| Peer-Reviewed

Molecular Mechanisms Associated with Virus-induced Oncogenesis and Oncolysis

Received: 4 June 2019    Accepted: 13 July 2019    Published: 15 August 2019
Views:       Downloads:
Abstract

Cancer is a leading cause of human deaths worldwide. Besides inherited genetic disorders, a diverse range of physical, chemical and biological agents may induce cancer. About 15-20% of cancers are known to be originated due to pathogens. Viruses are considered to be the second (after smoking) most important risk factor in inducing human cancer. Viruses may either harbour a copy of oncogene or have an ability to alter the expression of cellular copy of the oncogenes. Both RNA and DNA viruses are can induce oncogenesis. Most of the DNA tumour viruses either integrate their genome (complete or part of it) into the host genome or express early genes that are required for early event of virus replication. These early genes are responsible for oncogenic transformation of host cells. Based upon the mechanism involved, oncogenic RNA viruses are divided into two groups-transforming and non-transforming RNA viruses. Transforming RNA viruses carry viral oncogenes that are homologous to the host oncogene, their expression in infected cells results in oncogenic transformation of the cell. Non-transforming RNA viruses induce oncogenesis similar to the DNA viruses. Contrary, oncolytic viruses selectively replicate in cancerous cells and induce cell death without any damage to the normal tissues. Typically, oncolytic viruses are nonpathogenic to humans that can naturally replicate in cancer cells by exploiting oncogenic cell signalling pathways. Pathogenic viruses can also be genetically manipulated which allow them to replicate in cancerous but not in normal cells. This review review describes the molecular mechanisms associated with virus induced oncogenesis and oncolysis.

Published in Cancer Research Journal (Volume 7, Issue 3)
DOI 10.11648/j.crj.20190703.13
Page(s) 87-100
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Oncogenic, Oncolytic, Virus

References
[1] Wittekind C, Neid M. 2005. Cancer invasion and metastasis. Oncology 69 Suppl 1: 14-16.
[2] Sever R, Brugge JS. 2015. Signal transduction in cancer. Cold Spring Harbor Perspectives in Medicine 5.
[3] Cantley LC, Auger KR, Carpenter C, Duckworth B, Graziani A, Kapeller R, Soltoff S. 1991. Oncogenes and signal transduction. Cell 64: 281-302.
[4] Qin J, Xie LP, Zheng XY, Wang YB, Bai Y, Shen HF, Li LC, Dahiya R. 2007. A component of green tea, (-)-epigallocatechin-3-gallate, promotes apoptosis in T24 human bladder cancer cells via modulation of the PI3K/Akt pathway and Bcl-2 family proteins. Biochemical and Biophysical Research Communications 354: 852-857.
[5] Ingraham BA, Bragdon B, Nohe A. 2008. Molecular basis of the potential of vitamin D to prevent cancer. Current Medical Research and Opinion 24: 139-149.
[6] Zhang XK, Huang DP, Chiu DK, Chiu JF. 1987. The expression of oncogenes in human developing liver and hepatomas. Biochemical and Biophysical Research Communications 142: 932-938.
[7] Miyashita T, Krajewski S, Krajewska M, Wang HG, Lin HK, Liebermann DA, Hoffman B, Reed JC. 1994. Tumor suppressor p53 is a regulator of bcl-2 and bax gene expression in vitro and in vivo. Oncogene 9: 1799-1805.
[8] Szekely L, Selivanova G, Magnusson KP, Klein G, Wiman KG. 1993. EBNA-5, an Epstein-Barr virus-encoded nuclear antigen, binds to the retinoblastoma and p53 proteins. Proceedings of the National Academy of Sciences of the United States of America 90: 5455-5459.
[9] Parsa N. 2012. Environmental factors inducing human cancers. Iranian journal of public health 41: 1-9.
[10] De Flora S, La Maestra S. 2015. Epidemiology of cancers of infectious origin and prevention strategies. Journal of Preventive Medicine and Hygiene 56: E15-20.
[11] Justice Jt, Beemon KL. 2013. Avian retroviral replication. Current Opinion in Virology 3: 664-669.
[12] Moore PS, Chang Y. 2010. Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nature reviews. Cancer 10: 878-889.
[13] Butel JS. 2000. Viral carcinogenesis: revelation of molecular mechanisms and etiology of human disease. Carcinogenesis 21: 405-426.
[14] Luo GG, Ou JH. 2015. Oncogenic viruses and cancer. Virologica Sinica 30: 83-84.
[15] Ljubojevic S. 2006. The human papillomavirus vaccines. Acta Dermatovenerologica Croatica: ADC 14: 208.
[16] Chan PK, Chang AR, Cheung JL, Chan DP, Xu LY, Tang NL, Cheng AF. 2002. Determinants of cervical human papillomavirus infection: differences between high- and low-oncogenic risk types. The Journal of Infectious Diseases 185: 28-35.
[17] Campo MS. 2003. Papillomavirus and disease in humans and animals. Veterinary and Comparative Oncology 1: 3-14.
[18] Nguyen KD, Lee EE, Yue Y, Stork J, Pock L, North JP, Vandergriff T, Cockerell C, Hosler GA, Pastrana DV, Buck CB, Wang RC. 2017. Human polyomavirus 6 and 7 are associated with pruritic and dyskeratotic dermatoses. Journal of the American Academy of Dermatology 76: 932-940 e933.
[19] Ali N, Shaikh MU, Hasan S. 2011. BK Virus Associated Late Onset Haemorrhagic Cystitis After Allogeneic Peripheral Blood Stem Cell Transplant. Indian journal of hematology & blood transfusion: An Official Journal of Indian Society of Hematology and Blood Transfusion 27: 177-179.
[20] Elgui de Oliveira D. 2007. DNA viruses in human cancer: an integrated overview on fundamental mechanisms of viral carcinogenesis. Cancer Letters 247: 182-196.
[21] Hinze HC. 1971. Induction of lymphoid hyperplasia and lymphoma-like disease in rabbits by Herpesvirus sylvilagus. International Journal of Cancer 8: 514-522.
[22] Mathew S, Ali A, Abdel-Hafiz H, Fatima K, Suhail M, Archunan G, Begum N, Jahangir S, Ilyas M, Chaudhary AG, Al Qahtani M, Mohamad Bazarah S, Qadri I. 2014. Biomarkers for virus-induced hepatocellular carcinoma (HCC). Infection, genetics and evolution: Journal of Molecular Epidemiology and Evolutionary Genetics in Infectious Diseases 26: 327-339.
[23] Giacinti C, Giordano A. 2006. RB and cell cycle progression. Oncogene 25: 5220-5227.
[24] Ohtani N, Yamakoshi K, Takahashi A, Hara E. 2004. The p16INK4a-RB pathway: molecular link between cellular senescence and tumor suppression. The Journal of Medical Investigation: JMI 51: 146-153.
[25] Hwang SG, Lee D, Kim J, Seo T, Choe J. 2002. Human papillomavirus type 16 E7 binds to E2F1 and activates E2F1-driven transcription in a retinoblastoma protein-independent manner. The Journal of Biological Chemistry 277: 2923-2930.
[26] Seifried LA, Talluri S, Cecchini M, Julian LM, Mymryk JS, Dick FA. 2008. pRB-E2F1 complexes are resistant to adenovirus E1A-mediated disruption. Journal of Virology 82: 4511-4520.
