473
Views
21
CrossRef citations to date
0
Altmetric
REVIEW ARTICLE

Oncolytic Herpes Viruses as a Potential Mechanism for Cancer Therapy

Pages 660-671 | Received 18 Nov 2002, Accepted 04 Apr 2003, Published online: 08 Jul 2009

References

  • Dock G. The influence of complicating diseases upon leukaemia. Am J Med Sci 1904; 127: 563–92.
  • Webb HE, Smith CE. Viruses in the treatment of cancer. Lancet 1970; 1: 1206–8.
  • Moore AE. Effects of viruses on tumors. Ann Rev Microbiol 1954; 8: 393–410.
  • Smith RRHRJ, Rowe WP, Schatten WE, Thomas LB. Studies on the use of viruses in the treatment of carcinoma of the cervix. Cancer 1956; 9: 1211–8.
  • Southam C. Present status of oncolytic virus studies. Trans NY Acad Sci 1960; 22: 657–73.
  • Hermiston T. Gene delivery from replication-selective viruses: arming guided missiles in the war against cancer. J Clin Invest 2000; 105: 1169–72.
  • Kenney S, Pagano JS. Viruses as oncolytic agents: a new age for 'therapeutic' viruses? J Natl Cancer Inst 1994; 86: 1185–6.
  • Kim D, Martuza RL, Zwiebel J. Replication-selective vir-otherapy for cancer: biological principles, risk management and future directions. Nat Med 2001; 7: 781–7.
  • Kim DH. Replication-selective microbiological agents: fight-ing cancer with targeted germ warfare. J Clin Invest 2000; 105: 837–9.
  • Cassel WAG. Newcastle disease virus as an antineoplastic agent. Cancer 1965; 18: 863–8.
  • Lorence RM, Katubig BB, Reichard KW, et al. Complete regression of human fibrosarcoma xenografts after local Newcastle disease virus therapy. Cancer Res 1994; 54: 6017–21.
  • Lorence RM, Reichard KW, Katubig BB, et al. Complete regression of human neuroblastoma xenografts in athymic mice after local Newcastle disease virus therapy. J Natl Cancer Inst 1994; 86: 1228–33.
  • Pecora AL, Rizvi N, Cohen GI, et al. Phase I trial of intravenous administration of PV701, an oncolytic virus, in patients with advanced solid cancers. J Clin Oncol 2002; 20: 2251–66.
  • Richard KW, Lorence RM, Cascino CJ, et al. Newcastle disease virus selectively kills human tumor cells. J Surg Res 1992; 52: 448–53.
  • Coffey MC, Strong JE, Forsyth PA, Lee PW. Reovirus therapy of tumors with activated Ras pathway. Science 1998; 282: 1332–4.
  • Gross S. Measles and leukaemia. Lancet 1971; 1: 397–8.
  • Asada T. Treatment of human cancer with mumps virus. Cancer 1974; 34: 1907–28.
  • Levaditi CNS. Affinité du virus herpétique pour les néo-plasmes épithéliaux. Comptes Rendus des seances de la Société de Biologie 1922; 87: 498–500.
  • Hu Y, Lee J, McCart JA, et al. Yaba-like disease virus: an alternative replicating poxvirus vector for cancer gene therapy. J Virol 2001; 75: 10300–8.
  • Taylor MW, Cordell B, Souhrada M, Prather S. Viruses as an aid to cancer therapy: regression of solid and ascites tumors in rodents after treatment with bovine enterovirus. Proc Natl Acad Sci USA 1971; 68: 836–40.
  • Fujiwara T, Grimm EA, Mukhopadhyay T, Cai DW, Owen-Schaub LB, Roth JA. A retroviral wild-type p53 expression vector penetrates human lung cancer spheroids and inhibits growth by inducing apoptosis. Cancer Res 1993; 53: 4129–33.
  • Roth JA, Nguyen D, Lawrence DD, et al. Retrovirus-mediated wild-type p53 gene transfer to tumors of patients with lung cancer. Nat Med 1996; 2: 985–91.
  • Lohr F, Huang Q, Hu K, Dewhirst MW, Li CY. Systemic vector leakage and transgene expression by intratumorally injected recombinant adenovirus vectors. Clin Cancer Res 2001; 7: 3625–8.
  • Wang S, Baum BJ, Yamano S, et al. Adenoviral-mediated gene transfer to mouse salivary glands. J Dent Res 2000; 79: 701–8.
  • Xia H, Anderson B, Mao Q, Davidson BL. Recombinant human adenovirus: targeting to the human transferrin recep-tor improves gene transfer to brain microcapillary endothe-lium. J Virol 2000; 74: 11359–66.
  • Kaiser J. Gene therapy. Seeking the cause of induced leukemias in X-SCID trial. Science 2003; 299: 495.
  • Marshall E. Gene therapy a suspect in leukemia-like disease. Science 2002; 298: 34–5.
  • Marshall E. Gene therapy. Second child in French trial is found to have leukemia. Science 2003; 299: 320.
  • Bergmann M, Romirer I, Sachet M, et al. A genetically engineered influenza A virus with ras-dependent oncolytic properties. Cancer Res 2001; 61: 8188–93.
  • Zheng H, Palese P, Garcia-Sastre A. Antitumor properties of influenza virus vectors. Cancer Res 2000; 60: 6972–6.
  • Ansardi DC, Porter DC, Jackson CA, Gillespie GY, Morrow CD. RNA replicons derived from poliovirus are directly oncolytic for human tumor cells of diverse origins. Cancer Res 2001; 61: 8470–9.
  • Agha-Mohammadi S, Lotze MT. Immunomodulation of cancer: potential use of selectively replicating agents. J Clin Invest 2000; 105: 1173–6.
  • Bennett JJ, Tjuvajev J, Johnson P, et al. Positron emission tomography imaging for herpes virus infection: implications for oncolytic viral treatments of cancer. Nat Med 2001; 7: 859–63.
  • Culver KW, Ram Z, Wallbridge S, Ishii H, Oldfield EH, Blaese RM. In vivo gene transfer with retroviral vector-producer cells for treatment of experimental brain tumors. Science 1992; 256: 1550–2.
  • Moolten FL. Tumor chemosensitivity conferred by inserted herpes thymidine kinase genes: paradigm for a prospective cancer control strategy. Cancer Res 1986; 46: 5276–81.
  • Paillard F. Suicide genes against brain tumors. Hum Gene Ther 1998; 9: 3–4.
  • Coen DM, Kosz-Vnenchak M, Jacobson JG, et al. Thymidine kinase-negative herpes simplex virus mutants establish latency in mouse trigeminal ganglia but do not reactivate. Proc Natl Acad Sci USA 1989; 86: 4736–40.
  • Markert JM, Malick A, Coen DM, Martuza RL. Reduction and elimination of encephalitis in an experimental glioma therapy model with attenuated herpes simplex mutants that retain susceptibility to acyclovir. Neurosurgery 1993; 32: 597–603.
  • Bauerschmitz GJ, Lam JT, Kanerva A, et al. Treatment of ovarian cancer with a tropism modified oncolytic adenovirus. Cancer Res 2002; 62: 1266–70.
  • Douglas JT, Kim M, Sumerel LA, Carey DE, Curiel DT. Efficient oncolysis by a replicating adenovirus (ad) in vivo is critically dependent on tumor expression of primary ad receptors. Cancer Res 2001; 61: 813–7.
  • Suzuki K, Fueyo J, Krasnykh V, Reynolds PN, Curiel DT, Alemany R. A conditionally replicative adenovirus with enhanced infectivity shows improved oncolytic potency. Clin Cancer Res 2001; 7: 120–6.
  • Hemminki A, Dmitriev I, Liu B, Desmond RA, Alemany R, Curiel DT. Targeting oncolytic adenoviral agents to the epidermal growth factor pathway with a secretory fusion molecule. Cancer Res 2001; 61: 6377–81.
  • Bischoff JR, Kim DH, Williams A, et al. An adenovirus mutant that replicates selectively in p53-deficient human tumor cells. Science 1996; 274: 373–6.
  • Ramachandra M, Rahman A, Zou A, et al. Re-engineering adenovirus regulatory pathways to enhance oncolytic specifi-city and efficacy. Nat Biotechnol 2001; 19: 1035–41.
  • Brunori M, Malerba M, Kashiwazaki H, Iggo R. Replicating adenoviruses that target tumors with constitutive activation of the wnt signaling pathway. J Virol 2001; 75: 2857–65.
  • Matsubara S, Wada Y, Gardner TA, et al. A conditional replication-competent adenoviral vector, Ad-DC-El a, to co-target prostate cancer and bone stroma in an experimental model of androgen-independent prostate cancer bone metas-tasis. Cancer Res 2001; 61: 6012–9.
  • Adachi Y, Reynolds PN, Yamamoto M, et al. A midkine promoter-based conditionally replicative adenovirus for treat-ment of pediatric solid tumors and bone marrow tumor purging. Cancer Res 2001; 61: 7882–8.
  • Chen Y, DeWeese T, Dilley J, et al. CV706, a prostate cancer-specific adenovirus variant, in combination with radiotherapy produces synergistic antitumor efficacy without increasing toxicity. Cancer Res 2001; 61: 5453–60.
  • DeWeese TL, van der Poel H, Li S, et al. A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy. Cancer Res 2001; 61: 7464–72.
  • Yu DC, Chen Y, Dilley J, et al. Antitumor synergy of CV787, a prostate cancer-specific adenovirus, and paclitaxel and docetaxel. Cancer Res 2001; 61: 517–25.
  • Yang Y, Li Q, Ertl HC, Wilson JM. Cellular and humoral immune responses to viral antigens create barriers to lung-directed gene therapy with recombinant adenoviruses. J Virol 1995; 69: 2004–15.
  • Maione D, Della Rocca C, Giannetti P, et al. An improved helper-dependent adenoviral vector allows persistent gene expression after intramuscular delivery and overcomes pre-existing immunity to adenovirus. Proc Natl Acad Sci USA 2001; 98: 5986–91.
  • Thomas CE, Schiedner G, Kochanek S, Castro MG, Low-enstein PR. Preexisting antiadenoviral immunity is not a barrier to efficient and stable transduction of the brain, mediated by novel high-capacity adenovirus vectors. Hum Gene Ther 2001; 12: 839–46.
  • Mesnil M, Yamasaki H. Bystander effect in herpes simplex virus-thymidine kinase/ganciclovir cancer gene therapy: role of gap-junctional intercellular communication. Cancer Res 2000; 60: 3989–99.
  • Freeman SM, Abboud CN, Whartenby KA, et al. The 'bystander effect': tumor regression when a fraction of the tumor mass is genetically modified. Cancer Res 1993; 53: 5274–83.
  • Nagamachi Y, Tani M, Shimizu K, Yoshida T, Yokota J. Suicidal gene therapy for pleural metastasis of lung cancer by liposome-mediated transfer of herpes simplex virus thymidine kinase gene. Cancer Gene Ther 1999; 6: 546–53.
  • Nanda D, Vogels R, Havenga M, Avezaat CJ, Bout A, Smitt PS. Treatment of malignant gliomas with a replicating adenoviral vector expressing herpes simplex virus-thymidine kinase. Cancer Res 2001; 61: 8743–50.
  • Wildner 0, Blaese RM, Morris JC. Therapy of colon cancer with oncolytic adenovirus is enhanced by the addition of herpes simplex virus-thymidine kinase. Cancer Res 1999; 59: 410–3.
  • Wildner O, Morris JC. The role of the ElB 55 kDa gene product in oncolytic adenoviral vectors expressing herpes simplex virus-tk: assessment of antitumor efficacy and toxi-city. Cancer Res 2000; 60: 4167–74.
  • Kucharczuk JC, Raper S, Elshami AA, et al. Safety of intrapleurally administered recombinant adenovirus carrying herpes simplex thymidine kinase DNA followed by ganciclovir therapy in nonhuman primates. Hum Gene Ther 1996; 7: 2225–33.
  • Sterman DH, Treat J, Litzky LA, et al. Adenovirus-mediated herpes simplex virus thymidine kinase/ganciclovir gene ther-apy in patients with localized malignancy: results of a phase I clinical trial in malignant mesothelioma. Hum Gene Ther 1998; 9: 1083–92.
  • Wildner 0, Morris JC, Vahanian NN, Ford H Jr, Ramsey WJ, Blaese RM. Adenoviral vectors capable of replication improve the efficacy of HSVtk/GCV suicide gene therapy of cancer. Gene Ther 1999; 6: 57–62.
  • Lambright ES, Amin K, Wiewrodt R, et al. Inclusion of the herpes simplex thymidine kinase gene in a replicating adeno-virus does not augment antitumor efficacy. Gene Ther 2001; 8: 946–53.
  • Wygoda MR, Wilson MR, Davis MA, Trosko JE, Rehemtulla A, Lawrence TS. Protection of herpes simplex virus thymidine kinase-transduced cells from ganciclovir-mediated cytotoxi-city by bystander cells: the Good Samaritan effect. Cancer Res 1997; 57: 1699–703.
  • Chen SH, Shine HD, Goodman JC, Grossman RG, Woo SL. Gene therapy for brain tumors: regression of experimental gliomas by adenovirus-mediated gene transfer in vivo. Proc Natl Acad Sci USA 1994; 91: 3054–7.
  • Freytag SO, Rogulski KR, Paielli DL, Gilbert JD, Kim JH. A novel three-pronged approach to kill cancer cells selectively: concomitant viral, double suicide gene, and radiotherapy. Hum Gene Ther 1998; 9: 1323–33.
  • Rogulski KR, Freytag SO, Zhang K, et al. In vivo antitumor activity of ONYX-015 is influenced by p53 status and is augmented by radiotherapy. Cancer Res 2000; 60: 1193–6.
  • Rogulski KR, Wing MS, Paielli DL, Gilbert JD, Kim JH, Freytag SO. Double suicide gene therapy augments the antitumor activity of a replication-competent lytic adenovirus through enhanced cytotoxicity and radiosensitization. Hum Gene Ther 2000; 11: 67–76.
  • Elshami AA, Kucharczuk JC, Sterman DH, et al. The role of immunosuppression in the efficacy of cancer gene therapy using adenovirus transfer of the herpes simplex thymidine kinase gene. Ann Surg 1995; 222: 298–310.
  • Yang L, Chiang Y, Lenz HJ, et al. Intercellular communica-tion mediates the bystander effect during herpes simplex thymidine kinase/ganciclovir-based gene therapy of human gastrointestinal tumor cells. Hum Gene Ther 1998; 9: 719–28.
  • Bi WL, Parysek LM, Warnick R, Stambrook PJ. In vitro evidence that metabolic cooperation is responsible for the bystander effect observed with HSV tk retroviral gene therapy. Hum Gene Ther 1993; 4: 725–31.
  • Bi W, Kim YG, Feliciano ES, et al. An HSVtk-mediated local and distant antitumor bystander effect in tumors of head and neck origin in athymic mice. Cancer Gene Ther 1997; 4: 246–52.
