Gemcitabine in advanced pancreatic cancers. J Gastrointest Cancer 2019; 50(1): 62-8.
[http://dx.doi.org/10.1007/s12029-017-0028-5] [PMID: 29143916]
[63]
Cheng PH, Wechman SL, McMasters KM, Zhou HS. Oncolytic replication of E1b-deleted adenoviruses. Viruses 2015; 7(11): 5767-79.
[http://dx.doi.org/10.3390/v7112905] [PMID: 26561828]
[64]
Mulvihill S, Warren R, Venook A, et al. Safety and feasibility of injection with an E1B-55 kDa gene-deleted, replication-selective adenovirus (ONYX-015) into primary carcinomas of the pancreas: a phase I trial. Gene Ther 2001; 8(4): 308-15.
[http://dx.doi.org/10.1038/sj.gt.3301398] [PMID: 11313805]
[65]
Hecht JR, Bedford R, Abbruzzese JL, et al. A phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin Cancer Res 2003; 9(2): 555-61.
[PMID: 12576418]
[66]
Kasloff SB, Pizzuto MS, Silic-Benussi M, Pavone S, Ciminale V, Capua I. Oncolytic activity of avian influenza virus in human pancreatic ductal adenocarcinoma cell lines. J Virol 2014; 88(16): 9321-34.
[http://dx.doi.org/10.1128/JVI.00929-14] [PMID: 24899201]
[67]
Awano M, Fujiyuki T, Shoji K, et al. Measles virus selectively blind to signaling lymphocyte activity molecule has oncolytic efficacy against nectin-4-expressing pancreatic cancer cells. Cancer Sci 2016; 107(11): 1647-52.
[http://dx.doi.org/10.1111/cas.13064] [PMID: 27561180]
[68]
Jung KH, Choi IK, Lee HS, et al. Oncolytic adenovirus expressing relaxin (YDC002) enhances therapeutic efficacy of gemcitabine against pancreatic cancer. Cancer Lett 2017; 396: 155-66.
[http://dx.doi.org/10.1016/j.canlet.2017.03.009] [PMID: 28315430]
[69]
Hirooka Y, Kasuya H, Ishikawa T, et al. A Phase I clinical trial of EUS-guided intratumoral injection of the oncolytic virus, HF10 for unresectable locally advanced pancreatic cancer. BMC Cancer 2018; 18(1): 596.
[http://dx.doi.org/10.1186/s12885-018-4453-z] [PMID: 29801474]
[70]
Cheng X, Wang W, Xu Q, et al. Genetic modification of oncolytic newcastle disease virus for cancer therapy. J Virol 2016; 90(11): 5343-52.
[http://dx.doi.org/10.1128/JVI.00136-16] [PMID: 27009956]
[71]
Schwaiger T, Knittler MR, Grund C, et al. Newcastle disease virus mediates pancreatic tumor rejection via NK cell activation and prevents cancer relapse by prompting adaptive immunity. Int J Cancer 2017; 141(12): 2505-16.
[http://dx.doi.org/10.1002/ijc.31026] [PMID: 28857157]
[72]
Wu Y, Mou X, Wang S, Liu XE, Sun X. ING4 expressing oncolytic vaccinia virus promotes anti-tumor efficiency and synergizes with gemcitabine in pancreatic cancer. Oncotarget 2017; 8(47): 82728-39.
[http://dx.doi.org/10.18632/oncotarget.21095] [PMID: 29137298]
[73]
Tremont A, Lu J, Cole JT. Endocrine Therapy for Early Breast Cancer: Updated Review. Ochsner J 2017; 17(4): 405-11.
[PMID: 29230126]
[74]
Ranjbar S, Hashemzadeh MS, NIKKHOI SK, Farasat A, Tat M, Ghalavand M. Selective suppression of tumor cells by a tumorspecific bicistronic lentiviral vector 2016; 40(6): 1289-94.
[75]
Chen X, Han J, Chu J, et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget 2016; 7(19): 27764-77.
[http://dx.doi.org/10.18632/oncotarget.8526] [PMID: 27050072]
[76]
Cody JJ, Hurst DR. Promising oncolytic agents for metastatic breast cancer treatment. Oncolytic Virother 2015; 4: 63-73.
[PMID: 27512671]
[77]
Nokisalmi P, Pesonen S, Escutenaire S, et al. Oncolytic adenovirus ICOVIR-7 in patients with advanced and refractory solid tumors. Clin Cancer Res 2010; 16(11): 3035-43.
[http://dx.doi.org/10.1158/1078-0432.CCR-09-3167 ] [PMID: 20501623]
[78]
Wang N, Lu Y, Pinard M, et al. Sustained production of a soluble IGF-I receptor by gutless adenovirus-transduced host cells protects from tumor growth in the liver. Cancer Gene Ther 2013; 20(4): 229-36.