[27] Brooks CL, Gu W. 2006. p53 ubiquitination: Mdm2 and beyond. Molecular Cell 21: 307-315.
[28] Amaral JD, Xavier JM, Steer CJ, Rodrigues CM. 2010. The role of p53 in apoptosis. Discovery Medicine 9: 145-152.
[29] Brown VL, Harwood CA, Crook T, Cronin JG, Kelsell DP, Proby CM. 2004. p16INK4a and p14ARF tumor suppressor genes are commonly inactivated in cutaneous squamous cell carcinoma. The Journal of Investigative Dermatology 122: 1284-1292.
[30] Cory S, Adams JM. 2002. The Bcl2 family: regulators of the cellular life-or-death switch. Nature reviews. Cancer 2: 647-656.
[31] Haupt S, Berger M, Goldberg Z, Haupt Y. 2003. Apoptosis - the p53 network. Journal of Cell Science 116: 4077-4085.
[32] White MK, Khalili K. 2004. Polyomaviruses and human cancer: molecular mechanisms underlying patterns of tumorigenesis. Virology 324: 1-16.
[33] Querido E, Blanchette P, Yan Q, Kamura T, Morrison M, Boivin D, Kaelin WG, Conaway RC, Conaway JW, Branton PE. 2001. Degradation of p53 by adenovirus E4orf6 and E1B55K proteins occurs via a novel mechanism involving a Cullin-containing complex. Genes and Development 15: 3104-3117.
[34] Scheffner M, Werness BA, Huibregtse JM, Levine AJ, Howley PM. 1990. The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of p53. Cell 63: 1129-1136.
[35] Scheffner M, Huibregtse JM, Vierstra RD, Howley PM. 1993. The HPV-16 E6 and E6-AP complex functions as a ubiquitin-protein ligase in the ubiquitination of p53. Cell 75: 495-505.
[36] Chen Y, Williams V, Filippova M, Filippov V, Duerksen-Hughes P. 2014. Viral carcinogenesis: factors inducing DNA damage and virus integration. Cancers 6: 2155-2186.
[37] Kim CM, Koike K, Saito I, Miyamura T, Jay G. 1991. HBx gene of hepatitis B virus induces liver cancer in transgenic mice. Nature 351: 317-320.
[38] Cacciotti P, Libener R, Betta P, Martini F, Porta C, Procopio A, Strizzi L, Penengo L, Tognon M, Mutti L, Gaudino G. 2001. SV40 replication in human mesothelial cells induces HGF/Met receptor activation: a model for viral-related carcinogenesis of human malignant mesothelioma. Proceedings of the National Academy of Sciences of the United States of America 98: 12032-12037.
[39] Bocchetta M, Miele L, Pass HI, Carbone M. 2003. Notch-1 induction, a novel activity of SV40 required for growth of SV40-transformed human mesothelial cells. Oncogene 22: 81-89.
[40] Pass HI, Mew DJ, Carbone M, Matthews WA, Donington JS, Baserga R, Walker CL, Resnicoff M, Steinberg SM. 1996. Inhibition of hamster mesothelioma tumorigenesis by an antisense expression plasmid to the insulin-like growth factor-1 receptor. Cancer Research 56: 4044-4048.
[41] Mosialos G, Birkenbach M, Yalamanchili R, VanArsdale T, Ware C, Kieff E. 1995. The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family. Cell 80: 389-399.
[42] Yasunaga J, Matsuoka M. 2007. Leukaemogenic mechanism of human T-cell leukaemia virus type I. Reviews in Medical Virology 17: 301-311.
[43] Karin M, Greten FR. 2005. NF-kappaB: linking inflammation and immunity to cancer development and progression. Nature Reviews. Immunology 5: 749-759.
[44] Moore PS, Chang Y. 1998. Antiviral activity of tumor-suppressor pathways: clues from molecular piracy by KSHV. Trends in Genetics: TIG 14: 144-150.
[45] Jin X, Beck S, Sohn YW, Kim JK, Kim SH, Yin J, Pian X, Kim SC, Choi YJ, Kim H. 2010. Human telomerase catalytic subunit (hTERT) suppresses p53-mediated anti-apoptotic response via induction of basic fibroblast growth factor. Experimental and Molecular Medicine 42: 574-582.
[46] Veldman T, Horikawa I, Barrett JC, Schlegel R. 2001. Transcriptional activation of the telomerase hTERT gene by human papillomavirus type 16 E6 oncoprotein. Journal of Virology 75: 4467-4472.
[47] Terrin L, Dal Col J, Rampazzo E, Zancai P, Pedrotti M, Ammirabile G, Bergamin S, Rizzo S, Dolcetti R, De Rossi A. 2008. Latent membrane protein 1 of Epstein-Barr virus activates the hTERT promoter and enhances telomerase activity in B lymphocytes. Journal of Virology 82: 10175-10187.
[48] Gabet AS, Mortreux F, Charneau P, Riou P, Duc-Dodon M, Wu Y, Jeang KT, Wattel E. 2003. Inactivation of hTERT transcription by Tax. Oncogene 22: 3734-3741.
[49] Toyooka S, Carbone M, Toyooka KO, Bocchetta M, Shivapurkar N, Minna JD, Gazdar AF. 2002. Progressive aberrant methylation of the RASSF1A gene in simian virus 40 infected human mesothelial cells. Oncogene 21: 4340-4344.
[50] De Bolle L, Naesens L, De Clercq E. 2005. Update on human herpesvirus 6 biology, clinical features, and therapy. Clinical Microbiology Reviews 18: 217-245.