  • Glorioso JC, DeLuca NA, Fink DJ. Development and application of herpes simplex virus vectors for human gene therapy. Annu Rev Microbiol 1995; 49: 675–710.
  • Cotter MA, Robertson ES. Molecular genetic analysis of herpesviruses and their potential use as vectors for gene therapy applications. Curr Opin Mol Ther 1999; 1: 633–44.
  • Boviatsis EJ, Chase M, Wei MX, et al. Gene transfer into experimental brain tumors mediated by adenovirus, herpes simplex virus, and retrovirus vectors. Hum Gene Ther 1994; 5: 183–91.
  • Markert JM, Gillespie GY, Weichselbaum RR, Roizman B, Whitley RJ. Genetically engineered HSV in the treatment of glioma: a review. Rev Med Virol 2000; 10: 17–30.
  • Wolfe D, Goins WF, Yamada M, et al. Engineering herpes simplex virus vectors for CNS applications. Exp Neurol 1999; 159: 34–46.
  • Goins WF, Krisky D, Marconi P, et al. Herpes simplex virus vectors for gene transfer to the nervous system. J Neurovirol 1997; 3(Suppl 1): S80–8.
  • Roizman B. The function of herpes simplex virus genes: a primer for genetic engineering of novel vectors. Proc Natl Acad Sci USA 1996; 93: 11307–12.
  • Geller Al, Keyomarsi K, Bryan J, Pardee AB. An efficient deletion mutant packaging system for defective herpes simplex virus vectors: potential applications to human gene therapy and neuronal physiology. Proc Natl Acad Sci USA 1990; 87: 8950–4.
  • Marconi P, Krisky D, Oligino T, et al. Replication-defective herpes simplex virus vectors for gene transfer in vivo. Proc Natl Acad Sci USA 1996; 93: 11319–20.
  • Pechan PA, Fotaki M, Thompson RL, et al. A novel 'piggy-back' packaging system for herpes simplex virus amplicon vectors. Hum Gene Ther 1996; 7: 2003–13.
  • Todryk S, McLean C, Ali S, et al. Disabled infectious single-cycle herpes simplex virus as an oncolytic vector for immu-notherapy of colorectal cancer. Hum Gene Ther 1999; 10: 2757–68.
  • Yanagi K, Talavera A, Nishimoto T, Rush MG. Inhibition of herpes simplex virus type 1 replication in temperature-sensitive cell cycle mutants. J Virol 1978; 25: 42–50.
  • Ichikawa T, Chiocca EA. Comparative analyses of transgene delivery and expression in tumors inoculated with a replica-tion-conditional or -defective viral vector. Cancer Res 2001; 61: 5336–9.
  • Markert JM, Coen DM, Malick A, Mineta T, Martuza RL. Expanded spectrum of viral therapy in the treatment of nervous system tumors. J Neurosurg 1992; 77: 590–4.
  • Markert JM, Medlock MD, Rabkin SD, et al. Conditionally replicating herpes simplex virus mutant, G207 for the treat-ment of malignant glioma: results of a phase I trial. Gene Ther 2000; 7: 867–74.
  • Martuza RL, Malick A, Markert JM, Ruffner KL, Coen DM. Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science 1991; 252: 854–6.
  • Mineta T, Markert JM, Takamiya Y, Coen DM, Rabkin SD, Martuza RL. CNS tumor therapy by attenuated herpes simplex viruses. Gene Ther 1994; 1: S78.
  • Mineta T, Rabkin SD, Martuza RL. Treatment of malignant gliomas using ganciclovir-hypersensitive, ribonucleotide re- ductase-deficient herpes simplex viral mutant. Cancer Res 1994; 54: 3963–6.
  • Mineta T, Rabkin SD, Yazaki T, Hunter WD, Martuza RL. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med 1995; 1: 938–43.
  • Chahlavi A, Todo T, Martuza RL, Rabkin SD. Replication-competent herpes simplex virus vector G207 and cisplatin combination therapy for head and neck squamous cell carcinoma. Neoplasia 1999; 1: 162–9.
  • Coukos G, Makrigiannakis A, Kang EH, Rubin SC, Albelda SM, Molnar-Kimber KL. Oncolytic herpes simplex virus-1 lacking ICP34.5 induces p53-independent death and is efficacious against chemotherapy-resistant ovarian cancer. Clin Cancer Res 2000; 6: 3342–53.
  • Coukos G, Makrigiannakis A, Montas S, et al. Multi-attenuated herpes simplex virus-1 mutant G207 exerts cyto-toxicity against epithelial ovarian cancer but not normal mesothelium and is suitable for intraperitoneal oncolytic therapy. Cancer Gene Ther 2000; 7: 275–83.
  • Randazzo BP, Bhat MG, Kesari S, Fraser NW, Brown SM. Treatment of experimental subcutaneous human melanoma with a replication-restricted herpes simplex virus mutant. J Invest Dermatol 1997; 108: 933–7.
  • Wong RJ, Kim SH, Joe JK, Shah JP, Johnson PA, Fong Y Effective treatment of head and neck squamous cell carci-noma by an oncolytic herpes simplex virus. J Am Coll Surg 2001; 193: 12–21.
  • Yoon SS, Carroll NM, Chiocca EA, Tanabe KK. Cancer gene therapy using a replication-competent herpes simplex virus type 1 vector. Ann Surg 1998; 228: 366–74.
  • Boeckh M, Zaia JA, Jung D, Skettino S, Chauncey TR, Bowden RA. A study of the pharmacokinetics, antiviral activity, and tolerability of oral ganciclovir for CMV prophy-laxis in marrow transplantation. Biol Blood Marrow Trans-plant 1998; 4: 13–9.
  • Kennedy GA, Kay TD, Johnson DW, et al. Neutrophil dysplasia characterised by a pseudo-Pelger-Huet anomaly occurring with the use of mycophenolate mofetil and ganci-clovir following renal transplantation: a report of five cases. Pathology 2002; 34: 263–6.
  • Sharathkumar A, Shaw P. Ganciclovir-induced encephalopa-thy in a bone marrow transplant recipient. Bone Marrow Transplant 1999; 24: 421–3.
  • Turgeon N, Hovingh GK, Fishman JA, et al. Safety and efficacy of granulocyte colony-stimulating factor in kidney and liver transplant recipients. Transpl Infect Dis 2000; 2: 15–21.
  • Black ME, Kokoris MS, Sabo P. Herpes simplex virus-1 thymidine kinase mutants created by semi-random sequence mutagenesis improve prodrug-mediated tumor cell killing. Cancer Res 2001; 61: 3022–6.
  • Miyatake S, Martuza RL, Rabkin SD. Defective herpes simplex virus vectors expressing thymidine kinase for the treatment of malignant glioma. Cancer Gene Ther 1997; 4: 222–8.
  • Moriuchi S, Krisky DM, Marconi PC, et al. HSV vector cytotoxicity is inversely correlated with effective TK/GCV suicide gene therapy of rat gliosarcoma. Gene Ther 2000; 7: 1483–90.
  • Nakamura H, Mullen JT, Chandrasekhar S, Pawlik TM, Yoon SS, Tanabe KK. Multimodality therapy with a replica-tion-conditional herpes simplex virus 1 mutant that expresses yeast cytosine deaminase for intratumoral conversion of 5-fluorocytosine to 5-fluorouracil. Cancer Res 2001; 61: 5447–52.
  • Chase M, Chung RY, Chiocca EA. An oncolytic viral mutant that delivers the CYP2B1 transgene and augments cyclopho-sphamide chemotherapy. Nat Biotechnol 1998; 16: 444–8.
  • Herrlinger U, Woiciechowski C, Sena-Esteves M, et al. Neural precursor cells for delivery of replication-conditional HSV-1 vectors to intracerebral gliomas. Mol Ther 2000; 1: 347–57.
  • Ichikawa T, Petros WP, Ludeman SM, et al. Intraneoplastic polymer-based delivery of cyclophosphamide for intratumoral bioconversion by a replicating oncolytic viral vector. Cancer Res 2001; 61: 864–8.
  • Pawlik TM, Nakamura H, Yoon SS, et al. Oncolysis of diffuse hepatocellular carcinoma by intravascular administration of a replication-competent, genetically engineered herpesvirus. Cancer Res 2000; 60: 2790–5.
  • Jia WW, McDermott M, Goldie J, Cynader M, Tan J, Tufaro F. Selective destruction of gliomas in immunocompetent rats by thymidine kinase-defective herpes simplex virus type 1. J Natl Cancer Inst 1994; 86: 1209–15.
  • Jia WW, Tan J, Redekop GJ, Goldie JH. Toxicity studies in thymidine kinase-deficient herpes simplex virus therapy for malignant astrocytoma. J Neurosurg 1996; 85: 662–6.
  • Valyi-Nagy T, Fareed MU, O'Keefe JS, et al. The herpes simplex virus type 1 strain 17+ gamma 34.5 deletion mutant 1716 is avirulent in SCID mice. J Gen Virol 1994; 75: 2059–63.
  • Valyi-Nagy T, Gesser RM, Raengsakulrach B, et al. A thymidine kinase-negative HSV-1 strain establishes a persis-tent infection in SCID mice that features uncontrolled peripheral replication but only marginal nervous system involvement. Virology 1994; 199: 484–90.
  • Goldstein DJ, Weller SK. Herpes simplex virus type 1-induced ribonucleotide reductase activity is dispensable for virus growth and DNA synthesis: isolation and characteriza-tion of an ICP6 lacZ insertion mutant. J Virol 1988; 62: 196–205.
  • Petrowsky H, Roberts GD, Kooby DA, et al. Functional interaction between fluorodeoxyuridine-induced cellular al-terations and replication of a ribonucleotide reductase-nega-tive herpes simplex virus. J Virol 2001; 75: 7050–8.
  • Jacobs A, Tjuvajev JG, Dubrovin M, et al. Positron emission tomography-based imaging of transgene expression mediated by replication-conditional, oncolytic herpes simplex virus type 1 mutant vectors in vivo. Cancer Res 2001; 61: 2983–95.
