39
Views
2
CrossRef citations to date
0
Altmetric
Review

Oncolytic viruses: a step into cancer immunotherapy

, &
Pages 1-21 | Published online: 28 Dec 2011

References

  • Lopez AD, Mathers CD, Ezzati M, et al. Global burden of disease and risk factors. Washington (DC): World Bank; 2006.
  • Danaei G, Vander Hoorn S, Lopez AD, et al. Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet. 2005;366:1784–1793.
  • McLaughlin JR, Risch HA, Lubinski J, et al. Reproductive risk factors for ovarian cancer in carriers of BRCA1 or BRCA2 mutations: a case- control study. Lancet Oncol. 2007;8:26–34.
  • Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3:e442.
  • Dixon K, Kopras E. Genetic alterations and DNA repair in human carcinogenesis. Semin Cancer Biol. 2004;14:441–448.
  • Borlado LR, Mendez J. CDC6: from DNA replication to cell cycle checkpoints and oncogenesis. Carcinogenesis. 2008;29:237–243.
  • Pearson CA, Prozialeck WC. E-cadherin, beta-catenin and cadmium carcinogenesis. Med Hypotheses. 2001;56:573–581.
  • Lengauer C, Wang Z. From spindle checkpoint to cancer. Nat Genet. 2004;36:1144–1145.
  • Hanks S, Coleman K, Reid S, et al. Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B. Nat Genet 2004;36:1159–1161.
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
  • Shankaran V Ikeda H, Bruce AT, et al. IFNgamma and lymphocytes prevent primary tumor development and shape tumor immunogenicity. Nature. 2001;410:1107–1111.
  • Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–998.
  • Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ. Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 2011 ;29: 235–271.
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570.
  • Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 2007;7:834–846.
  • Bernstein SH, Burack WR. The incidence, natural history, biology, and treatment of transformed lymphomas. Hematology Am Soc Hematol Educ Program. 2009:532–541.
  • Taylor HS, Manson JE. Update in hormone therapy use in menopause. J Clin Endocrinol Metab. 2011;96:255–264.
  • Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol. 2009;27:83–117.
  • Wierda WG, Kipps TJ, Durig J, et al. Chemoimmunotherapy with ofatumumab, fludarabine, and cyclophosphamide (O-FC) in previously untreated patients with chronic lymphocytic leukemia. Blood. 2011;117:6450–6458.
  • Stopeck AT, Lipton A, Body JJ, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol. 2010;28:5132–5139.
  • Culver ME, Gatesman ML, Mancl EE, Lowe DK. Ipilimumab: a novel treatment for metastatic melanoma. Ann Pharmacother. 2011 ;45: 510–519.
  • Di Lorenzo G, Perdona S, Damiano R, et al. Gemcitabine versus bacille Calmette-Guerin after initial bacille Calmette-Guerin failure in nonmuscle-invasive bladder cancer: a multicenter prospective randomized trial. Cancer. 2010;116:1893–1900.
  • Boozari B, Mundt B, Woller N, et al. Antitumoral immunity by virusmediated immunogenic apoptosis inhibits metastatic growth of hepatocellular carcinoma. Gut. 2010;59:1416–1426.
  • Kirkwood JM, Tarhini AA, Moschos SJ, Panelli MC. Adjuvant therapy with high-dose interferon alpha 2b in patients with high-risk stage IIB/III melanoma. Nat Clin Pract Oncol. 2008;5:2–3.
  • Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer JSci Am. 2000;6(Suppl 1):S11-S14.
  • Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol. 1999;17:2105–2116.
  • Hurteau JA, Brady MF, Darcy KM, et al. Randomized phase III trial of tamoxifen versus thalidomide in women with biochemical-recurrent- only epithelial ovarian, fallopian tube or primary peritoneal carcinoma after a complete response to first-line platinum/taxane chemotherapy with an evaluation of serum vascular endothelial growth factor (VEGF): a Gynecologic Oncology Group Study. Gynecol Oncol. 2010;119:444–450.
  • Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreted patients with metastatic melanoma using T cell transfer immunotherapy. Clin Cancer Res. April 15, 2011. [Epub ahead of print.]
  • Rosenberg SA, Dudley ME. Adoptive cell therapy for the treatment of patients with metastatic melanoma. Curr Opin Immunol. 2009;21:233–240.
  • Gilboa E. DC-based cancer vaccines. J Clin Invest. 2007;117:1195–1203.
  • Valmori D, Souleimanian NE, Tosello V, et al. Vaccination with NY- ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming. Proc Natl Acad Sci U S A. 2007;104:8947–8952.
  • Harada N, Hoshiai K, Takahashi Y, et al. Preclinical safety pharmacology study of a novel protein-based cancer vaccine CHP-NY-ESO-1. Kobe J Med Sci. 2008;54:E23-E34.
  • Odunsi K, Qian F, Matsuzaki J, et al. Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci U SA. 2007;104:12837–12842.
  • Gnjatic S, Altorki NK, Tang DN, et al. NY-ESO-1 DNA vaccine induces T-cell responses that are suppressed by regulatory T cells. Clin Cancer Res. 2009;15:2130–2139.
  • Nishikawa H, Sato E, Briones G, et al. In vivo antigen delivery by a Salmonella typhimurium type III secretion system for therapeutic cancer vaccines. J Clin Invest. 2006;116:1946–1954.
  • Jager E, Karbach J, Gnjatic S, et al. Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1- specific immune responses in cancer patients. Proc Natl Acad Sci U S A. 2006;103:14453–14458.
  • Almeida LG, Sakabe NJ, deOliveira AR, et al. CT database: a knowledge-base of high-throughput and curated data on cancer-testis antigens. Nucleic Acids Res. 2009;37(database issue):D816-D819.
  • Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets for immunotherapy. Cancer Sci. 2009;100:2014–2021.
  • Brichard VG, Lejeune D. GSK’s antigen-specific cancer immunotherapy programme: pilot results leading to phase III clinical development. Vaccine. 2007;25(Suppl 2):B61-B71.
  • Gnjatic S, Nishikawa H, Jungbluth AA, et al. NY-ESO-1: review of an immunogenic tumor antigen. Adv Cancer Res. 2006;95:1–30.
  • Davis ID, Chen W, Jackson H, et al. Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4(+) and CD8(+) T cell responses in humans. Proc Natl Acad Sci USA. 2004;101:10697–10702.
  • Marchand M, van Baren N, Weynants P, et al. Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer. 1999;80:219–230.
  • Alvaro T, de la Cruz-Merino L, Henao-Carrasco F, et al. Tumor microenvironment and immune effects of antineoplastic therapy in lymphoproliferative syndromes. J Biomed Biotechnol. August 12, 2010. [Epub ahead of print.]
  • Alvarez-Rueda N, Ladjemi MZ, Behar G, et al. A llama single domain anti-idiotypic antibody mimicking HER2 as a vaccine: Immunogenicity and efficacy. Vaccine. 2009;27:4826–4833.
  • Egri G, Takats A. Monoclonal antibodies in the treatment of lung cancer. Eur J Surg Oncol. 2006;32:385–394.
  • Palucka K, Ueno H, Banchereau J. Recent developments in cancer vaccines. J Immunol 2011;186:1325–1331.
  • Nair S, McLaughlin C, Weizer A, et al. Injection of immature dendritic cells into adjuvant-treated skin obviates the need for ex vivo maturation. J Immunol. 2003;171:6275–6282.
  • Martin-Fontecha A, Sebastiani S, Hopken UE, et al. Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J Exp Med. 2003;198:615–621.
  • Schuler G. Dendritic cells in cancer immunotherapy. Eur J Immunol. 2010;40:2123–2130.
  • Boudreau JE, Bonehill A, Thielemans K, Wan Y. Engineering dendritic cells to enhance cancer immunotherapy. Mol Ther. 2011;19:841–853.
  • Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422.
  • Hovden AO, Appel S. The first dendritic cell-based therapeutic cancer vaccine is approved by the FDA. Scand J Immunol. 2010;72:554.
  • Mackiewicz J, Mackiewicz A. Gene-modified cellular vaccines: technologic aspects and clinical problems. Transplant Proc. 2010;42:3287–3292.
  • Moschella F, Proietti E, Capone I, Belardelli F. Combination strategies for enhancing the efficacy of immunotherapy in cancer patients. Ann N Y Acad Sci. 2010;1194:169–178.
  • Nowak AK, Lake RA, Robinson BW. Combined chemoimmunotherapy of solid tumors: improving vaccines? Adv Drug Deliv Rev. 2006;58:975–990.
  • Moschella F, Valentini M, Arico E, et al. Unraveling cancer chemoim- munotherapy mechanisms by gene and protein expression profiling of responses to cyclophosphamide. Cancer Res. 2011;71:3528–3539.
  • Antonarakis ES, Carducci MA. Combining low-dose cyclophosphamide with GM-CSF-secreting prostate cancer immunotherapy enhances antitumor immune effects. Expert Opin InvestigDrugs. 2010;19:311–314.
  • Orlandi F, Guevara-Patino JA, Merghoub T, et al. Combination of epitope-optimized DNA vaccination and passive infusion of monoclonal antibody against HER2/neu leads to breast tumor regression in mice. Vaccine. 2011;29:3646–3654.
  • Ladjemi MZ, Chardes T, Corgnac S, et al. Vaccination with human anti-trastuzumab anti-idiotype scFv reverses HER2 immunological tolerance and induces tumor immunity in MMTV.f.huHER2(Fo5) mice. Breast Cancer Res. 2011;13:R17.
  • Ribas A, Weber JS, Chmielowski B, et al. Intra-lymph node prime- boost vaccination against melan A and tyrosinase for the treatment of metastatic melanoma: results of a phase 1 clinical trial. Clin Cancer Res. 2011;17:2987–2996.
  • Akbulut H, Tang Y, Akbulut KG, et al. Addition of adenoviral vector targeting of chemotherapy to the MUC-1/ecdCD40L VPPP vector prime protein boost vaccine prolongs survival of mice carrying growing subcutaneous deposits of Lewis lung cancer cells. Gene Ther. 2010;17:1333–1340.
  • Ishizaki H, Song GY, Srivastava T, et al. Heterologous prime/boost immunization with p53-based vaccines combined with toll-like receptor stimulation enhances tumor regression. JImmunother. 2010;33:609–617.
  • Dangoor A, Lorigan P, Keilholz U, et al. Clinical and immunological responses in metastatic melanoma patients vaccinated with a highdose poly-epitope vaccine. Cancer Immunol Immunother. 2010;59:863–873.
  • Hodge JW, Higgins J, Schlom J. Harnessing the unique local immunostimulatory properties of modified vaccinia Ankara (MVA) virus to generate superior tumor-specific immune responses and antitumor activity in a diversified prime and boost vaccine regimen. Vaccine. 2009;27:4475–4482.
  • Adamina M, Rosenthal R, Weber WP, et al. Intranodal immunization with a vaccinia virus encoding multiple antigenic epitopes and costimulatory molecules in metastatic melanoma. Mol Ther. 2010;18:651–659.
  • Kono K, Mizukami Y, Daigo Y, et al. Vaccination with multiple peptides derived from novel cancer-testis antigens can induce specific T-cell responses and clinical responses in advanced esophageal cancer. Cancer Sci. 2009;100:1502–1509.
  • Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Mol Ther. 2007;15:651–659.
  • Moore AE. The destructive effect of the virus of Russian Far East encephalitis on the transplantable mouse sarcoma 180. Cancer. 1949;2:525–534.
  • Moore AE. Inhibition of growth of five transplantable mouse tumors by the virus of Russian Far East encephalitis. Cancer. 1951;4:375–382.
  • Martuza RL, Malick A, Markert JM, et al. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science. 1991;252:854–856.
  • Rowan K. Oncolytic viruses move forward in clinical trials. J Natl Cancer Inst. 2010;102:590–595.
  • Garber K. China approves world’s first oncolytic virus therapy for cancer treatment. J Natl Cancer Inst. 2006;98:298–300.
  • Pesonen S, Kangasniemi L, Hemminki A. Oncolytic adenoviruses for the treatment of human cancer: focus on translational and clinical data. Mol Pharm. 2011;8:12–28.
  • Shashkova EV May SM, Barry MA. Characterization of human adenovirus serotypes 5, 6, 11, and 35 as anticancer agents. Virology. 2009;394:311–320.
  • Hemminki O, Bauerschmitz G, Hemmi S, et al. Oncolytic adenovirus based on serotype 3. Cancer Gene Ther. 2011;18:288–296.
  • Silver J, Mei YF. Transduction and oncolytic profile of a potent replication-competent adenovirus 11p vector (RCAd11pGFP) in colon carcinoma cells. PLoS One. 2011;6:e17532.
  • Kaufman HL, Bines SD. OPTIM trial: a phase III trial of an oncolytic herpes virus encoding GM-CSF for unresectable stage III or IV melanoma. Future Oncol. 2010;6:941–949.
  • Li H, Zeng Z, Fu X, Zhang X. Coadministration of a herpes simplex virus-2 based oncolytic virus and cyclophosphamide produces a synergistic antitumor effect and enhances tumor-specific immune responses. Cancer Res. 2007;67:7850–7855.
  • Li H, Dutuor A, Tao L, et al. Virotherapy with a type 2 herpes simplex virus-derived oncolytic virus induces potent antitumor immunity against neuroblastoma. Clin Cancer Res. 2007;13:316–322.
  • Rodrigues R, Cuddington B, Mossman K. Bovine herpesvirus type 1 as a novel oncolytic virus. Cancer Gene Ther. 2010;17:344–355.
  • Shiau AL, Lin YP, Shieh GS, et al. Development of a conditionally replicating pseudorabies virus for HER-2/neu-overexpressing bladder cancer therapy. Mol Ther. 2007;15:131–138.
  • Redaelli M, Mucignat-Caretta C, Cavaggioni A, et al. Bovine herpesvirus 4 based vector as a potential oncolytic-virus for treatment of glioma. Virol J. 2010;7:298.
  • Stevenson AJ, Giles MS, Hall KT, et al. Specific oncolytic activity of herpesvirus saimiri in pancreatic cancer cells. Br J Cancer. August 2000;83:329–332.
  • Wollmann G, Tattersall P, van den Pol AN. Targeting human glioblastoma cells: comparison of nine viruses with oncolytic potential. J Virol. 2005;79:6005–6022.
  • Breitbach CJ, Burke J, Jonker D, et al. Intravenous delivery of a multi- mechanistic cancer-targeted oncolytic poxvirus in humans. Nature. 2011;477:99–102.
  • Evgin L, Vaha-Koskela M, Rintoul J, et al. Potent oncolytic activity of raccoonpox virus in the absence of natural pathogenicity. Mol Ther. 2010;18:896–902.
  • Lun X, Alain T, Zemp FJ, et al. Myxoma virus virotherapy for glioma in immunocompetent animal models: optimizing administration routes and synergy with rapamycin. Cancer Res. 2010;70:598–608.
  • Wurdinger T, Verheije MH, Raaben M, et al. Targeting non-human coronaviruses to human cancer cells using a bispecific single-chain antibody. Gene Ther. 2005;12:1394–1404.
  • Verheije MH, Lamfers ML, Wurdinger T, et al. Coronavirus genetically redirected to the epidermal growth factor receptor exhibits effective antitumor activity against a malignant glioblastoma. J Virol. 2009;83:7507–7516.
  • Sturlan S, Stremitzer S, Bauman S, et al. Endogenous expression of proteases in colon cancer cells facilitate influenza A viruses mediated oncolysis. Cancer Biol Ther. 2010;10:592–599.
  • Galanis E, Hartmann LC, Cliby WA, et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res. 2010;70:875–882.
  • Hasegawa Y, Kinoh H, Iwadate Y, et al. Urokinase-targeted fusion by oncolytic Sendai virus eradicates orthotopic glioblastomas by pronounced synergy with interferon-beta gene. Mol Ther. 2010;18:1778–1786.
  • Myers R, Greiner S, Harvey M, et al. Oncolytic activities of approved mumps and measles vaccines for therapy of ovarian cancer. Cancer Gene Ther. 2005;12:593–599.
  • Roos FC, Roberts AM, Hwang II, et al. Oncolytic targeting of renal cell carcinoma via encephalomyocarditis virus. EMBO Mol Med. 2010;2:275–288.
  • Adachi M, Brooks SE, Stein MR, et al. Destruction of human retinoblastoma after treatment by the E variant of encephalomyocarditis virus. J Neurooncol. 2006;77:233–240.
  • Au GG, Beagley LG, Haley ES, et al. Oncolysis of malignant human melanoma tumors by Coxsackieviruses A13, A15 and A18. Virol J. 2011;8:22.
  • Toyoda H, Wimmer E, Cello J. Oncolytic poliovirus therapy and immunization with poliovirus-infected cell lysate induces potent antitumor immunity against neuroblastoma in vivo. Int J Oncol. 2011;38:81–87.
  • Haley ES, Au GG, Carlton BR, et al. Regional administration of oncolytic Echovirus 1 as a novel therapy for the peritoneal dissemination of gastric cancer. J Mol Med. 2009;87:385–399.
  • Berry LJ, Au GG, Barry RD, Shafren DR. Potent oncolytic activity of human enteroviruses against human prostate cancer. Prostate. 2008;68:577–587.
  • Shafren DR, Sylvester D, Johansson ES, et al. Oncolysis of human ovarian cancers by echovirus type 1. Int J Cancer. 2005;115:320–328.
  • Smyth M, Symonds A, Brazinova S, Martin J. Bovine enterovirus as an oncolytic virus: foetal calf serum facilitates its infection of human cells. Int J Mol Med. 2002;10:49–53.
  • Hu J, Dong CY, Li JK, et al. Selective in vitro cytotoxic effect of human cancer cells by bluetongue virus-10. Acta Oncol. 2008;47:124–134.
  • Kubo S, Haga K, Tamamoto A, et al. Adenovirus-retrovirus hybrid vectors achieve highly enhanced tumor transduction and antitumor efficacy in vivo. Mol Ther. 2011;19:76–82.
  • Duerner LJ, Schwantes A, Schneider IC, et al. Cell entry targeting restricts biodistribution of replication-competent retroviruses to tumor tissue. Gene Ther. 2008;15:1500–1510.
  • Heinkelein M, Hoffmann U, Lucke M, et al. Experimental therapy of allogeneic solid tumors induced in athymic mice with suicide gene- transducing replication-competent foamy virus vectors. Cancer Gene Ther. 2005;12:947–953.
  • Ausubel LJ, Meseck M, Derecho I, et al. Current good manufacturing practice production of an oncolytic recombinant vesicular stomatitis viral vector for cancer treatment. Hum Gene Ther. 2011 ;22: 489–497.
  • Brun J, McManus D, Lefebvre C, et al. Identification of genetically modified Maraba virus as an oncolytic rhabdovirus. Mol Ther. 2010;18:1440–1449.
  • Zhang YQ, Tsai YC, Monie A, et al. Enhancing the therapeutic effect against ovarian cancer through a combination of viral oncolysis and antigen-specific immunotherapy. Mol Ther. 2010;18:692–699.
  • Quetglas JI, Ruiz-Guillen M, Aranda A, et al. Alphavirus vectors for cancer therapy. Virus Res. 2010;153:179–196.
  • Tseng JC, Granot T, Digiacomo V et al. Enhanced specific delivery and targeting of oncolytic Sindbis viral vectors by modulating vascular leakiness in tumor. Cancer Gene Ther. 2010;17:244–255.
  • Unno Y, Shino Y, Kondo F, et al. Oncolytic viral therapy for cervical and ovarian cancer cells by Sindbis virus AR339 strain. Clin Cancer Res. 2005;11:4553–4560.
  • Berno V Porrini D, Castiglioni F, et al. The 67 kDa laminin receptor increases tumor aggressiveness by remodeling laminin-1. Endocr Relat Cancer. 2005;12:393–406.
  • Wang KS, Kuhn RJ, Strauss EG, et al. High-affinity laminin receptor is a receptor for Sindbis virus in mammalian cells. J Virol. 1992;66:4992–5001.
  • Shafren DR, Au GG, Nguyen T, et al. Systemic therapy of malignant human melanoma tumors by a common cold-producing enterovirus, coxsackievirus a21. Clin Cancer Res. 2004;10:53–60.
  • Bartee E, McFadden G. Human cancer cells have specifically lost the ability to induce the synergistic state caused by tumor necrosis factor plus interferon-beta. Cytokine. 2009;47:199–205.
  • Noser JA, Mael AA, Sakuma R, et al. The RAS/Raf1 /MEK/ERK signaling pathway facilitates VSV-mediated oncolysis: implication for the defective interferon response in cancer cells. Mol Ther. 2007;15:1531–1536.
  • Rommelaere J, Geletneky K, Angelova AL, et al. Oncolytic parvoviruses as cancer therapeutics. Cytokine Growth Factor Rev. 2010;21:185–195.
  • Mansour M, Palese P, Zamarin D. Oncolytic specificity of Newcastle disease virus is mediated by selectivity for apoptosis-resistant cells. J Virol. 2011;85:6015–6023.
  • Kelly K, Nawrocki S, Mita A, et al. Reovirus-based therapy for cancer. Expert Opin Biol Ther. 2009;9:817–830.
  • Stanford MM, McFadden G. Myxoma virus and oncolytic virotherapy: a new biologic weapon in the war against cancer. Expert Opin Biol Ther. 2007;7:1415–1425.
  • Kolodkin-Gal D, Zamir G, Edden Y, et al. Herpes simplex virus type 1 preferentially targets human colon carcinoma: role of extracellular matrix. J Virol. 2008;82:999–1010.
  • Coughlan L, Vallath S, Saha A, et al. In vivo retargeting of adenovirus type 5 to alphavbeta6 integrin results in reduced hepatotoxicity and improved tumor uptake following systemic delivery. J Virol. 2009;83:6416–6428.
  • Grandi P, Fernandez J, Szentirmai O, et al. Targeting HSV-1 virions for specific binding to epidermal growth factor receptor-vIII-bearing tumor cells. Cancer Gene Ther. 2010;17:655–663.
  • Menotti L, Nicoletti G, Gatta V et al. Inhibition of human tumor growth in mice by an oncolytic herpes simplex virus designed to target solely HER-2-positive cells. Proc Natl Acad Sci U S A. 2009;106:9039–9044.
  • Liu C, Hasegawa K, Russell SJ, et al. Prostate-specific membrane antigen retargeted measles virotherapy for the treatment of prostate cancer. Prostate. 2009;69:1128–1141.
  • Jing Y, Tong C, Zhang J, et al. Tumor and vascular targeting of a novel oncolytic measles virus retargeted against the urokinase receptor. Cancer Res. 2009;69:1459–1468.
  • Hasegawa K, Nakamura T, Harvey M, et al. The use of a tropism- modified measles virus in folate receptor-targeted virotherapy of ovarian cancer. Clin Cancer Res. 2006;12:6170–6178.
  • Nakamura T, Peng KW, Harvey M, et al. Rescue and propagation of fully retargeted oncolytic measles viruses. Nat Biotechnol. 2005;23:209–214.
  • Menotti L, Cerretani A, Campadelli-Fiume G. A herpes simplex virus recombinant that exhibits a single-chain antibody to HER2/neu enters cells through the mammary tumor receptor, independently of the gD receptors. J Virol. 2006;80:5531–5539.
  • Kinoh H, Inoue M, Komaru A, et al. Generation of optimized and urokinase-targeted oncolytic Sendai virus vectors applicable for various human malignancies. Gene Ther. 2009;16:392–403.
  • Springfeld C, von Messling V Frenzke M, et al. Oncolytic efficacy and enhanced safety of measles virus activated by tumor-secreted matrix metalloproteinases. Cancer Res. 2006;66:7694–7700.
  • Wakimoto H, Kesari S, Farrell CJ, et al. Human glioblastoma-derived cancer stem cells: establishment of invasive glioma models and treatment with oncolytic herpes simplex virus vectors. Cancer Res. 2009;69:3472–3481.
  • Nakamura H, Kasuya H, Mullen JT, et al. Regulation of herpes simplex virus gamma(1)34.5 expression and oncolysis of diffuse liver metastases by Myb34.5. J Clin Invest. 2002;109:871–882.
  • Fukuda K, Abei M, Ugai H, et al. E1A, E1B double-restricted replicative adenovirus at low dose greatly augments tumor-specific suicide gene therapy for gallbladder cancer. Cancer Gene Ther. 2009;16:126–136.
  • Fukuda K, Abei M, Ugai H, et al. E1A, E1B double-restricted adenovirus for oncolytic gene therapy of gallbladder cancer. Cancer Res. 2003;63:4434–4440.
  • Thorne SH, Hwang TH, O’Gorman WE, et al. Rational strain selection and engineering creates a broad-spectrum, systemically effective oncolytic poxvirus, JX-963. J Clin Invest. 2007;117:3350–3358.
  • Doloff JC, Jounaidi Y, Waxman DJ. Dual E1A oncolytic adenovirus: targeting tumor heterogeneity with two independent cancer-specific promoter elements, DF3/MUC1 and hTERT. Cancer Gene Ther. 2011;18:153–166.
  • Lee CY, Bu LX, DeBenedetti A, et al. Transcriptional and translational dual-regulated oncolytic herpes simplex virus type 1 for targeting prostate tumors. Mol Ther. 2010;18:929–935.
  • Zhang KJ, Wang YG, Cao X, et al. Potent antitumor effect of interleukin-24 gene in the survivin promoter and retinoblastoma double-regulated oncolytic adenovirus. Hum Gene Ther. 2009;20:818–830.
  • Hardcastle J, Kurozumi K, Chiocca EA, Kaur B. Oncolytic viruses driven by tumor-specific promoters. Curr Cancer Drug Targets. 2007;7:181–189.
  • Hamada K, Zhang T, Desaki J, et al. Carrier cell-mediated cell lysis of squamous cell carcinoma cells by squamous cell carcinoma antigen 1 promoter-driven oncolytic adenovirus. J Gene Med. 2010;12:545–554.
  • Hikichi M, Kidokoro M, Haraguchi T, et al. MicroRNA regulation of glycoprotein B5R in oncolytic vaccinia virus reduces viral pathogenicity without impairing its antitumor efficacy. Mol Ther. 2011;19:1107–1115.
  • Kelly EJ, Hadac EM, Greiner S, Russell SJ. Engineering microRNA responsiveness to decrease virus pathogenicity. Nat Med. 2008;14:1278–1283.
  • Ylosmaki E, Hakkarainen T, Hemminki A, et al. Generation of a conditionally replicating adenovirus based on targeted destruction of E1A mRNA by a cell type-specific MicroRNA. J Virol. 2008;82:11009–11015.
  • Edge RE, Falls TJ, Brown CW, et al. A let-7 microRNA-sensitive vesicular stomatitis virus demonstrates tumor-specific replication. Mol Ther. 2008;16:1437–1443.
  • Bateman A, Bullough F, Murphy S, et al. Fusogenic membrane glycoproteins as a novel class of genes for the local and immune-mediated control of tumor growth. Cancer Res. 2000;60:1492–1497.
  • Heise C, Kirn DH. Replication-selective adenoviruses as oncolytic agents. J Clin Invest. 2000;105:847–851.
  • Jiang H, Gomez-Manzano C, Aoki H, et al. Examination of the therapeutic potential of Delta-24-RGD in brain tumor stem cells: role of autophagic cell death. J Natl Cancer Inst. 2007;99:1410–1414.
  • Wang H, Song X, Zhang H, et al. Potentiation of tumor radiotherapy by a radiation-inducible oncolytic and oncoapoptotic adenovirus in cervical cancer xenografts. Int J Cancer. 2012;130:443–453.
  • Chen L, Chen D, Gong M, et al. Concomitant use of Ad5/35 chimeric oncolytic adenovirus with TRAIL gene and taxol produces synergistic cytotoxicity in gastric cancer cells. Cancer Lett. 2009;284:141–148.
  • Wohlfahrt ME, Beard BC, Lieber A, Kiem HP. A capsid-modified, conditionally replicating oncolytic adenovirus vector expressing TRAIL Leads to enhanced cancer cell killing in human glioblastoma models. Cancer Res. 2007;67:8783–8790.
  • Zhao L, Dong A, Gu J, et al. The antitumor activity of TRAIL and IL-24 with replicating oncolytic adenovirus in colorectal cancer. Cancer Gene Ther. 2006;13:1011–1022.
  • Ebert O, Shinozaki K, Kournioti C, et al. Syncytia induction enhances the oncolytic potential of vesicular stomatitis virus in virotherapy for cancer. Cancer Res. 2004;64:3265–3270.
  • van Beusechem VW, van den Doel PB, Grill J, et al. Conditionally replicative adenovirus expressing p53 exhibits enhanced oncolytic potency. Cancer Res. 2002;62:6165–6171.
  • Ma J, He X, Wang W, et al. E2F promoter-regulated oncolytic adenovirus with p16 gene induces cell apoptosis and exerts antitumor effect on gastric cancer. DigDis Sci. 2009;54:1425–1431.
  • Cui Q, Jiang W, Wang Y, et al. Transfer of suppressor of cytokine signaling 3 by an oncolytic adenovirus induces potential antitumor activities in hepatocellular carcinoma. Hepatology. 2008;47:105–112.
  • Liu XR, Cai Y, Cao X, et al. A new oncolytic adenoviral vector carrying dual tumor suppressor genes shows potent antitumor effect. J Cell Mol Med. July 27, 2011. [Epub ahead of print.]
  • Zheng JN, Pei DS, Sun FH, et al. Inhibition of renal cancer cell growth by oncolytic adenovirus armed short hairpin RNA targeting hTERT gene. Cancer Biol Ther. 2009;8:84–91.
  • Zheng JN, Pei DS, Mao LJ, et al. Inhibition of renal cancer cell growth in vitro and in vivo with oncolytic adenovirus armed short hairpin RNA targeting Ki-67 encoding mRNA. Cancer Gene Ther. 2009;16:20–32.
  • Shen W, Wang CY, Wang XH, Fu ZX. Oncolytic adenovirus mediated Survivin knockdown by RNA interference suppresses human colorectal carcinoma growth in vitro and in vivo. J Exp Clin Cancer Res. 2009;28:81.
  • Chu L, Gu J, Sun L, et al. Oncolytic adenovirus-mediated shRNA against Apollon inhibits tumor cell growth and enhances antitumor effect of 5-fluorouracil. Gene Ther. 2008;15:484–494.
  • Breitbach CJ, De Silva NS, Falls TJ, et al. Targeting tumor vasculature with an oncolytic virus. Mol Ther. 2011;19:886–894.
  • Liu TC, Hwang T, Park BH, et al. The targeted oncolytic poxvirus JX-594 demonstrates antitumoral, antivascular, and anti-HBV activities in patients with hepatocellular carcinoma. Mol Ther. 2008;16:1637–1642.
  • Ahn M, Lee SJ, Li X, et al. Enhanced combined tumor-specific oncolysis and suicide gene therapy for prostate cancer using M6 promoter. Cancer Gene Ther. 2009;16:73–82.
  • Zheng FQ, Xu Y, Yang RJ, et al. Combination effect of oncolytic adenovirus therapy and herpes simplex virus thymidine kinase/ganciclovir in hepatic carcinoma animal models. Acta Pharmacol Sin. 2009;30:617–627.
  • Bossow S, Grossardt C, Temme A, et al. Armed and targeted measles virus for chemovirotherapy of pancreatic cancer. Cancer Gene Ther. 2011;18:598–608.
  • Chalikonda S, Kivlen MH, O’Malley ME, et al. Oncolytic virotherapy for ovarian carcinomatosis using a replication-selective vaccinia virus armed with a yeast cytosine deaminase gene. Cancer Gene Ther. 2008;15:115–125.
  • Foloppe J, Kintz J, Futin N, et al. Targeted delivery of a suicide gene to human colorectal tumors by a conditionally replicating vaccinia virus. Gene Ther. 2008;15:1361–1371.
  • Braidwood L, Dunn PD, Hardy S, et al. Antitumor activity of a selectively replication competent herpes simplex virus (HSV) with enzyme prodrug therapy. Anticancer Res. 2009;29:2159–2166.
  • Hakkarainen T, Rajecki M, Sarparanta M, et al. Targeted radiotherapy for prostate cancer with an oncolytic adenovirus coding for human sodium iodide symporter. Clin Cancer Res. 2009;15:5396–5403.
  • Goel A, Carlson SK, Classic KL, et al. Radioiodide imaging and radiovirotherapy of multiple myeloma using VSV (Delta51)-NIS, an attenuated vesicular stomatitis virus encoding the sodium iodide symporter gene. Blood. 2007;110:2342–2350.
  • Li H, Peng KW, Dingli D, et al. Oncolytic measles viruses encoding interferon beta and the thyroidal sodium iodide symporter gene for mesothelioma virotherapy. Cancer Gene Ther. 2010;17:550–558.
  • Blechacz B, Splinter PL, Greiner S, et al. Engineered measles virus as a novel oncolytic viral therapy system for hepatocellular carcinoma. Hepatology. 2006;44:1465–1477.
  • Hasegawa K, Pham L, O’Connor MK, et al. Dual therapy of ovarian cancer using measles viruses expressing carcinoembryonic antigen and sodium iodide symporter. Clin Cancer Res. 2006;12:1868–1875.
  • Dingli D, Peng KW, Harvey ME, et al. Image-guided radiovirotherapy for multiple myeloma using a recombinant measles virus expressing the thyroidal sodium iodide symporter. Blood. 2004;103:1641–1646.
  • Benencia F, Courreges MC, Conejo-Garcia JR, et al. HSV oncolytic therapy upregulates interferon-inducible chemokines and recruits immune effector cells in ovarian cancer. Mol Ther. 2005;12:789–802.
  • Endo Y, Sakai R, Ouchi M, et al. Virus-mediated oncolysis induces danger signal and stimulates cytotoxic T-lymphocyte activity via proteasome activator upregulation. Oncogene. 2008;27:2375–2381.
  • Errington F, Steele L, Prestwich R, et al. Reovirus activates human dendritic cells to promote innate antitumor immunity. J Immunol. 2008;180:6018–6026.
  • Steele L, Errington F, Prestwich R, et al. Pro-inflammatory cytokine/ chemokine production by reovirus treated melanoma cells is PKR/ NF-kappaB mediated and supports innate and adaptive anti-tumor immune priming. Mol Cancer. 2011;10:20.
  • Gujar SA, Marcato P, Pan D, Lee PW. Reovirus virotherapy overrides tumor antigen presentation evasion and promotes protective antitumor immunity. Mol Cancer Ther. 2010;9:2924–2933.
  • Errington F, White CL, Twigger KR, et al. Inflammatory tumor cell killing by oncolytic reovirus for the treatment of melanoma. Gene Ther. 2008;15:1257–1270.
  • Kepp O, Tesniere A, Schlemmer F, et al. Immunogenic cell death modalities and their impact on cancer treatment. Apoptosis. 2009;14:364–375.
  • Tesniere A, Apetoh L, Ghiringhelli F, et al. Immunogenic cancer cell death: a key-lock paradigm. Curr Opin Immunol. 2008;20:504–511.
  • Huang B, Sikorski R, Kirn DH, Thorne SH. Synergistic anti-tumor effects between oncolytic vaccinia virus and paclitaxel are mediated by the IFN response and HMGB1. Gene Ther. 2011;18:164–172.
  • Gauvrit A, Brandler S, Sapede-Peroz C, et al. Measles virus induces oncolysis of mesothelioma cells and allows dendritic cells to cross-prime tumor-specific CD8 response. Cancer Res. 2008;68:4882–4892.
  • Guo ZS, Naik A, O’Malley ME, et al. The enhanced tumor selectivity of an oncolytic vaccinia lacking the host range and antiapoptosis genes SPI-1 and SPI-2. Cancer Res. 2005;65:9991–9998.
  • Bai L, Koopmann J, Fiola C, et al. Dendritic cells pulsed with viral oncolysates potently stimulate autologous T cells from cancer patients. Int J Oncol. 2002;21:685–694.
  • Bridle BW, Stephenson KB, Boudreau JE, et al. Potentiating cancer immunotherapy using an oncolytic virus. Mol Ther. 2010;184:4269–4275.
  • Savina A, Amigorena S. Phagocytosis and antigen presentation in dendritic cells. Immunol Rev. 2007;219:143–156.
  • Santambrogio L, Sato AK, Carven GJ, et al. Extracellular antigen processing and presentation by immature dendritic cells. Proc Natl Acad Sci U S A. 1999;96:15056–15061.
  • Prestwich RJ, Errington F, Steele LP, et al. Reciprocal human dendritic cell-natural killer cell interactions induce antitumor activity following tumor cell infection by oncolytic reovirus. J Immunol. 2009;183:4312–4321.
  • White CL, Twigger KR, Vidal L, et al. Characterization of the adaptive and innate immune response to intravenous oncolytic reovirus (Dearing type 3) during a phase I clinical trial. Gene Ther. 2008;15:911–920.
  • Prestwich RJ, Errington F, Ilett EJ, et al. Tumor infection by oncolytic reovirus primes adaptive antitumor immunity. Clin Cancer Res. 2008;14:7358–7366.
  • Sobol PT, Boudreau JE, Stephenson K, et al. Adaptive antiviral immunity is a determinant of the therapeutic success of oncolytic virotherapy. Mol Ther. 2011;19:335–344.
  • Li H, Dutuor A, Fu X, Zhang X. Induction of strong antitumor immunity by an HSV-2-based oncolytic virus in a murine mammary tumor model. J Gene Med. 2007;9:161–169.
  • Hummel JL, Safroneeva E, Mossman KL. The role of ICP0-Null HSV-1 and interferon signaling defects in the effective treatment of breast adenocarcinoma. Mol Ther. 2005;12:1101–1110.
  • Nakamori M, Fu X, Rousseau R, et al. Destruction of nonimmunogenic mammary tumor cells by a fusogenic oncolytic herpes simplex virus induces potent antitumor immunity. Mol Ther. 2004;9:658–665.
  • Toda M, Iizuka Y, Kawase T, et al. Immuno-viral therapy of brain tumors by combination of viral therapy with cancer vaccination using a replication-conditional HSV. Cancer Gene Ther. 2002;9:356–364.
  • Moehler MH, Zeidler M, Wilsberg V, et al. Parvovirus H-1-induced tumor cell death enhances human immune response in vitro via increased phagocytosis, maturation, and cross-presentation by dendritic cells. Hum Gene Ther. 2005;16:996–1005.
  • Bhat R, Dempe S, Dinsart C, Rommelaere J. Enhancement of NK cell anti-tumor responses using an oncolytic parvovirus. Int J Cancer. 2011;128:908–919.
  • Diaz RM, Galivo F, Kottke T, et al. Oncolytic immunovirotherapy for melanoma using vesicular stomatitis virus. Cancer Res. 2007;67:2840–2848.
  • Wongthida P, Diaz RM, Galivo F, et al. Type III IFN interleukin-28 mediates the antitumor efficacy of oncolytic virus VSV in immune-competent mouse models of cancer. Cancer Res. 2010;70:4539–4549.
  • Liu BL, Robinson M, Han ZQ, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumor properties. Gene Ther. 2003;10:292–303.
  • Kottke T, Galivo F, Wongthida P, et al. Treg depletion-enhanced IL-2 treatment facilitates therapy of established tumors using systemically delivered oncolytic virus. Mol Ther. 2008;16:1217–1226.
  • Bronte V, Tsung K, Rao JB, et al. IL-2 enhances the function of recombinant poxvirus-based vaccines in the treatment of established pulmonary metastases. J Immunol. 15 1995;154:5282–5292.
  • Lee YS, Kim JH, Choi KJ, et al. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B7–1 in an immunocompetent murine model. Clin Cancer Res. 2006;12:5859–5868.
  • Senzer NN, Kaufman HL, Amatruda T, 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;27:5763–5771.
  • Hu JC, Coffin RS, Davis CJ, et al. A phase I study of OncoVEXGM- CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor. Clin Cancer Res. 2006;12:6737–6747.
  • Harrington KJ, Hingorani M, Tanay MA, et al. Phase I/II study of oncolytic HSV GM-CSF in combination with radiotherapy and cis- platin in untreated stage III/IV squamous cell cancer of the head and neck. Clin Cancer Res. 2010;16:4005–4015.
  • Kaufman HL, Kim DW, Deraffele G, et al. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Ann Surg Oncol. 2010;17:718–730.
  • Park BH, Hwang T, Liu TC, 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;9:533–542.
  • Chang J, Zhao X, Wu X, et al. A phase I study of KH901, a conditionally replicating granulocyte-macrophage colony-stimulating factor: armed oncolytic adenovirus for the treatment of head and neck cancers. Cancer Biol Ther. 2009;8:676–682.
  • Ramakrishna E, Woller N, Mundt B, et al. Antitumoral immune re spon se by recruitment and expansion of dendritic cells in tumors infected with telomerase-dependent oncolytic viruses. Cancer Res. 2009;69:1448–1458.
  • Bernt KM, Ni S, Tieu AT, Lieber A. Assessment of a combined, adenovirus-mediated oncolytic and immunostimulatory tumor therapy. Cancer Res. 2005;65:4343–4352.
  • Li J, O’Malley M, Urban J, et al. Chemokine expression from oncolytic vaccinia virus enhances vaccine therapies of cancer. Mol Ther. 2011;19:650–657.
  • Lapteva N, Aldrich M, Weksberg D, et al. Targeting the intratumoral dendritic cells by the oncolytic adenoviral vaccine expressing RANTES elicits potent antitumor immunity. J Immunother. 2009;32:145–156.
  • Wong RJ, Patel SG, Kim S, et al. Cytokine gene transfer enhances herpes oncolytic therapy in murine squamous cell carcinoma. Hum Gene Ther. 2001;12:253–265.
  • Kim JH, Oh JY, Park BH, et al. Systemic armed oncolytic and immunologic therapy for cancer with JX-594, a targeted poxvirus expressing GM-CSF. Mol Ther. 2006;14:361–370.
  • Lee JH, Roh MS, Lee YK, et al. Oncolytic and immunostimulatory efficacy of a targeted oncolytic poxvirus expressing human GM-CSF following intravenous administration in a rabbit tumor model. Cancer Gene Ther. 2010;17:73–79.
  • Cerullo V Pesonen S, Diaconu I, et al. Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res. 2010;70:4297–4309.
  • Robinson M, Li B, Ge Y, et al. Novel immunocompetent murine tumor model for evaluation of conditionally replication-competent (oncolytic) murine adenoviral vectors. J Virol. 2009;83:3450–3462.
  • Vigil A, Park MS, Martinez O, et al. Use of reverse genetics to enhance the oncolytic properties of Newcastle disease virus. Cancer Res. 2007;67:8285–8292.
  • Grote D, Cattaneo R, Fielding AK. Neutrophils contribute to the measles virus-induced antitumor effect: enhancement by granulocyte macrophage colony-stimulating factor expression. Cancer Res. 2003;63:6463–6468.
  • Bergman I, Griffin JA, Gao Y, Whitaker-Dowling P. Treatment of implanted mammary tumors with recombinant vesicular stomatitis virus targeted to Her2/neu. Int J Cancer. 2007;121:425–430.
  • Choi IK, Lee JS, Zhang SN, et al. Oncolytic adenovirus co-expressing IL-12 and IL-18 improves tumor-specific immunity via differentiation of T cells expressing IL-12Rbeta(2) or IL-18Ralpha. Gene Ther. 2011;18:898–909.
  • Varghese S, Rabkin SD, Liu R, et al. Enhanced therapeutic efficacy of IL-12, but not GM-CSF, expressing oncolytic herpes simplex virus for transgenic mouse derived prostate cancers. Cancer Gene Ther. 2006;13:253–265.
  • Derubertis BG, Stiles BM, Bhargava A, et al. Cytokine-secreting herpes viral mutants effectively treat tumor in a murine metastatic colorectal liver model by oncolytic and T-cell-dependent mechanisms. Cancer Gene Ther. 2007;14:590–597.
  • Shin EJ, Wanna GB, Choi B, et al. Interleukin-12 expression enhances vesicular stomatitis virus oncolytic therapy in murine squamous cell carcinoma. Laryngoscope. 2007;117:210–214.
  • Zhao H, Janke M, Fournier P, Schirrmacher V. Recombinant Newcastle disease virus expressing human interleukin-2 serves as a potential candidate for tumor therapy. Virus Res. 2008;136:75–80.
  • Carew JF, Kooby DA, Halterman MW, et al. A novel approach to cancer therapy using an oncolytic herpes virus to package amplicons containing cytokine genes. Mol Ther. 2001;4:250–256.
  • Post DE, Sandberg EM, Kyle MM, et al. Targeted cancer gene therapy using a hypoxia inducible factor dependent oncolytic adenovirus armed with interleukin-4. Cancer Res. 2007;67:6872–6881.
  • Terada K, Wakimoto H, Tyminski E, et al. Development of a rapid method to generate multiple oncolytic HSV vectors and their in vivo evaluation using syngeneic mouse tumor models. Gene Ther. 2006 ;13: 705–714.
  • Fukuhara H, Ino Y, Kuroda T, et al. Triple gene-deleted oncolytic herpes simplex virus vector double-armed with interleukin-18 and soluble B7–1 constructed by bacterial artificial chromosome-mediated system. Cancer Res. 2005;65:10663–10668.
  • Ino Y, Saeki Y, Fukuhara H, Todo T. Triple combination of oncolytic herpes simplex virus-1 vectors armed with interleukin-12, interleukin-18, or soluble B7–1 results in enhanced antitumor efficacy. Clin Cancer Res. 2006;12:643–652.
  • Shashkova EV, Kuppuswamy MN, Wold WS, Doronin K. Anticancer activity of oncolytic adenovirus vector armed with IFN-alpha and ADP is enhanced by pharmacologically controlled expression of TRAIL. Cancer Gene Ther. 2008;15:61–72.
  • Shashkova EV, Spencer JF, Wold WS, Doronin K. Targeting interferon-alpha increases antitumor efficacy and reduces hepatotoxic- ity of E1A-mutated spread-enhanced oncolytic adenovirus. Mol Ther. 2007;15:598–607.
  • Kirn DH, Wang Y, Le Boeuf F, et al. Targeting of interferon-beta to produce a specific, multi-mechanistic oncolytic vaccinia virus. PLoS Med. 2007;4:e353.
  • Willmon CL, Saloura V Fridlender ZG, et al. Expression of IFN-beta enhances both efficacy and safety of oncolytic vesicular stomatitis virus for therapy of mesothelioma. Cancer Res. 2009;69:7713–7720.
  • Su C, Peng L, Sham J, et al. Immune gene-viral therapy with triplex efficacy mediated by oncolytic adenovirus carrying an interferongamma gene yields efficient antitumor activity in immunodeficient and immunocompetent mice. Mol Ther. 2006;13:918–927.
  • Han ZQ, Assenberg M, Liu BL, et al. Development of a second- generation oncolytic Herpes simplex virus expressing TNFalpha for cancer therapy. J Gene Med. 2007;9:99–106.
  • Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. 2003;3:133–146.
  • Nishimura T, Iwakabe K, Sekimoto M, et al. Distinct role of antigen- specific T helper type 1 (Th1) and Th2 cells in tumor eradication in vivo. J Exp Med. 1999;190:617–627.
  • de Jong EC, Smits HH, Kapsenberg ML. Dendritic cell-mediated T cell polarization. Springer Semin Immunopathol. 2005;26:289–307.
  • Voest EE, Kenyon BM, O’Reilly MS, et al. Inhibition of angiogenesis in vivo by interleukin-12. J Natl Cancer Inst. 1995;87:581–586.
  • Cavallo F, Quaglino E, Cifaldi L, et al. Interleukin-12-activated lymphocytes influence tumor genetic programs. Cancer Res. 2001;61:3518–3523.
  • Yao L, Sgadari C, Furuke K, et al. Contribution of natural killer cells to inhibition of angiogenesis by interleukin-12. Blood. 1999;93:1612–1621.
  • Coughlin CM, Salhany KE, Gee MS, et al. Tumor cell responses to IFNgamma affect tumorigenicity and response to IL-12 therapy and antiangiogenesis. Immunity. 1998;9:25–34.
  • Nastala CL, Edington HD, McKinney TG, et al. Recombinant IL-12 administration induces tumor regression in association with IFN- gamma production. J Immunol. 1994;153:1697–1706.
  • Tahara H, Zeh HJ 3rd, Storkus WJ, et al. Fibroblasts genetically engineered to secrete interleukin-12 can suppress tumor growth and induce antitumor immunity to a murine melanoma in vivo. Cancer Res. 1994;54:182–189.
  • Noguchi Y, Jungbluth A, Richards EC, Old LJ. Effect of interleukin-12 on tumor induction by 3-methylcholanthrene. Proc Natl Acad Sci U S A. 1996;93:11798–11801.
  • Del Vecchio M, Bajetta E, Canova S, et al. Interleukin-12: biological properties and clinical application. Clin Cancer Res. 2007;13:4677–4685.
  • Leonard JP, Sherman ML, Fisher GL, et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90:2541–2548.
  • Sarmiento UM, Riley JH, Knaack PA, et al. Biologic effects of recombinant human interleukin-12 in squirrel monkeys (Sciureus saimiri). Lab Invest. 1994;71:862–873.
  • Cohen J. IL-12 deaths: explanation and a puzzle. Science. 1995;270:908.
  • Tare NS, Bowen S, Warrier RR, et al. Administration of recombinant interleukin-12 to mice suppresses hematopoiesis in the bone marrow but enhances hematopoiesis in the spleen. J Interferon Cytokine Res.1995;15:377–383.
  • Lui VW, He Y, Falo L, Huang L. Systemic administration of naked DNA encoding interleukin-12 for the treatment of human papillomavirus DNA-positive tumor. Hum Gene Ther. 2002;13:177–185.
  • Parker JN, Gillespie GY, Love CE, et al. Engineered herpes simplex virus expressing IL-12 in the treatment of experimental murine brain tumors. Proc Natl Acad Sci U S A. 2000;97:2208–2213.
  • Jarnagin WR, Zager JS, Klimstra D, et al. Neoadjuvant treatment of hepatic malignancy: an oncolytic herpes simplex virus expressing IL-12 effectively treats the parent tumor and protects against recurrence-after resection. Cancer Gene Ther. 2003;10:215–223.