[51] Duensing S, Lee LY, Duensing A, Basile J, Piboonniyom S, Gonzalez S, Crum CP, Munger K. 2000. The human papillomavirus type 16 E6 and E7 oncoproteins cooperate to induce mitotic defects and genomic instability by uncoupling centrosome duplication from the cell division cycle. Proceedings of the National Academy of Sciences of the United States of America 97: 10002-10007.
[52] Butel JS. 2000. Viral carcinogenesis: revelation of molecular mechanisms and etiology of human disease. Carcinogenesis 21: 405-426.
[53] Iba H, Takeya T, Cross FR, Hanafusa T, Hanafusa H. 1984. Rous sarcoma virus variants that carry the cellular src gene instead of the viral src gene cannot transform chicken embryo fibroblasts. Proceedings of the National Academy of Sciences of the United States of America 81: 4424-4428.
[54] Graf T, Beug H. 1983. Role of the v-erbA and v-erbB oncogenes of avian erythroblastosis virus in erythroid cell transformation. Cell 34: 7-9.
[55] Gojobori T, Yokoyama S. 1985. Rates of evolution of the retroviral oncogene of Moloney murine sarcoma virus and of its cellular homologues. Proceedings of the National Academy of Sciences of the United States of America 82: 4198-4201.
[56] Manly KF, Anderson GR, Stoler DL. 1988. Harvey sarcoma virus genome contains no extensive sequences unrelated to those of other retroviruses except ras. Journal of Virology 62: 3540-3543.
[57] Enrietto PJ, Payne LN, Hayman MJ. 1983. A recovered avian myelocytomatosis virus that induces lymphomas in chickens: pathogenic properties and their molecular basis. Cell 35: 369-379.
[58] Vogt PK. 2012. Retroviral oncogenes: a historical primer. Nature reviews. Cancer 12: 639-648.
[59] Vescovo T, Refolo G, Vitagliano G, Fimia GM, Piacentini M. 2016. Molecular mechanisms of hepatitis C virus-induced hepatocellular carcinoma. Clinical microbiology and infection: The Official Publication of the European Society of Clinical Microbiology and Infectious Diseases 22: 853-861.
[60] Watanabe H, Saitoh K, Kameda T, Murakami M, Niikura Y, Okazaki S, Morishita Y, Mori S, Yokouchi Y, Kuroiwa A, Iba H. 1997. Chondrocytes as a specific target of ectopic Fos expression in early development. Proceedings of the National Academy of Sciences of the United States of America 94: 3994-3999.
[61] Russell SJ, Peng KW, Bell JC. 2012. Oncolytic virotherapy. Nature Biotechnology 30: 658-670.
[62] Merrill MK, Bernhardt G, Sampson JH, Wikstrand CJ, Bigner DD, Gromeier M. 2004. Poliovirus receptor CD155-targeted oncolysis of glioma. Neuro-oncology 6: 208-217.
[63] Singh PK, Doley J, Kumar GR, Sahoo AP, Tiwari AK. 2012. Oncolytic viruses & their specific targeting to tumour cells. The Indian Journal of Medical Research 136: 571-584.
[64] Yu Z, Adusumilli PS, Eisenberg DP, Darr E, Ghossein RA, Li S, Liu S, Singh B, Shah JP, Fong Y, Wong RJ. 2007. Nectin-1 expression by squamous cell carcinoma is a predictor of herpes oncolytic sensitivity. Molecular Therapy: The Journal of the American Society of Gene Therapy 15: 103-113.
[65] Grunwald GK, Vetter A, Klutz K, Willhauck MJ, Schwenk N, Senekowitsch-Schmidtke R, Schwaiger M, Zach C, Wagner E, Goke B, Holm PS, Ogris M, Spitzweg C. 2013. EGFR-Targeted Adenovirus Dendrimer Coating for Improved Systemic Delivery of the Theranostic NIS Gene. Molecular Therapy. Nucleic Acids 2: e131.
[66] Petrovic B, Gianni T, Gatta V, Campadelli-Fiume G. 2017. Insertion of a ligand to HER2 in gB retargets HSV tropism and obviates the need for activation of the other entry glycoproteins. PLoS Pathogens 13: e1006352.
[67] Leja J, Yu D, Nilsson B, Gedda L, Zieba A, Hakkarainen T, Akerstrom G, Oberg K, Giandomenico V, Essand M. 2011. Oncolytic adenovirus modified with somatostatin motifs for selective infection of neuroendocrine tumor cells. Gene Therapy 18: 1052-1062.
[68] Jiang H, Gomez-Manzano C, Lang FF, Alemany R, Fueyo J. 2009. Oncolytic adenovirus: preclinical and clinical studies in patients with human malignant gliomas. Current Gene Therapy 9: 422-427.
[69] Sengupta S, Ulasov IV, Thaci B, Ahmed AU, Lesniak MS. 2011. Enhanced transduction and replication of RGD-fiber modified adenovirus in primary T cells. PloS One 6: e18091.
[70] Fujiwara T, Tanaka N. 2007. [Theranostic application of telomerase-specific oncolytic adenovirus for human cancer]. Nihon rinsho. Japanese Journal of Clinical Medicine 65: 1913-1922.
[71] Small EJ, Carducci MA, Burke JM, Rodriguez R, Fong L, van Ummersen L, Yu DC, Aimi J, Ando D, Working P, Kirn D, Wilding G. 2006. 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. Molecular Therapy: The Journal of the American Society of Gene Therapy 14: 107-117.
[72] Balachandran S, Porosnicu M, Barber GN. 2001. Oncolytic activity of vesicular stomatitis virus is effective against tumors exhibiting aberrant p53, Ras, or myc function and involves the induction of apoptosis. Journal of Virology 75: 3474-3479.
[73] Garant KA, Shmulevitz M, Pan L, Daigle RM, Ahn DG, Gujar SA, Lee PW. 2016. Oncolytic reovirus induces intracellular redistribution of Ras to promote apoptosis and progeny virus release. Oncogene 35: 771-782.
[74] Pan W, Bodempudi V, Esfandyari T, Farassati F. 2009. Utilizing ras signaling pathway to direct selective replication of herpes simplex virus-1. PloS One 4: e6514.
[75] McCart JA, Ward JM, Lee J, Hu Y, Alexander HR, Libutti SK, Moss B, Bartlett DL. 2001. Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes. Cancer Research 61: 8751-8757.
[76] Puhlmann M, Brown CK, Gnant M, Huang J, Libutti SK, Alexander HR, Bartlett DL. 2000. Vaccinia as a vector for tumor-directed gene therapy: biodistribution of a thymidine kinase-deleted mutant. Cancer Gene Therapy 7: 66-73.
[77] Callegari E, Elamin BK, D'Abundo L, Falzoni S, Donvito G, Moshiri F, Milazzo M, Altavilla G, Giacomelli L, Fornari F, Hemminki A, Di Virgilio F, Gramantieri L, Negrini M, Sabbioni S. 2013. Anti-tumor activity of a miR-199-dependent oncolytic adenovirus. PloS One 8: e73964.
[78] Baertsch MA, Leber MF, Bossow S, Singh M, Engeland CE, Albert J, Grossardt C, Jager D, von Kalle C, Ungerechts G. 2014. MicroRNA-mediated multi-tissue detargeting of oncolytic measles virus. Cancer Gene Therapy 21: 373-380.
[79] Palosaari H, Parisien JP, Rodriguez JJ, Ulane CM, Horvath CM. 2003. STAT protein interference and suppression of cytokine signal transduction by measles virus V protein. Journal of Virology 77: 7635-7644.
[80] Learmonth K, Braidwood L, Woll P, Conner J. 2015. Immune responses following intrapleural administration of oncolytic SEPREHVIR in patients with malignant pleural mesothelioma. Journal for Immunotherapy of Cancer 3: P335-P335.
[81] Stojdl DF, Lichty BD, tenOever BR, Paterson JM, Power AT, Knowles S, Marius R, Reynard J, Poliquin L, Atkins H, Brown EG, Durbin RK, Durbin JE, Hiscott J, Bell JC. 2003. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. Cancer Cell 4: 263-275.
[82] Takeuchi K, Kadota SI, Takeda M, Miyajima N, Nagata K. 2003. Measles virus V protein blocks interferon (IFN)-alpha/beta but not IFN-gamma signaling by inhibiting STAT1 and STAT2 phosphorylation. FEBS Letters 545: 177-182.
[83] Liu BL, Robinson M, Han ZQ, Branston RH, English C, Reay P, McGrath Y, Thomas SK, Thornton M, Bullock P, Love CA, Coffin RS. 2003. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Therapy 10: 292-303.
[84] Goldsmith K, Chen W, Johnson DC, Hendricks RL. 1998. Infected cell protein (ICP) 47 enhances herpes simplex virus neurovirulence by blocking the CD8+ T cell response. The Journal of Experimental Medicine 187: 341-348.
[85] Gonzalez R, Huang W, Finnen R, Bragg C, Flint SJ. 2006. Adenovirus E1B 55-kilodalton protein is required for both regulation of mRNA export and efficient entry into the late phase of infection in normal human fibroblasts. Journal of Virology 80: 964-974.
[86] Wang JN, Hu P, Zeng MS, Liu RB. 2011. Anti-tumor effect of oncolytic herpes simplex virus G47delta on human nasopharyngeal carcinoma. Chinese Journal of Cancer 30: 831-841.
[87] Liang M. 2018. Oncorine, the World First Oncolytic Virus Medicine and its Update in China. Current Cancer Drug Targets 18: 171-176.
[88] Ries S, Korn WM. 2002. ONYX-015: mechanisms of action and clinical potential of a replication-selective adenovirus. British Journal of Cancer 86: 5-11.
[89] DeWeese TL, van der Poel H, Li S, Mikhak B, Drew R, Goemann M, Hamper U, DeJong R, Detorie N, Rodriguez R, Haulk T, DeMarzo AM, Piantadosi S, Yu DC, Chen Y, Henderson DR, Carducci MA, Nelson WG, Simons JW. 2001. 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 Research 61: 7464-7472.
[90] Burke JM, Lamm DL, Meng MV, Nemunaitis JJ, Stephenson JJ, Arseneau JC, Aimi J, Lerner S, Yeung AW, Kazarian T, Maslyar DJ, McKiernan JM. 2012. A first in human phase 1 study of CG0070, a GM-CSF expressing oncolytic adenovirus, for the treatment of nonmuscle invasive bladder cancer. The Journal of Urology 188: 2391-2397.
[91] Freytag SO, Stricker H, Pegg J, Paielli D, Pradhan DG, Peabody J, DePeralta-Venturina M, Xia X, Brown S, Lu M, Kim JH. 2003. Phase I study of replication-competent adenovirus-mediated double-suicide gene therapy in combination with conventional-dose three-dimensional conformal radiation therapy for the treatment of newly diagnosed, intermediate- to high-risk prostate cancer. Cancer Research 63: 7497-7506.
[92] Freytag SO, Stricker H, Lu M, Elshaikh M, Aref I, Pradhan D, Levin K, Kim JH, Peabody J, Siddiqui F, Barton K, Pegg J, Zhang Y, Cheng J, Oja-Tebbe N, Bourgeois R, Gupta N, Lane Z, Rodriguez R, DeWeese T, Movsas B. 2014. Prospective randomized phase 2 trial of intensity modulated radiation therapy with or without oncolytic adenovirus-mediated cytotoxic gene therapy in intermediate-risk prostate cancer. International Journal of Radiation Oncology, Biology, Physics 89: 268-276.
[93] Pol J, Buque A, Aranda F, Bloy N, Cremer I, Eggermont A, Erbs P, Fucikova J, Galon J, Limacher JM, Preville X, Sautes-Fridman C, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. 2016. Trial Watch-Oncolytic viruses and cancer therapy. Oncoimmunology 5: e1117740.
[94] Lin CZ, Xiang GL, Zhu XH, Xiu LL, Sun JX, Zhang XY. 2018. Advances in the mechanisms of action of cancer-targeting oncolytic viruses. Oncology Letters 15: 4053-4060.
[95] Kim KH, Dmitriev I, O'Malley JP, Wang M, Saddekni S, You Z, Preuss MA, Harris RD, Aurigemma R, Siegal GP, Zinn KR, Curiel DT, Alvarez RD. 2012. A phase I clinical trial of Ad5.SSTR/TK.RGD, a novel infectivity-enhanced bicistronic adenovirus, in patients with recurrent gynecologic cancer. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research 18: 3440-3451.
[96] Dhar D, Spencer JF, Toth K, Wold WS. 2009. Effect of preexisting immunity on oncolytic adenovirus vector INGN 007 antitumor efficacy in immunocompetent and immunosuppressed Syrian hamsters. Journal of Virology 83: 2130-2139.
[97] Alonso MM, Gomez-Manzano C, Jiang H, Bekele NB, Piao Y, Yung WK, Alemany R, Fueyo J. 2007. Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo. Cancer Gene Therapy 14: 756-761.
[98] Andtbacka RH, Ross M, Puzanov I, Milhem M, Collichio F, Delman KA, Amatruda T, Zager JS, Cranmer L, Hsueh E, Chen L, Shilkrut M, Kaufman HL. 2016. Patterns of Clinical Response with Talimogene Laherparepvec (T-VEC) in Patients with Melanoma Treated in the OPTiM Phase III Clinical Trial. Annals of Surgical Oncology 23: 4169-4177.
[99] Kelly KJ, Wong J, Fong Y. 2008. Herpes simplex virus NV1020 as a novel and promising therapy for hepatic malignancy. Expert Opinion on Investigational Drugs 17: 1105-1113.
[100] Nawa A, Luo C, Zhang L, Ushjima Y, Ishida D, Kamakura M, Fujimoto Y, Goshima F, Kikkawa F, Nishiyama Y. 2008. Non-engineered, naturally oncolytic herpes simplex virus HSV1 HF-10: applications for cancer gene therapy. Current Gene Therapy 8: 208-221.
[101] Nakamura T, Peng KW, Harvey M, Greiner S, Lorimer IA, James CD, Russell SJ. 2005. Rescue and propagation of fully retargeted oncolytic measles viruses. Nature Biotechnology 23: 209-214.
[102] Csatary LK, Moss RW, Beuth J, Torocsik B, Szeberenyi J, Bakacs T. 1999. Beneficial treatment of patients with advanced cancer using a Newcastle disease virus vaccine (MTH-68/H). Anticancer Research 19: 635-638.
[103] Gollamudi R, Ghalib MH, Desai KK, Chaudhary I, Wong B, Einstein M, Coffey M, Gill GM, Mettinger K, Mariadason JM, Mani S, Goel S. 2010. Intravenous administration of Reolysin, a live replication competent RNA virus is safe in patients with advanced solid tumors. Investigational New Drugs 28: 641-649.
[104] Diaz RM, Galivo F, Kottke T, Wongthida P, Qiao J, Thompson J, Valdes M, Barber G, Vile RG. 2007. Oncolytic immunovirotherapy for melanoma using vesicular stomatitis virus. Cancer Research 67: 2840-2848.
[105] Bell J, McFadden G. 2014. Viruses for tumor therapy. Cell Host and Microbe 15: 260-265.
[106] Mell LK, Brumund KT, Daniels GA, Advani SJ, Zakeri K, Wright ME, Onyeama SJ, Weisman RA, Sanghvi PR, Martin PJ, Szalay AA. 2017. Phase I Trial of Intravenous Oncolytic Vaccinia Virus (GL-ONC1) with Cisplatin and Radiotherapy in Patients with Locoregionally Advanced Head and Neck Carcinoma. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research 23: 5696-5702.
[107] Downs-Canner S, Guo ZS, Ravindranathan R, Breitbach CJ, O'Malley ME, Jones HL, Moon A, McCart JA, Shuai Y, Zeh HJ, Bartlett DL. 2016. Phase 1 Study of Intravenous Oncolytic Poxvirus (vvDD) in Patients With Advanced Solid Cancers. Molecular Therapy: the Journal of the American Society of Gene Therapy 24: 1492-1501.
[108] Burke MJ, Ahern C, Weigel BJ, Poirier JT, Rudin CM, Chen Y, Cripe TP, Bernhardt MB, Blaney SM. 2015. Phase I trial of Seneca Valley Virus (NTX-010) in children with relapsed/refractory solid tumors: a report of the Children's Oncology Group. Pediatric Blood and Cancer 62: 743-750.
[109] Merchan JR, Ostertag D, Jolly DJ, Tejera D, Gruber HE, Shorr J. 2018. Toca 511 and Toca FC in patients with gastrointestinal tumors in the Toca 6 study. Journal of Clinical Oncology 36: TPS880-TPS880.
[110] Nakajima TI, Toshimitsu & Wada, Hiroshi & Yamauchi, Toshie & Kiyohara, Eiji & Kane, Yasufumi.. 2013. A Novel Therapy for Melanoma and Prostate Cancer Using a Non-Replicating Sendai Virus Particle (HVJ-E)
[111] Danen-van Oorschot AA, van Der Eb AJ, Noteborn MH. 2000. The chicken anemia virus-derived protein apoptin requires activation of caspases for induction of apoptosis in human tumor cells. Journal of Virology 74: 7072-7078.
[112] Jhawar SR, Thandoni A, Bommareddy PK, Hassan S, Kohlhapp FJ, Goyal S, Schenkel JM, Silk AW, Zloza A. 2017. Oncolytic Viruses-Natural and Genetically Engineered Cancer Immunotherapies. Frontiers in Oncology 7: 202.
[113] Moehler MH, Zeidler M, Wilsberg V, Cornelis JJ, Woelfel T, Rommelaere J, Galle PR, Heike M. 2005. Parvovirus H-1-induced tumor cell death enhances human immune response in vitro via increased phagocytosis, maturation, and cross-presentation by dendritic cells. Human Gene Therapy 16: 996-1005.
[114] Cattaneo R, Miest T, Shashkova EV, Barry MA. 2008. Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nature reviews. Microbiology 6: 529-540.
[115] Iankov ID, Allen C, Federspiel MJ, Myers RM, Peng KW, Ingle JN, Russell SJ, Galanis E. 2012. Expression of immunomodulatory neutrophil-activating protein of Helicobacter pylori enhances the antitumor activity of oncolytic measles virus. Molecular Therapy: The Journal of the American Society of Gene Therapy 20: 1139-1147.
[116] Howells A, Marelli G, Lemoine NR, Wang Y. 2017. Oncolytic Viruses-Interaction of Virus and Tumor Cells in the Battle to Eliminate Cancer. Frontiers in Oncology 7: 195.
[117] Zhao Y, Li H, Wu R, Li S, Wang P, Wang H, Wang J, Zhou J. 2015. Antitumor Effects of Oncolytic Adenovirus-Carrying siRNA Targeting Potential Oncogene EphA3. PloS One 10: e0126726.
[118] Yoo JY, Kim JH, Kwon YG, Kim EC, Kim NK, Choi HJ, Yun CO. 2007. VEGF-specific short hairpin RNA-expressing oncolytic adenovirus elicits potent inhibition of angiogenesis and tumor growth. Molecular Therapy: The Journal of the American Society of Gene Therapy 15: 295-302.
[119] Sun A, Tang J, Terranova PF, Zhang X, Thrasher JB, Li B. 2010. Adeno-associated virus-delivered short hairpin-structured RNA for androgen receptor gene silencing induces tumor eradication of prostate cancer xenografts in nude mice: a preclinical study. International Journal of Cancer 126: 764-774.
[120] Smith JS, Herrero R, Bosetti C, Munoz N, Bosch FX, Eluf-Neto J, Castellsague X, Meijer CJ, Van den Brule AJ, Franceschi S, Ashley R, International Agency for Research on Cancer Multicentric Cervical Cancer Study G. 2002. Herpes simplex virus-2 as a human papillomavirus cofactor in the etiology of invasive cervical cancer. Journal of the National Cancer Institute 94: 1604-1613.
[121] Cao S, Wendl MC, Wyczalkowski MA, Wylie K, Ye K, Jayasinghe R, Xie M, Wu S, Niu B, Grubb R, 3rd, Johnson KJ, Gay H, Chen K, Rader JS, Dipersio JF, Chen F, Ding L. 2016. Divergent viral presentation among human tumors and adjacent normal tissues. Scientific Reports 6: 28294.
[122] Salmons B, Gunzburg WH. 2013. Revisiting a role for a mammary tumor retrovirus in human breast cancer. International Journal of Cancer 133: 1530-1535.
[123] Tang KW, Alaei-Mahabadi B, Samuelsson T, Lindh M, Larsson E. 2013. The landscape of viral expression and host gene fusion and adaptation in human cancer. Nature Communications 4: 2513.
[124] Cobbs CS, Harkins L, Samanta M, Gillespie GY, Bharara S, King PH, Nabors LB, Cobbs CG, Britt WJ. 2002. Human cytomegalovirus infection and expression in human malignant glioma. Cancer Research 62: 3347-3350.
[125] Tang KW, Hellstrand K, Larsson E. 2015. Absence of cytomegalovirus in high-coverage DNA sequencing of human glioblastoma multiforme. International Journal of Cancer 136: 977-981.
[126] Strong MJ, O'Grady T, Lin Z, Xu G, Baddoo M, Parsons C, Zhang K, Taylor CM, Flemington EK. 2013. Epstein-Barr virus and human herpesvirus 6 detection in a non-Hodgkin's diffuse large B-cell lymphoma cohort by using RNA sequencing. Journal of Virology 87: 13059-13062.
[127] Urisman A, Molinaro RJ, Fischer N, Plummer SJ, Casey G, Klein EA, Malathi K, Magi-Galluzzi C, Tubbs RR, Ganem D, Silverman RH, DeRisi JL. 2006. Identification of a novel Gammaretrovirus in prostate tumors of patients homozygous for R462Q RNASEL variant. PLoS Pathogens 2: e25.
[128] Johnson AD, Cohn CS. 2016. Xenotropic Murine Leukemia Virus-Related Virus (XMRV) and the Safety of the Blood Supply. Clinical Microbiology Reviews 29: 749-757.
Cite This Article
  • APA Style

    Ram Kumar, Riyesh Thachamvally, Sunil Maherchandani, Bhupendra Nath Tripathi, Sanjay Barua, et al. (2019). Molecular Mechanisms Associated with Virus-induced Oncogenesis and Oncolysis. Cancer Research Journal, 7(3), 87-100. https://doi.org/10.11648/j.crj.20190703.13

    Copy | Download

    ACS Style

    Ram Kumar; Riyesh Thachamvally; Sunil Maherchandani; Bhupendra Nath Tripathi; Sanjay Barua, et al. Molecular Mechanisms Associated with Virus-induced Oncogenesis and Oncolysis. Cancer Res. J. 2019, 7(3), 87-100. doi: 10.11648/j.crj.20190703.13

    Copy | Download

    AMA Style

    Ram Kumar, Riyesh Thachamvally, Sunil Maherchandani, Bhupendra Nath Tripathi, Sanjay Barua, et al. Molecular Mechanisms Associated with Virus-induced Oncogenesis and Oncolysis. Cancer Res J. 2019;7(3):87-100. doi: 10.11648/j.crj.20190703.13

    Copy | Download

  • @article{10.11648/j.crj.20190703.13,
      author = {Ram Kumar and Riyesh Thachamvally and Sunil Maherchandani and Bhupendra Nath Tripathi and Sanjay Barua and Naveen Kumar},
      title = {Molecular Mechanisms Associated with Virus-induced Oncogenesis and Oncolysis},
      journal = {Cancer Research Journal},
      volume = {7},
      number = {3},
      pages = {87-100},
      doi = {10.11648/j.crj.20190703.13},
      url = {https://doi.org/10.11648/j.crj.20190703.13},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.crj.20190703.13},
      abstract = {Cancer is a leading cause of human deaths worldwide. Besides inherited genetic disorders, a diverse range of physical, chemical and biological agents may induce cancer. About 15-20% of cancers are known to be originated due to pathogens. Viruses are considered to be the second (after smoking) most important risk factor in inducing human cancer. Viruses may either harbour a copy of oncogene or have an ability to alter the expression of cellular copy of the oncogenes. Both RNA and DNA viruses are can induce oncogenesis. Most of the DNA tumour viruses either integrate their genome (complete or part of it) into the host genome or express early genes that are required for early event of virus replication. These early genes are responsible for oncogenic transformation of host cells. Based upon the mechanism involved, oncogenic RNA viruses are divided into two groups-transforming and non-transforming RNA viruses. Transforming RNA viruses carry viral oncogenes that are homologous to the host oncogene, their expression in infected cells results in oncogenic transformation of the cell. Non-transforming RNA viruses induce oncogenesis similar to the DNA viruses. Contrary, oncolytic viruses selectively replicate in cancerous cells and induce cell death without any damage to the normal tissues. Typically, oncolytic viruses are nonpathogenic to humans that can naturally replicate in cancer cells by exploiting oncogenic cell signalling pathways. Pathogenic viruses can also be genetically manipulated which allow them to replicate in cancerous but not in normal cells. This review review describes the molecular mechanisms associated with virus induced oncogenesis and oncolysis.},
     year = {2019}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Molecular Mechanisms Associated with Virus-induced Oncogenesis and Oncolysis
    AU  - Ram Kumar
    AU  - Riyesh Thachamvally
    AU  - Sunil Maherchandani
    AU  - Bhupendra Nath Tripathi
    AU  - Sanjay Barua
    AU  - Naveen Kumar
    Y1  - 2019/08/15
    PY  - 2019
    N1  - https://doi.org/10.11648/j.crj.20190703.13
    DO  - 10.11648/j.crj.20190703.13
    T2  - Cancer Research Journal
    JF  - Cancer Research Journal
    JO  - Cancer Research Journal
    SP  - 87
    EP  - 100
    PB  - Science Publishing Group
    SN  - 2330-8214
    UR  - https://doi.org/10.11648/j.crj.20190703.13
    AB  - Cancer is a leading cause of human deaths worldwide. Besides inherited genetic disorders, a diverse range of physical, chemical and biological agents may induce cancer. About 15-20% of cancers are known to be originated due to pathogens. Viruses are considered to be the second (after smoking) most important risk factor in inducing human cancer. Viruses may either harbour a copy of oncogene or have an ability to alter the expression of cellular copy of the oncogenes. Both RNA and DNA viruses are can induce oncogenesis. Most of the DNA tumour viruses either integrate their genome (complete or part of it) into the host genome or express early genes that are required for early event of virus replication. These early genes are responsible for oncogenic transformation of host cells. Based upon the mechanism involved, oncogenic RNA viruses are divided into two groups-transforming and non-transforming RNA viruses. Transforming RNA viruses carry viral oncogenes that are homologous to the host oncogene, their expression in infected cells results in oncogenic transformation of the cell. Non-transforming RNA viruses induce oncogenesis similar to the DNA viruses. Contrary, oncolytic viruses selectively replicate in cancerous cells and induce cell death without any damage to the normal tissues. Typically, oncolytic viruses are nonpathogenic to humans that can naturally replicate in cancer cells by exploiting oncogenic cell signalling pathways. Pathogenic viruses can also be genetically manipulated which allow them to replicate in cancerous but not in normal cells. This review review describes the molecular mechanisms associated with virus induced oncogenesis and oncolysis.
    VL  - 7
    IS  - 3
    ER  - 

    Copy | Download

Author Information
  • National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India; Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Rajasthan, India

  • National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India

  • Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Rajasthan, India

  • National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India

  • National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India

  • National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, India

  • Sections