  • Kramm CM, Rainov NG, Sena-Esteves M, et al. Long-term survival in a rodent model of disseminated brain tumors by combined intrathecal delivery of herpes vectors and ganciclo-vir treatment. Hum Gene Ther 1996; 7: 1989–94.
  • Kasuya H, Nishiyama Y, Nomoto S, Hosono J, Takeda S, Nakao A. Intraperitoneal delivery of hrR3 and ganciclovir prolongs survival in mice with disseminated pancreatic cancer. J Surg Oncol 1999; 72: 136–41.
  • 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: 881–7.
  • 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: 4765–75.
  • Lasner TM, Kesari S, Brown SM, Lee VM, Fraser NW, Trojanowski JQ. Therapy of a murine model of pediatric brain tumors using a herpes simplex virus type-1 ICP34.5 mutant and demonstration of viral replication within the CNS. J Neuropathol Exp Neurol 1996; 55: 1259–69.
  • Kesari S, Randazzo BP, Valyi-Nagy T, et al. Therapy of experimental human brain tumors using a neuroattenuated herpes simplex virus mutant. Lab Invest 1995; 73: 636–48.
  • Kucharczuk JC, Randazzo B, Chang MY, et al. Use of a 'replication-restricted' herpes virus to treat experimental hu-man malignant mesothelioma. Cancer Res 1997; 57: 466–71.
  • Rampling R, Cruickshank G, Papanastassiou V, et al. Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther 2000; 7: 859–66.
  • Papanastassiou V, Rampling R, Fraser M, et al. The potential for efficacy of the modified (ICP 34.5(-)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study. Gene Ther 2002; 9: 398–406.
  • Andreansky SS, He B, Gillespie GY, et al. The application of genetically engineered herpes simplex viruses to the treatment of experimental brain tumors. Proc Natl Acad Sci USA 1996; 93: 11313–8.
  • Chambers R, Gillespie GY, Soroceanu L, et al. Comparison of genetically engineered herpes simplex viruses for the treatment of brain tumors in a scid mouse model of human malignant glioma. Proc Natl Acad Sci USA 1995; 92: 1411–5.
  • Randazzo BP, Kucharczuk JC, Litzky LA, et al. Herpes simplex 1716-an ICP 34.5 mutant-is severely replication restricted in human skin xenografts in vivo. Virology 1996; 223: 392–5.
  • Andreansky S, Soroceanu L, Flotte ER, et al. Evaluation of genetically engineered herpes simplex viruses as oncolytic agents for human malignant brain tumors. Cancer Res 1997; 57: 1502–9.
  • Taneja S, MacGregor J, Markus S, Ha S, Mohr I. Enhanced antitumor efficacy of a herpes simplex virus mutant isolated by genetic selection in cancer cells. Proc Natl Acad Sci USA 2001; 98: 8804–8.
  • Advani SJ, Sibley GS, Song PY, et al. Enhancement of replication of genetically engineered herpes simplex viruses by ionizing radiation: a new paradigm for destruction of therapeutically intractable tumors. Gene Ther 1998; 5: 160–5.
  • Bradley JD, Kataoka Y, Advani S, et al. Ionizing radiation improves survival in mice bearing intracranial high-grade gliomas injected with genetically modified herpes simplex virus. Clin Cancer Res 1999; 5: 1517–22.
  • Pyles RB, Wamick RE, Chalk CL, Szanti BE, Parysek LM. A novel multiple-mutated HSV-1 strain for the treatment of human brain tumors. Hum Gene Ther 1997; 8: 533–44.
  • Andreansky S, He B, van Cott J, et al. Treatment of intracranial gliomas in immunocompetent mice using herpes simplex viruses that express murine interleukins. Gene Ther 1998; 5: 121–30.
  • 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: 871–82.
  • Kramm CM, Chase M, Herrlinger U, et al. Therapeutic efficiency and safety of a second-generation replication-conditional HSV1 vector for brain tumor gene therapy. Hum Gene Ther 1997; 8: 2057–68.
  • Yazaki T, Manz HJ, Rabkin SD, Martuza RL. Treatment of human malignant meningiomas by G207, a replication-competent multimutated herpes simplex virus 1. Cancer Res 1995; 55: 4752–6.
  • Mashour GA, Moulding HD, Chahlavi A, et al. Therapeutic efficacy of G207 in a novel peripheral nerve sheath tumor model. Exp Neurol 2001; 169: 64–71.
  • Todo T, Rabkin SD, Martuza RL. Evaluation of ganciclovir-mediated enhancement of the antitumoral effect in oncolytic, multimutated herpes simplex virus type 1 (G207) therapy of brain tumors. Cancer Gene Ther 2000; 7: 939–46.
  • Toda M, Iizuka Y, Kawase T, Uyemura K, Kawakami Y. Immuno-viral therapy of brain tumors by combination of viral therapy with cancer vaccination using a replication-conditional HSV. Cancer Gene Ther 2002; 9: 356–64.
  • Toda M, Rabkin SD, Kojima H, Martuza RL. Herpes simplex virus as an in situ cancer vaccine for the induction of specific anti-tumor immunity. Hum Gene Ther 1999; 10: 385–93.