[http://dx.doi.org/10.1038/cgt.2013.10] [PMID: 23470563]
[79]
Holgado MP, Falivene J, Maeto C, et al. Deletion of A44L, A46R and C12L vaccinia virus genes from the mva genome improved the vector immunogenicity by modifying the innate immune response generating enhanced and optimized specific T-Cell responses. Viruses 2016; 8(5)E139
[http://dx.doi.org/10.3390/v8050139] [PMID: 27223301]
[80]
Harrop R, Connolly N, Redchenko I, et al. Vaccination of colorectal cancer patients with modified vaccinia Ankara delivering the tumor antigen 5T4 (TroVax) induces immune responses which correlate with disease control: A phase I/II trial. Clin Cancer Res 2006; 12(11 Pt 1): 3416-24.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-2732] [PMID: 16740766]
[81]
Amato RJ, Hawkins RE, Kaufman HL, et al. Vaccination of metastatic renal cancer patients with MVA-5T4: A randomized, double-blind, placebo-controlled phase III study. Clin Cancer Res 2010; 16(22): 5539-47.
[http://dx.doi.org/10.1158/1078-0432.CCR-10-2082] [PMID: 20881001]
[82]
Gholami S, Marano A, Chen NG, et al. A novel vaccinia virus with dual oncolytic and anti-angiogenic therapeutic effects against triple-negative breast cancer. Breast Cancer Res Treat 2014; 148(3): 489-99.
[http://dx.doi.org/10.1007/s10549-014-3180-7] [PMID: 25391896]
[83]
Gholami S, Chen CH, Lou E, et al. Vaccinia virus GLV-1h153 in combination with 131I shows increased efficiency in treating triple-negative breast cancer. FASEB J 2014; 28(2): 676-82.
[http://dx.doi.org/10.1096/fj.13-237222] [PMID: 24186964]
[84]
Choi AH, O’Leary MP, Chaurasiya S, et al. Novel chimeric parapoxvirus CF189 as an oncolytic immunotherapy in triple-negative breast cancer. Surgery 2018; 163(2): 336-42.
[http://dx.doi.org/10.1016/j.surg.2017.09.030] [PMID: 29174433]
[85]
Mao LJ, Zhang J, Liu N, et al. Oncolytic virus carrying shRNA targeting SATB1 inhibits prostate cancer growth and metastasis. Tumour Biol 2015; 36(11): 9073-81.
[http://dx.doi.org/10.1007/s13277-015-3658-x] [PMID: 26084613]
[86]
Hao J, Xie W, Li H, Li R. Prostate cancer-specific of DD3-driven oncolytic virus-harboring mK5 gene. Open Med (Wars) 2018; 14: 1-9.
[http://dx.doi.org/10.1515/med-2019-0001] [PMID: 30613790]
[87]
DeWeese TL, van der Poel H, Li S, et al. A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy. Cancer Res 2001; 61(20): 7464-72.
[PMID: 11606381]
[88]
Small EJ, Carducci MA, Burke JM, et al. A phase I trial of intravenous CG7870, a replication-selective, prostate-specific antigen-targeted oncolytic adenovirus, for the treatment of hormone-refractory, metastatic prostate cancer. Surgery 2006; 163(2): 336-42.
[http://dx.doi.org/10.1016/j.ymthe.2006.02.011]
[89]
Cui CX, Li YQ, Sun YJ, et al. Antitumor effect of a dual cancer-specific oncolytic adenovirus on prostate cancer PC-3 cells. Urol Oncol 2019; 37(6): 352.e1-e18.
[90]
Sakhawat A, Liu Y, Ma L, et al. Upregulation of coxsackie adenovirus receptor sensitizes cisplatin-resistant lung cancer cells to CRAd-induced inhibition. J Cancer 2017; 8(8): 1425-32.
[http://dx.doi.org/10.7150/jca.18371] [PMID: 28638457]
[91]
Hu J, Wang H, Gu J, Liu X, Zhou X. Trail armed oncolytic poxvirus suppresses lung cancer cell by inducing apoptosis. Acta Biochim Biophys Sin (Shanghai) 2018; 50(10): 1018-27.
[http://dx.doi.org/10.1093/abbs/gmy096] [PMID: 30137199]
[92]
Miyamoto S, Inoue H, Nakamura T, et al. Coxsackievirus B3 is an oncolytic virus with immunostimulatory properties that is active against lung adenocarcinoma. Cancer Res 2012; 72(10): 2609-21.
[http://dx.doi.org/10.1158/0008-5472.CAN-11-3185] [PMID: 22461509]
[93]
Ye T, Jiang K, Wei L, et al. Oncolytic Newcastle disease virus induces autophagy-dependent immunogenic cell death in lung cancer cells. Am J Cancer Res 2018; 8(8): 1514-27.
[PMID: 30210920]
[94]
Zhang R, Zhang X, Ma B, et al. Enhanced antitumor effect of combining TRAIL and MnSOD mediated by CEA-controlled oncolytic adenovirus in lung cancer. Cancer Gene Ther 2016; 23(6): 168-77.
[http://dx.doi.org/10.1038/cgt.2016.11] [PMID: 27080225]
[95]
Liu D, Kojima T, Ouchi M, et al. Preclinical evaluation of synergistic effect of telomerase-specific oncolytic virotherapy and gemcitabine for human lung cancer. Mol Cancer Ther 2009; 8(4): 980-7.
[http://dx.doi.org/10.1158/1535-7163.MCT-08-0901] [PMID: 19372571]
[96]
Patel MR, Dash A, Jacobson BA, et al. JAK/STAT inhibition with ruxolitinib enhances oncolytic virotherapy in non-small cell lung cancer models. Cancer Gene Ther 2019; 26(11-12): 411-8.
[http://dx.doi.org/10.1038/s41417-018-0074-6] [PMID: 30622322]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy