메뉴 건너뛰기




Volumn 4, Issue JUL, 2014, Pages

Pharmacological modulation of anti-tumor immunity induced by oncolytic viruses

Author keywords

Anti tumor immunity; Cancer; Chemotherapy; Combination therapy; Immuno modulatory therapy; Oncolytic virotherapy; Pharmacological therapy

Indexed keywords

BORTEZOMIB; CYCLOPHOSPHAMIDE; DECITABINE; ENTINOSTAT; GAMMA INTERFERON; GEMCITABINE; HISTONE DEACETYLASE INHIBITOR; I KAPPA B; I KAPPA B BETA; INTERLEUKIN 2; IRINOTECAN; MITOXANTRONE; ONCOLYTIC VIRUS; PHOSPHATIDYLINOSITOL 3 KINASE; PLATELET DERIVED GROWTH FACTOR RECEPTOR; RIBONUCLEOTIDE REDUCTASE; SUNITINIB; TEMOZOLOMIDE; TRICHOSTATIN A; TRIPTOLIDE; TUMOR NECROSIS FACTOR; VALPROIC ACID; VASCULOTROPIN RECEPTOR; VORINOSTAT;

EID: 84987984981     PISSN: None     EISSN: 2234943X     Source Type: Journal    
DOI: 10.3389/fonc.2014.00191     Document Type: Review
Times cited : (24)

References (138)
  • 1
    • 84885983827 scopus 로고    scopus 로고
    • Exploiting tumor epigenetics to improve oncolytic virotherapy
    • Forbes NE, Abdelbary H, Lupien M, Bell JC, Diallo JS. Exploiting tumor epigenetics to improve oncolytic virotherapy. Front Genet (2013) 4:184. doi: 10.3389/fgene.2013.00184
    • (2013) Front Genet , vol.4 , pp. 184
    • Forbes, N.E.1    Abdelbary, H.2    Lupien, M.3    Bell, J.C.4    Diallo, J.S.5
  • 2
    • 84893715844 scopus 로고    scopus 로고
    • From scourge to cure: tumour-selective viral pathogenesis as a new strategy against cancer
    • Ilkow CS, Swift SL, Bell JC, Diallo JS. From scourge to cure: tumour-selective viral pathogenesis as a new strategy against cancer. PLoS Pathog (2014) 10:e1003836. doi:10.1371/journal.ppat.1003836
    • (2014) PLoS Pathog , vol.10
    • Ilkow, C.S.1    Swift, S.L.2    Bell, J.C.3    Diallo, J.S.4
  • 4
    • 79953294511 scopus 로고    scopus 로고
    • Oncolytic virus-initiated protective immunity against prostate cancer
    • Gujar SA, Pan DA, Marcato P, Garant KA, Lee PW. Oncolytic virus-initiated protective immunity against prostate cancer. Mol Ther (2011) 19:797-804. doi:10.1038/mt.2010.297
    • (2011) Mol Ther , vol.19 , pp. 797-804
    • Gujar, S.A.1    Pan, D.A.2    Marcato, P.3    Garant, K.A.4    Lee, P.W.5
  • 5
    • 79952361112 scopus 로고    scopus 로고
    • Pro-inflammatory cytokine/chemokine production by reovirus treated melanoma cells is PKR/NF-kappaB mediated and supports innate and adaptive anti-tumour immune priming
    • Steele L, Errington F, Prestwich R, Ilett E, Harrington K, Pandha H, et al. Pro-inflammatory cytokine/chemokine production by reovirus treated melanoma cells is PKR/NF-kappaB mediated and supports innate and adaptive anti-tumour immune priming. Mol Cancer (2011) 10:20. doi:10.1186/1476-4598-10-20
    • (2011) Mol Cancer , vol.10 , pp. 20
    • Steele, L.1    Errington, F.2    Prestwich, R.3    Ilett, E.4    Harrington, K.5    Pandha, H.6
  • 7
    • 44249100151 scopus 로고    scopus 로고
    • Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial
    • Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol (2008) 9:533-42. doi:10.1016/S1470-2045(08)70107-4
    • (2008) Lancet Oncol , vol.9 , pp. 533-542
    • Park, B.H.1    Hwang, T.2    Liu, T.C.3    Sze, D.Y.4    Kim, J.S.5    Kwon, H.C.6
  • 8
    • 84867027824 scopus 로고    scopus 로고
    • Phase II trial of intravenous administration of Reolysin((R)) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma
    • Galanis E, Markovic SN, Suman VJ, Nuovo GJ, Vile RG, Kottke TJ, et al. Phase II trial of intravenous administration of Reolysin((R)) (Reovirus Serotype-3-dearing Strain) in patients with metastatic melanoma. Mol Ther (2012) 20:1998-2003. doi:10.1038/mt.2012.146
    • (2012) Mol Ther , vol.20 , pp. 1998-2003
    • Galanis, E.1    Markovic, S.N.2    Suman, V.J.3    Nuovo, G.J.4    Vile, R.G.5    Kottke, T.J.6
  • 9
    • 84882452568 scopus 로고    scopus 로고
    • A phase I clinical trial of Ad5/3-Delta24, a novel serotype-chimeric, infectivity-enhanced, conditionally-replicative adenovirus (CRAd), in patients with recurrent ovarian cancer
    • Kim KH, Dmitriev IP, Saddekni S, Kashentseva EA, Harris RD, Aurigemma R, et al. A phase I clinical trial of Ad5/3-Delta24, a novel serotype-chimeric, infectivity-enhanced, conditionally-replicative adenovirus (CRAd), in patients with recurrent ovarian cancer. Gynecol Oncol (2013) 130:518-24. doi:10.1016/j.ygyno.2013.06.003
    • (2013) Gynecol Oncol , vol.130 , pp. 518-524
    • Kim, K.H.1    Dmitriev, I.P.2    Saddekni, S.3    Kashentseva, E.A.4    Harris, R.D.5    Aurigemma, R.6
  • 10
    • 84879097715 scopus 로고    scopus 로고
    • REO-001: a phase I trial of percutaneous intralesional administration of reovirus type 3 dearing (Reolysin(R)) in patients with advanced solid tumors
    • Morris DG, Feng X, Difrancesco LM, Fonseca K, Forsyth PA, Paterson AH, et al. REO-001: a phase I trial of percutaneous intralesional administration of reovirus type 3 dearing (Reolysin(R)) in patients with advanced solid tumors. Invest New Drugs (2013) 31:696-706. doi:10.1007/s10637-012-9865-z
    • (2013) Invest New Drugs , vol.31 , pp. 696-706
    • Morris, D.G.1    Feng, X.2    Difrancesco, L.M.3    Fonseca, K.4    Forsyth, P.A.5    Paterson, A.H.6
  • 11
    • 33644852717 scopus 로고    scopus 로고
    • China approves world's first oncolytic virus therapy for cancer treatment
    • Garber K. China approves world's first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst (2006) 98:298-300. doi:10.1093/jnci/djj111
    • (2006) J Natl Cancer Inst , vol.98 , pp. 298-300
    • Garber, K.1
  • 12
    • 84884594361 scopus 로고    scopus 로고
    • OPTiM: a randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma
    • Andtbacka RH, Collichio FA, Amatruda T, Senzer NN, Chesney J, Delman KA, et al. OPTiM: a randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma. J Clin Oncol (2013) 31:LBA9008.
    • (2013) J Clin Oncol , vol.31
    • Andtbacka, R.H.1    Collichio, F.A.2    Amatruda, T.3    Senzer, N.N.4    Chesney, J.5    Delman, K.A.6
  • 13
    • 84988013272 scopus 로고    scopus 로고
    • Amgen to Pursue T-Vec Approval Despite PhIII Survival Miss [Online]
    • [cited 2014 Jun 24]
    • Helfand C. Amgen to Pursue T-Vec Approval Despite PhIII Survival Miss [Online]. FierceMarkets. (2014) [cited 2014 Jun 24]. Available from: http://www.fiercevaccines.com/story/amgen-pursue-t-vec-approval-despite-phiii-survival-miss/2014-06-03? utm_medium=nl&utm_source=internal
    • (2014) FierceMarkets
    • Helfand, C.1
  • 14
    • 73349133717 scopus 로고    scopus 로고
    • Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma
    • Senzer NN, Kaufman HL, Amatruda T, Nemunaitis M, Reid T, Daniels G, et al. Phase II clinical trial of a granulocyte-macrophage colony-stimulating factor-encoding, second-generation oncolytic herpesvirus in patients with unresectable metastatic melanoma. J Clin Oncol (2009) 27:5763-71. doi:10.1200/JCO.2009.24.3675
    • (2009) J Clin Oncol , vol.27 , pp. 5763-5771
    • Senzer, N.N.1    Kaufman, H.L.2    Amatruda, T.3    Nemunaitis, M.4    Reid, T.5    Daniels, G.6
  • 15
    • 80052406557 scopus 로고    scopus 로고
    • Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic poxvirus in humans
    • Breitbach CJ, Burke J, Jonker D, Stephenson J, Haas AR, Chow LQ, et al. Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic poxvirus in humans. Nature (2011) 477:99-102. doi:10.1038/nature10358
    • (2011) Nature , vol.477 , pp. 99-102
    • Breitbach, C.J.1    Burke, J.2    Jonker, D.3    Stephenson, J.4    Haas, A.R.5    Chow, L.Q.6
  • 16
    • 84875225339 scopus 로고    scopus 로고
    • Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer
    • Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med (2013) 19:329-36. doi:10.1038/nm.3089
    • (2013) Nat Med , vol.19 , pp. 329-336
    • Heo, J.1    Reid, T.2    Ruo, L.3    Breitbach, C.J.4    Rose, S.5    Bloomston, M.6
  • 18
    • 79952284127 scopus 로고    scopus 로고
    • Hallmarks of cancer: the next generation
    • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell (2011) 144:646-74. doi:10.1016/j.cell.2011.02.013
    • (2011) Cell , vol.144 , pp. 646-674
    • Hanahan, D.1    Weinberg, R.A.2
  • 19
    • 70350786392 scopus 로고    scopus 로고
    • Cyclophosphamide and cancer: golden anniversary
    • Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol (2009) 6:638-47. doi:10.1038/nrclinonc.2009.146
    • (2009) Nat Rev Clin Oncol , vol.6 , pp. 638-647
    • Emadi, A.1    Jones, R.J.2    Brodsky, R.A.3
  • 20
    • 0032814773 scopus 로고    scopus 로고
    • Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses
    • Ikeda K, Ichikawa T, Wakimoto H, Silver JS, Deisboeck TS, Finkelstein D, et al. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med (1999) 5:881-7. doi:10.1038/11320
    • (1999) Nat Med , vol.5 , pp. 881-887
    • Ikeda, K.1    Ichikawa, T.2    Wakimoto, H.3    Silver, J.S.4    Deisboeck, T.S.5    Finkelstein, D.6
  • 21
    • 0033997183 scopus 로고    scopus 로고
    • Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant
    • Ikeda K, Wakimoto H, Ichikawa T, Jhung S, Hochberg FH, Louis DN, et al. Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant. J Virol (2000) 74:4765-75. doi:10.1128/JVI.74.10.4765-4775.2000
    • (2000) J Virol , vol.74 , pp. 4765-4775
    • Ikeda, K.1    Wakimoto, H.2    Ichikawa, T.3    Jhung, S.4    Hochberg, F.H.5    Louis, D.N.6
  • 22
    • 0742271990 scopus 로고    scopus 로고
    • Altered expression of antiviral cytokine mRNAs associated with cyclophosphamide's enhancement of viral oncolysis
    • Wakimoto H, Fulci G, Tyminski E, Chiocca EA. Altered expression of antiviral cytokine mRNAs associated with cyclophosphamide's enhancement of viral oncolysis. Gene Ther (2004) 11:214-23. doi:10.1038/sj.gt.3302143
    • (2004) Gene Ther , vol.11 , pp. 214-223
    • Wakimoto, H.1    Fulci, G.2    Tyminski, E.3    Chiocca, E.A.4
  • 23
    • 33748088164 scopus 로고    scopus 로고
    • Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses
    • Fulci G, Breymann L, Gianni D, Kurozomi K, Rhee SS, Yu J, et al. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc Natl Acad Sci U S A (2006) 103:12873-8. doi:10.1073/pnas.0605496103
    • (2006) Proc Natl Acad Sci U S A , vol.103 , pp. 12873-12878
    • Fulci, G.1    Breymann, L.2    Gianni, D.3    Kurozomi, K.4    Rhee, S.S.5    Yu, J.6
  • 24
    • 42549146446 scopus 로고    scopus 로고
    • Efficacy and safety of the oncolytic herpes simplex virus rRp450 alone and combined with cyclophosphamide
    • Currier MA, Gillespie RA, Sawtell NM, Mahller YY, Stroup G, Collins MH, et al. Efficacy and safety of the oncolytic herpes simplex virus rRp450 alone and combined with cyclophosphamide. Mol Ther (2008) 16:879-85. doi:10.1038/mt.2008.49
    • (2008) Mol Ther , vol.16 , pp. 879-885
    • Currier, M.A.1    Gillespie, R.A.2    Sawtell, N.M.3    Mahller, Y.Y.4    Stroup, G.5    Collins, M.H.6
  • 25
    • 54749109597 scopus 로고    scopus 로고
    • Immunosuppression enhances oncolytic adenovirus replication and antitumor efficacy in the Syrian hamster model
    • Thomas MA, Spencer JF, Toth K, Sagartz JE, Phillips NJ, Wold WS. Immunosuppression enhances oncolytic adenovirus replication and antitumor efficacy in the Syrian hamster model. Mol Ther (2008) 16:1665-73. doi:10.1038/mt.2008.162
    • (2008) Mol Ther , vol.16 , pp. 1665-1673
    • Thomas, M.A.1    Spencer, J.F.2    Toth, K.3    Sagartz, J.E.4    Phillips, N.J.5    Wold, W.S.6
  • 26
    • 60049098649 scopus 로고    scopus 로고
    • Effect of preexisting immunity on oncolytic adenovirus vector INGN 007 antitumor efficacy in immunocompetent and immunosuppressed Syrian hamsters
    • Dhar D, Spencer JF, Toth K, Wold WS. Effect of preexisting immunity on oncolytic adenovirus vector INGN 007 antitumor efficacy in immunocompetent and immunosuppressed Syrian hamsters. J Virol (2009) 83:2130-9. doi:10.1128/JVI.02127-08
    • (2009) J Virol , vol.83 , pp. 2130-2139
    • Dhar, D.1    Spencer, J.F.2    Toth, K.3    Wold, W.S.4
  • 27
    • 80052420081 scopus 로고    scopus 로고
    • Immunological effects of low-dose cyclophosphamide in cancer patients treated with oncolytic adenovirus
    • Cerullo V, Diaconu I, Kangasniemi L, Rajecki M, Escutenaire S, Koski A, et al. Immunological effects of low-dose cyclophosphamide in cancer patients treated with oncolytic adenovirus. Mol Ther (2011) 19:1737-46. doi:10.1038/mt.2011.113
    • (2011) Mol Ther , vol.19 , pp. 1737-1746
    • Cerullo, V.1    Diaconu, I.2    Kangasniemi, L.3    Rajecki, M.4    Escutenaire, S.5    Koski, A.6
  • 28
    • 84879965657 scopus 로고    scopus 로고
    • Cyclophosphamide enhances antitumor efficacy of oncolytic adenovirus expressing uracil phosphoribosyltransferase (UPRT) in immunocompetent Syrian hamsters
    • Hasegawa N, Abei M, Yokoyama KK, Fukuda K, Seo E, Kawashima R, et al. Cyclophosphamide enhances antitumor efficacy of oncolytic adenovirus expressing uracil phosphoribosyltransferase (UPRT) in immunocompetent Syrian hamsters. Int J Cancer (2013) 133:1479-88. doi:10.1002/ijc.28132
    • (2013) Int J Cancer , vol.133 , pp. 1479-1488
    • Hasegawa, N.1    Abei, M.2    Yokoyama, K.K.3    Fukuda, K.4    Seo, E.5    Kawashima, R.6
  • 29
    • 65249177789 scopus 로고    scopus 로고
    • Efficacy of systemically administered oncolytic vaccinia virotherapy for malignant gliomas is enhanced by combination therapy with rapamycin or cyclophosphamide
    • Lun XQ, Jang JH, Tang N, Deng H, Head R, Bell JC, et al. Efficacy of systemically administered oncolytic vaccinia virotherapy for malignant gliomas is enhanced by combination therapy with rapamycin or cyclophosphamide. Clin Cancer Res (2009) 15:2777-88. doi:10.1158/1078-0432.CCR-08-2342
    • (2009) Clin Cancer Res , vol.15 , pp. 2777-2788
    • Lun, X.Q.1    Jang, J.H.2    Tang, N.3    Deng, H.4    Head, R.5    Bell, J.C.6
  • 30
    • 40749090005 scopus 로고    scopus 로고
    • Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus
    • Qiao J, Wang H, Kottke T, White C, Twigger K, Diaz RM, et al. Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus. Clin Cancer Res (2008) 14:259-69. doi:10.1158/1078-0432.CCR-07-1510
    • (2008) Clin Cancer Res , vol.14 , pp. 259-269
    • Qiao, J.1    Wang, H.2    Kottke, T.3    White, C.4    Twigger, K.5    Diaz, R.M.6
  • 31
    • 35549009347 scopus 로고    scopus 로고
    • An immunocompetent murine model for oncolysis with an armed and targeted measles virus
    • Ungerechts G, Springfeld C, Frenzke ME, Lampe J, Parker WB, Sorscher EJ, et al. An immunocompetent murine model for oncolysis with an armed and targeted measles virus. Mol Ther (2007) 15:1991-7. doi:10.1038/sj.mt.6300291
    • (2007) Mol Ther , vol.15 , pp. 1991-1997
    • Ungerechts, G.1    Springfeld, C.2    Frenzke, M.E.3    Lampe, J.4    Parker, W.B.5    Sorscher, E.J.6
  • 32
    • 78649985500 scopus 로고    scopus 로고
    • Mantle cell lymphoma salvage regimen: synergy between a reprogrammed oncolytic virus and two chemotherapeutics
    • Ungerechts G, Frenzke ME, Yaiw KC, Miest T, Johnston PB, Cattaneo R. Mantle cell lymphoma salvage regimen: synergy between a reprogrammed oncolytic virus and two chemotherapeutics. Gene Ther (2010) 17:1506-16. doi:10.1038/gt.2010.103
    • (2010) Gene Ther , vol.17 , pp. 1506-1516
    • Ungerechts, G.1    Frenzke, M.E.2    Yaiw, K.C.3    Miest, T.4    Johnston, P.B.5    Cattaneo, R.6
  • 33
    • 84874771300 scopus 로고    scopus 로고
    • Using clinically approved cyclophosphamide regimens to control the humoral immune response to oncolytic viruses
    • Peng KW, Myers R, Greenslade A, Mader E, Greiner S, Federspiel MJ, et al. Using clinically approved cyclophosphamide regimens to control the humoral immune response to oncolytic viruses. Gene Ther (2013) 20:255-61. doi:10.1038/gt.2012.31
    • (2013) Gene Ther , vol.20 , pp. 255-261
    • Peng, K.W.1    Myers, R.2    Greenslade, A.3    Mader, E.4    Greiner, S.5    Federspiel, M.J.6
  • 34
    • 59449108246 scopus 로고    scopus 로고
    • Improved systemic delivery of oncolytic reovirus to established tumors using preconditioning with cyclophosphamide-mediated Treg modulation and interleukin-2
    • Kottke T, Thompson J, Diaz RM, Pulido J, Willmon C, Coffey M, et al. Improved systemic delivery of oncolytic reovirus to established tumors using preconditioning with cyclophosphamide-mediated Treg modulation and interleukin-2. Clin Cancer Res (2009) 15:561-9. doi:10.1158/1078-0432.CCR-08-1688
    • (2009) Clin Cancer Res , vol.15 , pp. 561-569
    • Kottke, T.1    Thompson, J.2    Diaz, R.M.3    Pulido, J.4    Willmon, C.5    Coffey, M.6
  • 36
    • 79551708505 scopus 로고    scopus 로고
    • A phase I study of the combination of intravenous reovirus type 3 Dearing and gemcitabine in patients with advanced cancer
    • Lolkema MP, Arkenau HT, Harrington K, Roxburgh P, Morrison R, Roulstone V, et al. A phase I study of the combination of intravenous reovirus type 3 Dearing and gemcitabine in patients with advanced cancer. Clin Cancer Res (2011) 17:581-8. doi:10.1158/1078-0432.CCR-10-2159
    • (2011) Clin Cancer Res , vol.17 , pp. 581-588
    • Lolkema, M.P.1    Arkenau, H.T.2    Harrington, K.3    Roxburgh, P.4    Morrison, R.5    Roulstone, V.6
  • 37
    • 61349100687 scopus 로고    scopus 로고
    • Myeloid-derived suppressor cells as regulators of the immune system
    • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol (2009) 9:162-74. doi:10.1038/nri2506
    • (2009) Nat Rev Immunol , vol.9 , pp. 162-174
    • Gabrilovich, D.I.1    Nagaraj, S.2
  • 38
    • 63449117086 scopus 로고    scopus 로고
    • Oncolytic adenoviral mutants with E1B19K gene deletions enhance gemcitabine-induced apoptosis in pancreatic carcinoma cells and anti-tumor efficacy in vivo
    • Leitner S, Sweeney K, Oberg D, Davies D, Miranda E, Lemoine NR, et al. Oncolytic adenoviral mutants with E1B19K gene deletions enhance gemcitabine-induced apoptosis in pancreatic carcinoma cells and anti-tumor efficacy in vivo. Clin Cancer Res (2009) 15:1730-40. doi:10.1158/1078-0432.CCR-08-2008
    • (2009) Clin Cancer Res , vol.15 , pp. 1730-1740
    • Leitner, S.1    Sweeney, K.2    Oberg, D.3    Davies, D.4    Miranda, E.5    Lemoine, N.R.6
  • 39
    • 66449119930 scopus 로고    scopus 로고
    • Preclinical evaluation of synergistic effect of telomerase-specific oncolytic virotherapy and gemcitabine for human lung cancer
    • Liu D, Kojima T, Ouchi M, Kuroda S, Watanabe Y, Hashimoto Y, et al. Preclinical evaluation of synergistic effect of telomerase-specific oncolytic virotherapy and gemcitabine for human lung cancer. Mol Cancer Ther (2009) 8:980-7. doi:10.1158/1535-7163.MCT-08-0901
    • (2009) Mol Cancer Ther , vol.8 , pp. 980-987
    • Liu, D.1    Kojima, T.2    Ouchi, M.3    Kuroda, S.4    Watanabe, Y.5    Hashimoto, Y.6
  • 40
    • 77953359774 scopus 로고    scopus 로고
    • Combination with low-dose gemcitabine and hTERT-promoter-dependent conditionally replicative adenovirus enhances cytotoxicity through their crosstalk mechanisms in pancreatic cancer
    • Onimaru M, Ohuchida K, Nagai E, Mizumoto K, Egami T, Cui L, et al. Combination with low-dose gemcitabine and hTERT-promoter-dependent conditionally replicative adenovirus enhances cytotoxicity through their crosstalk mechanisms in pancreatic cancer. Cancer Lett (2010) 294:178-86. doi:10.1016/j.canlet.2010.01.034
    • (2010) Cancer Lett , vol.294 , pp. 178-186
    • Onimaru, M.1    Ohuchida, K.2    Nagai, E.3    Mizumoto, K.4    Egami, T.5    Cui, L.6
  • 41
    • 80052968635 scopus 로고    scopus 로고
    • An oncolytic adenovirus defective in pRb-binding (dl922-947) can efficiently eliminate pancreatic cancer cells and tumors in vivo in combination with 5-FU or gemcitabine
    • Bhattacharyya M, Francis J, Eddouadi A, Lemoine NR, Hallden G. An oncolytic adenovirus defective in pRb-binding (dl922-947) can efficiently eliminate pancreatic cancer cells and tumors in vivo in combination with 5-FU or gemcitabine. Cancer Gene Ther (2011) 18:734-43. doi:10.1038/cgt.2011.45
    • (2011) Cancer Gene Ther , vol.18 , pp. 734-743
    • Bhattacharyya, M.1    Francis, J.2    Eddouadi, A.3    Lemoine, N.R.4    Hallden, G.5
  • 42
    • 83255185076 scopus 로고    scopus 로고
    • The oncolytic adenovirus Addd enhances selective cancer cell killing in combination with DNA-damaging drugs in pancreatic cancer models
    • Cherubini G, Kallin C, Mozetic A, Hammaren-Busch K, Muller H, Lemoine NR, et al. The oncolytic adenovirus Addd enhances selective cancer cell killing in combination with DNA-damaging drugs in pancreatic cancer models. Gene Ther (2011) 18:1157-65. doi:10.1038/gt.2011.141
    • (2011) Gene Ther , vol.18 , pp. 1157-1165
    • Cherubini, G.1    Kallin, C.2    Mozetic, A.3    Hammaren-Busch, K.4    Muller, H.5    Lemoine, N.R.6
  • 43
    • 79954517666 scopus 로고    scopus 로고
    • E1A, E1B double-restricted adenovirus enhances the cytotoxicity and antitumor activity of gemcitabine to renal cell carcinoma
    • Wang H, Satoh M, Chen GP, Li DC, Hamada H, Arai Y. E1A, E1B double-restricted adenovirus enhances the cytotoxicity and antitumor activity of gemcitabine to renal cell carcinoma. Chin Med J (Engl) (2011) 124:1082-7.
    • (2011) Chin Med J (Engl) , vol.124 , pp. 1082-1087
    • Wang, H.1    Satoh, M.2    Chen, G.P.3    Li, D.C.4    Hamada, H.5    Arai, Y.6
  • 44
    • 84860217802 scopus 로고    scopus 로고
    • Effects of capsid-modified oncolytic adenoviruses and their combinations with gemcitabine or silica gel on pancreatic cancer
    • Kangasniemi L, Parviainen S, Pisto T, Koskinen M, Jokinen M, Kiviluoto T, et al. Effects of capsid-modified oncolytic adenoviruses and their combinations with gemcitabine or silica gel on pancreatic cancer. Int J Cancer (2012) 131:253-63. doi:10.1002/ijc.26370
    • (2012) Int J Cancer , vol.131 , pp. 253-263
    • Kangasniemi, L.1    Parviainen, S.2    Pisto, T.3    Koskinen, M.4    Jokinen, M.5    Kiviluoto, T.6
  • 45
    • 59449089665 scopus 로고    scopus 로고
    • Improvement of gemcitabine-based therapy of pancreatic carcinoma by means of oncolytic parvovirus H-1PV
    • Angelova AL, Aprahamian M, Grekova SP, Hajri A, Leuchs B, Giese NA, et al. Improvement of gemcitabine-based therapy of pancreatic carcinoma by means of oncolytic parvovirus H-1PV. Clin Cancer Res (2009) 15:511-9. doi:10.1158/1078-0432.CCR-08-1088
    • (2009) Clin Cancer Res , vol.15 , pp. 511-519
    • Angelova, A.L.1    Aprahamian, M.2    Grekova, S.P.3    Hajri, A.4    Leuchs, B.5    Giese, N.A.6
  • 46
    • 84899085760 scopus 로고    scopus 로고
    • Complementary induction of immunogenic cell death by oncolytic parvovirus H-1PV and gemcitabine in pancreatic cancer
    • Angelova AL, Grekova SP, Heller A, Kuhlmann O, Soyka E, Giese T, et al. Complementary induction of immunogenic cell death by oncolytic parvovirus H-1PV and gemcitabine in pancreatic cancer. J Virol (2014) 88:5263-76. doi:10.1128/JVI.03688-13
    • (2014) J Virol , vol.88 , pp. 5263-5276
    • Angelova, A.L.1    Grekova, S.P.2    Heller, A.3    Kuhlmann, O.4    Soyka, E.5    Giese, T.6
  • 47
    • 68849111614 scopus 로고    scopus 로고
    • Synergistic antitumor activity of oncolytic reovirus and chemotherapeutic agents in non-small cell lung cancer cells
    • Sei S, Mussio JK, Yang QE, Nagashima K, Parchment RE, Coffey MC, et al. Synergistic antitumor activity of oncolytic reovirus and chemotherapeutic agents in non-small cell lung cancer cells. Mol Cancer (2009) 8:47. doi:10.1186/1476-4598-8-47
    • (2009) Mol Cancer , vol.8 , pp. 47
    • Sei, S.1    Mussio, J.K.2    Yang, Q.E.3    Nagashima, K.4    Parchment, R.E.5    Coffey, M.C.6
  • 48
    • 84891859522 scopus 로고    scopus 로고
    • Gemcitabine enhances the efficacy of reovirus-based oncotherapy through anti-tumour immunological mechanisms
    • Gujar SA, Clements D, Dielschneider R, Helson E, Marcato P, Lee PW. Gemcitabine enhances the efficacy of reovirus-based oncotherapy through anti-tumour immunological mechanisms. Br J Cancer (2014) 110:83-93. doi:10.1038/bjc.2013.695
    • (2014) Br J Cancer , vol.110 , pp. 83-93
    • Gujar, S.A.1    Clements, D.2    Dielschneider, R.3    Helson, E.4    Marcato, P.5    Lee, P.W.6
  • 49
    • 84883274264 scopus 로고    scopus 로고
    • Oncolytic vesicular stomatitis virus in an immunocompetent model of MUC1-positive or MUC1-null pancreatic ductal adenocarcinoma
    • Hastie E, Besmer DM, Shah NR, Murphy AM, Moerdyk-Schauwecker M, Molestina C, et al. Oncolytic vesicular stomatitis virus in an immunocompetent model of MUC1-positive or MUC1-null pancreatic ductal adenocarcinoma. J Virol (2013) 87:10283-94. doi:10.1128/JVI.01412-13
    • (2013) J Virol , vol.87 , pp. 10283-10294
    • Hastie, E.1    Besmer, D.M.2    Shah, N.R.3    Murphy, A.M.4    Moerdyk-Schauwecker, M.5    Molestina, C.6
  • 50
    • 39749113117 scopus 로고    scopus 로고
    • Effects of tumor selective replication-competent herpes viruses in combination with gemcitabine on pancreatic cancer
    • Watanabe I, Kasuya H, Nomura N, Shikano T, Shirota T, Kanazumi N, et al. Effects of tumor selective replication-competent herpes viruses in combination with gemcitabine on pancreatic cancer. Cancer Chemother Pharmacol (2008) 61:875-82. doi:10.1007/s00280-007-0567-8
    • (2008) Cancer Chemother Pharmacol , vol.61 , pp. 875-882
    • Watanabe, I.1    Kasuya, H.2    Nomura, N.3    Shikano, T.4    Shirota, T.5    Kanazumi, N.6
  • 51
    • 84873097527 scopus 로고    scopus 로고
    • Enhanced antitumoral activity of oncolytic herpes simplex virus with gemcitabine using colorectal tumor models
    • Esaki S, Goshima F, Kimura H, Murakami S, Nishiyama Y. Enhanced antitumoral activity of oncolytic herpes simplex virus with gemcitabine using colorectal tumor models. Int J Cancer (2013) 132:1592-601. doi:10.1002/ijc.27823
    • (2013) Int J Cancer , vol.132 , pp. 1592-1601
    • Esaki, S.1    Goshima, F.2    Kimura, H.3    Murakami, S.4    Nishiyama, Y.5
  • 52
    • 58149495955 scopus 로고    scopus 로고
    • Regression of human pancreatic tumor xenografts in mice after a single systemic injection of recombinant vaccinia virus GLV-1h68
    • Yu YA, Galanis C, Woo Y, Chen N, Zhang Q, Fong Y, et al. Regression of human pancreatic tumor xenografts in mice after a single systemic injection of recombinant vaccinia virus GLV-1h68. Mol Cancer Ther (2009) 8:141-51. doi:10.1158/1535-7163.MCT-08-0533
    • (2009) Mol Cancer Ther , vol.8 , pp. 141-151
    • Yu, Y.A.1    Galanis, C.2    Woo, Y.3    Chen, N.4    Zhang, Q.5    Fong, Y.6
  • 53
    • 84862820881 scopus 로고    scopus 로고
    • Myxoma virus sensitizes cancer cells to gemcitabine and is an effective oncolytic virotherapeutic in models of disseminated pancreatic cancer
    • Wennier ST, Liu J, Li S, Rahman MM, Mona M, Mcfadden G. Myxoma virus sensitizes cancer cells to gemcitabine and is an effective oncolytic virotherapeutic in models of disseminated pancreatic cancer. Mol Ther (2012) 20:759-68. doi:10.1038/mt.2011.293
    • (2012) Mol Ther , vol.20 , pp. 759-768
    • Wennier, S.T.1    Liu, J.2    Li, S.3    Rahman, M.M.4    Mona, M.5    Mcfadden, G.6
  • 54
    • 33947356281 scopus 로고    scopus 로고
    • Bortezomib-mediated 26S proteasome inhibition causes cell-cycle arrest and induces apoptosis in CD-30+ anaplastic large cell lymphoma
    • Bonvini P, Zorzi E, Basso G, Rosolen A. Bortezomib-mediated 26S proteasome inhibition causes cell-cycle arrest and induces apoptosis in CD-30+ anaplastic large cell lymphoma. Leukemia (2007) 21:838-42. doi:10.1038/sj.leu.2404528
    • (2007) Leukemia , vol.21 , pp. 838-842
    • Bonvini, P.1    Zorzi, E.2    Basso, G.3    Rosolen, A.4
  • 55
    • 0035207110 scopus 로고    scopus 로고
    • The role of nuclear factor-kappaB in the biology and treatment of multiple myeloma
    • Berenson JR, Ma HM, Vescio R. The role of nuclear factor-kappaB in the biology and treatment of multiple myeloma. Semin Oncol (2001) 28:626-33. doi:10.1053/sonc.2001.29542
    • (2001) Semin Oncol , vol.28 , pp. 626-633
    • Berenson, J.R.1    Ma, H.M.2    Vescio, R.3
  • 56
    • 55949101420 scopus 로고    scopus 로고
    • Enhanced killing of androgen-independent prostate cancer cells using inositol hexakisphosphate in combination with proteasome inhibitors
    • Diallo JS, Betton B, Parent N, Peant B, Lessard L, Le Page C, et al. Enhanced killing of androgen-independent prostate cancer cells using inositol hexakisphosphate in combination with proteasome inhibitors. Br J Cancer (2008) 99:1613-22. doi:10.1038/sj.bjc.6604730
    • (2008) Br J Cancer , vol.99 , pp. 1613-1622
    • Diallo, J.S.1    Betton, B.2    Parent, N.3    Peant, B.4    Lessard, L.5    Le Page, C.6
  • 57
    • 29244470510 scopus 로고    scopus 로고
    • Bortezomib inhibits PKR-like endoplasmic reticulum (ER) kinase and induces apoptosis via ER stress in human pancreatic cancer cells
    • Nawrocki ST, Carew JS, Dunner K Jr., Boise LH, Chiao PJ, Huang P, et al. Bortezomib inhibits PKR-like endoplasmic reticulum (ER) kinase and induces apoptosis via ER stress in human pancreatic cancer cells. Cancer Res (2005) 65:11510-9. doi:10.1158/0008-5472.CAN-05-2370
    • (2005) Cancer Res , vol.65 , pp. 11510-11519
    • Nawrocki, S.T.1    Carew, J.S.2    Dunner, K.3    Boise, L.H.4    Chiao, P.J.5    Huang, P.6
  • 58
    • 0035284812 scopus 로고    scopus 로고
    • Proteasomes modulate balance among proapoptotic and antiapoptotic Bcl-2 family members and compromise functioning of the electron transport chain in leukemic cells
    • Marshansky V, Wang X, Bertrand R, Luo H, Duguid W, Chinnadurai G, et al. Proteasomes modulate balance among proapoptotic and antiapoptotic Bcl-2 family members and compromise functioning of the electron transport chain in leukemic cells. J Immunol (2001) 166:3130-42. doi:10.4049/jimmunol.166.5.3130
    • (2001) J Immunol , vol.166 , pp. 3130-3142
    • Marshansky, V.1    Wang, X.2    Bertrand, R.3    Luo, H.4    Duguid, W.5    Chinnadurai, G.6
  • 59
    • 84866183414 scopus 로고    scopus 로고
    • Immune mechanism of the antitumor effects generated by bortezomib
    • Chang CL, Hsu YT, Wu CC, Yang YC, Wang C, Wu TC, et al. Immune mechanism of the antitumor effects generated by bortezomib. J Immunol (2012) 189:3209-20. doi:10.4049/jimmunol.1103826
    • (2012) J Immunol , vol.189 , pp. 3209-3220
    • Chang, C.L.1    Hsu, Y.T.2    Wu, C.C.3    Yang, Y.C.4    Wang, C.5    Wu, T.C.6
  • 60
    • 43749098729 scopus 로고    scopus 로고
    • Proteasome inhibition suppresses essential immune functions of human CD4+ T cells
    • Berges C, Haberstock H, Fuchs D, Miltz M, Sadeghi M, Opelz G, et al. Proteasome inhibition suppresses essential immune functions of human CD4+ T cells. Immunology (2008) 124:234-46. doi:10.1111/j.1365-2567.2007.02761.x
    • (2008) Immunology , vol.124 , pp. 234-246
    • Berges, C.1    Haberstock, H.2    Fuchs, D.3    Miltz, M.4    Sadeghi, M.5    Opelz, G.6
  • 61
    • 84888864862 scopus 로고    scopus 로고
    • Oncolytic vesicular stomatitis virus and bortezomib are antagonistic against myeloma cells in vitro but have additive anti-myeloma activity in vivo
    • Yarde DN, Nace RA, Russell SJ. Oncolytic vesicular stomatitis virus and bortezomib are antagonistic against myeloma cells in vitro but have additive anti-myeloma activity in vivo. Exp Hematol (2013) 41:1038-49. doi:10.1016/j.exphem.2013.09.005
    • (2013) Exp Hematol , vol.41 , pp. 1038-1049
    • Yarde, D.N.1    Nace, R.A.2    Russell, S.J.3
  • 62
    • 84881061487 scopus 로고    scopus 로고
    • Reolysin is a novel reovirus-based agent that induces endoplasmic reticular stress-mediated apoptosis in pancreatic cancer
    • Carew JS, Espitia CM, Zhao W, Kelly KR, Coffey M, Freeman JW, et al. Reolysin is a novel reovirus-based agent that induces endoplasmic reticular stress-mediated apoptosis in pancreatic cancer. Cell Death Dis (2013) 4:e728. doi:10.1038/cddis.2013.259
    • (2013) Cell Death Dis , vol.4
    • Carew, J.S.1    Espitia, C.M.2    Zhao, W.3    Kelly, K.R.4    Coffey, M.5    Freeman, J.W.6
  • 63
    • 77957191110 scopus 로고    scopus 로고
    • Antitumoural immunity by virus-mediated immunogenic apoptosis inhibits metastatic growth of hepatocellular carcinoma
    • Boozari B, Mundt B, Woller N, Struver N, Gurlevik E, Schache P, et al. Antitumoural immunity by virus-mediated immunogenic apoptosis inhibits metastatic growth of hepatocellular carcinoma. Gut (2010) 59:1416-26. doi:10.1136/gut.2009.196519
    • (2010) Gut , vol.59 , pp. 1416-1426
    • Boozari, B.1    Mundt, B.2    Woller, N.3    Struver, N.4    Gurlevik, E.5    Schache, P.6
  • 64
    • 0031610288 scopus 로고    scopus 로고
    • The geometry of intercalation complex of antitumor mitoxantrone and ametantrone with DNA: molecular dynamics simulations
    • Mazerski J, Martelli S, Borowski E. The geometry of intercalation complex of antitumor mitoxantrone and ametantrone with DNA: molecular dynamics simulations. Acta Biochim Pol (1998) 45:1-11.
    • (1998) Acta Biochim Pol , vol.45 , pp. 1-11
    • Mazerski, J.1    Martelli, S.2    Borowski, E.3
  • 65
    • 0022635318 scopus 로고
    • Selective immunomodulation by the antineoplastic agent mitoxantrone. II. Nonspecific adherent suppressor cells derived from mitoxantrone-treated mice
    • Fidler JM, Dejoy SQ, Smith FR III, Gibbons JJ Jr. Selective immunomodulation by the antineoplastic agent mitoxantrone. II. Nonspecific adherent suppressor cells derived from mitoxantrone-treated mice. J Immunol (1986) 136:2747-54.
    • (1986) J Immunol , vol.136 , pp. 2747-2754
    • Fidler, J.M.1    Dejoy, S.Q.2    Smith, F.R.3    Gibbons J.J, Jr.4
  • 66
    • 20444434782 scopus 로고    scopus 로고
    • Multiple sclerosis: mitoxantrone promotes differential effects on immunocompetent cells in vitro
    • Neuhaus O, Wiendl H, Kieseier BC, Archelos JJ, Hemmer B, Stuve O, et al. Multiple sclerosis: mitoxantrone promotes differential effects on immunocompetent cells in vitro. J Neuroimmunol (2005) 168:128-37. doi:10.1016/j.jneuroim.2005.01.024
    • (2005) J Neuroimmunol , vol.168 , pp. 128-137
    • Neuhaus, O.1    Wiendl, H.2    Kieseier, B.C.3    Archelos, J.J.4    Hemmer, B.5    Stuve, O.6
  • 67
    • 33751010179 scopus 로고    scopus 로고
    • Inhibition by mitoxantrone of in vitro migration of immunocompetent cells: a possible mechanism for therapeutic efficacy in the treatment of multiple sclerosis
    • Kopadze T, Dehmel T, Hartung HP, Stuve O, Kieseier BC. Inhibition by mitoxantrone of in vitro migration of immunocompetent cells: a possible mechanism for therapeutic efficacy in the treatment of multiple sclerosis. Arch Neurol (2006) 63:1572-8. doi:10.1001/archneur.63.11.1572
    • (2006) Arch Neurol , vol.63 , pp. 1572-1578
    • Kopadze, T.1    Dehmel, T.2    Hartung, H.P.3    Stuve, O.4    Kieseier, B.C.5
  • 68
    • 84893483687 scopus 로고    scopus 로고
    • Immunosuppressive treatments in multiple sclerosis
    • Okuda DT. Immunosuppressive treatments in multiple sclerosis. Handb Clin Neurol (2014) 122:503-11. doi:10.1016/B978-0-444-52001-2.00022-4
    • (2014) Handb Clin Neurol , vol.122 , pp. 503-511
    • Okuda, D.T.1
  • 69
    • 84891828604 scopus 로고    scopus 로고
    • Combining oncolytic HSV-1 with immunogenic cell death-inducing drug mitoxantrone breaks cancer immune tolerance and improves therapeutic efficacy
    • Workenhe S, Pol JG, Lichty BD, Cummings DT, Mossman KL. Combining oncolytic HSV-1 with immunogenic cell death-inducing drug mitoxantrone breaks cancer immune tolerance and improves therapeutic efficacy. Cancer Immunol Res (2013) 1:1-11. doi:10.1158/2326-6066.CIR-13-0059-T
    • (2013) Cancer Immunol Res , vol.1 , pp. 1-11
    • Workenhe, S.1    Pol, J.G.2    Lichty, B.D.3    Cummings, D.T.4    Mossman, K.L.5
  • 70
    • 74549180404 scopus 로고    scopus 로고
    • Improved potency and selectivity of an oncolytic E1ACR2 and E1B19K deleted adenoviral mutant in prostate and pancreatic cancers
    • Oberg D, Yanover E, Adam V, Sweeney K, Costas C, Lemoine NR, et al. Improved potency and selectivity of an oncolytic E1ACR2 and E1B19K deleted adenoviral mutant in prostate and pancreatic cancers. Clin Cancer Res (2010) 16:541-53. doi:10.1158/1078-0432.CCR-09-1960
    • (2010) Clin Cancer Res , vol.16 , pp. 541-553
    • Oberg, D.1    Yanover, E.2    Adam, V.3    Sweeney, K.4    Costas, C.5    Lemoine, N.R.6
  • 71
    • 77958108351 scopus 로고    scopus 로고
    • Efficacy of oncolytic mutants targeting pRb and p53 pathways is synergistically enhanced when combined with cytotoxic drugs in prostate cancer cells and tumor xenografts
    • Radhakrishnan S, Miranda E, Ekblad M, Holford A, Pizarro MT, Lemoine NR, et al. Efficacy of oncolytic mutants targeting pRb and p53 pathways is synergistically enhanced when combined with cytotoxic drugs in prostate cancer cells and tumor xenografts. Hum Gene Ther (2010) 21:1311-25. doi:10.1089/hum.2010.019
    • (2010) Hum Gene Ther , vol.21 , pp. 1311-1325
    • Radhakrishnan, S.1    Miranda, E.2    Ekblad, M.3    Holford, A.4    Pizarro, M.T.5    Lemoine, N.R.6
  • 72
    • 84867049698 scopus 로고    scopus 로고
    • Adenovirus-mediated sensitization to the cytotoxic drugs docetaxel and mitoxantrone is dependent on regulatory domains in the E1ACR1 gene-region
    • Miranda E, Maya Pineda H, Oberg D, Cherubini G, Garate Z, Lemoine NR, et al. Adenovirus-mediated sensitization to the cytotoxic drugs docetaxel and mitoxantrone is dependent on regulatory domains in the E1ACR1 gene-region. PLoS One (2012) 7:e46617. doi:10.1371/journal.pone.0046617
    • (2012) PLoS One , vol.7
    • Miranda, E.1    Maya Pineda, H.2    Oberg, D.3    Cherubini, G.4    Garate, Z.5    Lemoine, N.R.6
  • 73
    • 84855178795 scopus 로고    scopus 로고
    • Impact of chemotherapy for colorectal cancer on regulatory T-cells and tumor immunity
    • Maeda K, Hazama S, Tokuno K, Kan S, Maeda Y, Watanabe Y, et al. Impact of chemotherapy for colorectal cancer on regulatory T-cells and tumor immunity. Anticancer Res (2011) 31:4569-74.
    • (2011) Anticancer Res , vol.31 , pp. 4569-4574
    • Maeda, K.1    Hazama, S.2    Tokuno, K.3    Kan, S.4    Maeda, Y.5    Watanabe, Y.6
  • 74
    • 23044450359 scopus 로고    scopus 로고
    • Brain tumor oncolysis with replication-conditional herpes simplex virus type 1 expressing the prodrug-activating genes, CYP2B1 and secreted human intestinal carboxylesterase, in combination with cyclophosphamide and irinotecan
    • Tyminski E, Leroy S, Terada K, Finkelstein DM, Hyatt JL, Danks MK, et al. Brain tumor oncolysis with replication-conditional herpes simplex virus type 1 expressing the prodrug-activating genes, CYP2B1 and secreted human intestinal carboxylesterase, in combination with cyclophosphamide and irinotecan. Cancer Res (2005) 65:6850-7. doi:10.1158/0008-5472.CAN-05-0154
    • (2005) Cancer Res , vol.65 , pp. 6850-6857
    • Tyminski, E.1    Leroy, S.2    Terada, K.3    Finkelstein, D.M.4    Hyatt, J.L.5    Danks, M.K.6
  • 75
    • 84864117828 scopus 로고    scopus 로고
    • The role of natural killer cells in combinatorial anti-cancer therapy using Sindbis viral vectors and irinotecan
    • Granot T, Meruelo D. The role of natural killer cells in combinatorial anti-cancer therapy using Sindbis viral vectors and irinotecan. Cancer Gene Ther (2012) 19:588-91. doi:10.1038/cgt.2012.33
    • (2012) Cancer Gene Ther , vol.19 , pp. 588-591
    • Granot, T.1    Meruelo, D.2
  • 77
    • 0033989205 scopus 로고    scopus 로고
    • Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma
    • Middleton MR, Grob JJ, Aaronson N, Fierlbeck G, Tilgen W, Seiter S, et al. Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J Clin Oncol (2000) 18:158-66.
    • (2000) J Clin Oncol , vol.18 , pp. 158-166
    • Middleton, M.R.1    Grob, J.J.2    Aaronson, N.3    Fierlbeck, G.4    Tilgen, W.5    Seiter, S.6
  • 78
    • 84875068031 scopus 로고    scopus 로고
    • Myeloablative temozolomide enhances CD8(+) T-cell responses to vaccine and is required for efficacy against brain tumors in mice
    • Sanchez-Perez LA, Choi BD, Archer GE, Cui X, Flores C, Johnson LA, et al. Myeloablative temozolomide enhances CD8(+) T-cell responses to vaccine and is required for efficacy against brain tumors in mice. PLoS One (2013) 8:e59082. doi:10.1371/journal.pone.0059082
    • (2013) PLoS One , vol.8
    • Sanchez-Perez, L.A.1    Choi, B.D.2    Archer, G.E.3    Cui, X.4    Flores, C.5    Johnson, L.A.6
  • 79
    • 68549112974 scopus 로고    scopus 로고
    • Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model
    • Banissi C, Ghiringhelli F, Chen L, Carpentier AF. Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model. Cancer Immunol Immunother (2009) 58:1627-34. doi:10.1007/s00262-009-0671-1
    • (2009) Cancer Immunol Immunother , vol.58 , pp. 1627-1634
    • Banissi, C.1    Ghiringhelli, F.2    Chen, L.3    Carpentier, A.F.4
  • 80
    • 34447509647 scopus 로고    scopus 로고
    • Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo
    • Alonso MM, Gomez-Manzano C, Jiang H, Bekele NB, Piao Y, Yung WK, et al. Combination of the oncolytic adenovirus ICOVIR-5 with chemotherapy provides enhanced anti-glioma effect in vivo. Cancer Gene Ther (2007) 14:756-61. doi:10.1038/sj.cgt.7701067
    • (2007) Cancer Gene Ther , vol.14 , pp. 756-761
    • Alonso, M.M.1    Gomez-Manzano, C.2    Jiang, H.3    Bekele, N.B.4    Piao, Y.5    Yung, W.K.6
  • 81
    • 79959715402 scopus 로고    scopus 로고
    • YB-1 dependent virotherapy in combination with temozolomide as a multimodal therapy approach to eradicate malignant glioma
    • Holzmuller R, Mantwill K, Haczek C, Rognoni E, Anton M, Kasajima A, et al. YB-1 dependent virotherapy in combination with temozolomide as a multimodal therapy approach to eradicate malignant glioma. Int J Cancer (2011) 129:1265-76. doi:10.1002/ijc.25783
    • (2011) Int J Cancer , vol.129 , pp. 1265-1276
    • Holzmuller, R.1    Mantwill, K.2    Haczek, C.3    Rognoni, E.4    Anton, M.5    Kasajima, A.6
  • 82
    • 84878530852 scopus 로고    scopus 로고
    • Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients
    • Liikanen I, Ahtiainen L, Hirvinen ML, Bramante S, Cerullo V, Nokisalmi P, et al. Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients. Mol Ther (2013) 21:1212-23. doi:10.1038/mt.2013.51
    • (2013) Mol Ther , vol.21 , pp. 1212-1223
    • Liikanen, I.1    Ahtiainen, L.2    Hirvinen, M.L.3    Bramante, S.4    Cerullo, V.5    Nokisalmi, P.6
  • 83
    • 84883438714 scopus 로고    scopus 로고
    • The timing of neural stem cell-based virotherapy is critical for optimal therapeutic efficacy when applied with radiation and chemotherapy for the treatment of glioblastoma
    • Tobias AL, Thaci B, Auffinger B, Rincon E, Balyasnikova IV, Kim CK, et al. The timing of neural stem cell-based virotherapy is critical for optimal therapeutic efficacy when applied with radiation and chemotherapy for the treatment of glioblastoma. Stem Cells Transl Med (2013) 2:655-66. doi:10.5966/sctm.2013-0039
    • (2013) Stem Cells Transl Med , vol.2 , pp. 655-666
    • Tobias, A.L.1    Thaci, B.2    Auffinger, B.3    Rincon, E.4    Balyasnikova, I.V.5    Kim, C.K.6
  • 84
    • 30344448580 scopus 로고    scopus 로고
    • Effect of chemotherapy-induced DNA repair on oncolytic herpes simplex viral replication
    • Aghi M, Rabkin S, Martuza RL. Effect of chemotherapy-induced DNA repair on oncolytic herpes simplex viral replication. J Natl Cancer Inst (2006) 98:38-50. doi:10.1093/jnci/djj003
    • (2006) J Natl Cancer Inst , vol.98 , pp. 38-50
    • Aghi, M.1    Rabkin, S.2    Martuza, R.L.3
  • 85
    • 84855486345 scopus 로고    scopus 로고
    • Oncolytic virus-mediated manipulation of DNA damage responses: synergy with chemotherapy in killing glioblastoma stem cells
    • Kanai R, Rabkin SD, Yip S, Sgubin D, Zaupa CM, Hirose Y, et al. Oncolytic virus-mediated manipulation of DNA damage responses: synergy with chemotherapy in killing glioblastoma stem cells. J Natl Cancer Inst (2012) 104:42-55. doi:10.1093/jnci/djr509
    • (2012) J Natl Cancer Inst , vol.104 , pp. 42-55
    • Kanai, R.1    Rabkin, S.D.2    Yip, S.3    Sgubin, D.4    Zaupa, C.M.5    Hirose, Y.6
  • 86
    • 79960658055 scopus 로고    scopus 로고
    • Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity
    • Jha BK, Polyakova I, Kessler P, Dong B, Dickerman B, Sen GC, et al. Inhibition of RNase L and RNA-dependent protein kinase (PKR) by sunitinib impairs antiviral innate immunity. J Biol Chem (2011) 286:26319-26. doi:10.1074/jbc.M111.253443
    • (2011) J Biol Chem , vol.286 , pp. 26319-26326
    • Jha, B.K.1    Polyakova, I.2    Kessler, P.3    Dong, B.4    Dickerman, B.5    Sen, G.C.6
  • 87
    • 77951838524 scopus 로고    scopus 로고
    • Antiangiogenic cancer therapy combined with oncolytic virotherapy leads to regression of established tumors in mice
    • Kottke T, Hall G, Pulido J, Diaz RM, Thompson J, Chong H, et al. Antiangiogenic cancer therapy combined with oncolytic virotherapy leads to regression of established tumors in mice. J Clin Invest (2010) 120:1551-60. doi:10.1172/JCI41431
    • (2010) J Clin Invest , vol.120 , pp. 1551-1560
    • Kottke, T.1    Hall, G.2    Pulido, J.3    Diaz, R.M.4    Thompson, J.5    Chong, H.6
  • 88
    • 84883741789 scopus 로고    scopus 로고
    • Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy
    • Jha BK, Dong B, Nguyen CT, Polyakova I, Silverman RH. Suppression of antiviral innate immunity by sunitinib enhances oncolytic virotherapy. Mol Ther (2013) 21:1749-57. doi:10.1038/mt.2013.112
    • (2013) Mol Ther , vol.21 , pp. 1749-1757
    • Jha, B.K.1    Dong, B.2    Nguyen, C.T.3    Polyakova, I.4    Silverman, R.H.5
  • 89
    • 84902537301 scopus 로고    scopus 로고
    • Oncolytic vaccinia virus demonstrates antiangiogenic effects mediated by targeting of VEGF
    • Hou W, Chen H, Rojas J, Sampath P, Thorne SH. Oncolytic vaccinia virus demonstrates antiangiogenic effects mediated by targeting of VEGF. Int J Cancer (2014) 135:1238-46. doi:10.1002/ijc.28747
    • (2014) Int J Cancer , vol.135 , pp. 1238-1246
    • Hou, W.1    Chen, H.2    Rojas, J.3    Sampath, P.4    Thorne, S.H.5
  • 90
    • 34548062828 scopus 로고    scopus 로고
    • Targeted inflammation during oncolytic virus therapy severely compromises tumor blood flow
    • Breitbach CJ, Paterson JM, Lemay CG, Falls TJ, Mcguire A, Parato KA, et al. Targeted inflammation during oncolytic virus therapy severely compromises tumor blood flow. Mol Ther (2007) 15:1686-93. doi:10.1038/sj.mt.6300215
    • (2007) Mol Ther , vol.15 , pp. 1686-1693
    • Breitbach, C.J.1    Paterson, J.M.2    Lemay, C.G.3    Falls, T.J.4    Mcguire, A.5    Parato, K.A.6
  • 91
    • 50549091297 scopus 로고    scopus 로고
    • The targeted oncolytic poxvirus JX-594 demonstrates antitumoral, antivascular, and anti-HBV activities in patients with hepatocellular carcinoma
    • Liu TC, Hwang T, Park BH, Bell J, Kirn DH. The targeted oncolytic poxvirus JX-594 demonstrates antitumoral, antivascular, and anti-HBV activities in patients with hepatocellular carcinoma. Mol Ther (2008) 16:1637-42. doi:10.1038/mt.2008.143
    • (2008) Mol Ther , vol.16 , pp. 1637-1642
    • Liu, T.C.1    Hwang, T.2    Park, B.H.3    Bell, J.4    Kirn, D.H.5
  • 92
  • 93
    • 80054084614 scopus 로고    scopus 로고
    • Pioneer factors: directing transcriptional regulators within the chromatin environment
    • Magnani L, Eeckhoute J, Lupien M. Pioneer factors: directing transcriptional regulators within the chromatin environment. Trends Genet (2011) 27:465-74. doi:10.1016/j.tig.2011.07.002
    • (2011) Trends Genet , vol.27 , pp. 465-474
    • Magnani, L.1    Eeckhoute, J.2    Lupien, M.3
  • 94
    • 0036264330 scopus 로고    scopus 로고
    • Enhanced tumor development in mice lacking a functional type I interferon receptor
    • Picaud S, Bardot B, De Maeyer E, Seif I. Enhanced tumor development in mice lacking a functional type I interferon receptor. J Interferon Cytokine Res (2002) 22:457-62. doi:10.1089/10799900252952244
    • (2002) J Interferon Cytokine Res , vol.22 , pp. 457-462
    • Picaud, S.1    Bardot, B.2    De Maeyer, E.3    Seif, I.4
  • 95
    • 33750321707 scopus 로고    scopus 로고
    • Interferons, immunity and cancer immunoediting
    • Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol (2006) 6:836-48. doi:10.1038/nri1961
    • (2006) Nat Rev Immunol , vol.6 , pp. 836-848
    • Dunn, G.P.1    Koebel, C.M.2    Schreiber, R.D.3
  • 97
    • 10744223476 scopus 로고    scopus 로고
    • VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents
    • Stojdl DF, Lichty BD, Tenoever BR, Paterson JM, Power AT, Knowles S, et al. VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents. Cancer Cell (2003) 4:263-75. doi:10.1016/S1535-6108(03)00241-1
    • (2003) Cancer Cell , vol.4 , pp. 263-275
    • Stojdl, D.F.1    Lichty, B.D.2    Tenoever, B.R.3    Paterson, J.M.4    Power, A.T.5    Knowles, S.6
  • 98
    • 33645797382 scopus 로고    scopus 로고
    • Epigenetic silencing of interferon-inducible genes is implicated in interferon resistance of hepatitis C virus replicon-harboring cells
    • Naka K, Abe K, Takemoto K, Dansako H, Ikeda M, Shimotohno K, et al. Epigenetic silencing of interferon-inducible genes is implicated in interferon resistance of hepatitis C virus replicon-harboring cells. J Hepatol (2006) 44:869-78. doi:10.1016/j.jhep.2006.01.030
    • (2006) J Hepatol , vol.44 , pp. 869-878
    • Naka, K.1    Abe, K.2    Takemoto, K.3    Dansako, H.4    Ikeda, M.5    Shimotohno, K.6
  • 99
    • 54449089024 scopus 로고    scopus 로고
    • Chemical targeting of the innate antiviral response by histone deacetylase inhibitors renders refractory cancers sensitive to viral oncolysis
    • Nguyen TL, Abdelbary H, Arguello M, Breitbach C, Leveille S, Diallo JS, et al. Chemical targeting of the innate antiviral response by histone deacetylase inhibitors renders refractory cancers sensitive to viral oncolysis. Proc Natl Acad Sci U S A (2008) 105:14981-6. doi:10.1073/pnas.0803988105
    • (2008) Proc Natl Acad Sci U S A , vol.105 , pp. 14981-14986
    • Nguyen, T.L.1    Abdelbary, H.2    Arguello, M.3    Breitbach, C.4    Leveille, S.5    Diallo, J.S.6
  • 100
    • 84861722003 scopus 로고    scopus 로고
    • Histone H3 lysine 9 di-methylation as an epigenetic signature of the interferon response
    • Fang TC, Schaefer U, Mecklenbrauker I, Stienen A, Dewell S, Chen MS, et al. Histone H3 lysine 9 di-methylation as an epigenetic signature of the interferon response. J Exp Med (2012) 209:661-9. doi:10.1084/jem.20112343
    • (2012) J Exp Med , vol.209 , pp. 661-669
    • Fang, T.C.1    Schaefer, U.2    Mecklenbrauker, I.3    Stienen, A.4    Dewell, S.5    Chen, M.S.6
  • 101
    • 84871946952 scopus 로고    scopus 로고
    • Epigenetic silencing of antiviral genes renders clones of Huh-7 cells permissive for hepatitis C virus replication
    • Chen Q, Denard B, Huang H, Ye J. Epigenetic silencing of antiviral genes renders clones of Huh-7 cells permissive for hepatitis C virus replication. J Virol (2013) 87:659-65. doi:10.1128/JVI.01984-12
    • (2013) J Virol , vol.87 , pp. 659-665
    • Chen, Q.1    Denard, B.2    Huang, H.3    Ye, J.4
  • 102
    • 84877596501 scopus 로고    scopus 로고
    • Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses
    • Cho H, Proll SC, Szretter KJ, Katze MG, Gale M Jr, Diamond MS. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat Med (2013) 19:458-64. doi:10.1038/nm.3108
    • (2013) Nat Med , vol.19 , pp. 458-464
    • Cho, H.1    Proll, S.C.2    Szretter, K.J.3    Katze, M.G.4    Gale, M.5    Diamond, M.S.6
  • 103
    • 3042752799 scopus 로고    scopus 로고
    • Induction of interferon-stimulated gene expression and antiviral responses require protein deacetylase activity
    • Chang HM, Paulson M, Holko M, Rice CM, Williams BR, Marie I, et al. Induction of interferon-stimulated gene expression and antiviral responses require protein deacetylase activity. Proc Natl Acad Sci U S A (2004) 101:9578-83. doi:10.1073/pnas.0400567101
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 9578-9583
    • Chang, H.M.1    Paulson, M.2    Holko, M.3    Rice, C.M.4    Williams, B.R.5    Marie, I.6
  • 104
    • 60149097656 scopus 로고    scopus 로고
    • The effects of trichostatin A on the oncolytic ability of herpes simplex virus for oral squamous cell carcinoma cells
    • Katsura T, Iwai S, Ota Y, Shimizu H, Ikuta K, Yura Y. The effects of trichostatin A on the oncolytic ability of herpes simplex virus for oral squamous cell carcinoma cells. Cancer Gene Ther (2009) 16:237-45. doi:10.1038/cgt.2008.81
    • (2009) Cancer Gene Ther , vol.16 , pp. 237-245
    • Katsura, T.1    Iwai, S.2    Ota, Y.3    Shimizu, H.4    Ikuta, K.5    Yura, Y.6
  • 105
    • 44349124084 scopus 로고    scopus 로고
    • Trichostatin A and oncolytic HSV combination therapy shows enhanced antitumoral and antiangiogenic effects
    • Liu TC, Castelo-Branco P, Rabkin SD, Martuza RL. Trichostatin A and oncolytic HSV combination therapy shows enhanced antitumoral and antiangiogenic effects. Mol Ther (2008) 16:1041-7. doi:10.1038/mt.2008.58
    • (2008) Mol Ther , vol.16 , pp. 1041-1047
    • Liu, T.C.1    Castelo-Branco, P.2    Rabkin, S.D.3    Martuza, R.L.4
  • 106
    • 50549097330 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses
    • Otsuki A, Patel A, Kasai K, Suzuki M, Kurozumi K, Chiocca EA, et al. Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol Ther (2008) 16:1546-55. doi:10.1038/mt.2008.155
    • (2008) Mol Ther , vol.16 , pp. 1546-1555
    • Otsuki, A.1    Patel, A.2    Kasai, K.3    Suzuki, M.4    Kurozumi, K.5    Chiocca, E.A.6
  • 107
    • 84861406715 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor valproic acid lessens NK cell action against oncolytic virus-infected glioblastoma cells by inhibition of STAT5/T-BET signaling and generation of gamma interferon
    • Alvarez-Breckenridge CA, Yu J, Price R, Wei M, Wang Y, Nowicki MO, et al. The histone deacetylase inhibitor valproic acid lessens NK cell action against oncolytic virus-infected glioblastoma cells by inhibition of STAT5/T-BET signaling and generation of gamma interferon. J Virol (2012) 86:4566-77. doi:10.1128/JVI.05545-11
    • (2012) J Virol , vol.86 , pp. 4566-4577
    • Alvarez-Breckenridge, C.A.1    Yu, J.2    Price, R.3    Wei, M.4    Wang, Y.5    Nowicki, M.O.6
  • 108
    • 78651234760 scopus 로고    scopus 로고
    • Enhancement of vaccinia virus based oncolysis with histone deacetylase inhibitors
    • MacTavish H, Diallo JS, Huang B, Stanford M, Le Boeuf F, De Silva N, et al. Enhancement of vaccinia virus based oncolysis with histone deacetylase inhibitors. PLoS One (2010) 5:e14462. doi:10.1371/journal.pone.0014462
    • (2010) PLoS One , vol.5
    • MacTavish, H.1    Diallo, J.S.2    Huang, B.3    Stanford, M.4    Le Boeuf, F.5    De Silva, N.6
  • 109
    • 84878621268 scopus 로고    scopus 로고
    • HDAC inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity during tumor immunotherapy
    • Bridle BW, Chen L, Lemay CG, Diallo JS, Pol J, Nguyen A, et al. HDAC inhibition suppresses primary immune responses, enhances secondary immune responses, and abrogates autoimmunity during tumor immunotherapy. Mol Ther (2013) 21:887-94. doi:10.1038/mt.2012.265
    • (2013) Mol Ther , vol.21 , pp. 887-894
    • Bridle, B.W.1    Chen, L.2    Lemay, C.G.3    Diallo, J.S.4    Pol, J.5    Nguyen, A.6
  • 110
    • 67349190247 scopus 로고    scopus 로고
    • Linking DNA methylation and histone modification: patterns and paradigms
    • Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet (2009) 10:295-304. doi:10.1038/nrg2540
    • (2009) Nat Rev Genet , vol.10 , pp. 295-304
    • Cedar, H.1    Bergman, Y.2
  • 111
    • 84887116567 scopus 로고    scopus 로고
    • DNA demethylating agents synergize with oncolytic HSV1 against malignant gliomas
    • Okemoto K, Kasai K, Wagner B, Haseley A, Meisen H, Bolyard C, et al. DNA demethylating agents synergize with oncolytic HSV1 against malignant gliomas. Clin Cancer Res (2013) 19:5952-9. doi:10.1158/1078-0432.CCR-12-3588
    • (2013) Clin Cancer Res , vol.19 , pp. 5952-5959
    • Okemoto, K.1    Kasai, K.2    Wagner, B.3    Haseley, A.4    Meisen, H.5    Bolyard, C.6
  • 112
    • 75849118878 scopus 로고    scopus 로고
    • Targeting the PI3K signaling pathway in cancer
    • Wong KK, Engelman JA, Cantley LC. Targeting the PI3K signaling pathway in cancer. Curr Opin Genet Dev (2010) 20:87-90. doi:10.1016/j.gde.2009.11.002
    • (2010) Curr Opin Genet Dev , vol.20 , pp. 87-90
    • Wong, K.K.1    Engelman, J.A.2    Cantley, L.C.3
  • 114
    • 84889087725 scopus 로고    scopus 로고
    • Targeting PI3K in cancer: any good news?
    • Martini M, Ciraolo E, Gulluni F, Hirsch E. Targeting PI3K in cancer: any good news? Front Oncol (2013) 3:108. doi:10.3389/fonc.2013.00108
    • (2013) Front Oncol , vol.3 , pp. 108
    • Martini, M.1    Ciraolo, E.2    Gulluni, F.3    Hirsch, E.4
  • 115
    • 0028170210 scopus 로고
    • A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002)
    • Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem (1994) 269:5241-8.