  • Todo T, Rabkin SD, Sundaresan P, et al. Systemic antitumor immunity in experimental brain tumor therapy using a multimutated, replication-competent herpes simplex virus. Hum Gene Ther 1999; 10: 2741–55.
  • 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: 153–61.
  • Endo T, Toda M, Watanabe M, et al. In situ cancer vaccination with a replication-conditional HSV for the treatment of liver metastasis of colon cancer. Cancer Gene Ther 2002; 9: 142–8.
  • Fruh K, Ahn K, Djaballah H, et al. A viral inhibitor of peptide transporters for antigen presentation. Nature 1995; 375: 415–8.
  • Hill A, Jugovic P, York I, et al. Herpes simplex virus turns off the TAP to evade host immunity. Nature 1995; 375: 411–5.
  • Hunter WD, Martuza RL, Feigenbaum F, et al. Attenuated, replication-competent herpes simplex virus type 1 mutant G207: safety evaluation of intracerebral injection in nonhu-man primates. J Virol 1999; 73: 6319–26.
  • Sundaresan P, Hunter WD, Martuza RL, Rabkin SD. Attenuated, replication-competent herpes simplex virus type 1 mutant G207: safety evaluation in mice. J Virol 2000; 74: 3832–41.
  • Todo T, Feigenbaum F, Rabkin SD, et al. Viral shedding and biodistribution of G207, a multimutated, conditionally repli-cating herpes simplex virus type 1, after intracerebral inocu-lation in aotus. Mol Ther 2000; 2: 588–95.
  • Varghese S, Newsome JT, Rabkin SD, et al. Preclinical safety evaluation of G207, a replication-competent herpes simplex virus type 1, inoculated intraprostatically in mice and nonhu-man primates. Hum Gene Ther 2001; 12: 999–1010.
  • Kim DH. A tale of two trials: selectively replicating herpes-viruses for brain tumors. Gene Ther 2000; 7: 815–6.
  • Jorgensen TJ, Katz S, Wittmack EK, et al. Ionizing radiation does not alter the antitumor activity of herpes simplex virus vector G207 in subcutaneous tumor models of human and murine prostate cancer. Neoplasia 2001; 3: 451–6.
  • Todo T, Rabkin SD, Chahlavi A, Martuza RL. Corticosteroid administration does not affect viral oncolytic activity, but inhibits antitumor immunity in replication-competent herpes simplex virus tumor therapy. Hum Gene Ther 1999; 10: 2869–78.
  • Delman KA, Bennett JJ, Zager JS, et al. Effects of preexisting immunity on the response to herpes simplex-based oncolytic viral therapy. Hum Gene Ther 2000; 11: 2465–72.
  • Chahlavi A, Rabkin S, Todo T, Sundaresan P, Martuza R. Effect of prior exposure to herpes simplex virus 1 on viral vector-mediated tumor therapy in immunocompetent mice. Gene Ther 1999; 6: 1751–8.
  • Herrlinger U, Kramm CM, Aboody-Guterman KS, et al. Pre-existing herpes simplex virus 1 (HSV-1) immunity decreases, but does not abolish, gene transfer to experimental brain tumors by a HSV-1 vector. Gene Ther 2001; 5: 809–19.
  • Bennett JJ, Kooby DA, Delman K, et al. Antitumor efficacy of regional oncolytic viral therapy for peritoneally dissemi-nated cancer. J Mol Med 2000; 78: 166–74.
  • Blank SV, Rubin SC, Coukos G, Amin KM, Albelda SM, Molnar-Kimber KL. Replication-selective herpes simplex virus type 1 mutant therapy of cervical cancer is enhanced by low-dose radiation. Hum Gene Ther 2002; 13: 627–39.
  • Brandt CR, Imesch PD, Robinson NL, et al. Treatment of spontaneously arising retinoblastoma tumors in transgenic mice with an attenuated herpes simplex virus mutant. Virology 1997; 229: 283–91.
  • Coukos G, Makrigiannakis A, Kang EH, et al. Use of carrier cells to deliver a replication-selective herpes simplex virus-1 mutant for the intraperitoneal therapy of epithelial ovarian cancer. Clin Cancer Res 1999; 5: 1523–37.
  • Cozzi PJ, Malhotra S, McAuliffe P, et al. Intravesical oncolytic viral therapy using attenuated, replication-competent herpes simplex viruses G207 and Nv1020 is effective in the treatment of bladder cancer in an orthotopic syngeneic model. Faseb J 2001; 15: 1306–8.
  • Kooby DA, Carew JF, Halterman MW, et al. Oncolytic viral therapy for human colorectal cancer and liver metastases using a multi-mutated herpes simplex virus type-1 (G207). Faseb J 1999; 13: 1325–34.
  • Lee JH, Federoff HJ, Schoeniger LO. G207, modified herpes simplex virus type 1, kills human pancreatic cancer cells in vitro. J Gastrointest Surg 1999; 3: 127–31; discussion 32-3.
  • McAuliffe PF, Jarnagin WR, Johnson P, Delman KA, Feder-off H, Fong Y Effective treatment of pancreatic tumors with two multimutated herpes simplex oncolytic viruses. J Gastro-intest Surg 2000; 4: 580–8.