  • Wong RJ, Chan MK, Yu Z, et al. Effective intravenous therapy of murine pulmonary metastases with an oncolytic herpes virus expressing interleukin-12. Clin Cancer Res. 2004;10:251–259.
  • Choi KJ, Kim JH, Lee YS, et al. Concurrent delivery of GM-CSF and B7–1 using an oncolytic adenovirus elicits potent antitumor effect. Gene Ther. 2006;13:1010–1020.
  • Kim HS, Kim-Schulze S, Kim DW, Kaufman HL. Host lymphodeple- tion enhances the therapeutic activity of an oncolytic vaccinia virus expressing 4–1BB ligand. Cancer Res. 2009;69:8516–8525.
  • Huang JH, Zhang SN, Choi KJ, et al. Therapeutic and tumor-specific immunity induced by combination of dendritic cells and oncolytic adenovirus expressing IL-12 and 4–1BBL. Mol Ther. 2010;18:264–274.
  • Galivo F, Diaz RM, Thanarajasingam U, et al. Interference of CD40L- mediated tumor immunotherapy by oncolytic VSV. Hum Gene Ther. 2010;21:439–450.
  • Todo T, Martuza RL, Dallman MJ, Rabkin SD. In situ expression of soluble B7–1 in the context of oncolytic herpes simplex virus induces potent antitumor immunity. Cancer Res. 2001;61:153–161.
  • Li JL, Liu HL, Zhang XR, et al. A phase I trial of intratumoral administration of recombinant oncolytic adenovirus overexpressing HSP70 in advanced solid tumor patients. Gene Ther. 2009;16:376–382.
  • Yoo JY, Ryu J, Gao R, et al. Tumor suppression by apoptotic and anti-angiogenic effects of mortalin-targeting adeno-oncolytic virus. J Gene Med. 2010;12:586–595.
  • Di Paolo NC, Tuve S, Ni S, et al. Effect of adenovirus-mediated heat shock protein expression and oncolysis in combination with low-dose cyclophosphamide treatment on antitumor immune responses. Cancer Res. 2006;66:960–969.
  • Huang XF, Ren W, Rollins L, et al. A broadly applicable, personalized heat shock protein-mediated oncolytic tumor vaccine. Cancer Res. 2003;63:7321–7329.
  • Hu ZB, Wu CT, Wang H, et al. A simplified system for generating oncolytic adenovirus vector carrying one or two transgenes. Cancer Gene Ther. 2008;15:173–182.
  • Lonchay C, van der Bruggen P, Connerotte T, et al. Correlation between tumor regression and T cell responses in melanoma patients vaccinated with a MAGE antigen. Proc Natl Acad Sci U S A. 2004;101(Suppl 2):14631–14638.
  • Harrop R, Connolly N, Redchenko I, et al. Vaccination of colorectal cancer patients with modified vaccinia Ankara delivering the tumor antigen 5T4 (TroVax) induces immune responses which correlate with disease control: a phase I/II trial. Clin Cancer Res. 2006;12:3416–3424.
  • Amato RJ, Drury N, Naylor S, et al. Vaccination of prostate cancer patients with modified vaccinia ankara delivering the tumor antigen 5T4 (TroVax): a phase 2 trial. JImmunother. 2008;31:577–585.
  • Kaufman HL, Lenz HJ, Marshall J, et al. Combination chemotherapy and ALVAC-CEA/B7.1 vaccine in patients with metastatic colorectal cancer. Clin Cancer Res. 2008;14:4843–4849.
  • Horig H, Lee DS, Conkright W, et al. Phase I clinical trial of a recombinant canarypoxvirus (ALVAC) vaccine expressing human carcinoembryonic antigen and the B7.1 co-stimulatory molecule. Cancer Immunol Immunother. 2000;49:504–514.
  • Harrop R, John J, Carroll MW. Recombinant viral vectors: cancer vaccines. Adv Drug Deliv Rev. 2006;58:931–947.
  • Elzey BD, Siemens DR, Ratliff TL, Lubaroff DM. Immunization with type 5 adenovirus recombinant for a tumor antigen in combination with recombinant canarypox virus (ALVAC) cytokine gene delivery induces destruction of established prostate tumors. Int J Cancer. 2001;94:842–849.
  • Irvine KR, Chamberlain RS, Shulman EP, et al. Enhancing efficacy of recombinant anticancer vaccines with prime/boost regimens that use two different vectors. J Natl Cancer Inst. 1997;89:1595–1601.
  • Naslund TI, Uyttenhove C, Nordstrom EK, et al. Comparative prime-boost vaccinations using Semliki Forest virus, adenovirus, and ALVAC vectors demonstrate differences in the generation of a protective central memory CTL response against the P815 tumor. J Immunol. 2007;178:6761–6769.
  • Hodge JW, Poole DJ, Aarts WM, et al. Modified vaccinia virus ankara recombinants are as potent as vaccinia recombinants in diversified prime and boost vaccine regimens to elicit therapeutic antitumor responses. Cancer Res. 2003;63:7942–7949.
  • Hodge JW, McLaughlin JP, Kantor JA, Schlom J. Diversified prime and boost protocols using recombinant vaccinia virus and recombinant non-replicating avian pox virus to enhance T-cell immunity and antitumor responses. Vaccine. 1997;15:759–768.
  • Marshall JL, Hoyer RJ, Toomey MA, et al. Phase I study in advanced cancer patients of a diversified prime-and-boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avi- pox virus to elicit anti-carcinoembryonic antigen immune responses. J Clin Oncol. 2000;18:3964–3973.
  • Vigil A, Martinez O, Chua MA, Garcia-Sastre A. Recombinant Newcastle disease virus as a vaccine vector for cancer therapy. Mol Ther. 2008;16:1883–1890.
  • Power AT, Bell JC. Cell-based delivery of oncolytic viruses: a new strategic alliance for a biological strike against cancer. Mol Ther. 2007;15:660–665.
  • Ilett EJ, Barcena M, Errington-Mais F, et al. Internalization of oncolytic reovirus by human dendritic cell carriers protects the virus from neutralization. Clin Cancer Res. 2011;17:2767–2776.
  • Liu C, Russell SJ, Peng KW. Systemic therapy of disseminated myeloma in passively immunized mice using measles virus-infected cell carriers. Mol Ther. 2010;18:1155–1164.
  • Mader EK, Maeyama Y, Lin Y, et al. Mesenchymal stem cell carriers protect oncolytic measles viruses from antibody neutralization in an orthotopic ovarian cancer therapy model. Clin Cancer Res. 2009;15:7246–7255.
  • Qiao J, Kottke T, Willmon C, et al. Purging metastases in lymphoid organs using a combination of antigen-nonspecific adoptive T cell therapy, oncolytic virotherapy and immunotherapy. Nat Med. 2008;14:37–44.
  • Hoffmann D, Bayer W, Wildner O. Local and distant immune- mediated control of colon cancer growth with fusogenic membrane glycoproteins in combination with viral oncolysis. Hum Gene Ther. 2007;18:435–450.
  • Yotnda P, Savoldo B, Charlet-Berguerand N, et al. Targeted delivery of adenoviral vectors by cytotoxic T cells. Blood. 2004;104:2272–2280.
  • Thorne SH, Liang W, Sampath P, et al. Targeting localized immune suppression within the tumor through repeat cycles of immune cell- oncolytic virus combination therapy. Mol Ther. 2010;18:1698–1705.
  • Thomas DL, Doty R, Tosic V et al. Myxoma virus combined with rapamycin treatment enhances adoptive T cell therapy for murine melanoma brain tumors. Cancer Immunol Immunother. 2011;60:1461–1472.
  • Thorne SH, Negrin RS, Contag CH. Synergistic antitumor effects of immune cell-viral biotherapy. Science. 2006;311:1780–1784.
  • Raykov Z, Grekova S, Galabov AS, et al. Combined oncolytic and vaccination activities of parvovirus H-1 in a metastatic tumor model. Oncol Rep. 2007;17:1493–1499.
  • Schirrmacher V. Clinical trials of antitumor vaccination with an autologous tumor cell vaccine modified by virus infection: improvement of patient survival based on improved antitumor immune memory. Cancer Immunol Immunother. 2005;54:587–598.
  • Schirrmacher V, Haas C, Bonifer R, et al. Human tumor cell modification by virus infection: an efficient and safe way to produce cancer vaccine with pleiotropic immune stimulatory properties when using Newcastle disease virus. Gene Ther. 1999;6:63–73.