    • (1994) J Biol Chem , vol.269 , pp. 5241-5248
    • Vlahos, C.J.1    Matter, W.F.2    Hui, K.Y.3    Brown, R.F.4
  • 116
    • 79957881687 scopus 로고    scopus 로고
    • A novel oncolytic herpes simplex virus that synergizes with phosphoinositide 3-kinase/Akt pathway inhibitors to target glioblastoma stem cells
    • Kanai R, Wakimoto H, Martuza RL, Rabkin SD. A novel oncolytic herpes simplex virus that synergizes with phosphoinositide 3-kinase/Akt pathway inhibitors to target glioblastoma stem cells. Clin Cancer Res (2011) 17:3686-96. doi:10.1158/1078-0432.CCR-10-3142
    • (2011) Clin Cancer Res , vol.17 , pp. 3686-3696
    • Kanai, R.1    Wakimoto, H.2    Martuza, R.L.3    Rabkin, S.D.4
  • 117
    • 84908129327 scopus 로고    scopus 로고
    • PI3K inhibitor LY294002 inhibits activation of the Akt/mTOR pathway induced by an oncolytic adenovirus expressing TRAIL and sensitizes multiple myeloma cells to the oncolytic virus
    • Tong Y, Zhu W, Huang X, You L, Han X, Yang C, et al. PI3K inhibitor LY294002 inhibits activation of the Akt/mTOR pathway induced by an oncolytic adenovirus expressing TRAIL and sensitizes multiple myeloma cells to the oncolytic virus. Oncol Rep (2014) 31:1581-8. doi:10.3892/or.2014.3020
    • (2014) Oncol Rep , vol.31 , pp. 1581-1588
    • Tong, Y.1    Zhu, W.2    Huang, X.3    You, L.4    Han, X.5    Yang, C.6
  • 118
    • 33847251137 scopus 로고    scopus 로고
    • Regulatory effects of mammalian target of rapamycin-activated pathways in type I and II interferon signaling
    • Kaur S, Lal L, Sassano A, Majchrzak-Kita B, Srikanth M, Baker DP, et al. Regulatory effects of mammalian target of rapamycin-activated pathways in type I and II interferon signaling. J Biol Chem (2007) 282:1757-68. doi:10.1074/jbc.M607365200
    • (2007) J Biol Chem , vol.282 , pp. 1757-1768
    • Kaur, S.1    Lal, L.2    Sassano, A.3    Majchrzak-Kita, B.4    Srikanth, M.5    Baker, D.P.6
  • 119
    • 29244455327 scopus 로고    scopus 로고
    • Late expression of nitroreductase in an oncolytic adenovirus sensitizes colon cancer cells to the prodrug CB1954
    • Lukashev AN, Fuerer C, Chen MJ, Searle P, Iggo R. Late expression of nitroreductase in an oncolytic adenovirus sensitizes colon cancer cells to the prodrug CB1954. Hum Gene Ther (2005) 16:1473-83. doi:10.1089/hum.2005.16.1473
    • (2005) Hum Gene Ther , vol.16 , pp. 1473-1483
    • Lukashev, A.N.1    Fuerer, C.2    Chen, M.J.3    Searle, P.4    Iggo, R.5
  • 120
    • 84897920174 scopus 로고    scopus 로고
    • Rapamycin enhances adenovirus-mediated cancer imaging and therapy in pre-immunized murine hosts
    • Jiang ZK, Johnson M, Moughon DL, Kuo J, Sato M, Wu L. Rapamycin enhances adenovirus-mediated cancer imaging and therapy in pre-immunized murine hosts. PLoS One (2013) 8:e73650. doi:10.1371/journal.pone.0073650
    • (2013) PLoS One , vol.8
    • Jiang, Z.K.1    Johnson, M.2    Moughon, D.L.3    Kuo, J.4    Sato, M.5    Wu, L.6
  • 121
    • 79960924757 scopus 로고    scopus 로고
    • Rapamycin enhances the activity of oncolytic herpes simplex virus against tumor cells that are resistant to virus replication
    • Fu X, Tao L, Rivera A, Zhang X. Rapamycin enhances the activity of oncolytic herpes simplex virus against tumor cells that are resistant to virus replication. Int J Cancer (2011) 129:1503-10. doi:10.1002/ijc.25808
    • (2011) Int J Cancer , vol.129 , pp. 1503-1510
    • Fu, X.1    Tao, L.2    Rivera, A.3    Zhang, X.4
  • 122
    • 76549101731 scopus 로고    scopus 로고
    • Vesicular stomatitis virus oncolysis is potentiated by impairing mTORC1-dependent type I IFN production
    • Alain T, Lun X, Martineau Y, Sean P, Pulendran B, Petroulakis E, et al. Vesicular stomatitis virus oncolysis is potentiated by impairing mTORC1-dependent type I IFN production. Proc Natl Acad Sci U S A (2010) 107:1576-81. doi:10.1073/pnas.0912344107
    • (2010) Proc Natl Acad Sci U S A , vol.107 , pp. 1576-1581
    • Alain, T.1    Lun, X.2    Martineau, Y.3    Sean, P.4    Pulendran, B.5    Petroulakis, E.6
  • 123
    • 33846504772 scopus 로고    scopus 로고
    • Oncolytic virotherapy synergism with signaling inhibitors: rapamycin increases myxoma virus tropism for human tumor cells
    • Stanford MM, Barrett JW, Nazarian SH, Werden S, Mcfadden G. Oncolytic virotherapy synergism with signaling inhibitors: rapamycin increases myxoma virus tropism for human tumor cells. J Virol (2007) 81:1251-60. doi:10.1128/JVI.01408-06
    • (2007) J Virol , vol.81 , pp. 1251-1260
    • Stanford, M.M.1    Barrett, J.W.2    Nazarian, S.H.3    Werden, S.4    Mcfadden, G.5
  • 124
    • 84878783916 scopus 로고    scopus 로고
    • Treating brain tumor-initiating cells using a combination of myxoma virus and rapamycin
    • Zemp FJ, Lun X, Mckenzie BA, Zhou H, Maxwell L, Sun B, et al. Treating brain tumor-initiating cells using a combination of myxoma virus and rapamycin. Neuro Oncol (2013) 15:904-20. doi:10.1093/neuonc/not035
    • (2013) Neuro Oncol , vol.15 , pp. 904-920
    • Zemp, F.J.1    Lun, X.2    Mckenzie, B.A.3    Zhou, H.4    Maxwell, L.5    Sun, B.6
  • 125
    • 77953135826 scopus 로고    scopus 로고
    • A high-throughput pharmacoviral approach identifies novel oncolytic virus sensitizers
    • Diallo JS, Le Boeuf F, Lai F, Cox J, Vaha-Koskela M, Abdelbary H, et al. A high-throughput pharmacoviral approach identifies novel oncolytic virus sensitizers. Mol Ther (2010) 18:1123-9. doi:10.1038/mt.2010.67
    • (2010) Mol Ther , vol.18 , pp. 1123-1129
    • Diallo, J.S.1    Le Boeuf, F.2    Lai, F.3    Cox, J.4    Vaha-Koskela, M.5    Abdelbary, H.6
  • 126
    • 0036023433 scopus 로고    scopus 로고
    • Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells
    • Kiviharju TM, Lecane PS, Sellers RG, Peehl DM. Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells. Clin Cancer Res (2002) 8:2666-74.
    • (2002) Clin Cancer Res , vol.8 , pp. 2666-2674
    • Kiviharju, T.M.1    Lecane, P.S.2    Sellers, R.G.3    Peehl, D.M.4
  • 127
    • 67049110419 scopus 로고    scopus 로고
    • Phase I dose-escalation study of F60008, a novel apoptosis inducer, in patients with advanced solid tumours
    • Kitzen JJ, De Jonge MJ, Lamers CH, Eskens FA, Van Der Biessen D, Van Doorn L, et al. Phase I dose-escalation study of F60008, a novel apoptosis inducer, in patients with advanced solid tumours. Eur J Cancer (2009) 45:1764-72. doi:10.1016/j.ejca.2009.01.026
    • (2009) Eur J Cancer , vol.45 , pp. 1764-1772
    • Kitzen, J.J.1    De Jonge, M.J.2    Lamers, C.H.3    Eskens, F.A.4    Van Der Biessen, D.5    Van Doorn, L.6
  • 128
    • 84857381128 scopus 로고    scopus 로고
    • Tripterygium wilfordii bioactive compounds as anticancer and anti-inflammatory agents
    • Wong KF, Yuan Y, Luk JM. Tripterygium wilfordii bioactive compounds as anticancer and anti-inflammatory agents. Clin Exp Pharmacol Physiol (2012) 39:311-20. doi:10.1111/j.1440-1681.2011.05586.x
    • (2012) Clin Exp Pharmacol Physiol , vol.39 , pp. 311-320
    • Wong, K.F.1    Yuan, Y.2    Luk, J.M.3
  • 129
    • 84861144099 scopus 로고    scopus 로고
    • Triptolide: structural modifications, structure-activity relationships, bioactivities, clinical development and mechanisms
    • Zhou ZL, Yang YX, Ding J, Li YC, Miao ZH. Triptolide: structural modifications, structure-activity relationships, bioactivities, clinical development and mechanisms. Nat Prod Rep (2012) 29:457-75. doi:10.1039/c2np00088a
    • (2012) Nat Prod Rep , vol.29 , pp. 457-475
    • Zhou, Z.L.1    Yang, Y.X.2    Ding, J.3    Li, Y.C.4    Miao, Z.H.5
  • 130
    • 84887450116 scopus 로고    scopus 로고
    • Triptolide-mediated inhibition of interferon signaling enhances vesicular stomatitis virus-based oncolysis
    • Ben Yebdri F, Van Grevenynghe J, Tang VA, Goulet ML, Wu JH, Stojdl DF, et al. Triptolide-mediated inhibition of interferon signaling enhances vesicular stomatitis virus-based oncolysis. Mol Ther (2013) 21:2043-53. doi:10.1038/mt.2013.187
    • (2013) Mol Ther , vol.21 , pp. 2043-2053
    • Ben Yebdri, F.1    Van Grevenynghe, J.2    Tang, V.A.3    Goulet, M.L.4    Wu, J.H.5    Stojdl, D.F.6
  • 131
    • 77950684805 scopus 로고    scopus 로고
    • Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms
    • Quintas-Cardama A, Vaddi K, Liu P, Manshouri T, Li J, Scherle PA, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood (2010) 115:3109-17. doi:10.1182/blood-2009-04-214957
    • (2010) Blood , vol.115 , pp. 3109-3117
    • Quintas-Cardama, A.1    Vaddi, K.2    Liu, P.3    Manshouri, T.4    Li, J.5    Scherle, P.A.6
  • 133
    • 79551575494 scopus 로고    scopus 로고
    • Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond
    • Quintas-Cardama A, Kantarjian H, Cortes J, Verstovsek S. Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond. Nat Rev Drug Discov (2011) 10:127-40. doi:10.1038/nrd3264
    • (2011) Nat Rev Drug Discov , vol.10 , pp. 127-140
    • Quintas-Cardama, A.1    Kantarjian, H.2    Cortes, J.3    Verstovsek, S.4
  • 136
    • 80052475970 scopus 로고    scopus 로고
    • Vesicular stomatitis virus has extensive oncolytic activity against human sarcomas: rare resistance is overcome by blocking interferon pathways
    • Paglino JC, van den Pol AN. Vesicular stomatitis virus has extensive oncolytic activity against human sarcomas: rare resistance is overcome by blocking interferon pathways. J Virol (2011) 85:9346-58. doi:10.1128/JVI.00723-11
    • (2011) J Virol , vol.85 , pp. 9346-9358
    • Paglino, J.C.1    van den Pol, A.N.2
  • 137
    • 84883281279 scopus 로고    scopus 로고
    • Influence of the oncolytic parvovirus H-1, CTLA-4 antibody tremelimumab and cytostatic drugs on the human immune system in a human in vitro model of colorectal cancer cells
    • Heinrich B, Goepfert K, Delic M, Galle PR, Moehler M. Influence of the oncolytic parvovirus H-1, CTLA-4 antibody tremelimumab and cytostatic drugs on the human immune system in a human in vitro model of colorectal cancer cells. Onco Targets Ther (2013) 6:1119-27. doi:10.2147/OTT.S49371
    • (2013) Onco Targets Ther , vol.6 , pp. 1119-1127
    • Heinrich, B.1    Goepfert, K.2    Delic, M.3    Galle, P.R.4    Moehler, M.5
  • 138
    • 84897476768 scopus 로고    scopus 로고
    • Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy
    • Zamarin D, Holmgaard RB, Subudhi SK, Park JS, Mansour M, Palese P, et al. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci Transl Med (2014) 6:226ra232. doi:10.1126/scitranslmed.3008095
    • (2014) Sci Transl Med , vol.6
    • Zamarin, D.1    Holmgaard, R.B.2    Subudhi, S.K.3    Park, J.S.4    Mansour, M.5    Palese, P.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.