  • Nakano K, Todo T, Chijiiwa K, Tanaka M. Therapeutic efficacy of G207, a conditionally replicating herpes simplex virus type 1 mutant, for gallbladder carcinoma in immuno-competent hamsters. Mol Ther 2001; 3: 431–7.
  • Oyama M, Ohigashi T, Hoshi M, Murai M, Uyemura K, Yazaki T. Oncolytic viral therapy for human prostate cancer by conditionally replicating herpes simplex virus 1 vector G207. Jpn J Cancer Res 2000; 91: 1339–44.
  • Oyama M, Ohigashi T, Hoshi M, Murai M, Uyemura K, Yazaki T. Treatment of human renal cell carcinoma by a conditionally replicating herpes vector G207. J Urol 2001; 165: 1274–8.
  • Randazzo BP, Kesari S, Gesser RM, et al. Treatment of experimental intracranial murine melanoma with a neuroatte-nuated herpes simplex virus 1 mutant. Virology 1995; 211: 94–101.
  • Toda M, Rabkin SD, Martuza RL. Treatment of human breast cancer in a brain metastatic model by G207, a replication-competent multimutated herpes simplex virus 1. Hum Gene Ther 1998; 9: 2177–85.
  • Walker JR, McGeagh KG, Sundaresan P, Jorgensen TJ, Rabkin SD, Martuza RL. Local and systemic therapy of human prostate adenocarcinoma with the conditionally re-plicating herpes simplex virus vector G207. Hum Gene Ther 1999; 10: 2237–43.
  • 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: 253–65.
  • Wu A, Mazumder A, Martuza RL, et al. Biological purging of breast cancer cells using an attenuated replication-competent herpes simplex virus in human hematopoietic stem cell transplantation. Cancer Res 2001; 61: 3009–15.
  • Advani SJ, Chung SM, Yan SY, et al. Replication-competent, nonneuroinvasive genetically engineered herpes virus is highly effective in the treatment of therapy-resistant experimental human tumors. Cancer Res 1999; 59: 2055–8.
  • Chung SM, Advani SJ, Bradley JD, et al. The use of a genetically engineered herpes simplex virus (R7020) with ionizing radiation for experimental hepatoma. Gene Ther 2002; 9: 75–80.
  • Carew JF, Kooby DA, Halterman MW, Kim SH, Federoff HJ, Fong Y. A novel approach to cancer therapy using an oncolytic herpes virus to package amplicons containing cytokine genes. Mol Ther 2001; 4: 250–6.
  • Parker JN, Gillespie GY, Love CE, Randall S, Whitley RJ, Markert JM. Engineered herpes simplex virus expressing IL-12 in the treatment of experimental murine brain tumors. Proc Natl Acad Sci USA 2000; 97: 2208–13.
  • Toda M, Martuza RL, Kojima H, Rabkin SD. In situ cancer vaccination: an IL-12 defective vector/replication-competent herpes simplex virus combination induces local and systemic antitumor activity. J Immunol 1998; 160: 4457–64.
  • Zager JS, Delman KA, Malhotra S, et al. Combination vascular delivery of herpes simplex oncolytic viruses and amplicon mediated cytokine gene transfer is effective therapy for experimental liver cancer. Mol Med 2001; 7: 561–8.
  • Moriuchi S, Oligino T, Krisky D, et al. Enhanced tumor cell killing in the presence of ganciclovir by herpes simplex virus type 1 vector-directed coexpression of human tumor necrosis factor-alpha and herpes simplex virus thymidine kinase. Cancer Res 1998; 58: 5731–7.
  • Toda M, Martuza RL, Rabkin SD. Combination suicide/ cytokine gene therapy as adjuvants to a defective herpes simplex virus-based cancer vaccine. Gene Ther 2001; 8: 332–9.
  • Jacobs A, Breakefield XO, Fraefel C. HSV-1-based vectors for gene therapy of neurological diseases and brain tumors: part I. HSV-1 structure, replication and pathogenesis. Neoplasia 1999; 1: 387–401.
  • Jacobs A, Breakefield XO, Fraefel C. HSV-1-based vectors for gene therapy of neurological diseases and brain tumors: part II. Vector systems and applications. Neoplasia 1999; 1: 402–16.
  • MacLean AR, ul-Fareed M, Robertson L, Harland J, Brown SM. Herpes simplex virus type 1 deletion variants 1714 and 1716 pinpoint neurovirulence-related sequences in Glasgow strain 17 + between immediate early gene 1 and the 'a' sequence. J Gen Virol 1991; 72: 631–9.
  • Chou J, Kern ER, Whitley RJ, Roizman B. Mapping of herpes simplex virus-1 neurovirulence to gamma 134.5, a gene nonessential for growth in culture. Science 1990; 250: 1262–6.
  • Todo T, Martuza RL, Rabkin SD, Johnson PA. Oncolytic herpes simplex virus vector with enhanced MHC class I presentation and tumor cell killing. Proc Natl Acad Sci USA 2001; 98: 6396–401.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.