We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Oncolytic herpes simplex virus vectors and chemotherapy: are combinatorial strategies more effective for cancer?

    Ryuichi Kanai

    Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, & Harvard Medical School, Boston, MA, USA

    ,
    Hiroaki Wakimoto

    Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, & Harvard Medical School, Boston, MA, USA

    ,
    Tooba Cheema

    Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, & Harvard Medical School, Boston, MA, USA

    &
    Samuel D Rabkin

    † Author for correspondence

    Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, & Harvard Medical School, Boston, MA, USA.

    Published Online:https://doi.org/10.2217/fon.10.18

    >Despite aggressive treatments, including chemotherapy and radiotherapy, cancers often recur owing to resistance to conventional therapies. Oncolytic viruses such as oncolytic herpes simplex virus (oHSV) represent an exciting biological approach to cancer therapy. A range of viral mutations has been engineered into HSV to engender oncolytic activity. While oHSV as a single agent has been tested in a number of cancer clinical trials, preclinical studies have demonstrated enhanced efficacy when it is combined with cytotoxic anticancer drugs. Among the strategies that will be discussed in this article are combinations with standard-of-care chemotherapeutics, expression of prodrug-activating enzymes to enhance chemotherapy and small-molecule inhibitors. The combination of oHSV and chemotherapy can achieve much more efficient cancer cell killing than either single agent alone, often through synergistic interactions. This can be clinically important not just for improving efficacy but also for permitting lower and less toxic chemotherapeutic doses. The viral mutations in an oHSV vector often determine the favorability of its interactions with chemotherapy, just as different cancer cells, due to genetic alterations, vary in their response to chemotherapy. As chemotherapeutics are often the standard of care, combining them with an investigational new drug, such as oHSV, is clinically easier than combining multiple novel agents. As has become clear for most cancer therapies, multimodal treatments are usually more effective. In this article, we will discuss the recent progress of these combinatorial strategies between virotherapy and chemotherapy and future directions.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Guo ZS, Thorne SH, Bartlett DL: Oncolytic virotherapy: molecular targets in tumor-selective replication and carrier cell-mediated delivery of oncolytic viruses. Biochim. Biophys. Acta1785,217–231 (2008).
    • Kelly E, Russell SJ: History of oncolytic viruses: genesis to genetic engineering. Mol. Ther.15,651–659 (2007).
    • Russell SJ, Peng KW: Viruses as anticancer drugs. Trends Pharmacol. Sci.28,326–333 (2007).
    • Martuza RL, Malick A, Markert JM, Ruffner KL, Coen DM: Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science252,854–856 (1991).
    • Kirn D: Clinical research results with dl1520 (Onyx-015), a replication-selective adenovirus for the treatment of cancer: what have we learned? Gene Ther.8,89–98 (2001).
    • Liu TC, Galanis E, Kirn D: Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress. Nat. Clin.Pract. Oncol.4,101–117 (2007).
    • Roizman B, Knipe DM, Whitley RJ. Herpes simplex viruses. In: Fields Virology (5th Edition). Knipe DM, Howley PM (Eds). Wolters Kluwer Health/Lippincott Williams & Wilkins, PA, USA, 2501–2601 (2007).
    • Varghese S, Rabkin SD: Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther.9,967–978 (2002).
    • Shen Y, Nemunaitis J: Herpes simplex virus 1 (HSV-1) for cancer treatment. Cancer Gene Ther.13,975–992 (2006).
    • 10  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.1,938–943 (1995).
    • 11  Aghi M, Rabkin S: Viral vectors as therapeutic agents for glioblastoma. Curr. Opin. Mol. Ther.7,419–430 (2005).
    • 12  Reinblatt M, Pin RH, Fong Y: Herpes viral oncolysis: a novel cancer therapy. J. Am. Coll. Surg.205,S69–S75 (2007).
    • 13  Markert JM, Liechty PG, Wang W et al.: Phase Ib trial of mutant herpes simplex virus G207 inoculated pre-and post-tumor resection for recurrent GBM. Mol. Ther.17,199–207 (2009).
    • 14  Yun CO: Overcoming the extracellular matrix barrier to improve intratumoral spread and therapeutic potential of oncolytic virotherapy. Curr. Opin. Mol. Ther.10,356–361 (2008).
    • 15  Wakimoto H, Johnson PR, Knipe DM, Chiocca EA: Effects of innate immunity on herpes simplex virus and its ability to kill tumor cells. Gene Ther.10,983–990 (2003).