  • Thorne SH, Contag CH. Combining immune cell and viral therapy for the treatment of cancer. Cell Mol Life Sci. 2007;64:1449–1451.
  • Boudreau JE, Bridle BW, Stephenson KB, et al. Recombinant vesicular stomatitis virus transduction of dendritic cells enhances their ability to prime innate and adaptive antitumor immunity. Mol Ther. 2009;17:1465–1472.
  • Heinzerling L, Kunzi V, Oberholzer PA, et al. Oncolytic measles virus in cutaneous T-cell lymphomas mounts antitumor immune responses in vivo and targets interferon-resistant tumor cells. Blood. 2005;106:2287–2294.
  • Mineta T, Rabkin SD, Yazaki T, et al. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med. 1995;1:938–943.
  • Harrington KJ, Karapanagiotou EM, Roulstone V, et al. Two-stage phase I dose-escalation study of intratumoral reovirus type 3 dearing and palliative radiotherapy in patients with advanced cancers. Clin Cancer Res. 2010;16:3067–3077.
  • Smith E, Breznik J, Lichty BD. Strategies to enhance viral penetration of solid tumors. Hum Gene Ther. 2011;22:1053–1060.
  • Liu TC, Kirn D. Systemic efficacy with oncolytic virus therapeutics: clinical proof-of-concept and future directions. Cancer Res. 2007;67:429–432.
  • Law M, Hollinshead R, Smith GL. Antibody-sensitive and antibody- resistant cell-to-cell spread by vaccinia virus: role of the A33R protein in antibody-resistant spread. J Gen Virol. 2002;83:209–222.
  • Vanderplasschen A, Mathew E, Hollinshead M, et al. Extracellular enveloped vaccinia virus is resistant to complement because of incorporation of host complement control proteins into its envelope. Proc Natl Acad Sci U SA. 1998;95:7544–7549.
  • Ikeda K, Wakimoto H, Ichikawa T, et al. Complement depletion facilitates the infection of multiple brain tumors by an intravascular, replication-conditional herpes simplex virus mutant. J Virol. 2000;74:4765–4775.
  • Ikeda K, Ichikawa T, Wakimoto H, et al. Oncolytic virus therapy of multiple tumors in the brain requires suppression of innate and elicited antiviral responses. Nat Med. 1999;5:881–887.
  • Dhar D, Spencer JF, Toth K, Wold WS. Pre-existing immunity and passive immunity to adenovirus 5 prevents toxicity caused by an oncolytic adenovirus vector in the Syrian hamster model. Mol Ther. 2009;17:1724–1732.
  • Miest TS, Yaiw KC, Frenzke M, et al. Envelope-chimeric entry- targeted measles virus escapes neutralization and achieves oncolysis. Mol Ther. May 24, 2011.
  • Blechacz B, Russell SJ. Measles virus as an oncolytic vector platform. Curr Gene Ther. 2008;8:162–175.
  • Qiao J, Wang H, Kottke T, et al. Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus. Clin Cancer Res. 2008;14:259–269.
  • 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;11:214–223.
  • Smakman N, van der Bilt JD, van den Wollenberg DJ, et al. Immunosuppression promotes reovirus therapy of colorectal liver metastases. Cancer Gene Ther. 2006;13:815–818.
  • Ungerechts G, Springfeld C, Frenzke ME, et al. An immunocompetent murine model for oncolysis with an armed and targeted measles virus. Mol Ther. 2007;15:1991–1997.
  • MacTavish H, Diallo JS, Huang B, et al. Enhancement of vaccinia virus based oncolysis with histone deacetylase inhibitors. PLoS One. 2010;5:e14462.
  • Kurozumi K, Hardcastle J, Thakur R, et al. Effect of tumor microenvironment modulation on the efficacy of oncolytic virus therapy. J Natl Cancer Inst. 2007;99:1768–1781.
  • 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;16:1041–1047.
  • Frentzen A, Yu YA, Chen N, et al. Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy. Proc Natl Acad Sci U S A. 2009;106:12915–12920.
  • Tysome J, Wang P, Alusi G, et al. Lister vaccine strain of vaccinia virus armed with the endostatin-angiostatin fusion gene: a superior oncolytic virus to dl1520 (0NYX-015) for human head and neck cancer. Hum Gene Ther. 2011;22:1101–1108.
  • Tysome JR, Briat A, Alusi G, et al. Lister strain of vaccinia virus armed with endostatin-angiostatin fusion gene as a novel therapeutic agent for human pancreatic cancer. Gene Ther. 2009;16:1223–1233.
  • Tysome JR, Lemoine NR, Wang Y. Combination of anti-angiogenic therapy and virotherapy: arming oncolytic viruses with anti-angiogenic genes. Curr Opin Mol Ther. 2009;11:664–669.
  • Su C, Na M, Chen J, et al. Gene-viral cancer therapy using dualregulated oncolytic adenovirus with antiangiogenesis gene for increased efficacy. Mol Cancer Res. 2008;6:568–575.
  • Fang L, Pu YY, Hu XC, et al. Antiangiogenesis gene armed tumortargeting adenovirus yields multiple antitumor activities in human HCC xenografts in nude mice. Hepatol Res. 2009;40:216–228.
  • Guse K, Diaconu I, Rajecki M, et al. Ad5/3 -9HIF -delta24-VEGFR-1 -Ig, an infectivity enhanced, dual-targeted and antiangiogenic oncolytic adenovirus for kidney cancer treatment. Gene Ther. 2009;16:1009–1020.
  • Kottke T, Thompson J, Diaz RM, 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;15:561–569.
  • Willmon C, Diaz RM, Wongthida P, et al. Vesicular stomatitis virus-induced immune suppressor cells generate antagonism between intratumoral oncolytic virus and cyclophosphamide. Mol Ther. 2011;19:140–149.
  • Kottke T, Chester J, Ilett E, et al. Precise scheduling of chemotherapy primes VEGF-producing tumors for successful systemic oncolytic virotherapy. Mol Ther. July 26, 2011.
  • Bajzer Z, Carr T, Josic K, et al. Modeling of cancer virotherapy with recombinant measles viruses. JTheor Biol. 2008;252:109–122.
  • Biesecker M, Kimn JH, Lu H, et al. Optimization of virotherapy for cancer. Bull Math Biol. 2010;72:469–489.
  • Dingli D, Cascino MD, Josic K, et al. Mathematical modeling of cancer radiovirotherapy. Math Biosci. 2006;199:55–78.
  • Dingli D, Offord C, Myers R, et al. Dynamics of multiple myeloma tumor therapy with a recombinant measles virus. Cancer gene therapy. 2009;16:873–882.
  • Friedman A, Tian JP, Fulci G, et al. Glioma virotherapy: effects of innate immune suppression and increased viral replication capacity. Cancer research. 2006;66:2314–2319.
  • Karev GP, Novozhilov AS, Koonin EV. Mathematical modeling of tumor therapy with oncolytic viruses: effects of parametric heterogeneity on cell dynamics. Biol Direct. 2006;1:30.
  • Novozhilov AS, Berezovskaya FS, Koonin EV, Karev GP. Mathematical modeling of tumor therapy with oncolytic viruses: regimes with complete tumor elimination within the framework of deterministic models. Biol Direct. 2006;1:6.
  • Paiva LR, Binny C, Ferreira SC Jr, Martins ML. A multiscale mathematical model for oncolytic virotherapy. Cancer Res. 2009;69:1205–1211.
  • Rommelfanger DM, Offord CP, Dev J, et al. Dynamics of melanoma tumor therapy with vesicular stomatitis virus: explaining the variability in outcomes using mathematical modeling. Gene Ther. September 15, 2011. [Epub ahead of print.]
  • Wein LM, Wu JT, Kirn DH. Validation and analysis of a mathematical model of a replication-competent oncolytic virus for cancer treatment: implications for virus design and delivery. Cancer Res. 2003;63:1317–1324.
  • Wodarz D, Komarova N. Towards predictive computational models of oncolytic virus therapy: basis for experimental validation and model selection. PloS One. 2009;4:e4271.
  • Wu JT, Byrne HM, Kirn DH, Wein LM. Modeling and analysis of a virus that replicates selectively in tumor cells. Bull Math Biol. 2001;63:731–768.
  • Wu JT, Kirn DH, Wein LM. Analysis of a three-way race between tumor growth, a replication-competent virus and an immune response. Bull Math Biol. 2004;66:605–625.
  • Eftimie R, Dushoff J, Bridle BW, et al. Multi-stability and multiinstability phenomena in a mathematical model of tumor-immune- virus interactions. Bull Math Biol. 2011;73:2932–2961.