    • 16  Fukuhara H, Todo T: Oncolytic herpes simplex virus type 1 and host immune responses. Curr. Cancer Drug Targets7,149–155 (2007).
    • 17  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.27,5763–5771 (2009).▪ First report of successful Phase II clinical trial with oncolytic herpes simplex virus (oHSV).
    • 18  Kumar S, Gao L, Yeagy B, Reid T: Virus combinations and chemotherapy for the treatment of human cancers. Curr. Opin. Mol. Ther.10,371–379 (2008).
    • 19  Nguyen TL, Tumilasci VF, Singhroy D, Arguello M, Hiscott J: The emergence of combinatorial strategies in the development of RNA oncolytic virus therapies. Cell. Microbiol.11,889–897 (2009).
    • 20  Alvarez-Breckenridge C, Kaur B, Chiocca EA: Pharmacologic and chemical adjuvants in tumor virotherapy. Chem. Rev.109,3125–3140 (2009).
    • 21  Heise C, Sampson-Johannes A, Williams A, McCormick F, Von Hoff DD, Kirn DH: ONYX-015, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents. Nat. Med.3,639–645 (1997).
    • 22  Khuri FR, Nemunaitis J, Ganly I et al.: A controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat. Med.6,879–885 (2000).
    • 23  Reid T, Galanis E, Abbruzzese J et al.: Intra-arterial administration of a replication-selective adenovirus (dl1520) in patients with colorectal carcinoma metastatic to the liver: a Phase I trial. Gene Ther.8,1618–1626 (2001).
    • 24  Galanis E, Okuno SH, Nascimento AG et al.: Phase I-II trial of ONYX-015 in combination with MAP chemotherapy in patients with advanced sarcomas. Gene Ther.12,437–445 (2005).
    • 25  Nemunaitis J, Cunningham C, Tong AW et al.: Pilot trial of intravenous infusion of a replication-selective adenovirus (ONYX-015) in combination with chemotherapy or IL-2 treatment in refractory cancer patients. Cancer Gene Ther.10,341–352 (2003).
    • 26  Hecht JR, Bedford R, Abbruzzese JL et al.: A Phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinoma. Clin. Cancer Res.9,555–561 (2003).
    • 27  Yu W, Fang H: Clinical trials with oncolytic adenovirus in China. Curr. Cancer Drug Targets7,141–148 (2007).
    • 28  Quirin C, Mainka A, Hesse A, Nettelbeck DM: Combining adenoviral oncolysis with temozolomide improves cell killing of melanoma cells. Int. J. Cancer121,2801–2807 (2007).
    • 29  Leitner S, Sweeney K, Oberg D et al.: Oncolytic adenoviral mutants with E1B19K gene deletions enhance gemcitabine-induced apoptosis in pancreatic carcinoma cells and anti-tumor efficacy in vivo. Clin. Cancer Res.15,1730–1740 (2009).
    • 30  Nelson AR, Davydova J, Curiel DT, Yamamoto M: Combination of conditionally replicative adenovirus and standard chemotherapies shows synergistic antitumor effect in pancreatic cancer. Cancer Sci.100,2181–2187 (2009).
    • 31  Sei S, Mussio JK, Yang QE et al.: Synergistic antitumor activity of oncolytic reovirus and chemotherapeutic agents in non-small cell lung cancer cells. Mol. Cancer8,47 (2009).
    • 32  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.6,3342–3353 (2000).
    • 33  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. Neoplasia1,162–169 (1999).
    • 34  Gutermann A, Mayer E, von Dehn-Rothfelser K et al.: Efficacy of oncolytic herpesvirus NV1020 can be enhanced by combination with chemotherapeutics in colon carcinoma cells. Hum. Gene Ther.17,1241–1253 (2006).
    • 35  Aghi M, Rabkin S, Martuza RL: Effect of chemotherapy-induced DNA repair on oncolytic herpes simplex viral replication. J. Natl Cancer Inst.98,38–50 (2006).▪ Salient study demonstrating the interaction of DNA repair pathways with different oHSV mutants in generating synergy with temozolomide.
    • 36  Fukuhara H, Martuza RL, Rabkin SD, Ito Y, Todo T: Oncolytic herpes simplex virus vector G47Δ in combination with androgen ablation for the treatment of human prostate adenocarcinoma. Clin. Cancer Res.11,7886–7890 (2005).
    • 37  Chou TC, Talalay P: Quantitative analysis of dose–effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv. Enzyme Regul.22,27–55 (1984).
    • 38  Chou TC: Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol. Rev.58,621–681 (2006).
    • 39  Chou TC: Drug combination studies and their synergy quantification using the Chou–Talalay method. Cancer Res.70,440–446 (2010).
    • 40  Reynolds CP, Maurer BJ: Evaluating response to antineoplastic drug combinations in tissue culture models. Methods Mol. Med.110,173–183 (2005).
    • 41  Watanabe I, Kasuya H, Nomura N et al.: Effects of tumor selective replication-competent herpes viruses in combination with gemcitabine on pancreatic cancer. Cancer Chemother. Pharmacol.61,875–882 (2008).
    • 42  Nguyen ML, Blaho JA: Apoptosis during herpes simplex virus infection. Adv. Virus Res.69,67–97 (2007).
    • 43  Hibma MH, Real NC, Wiles A et al.: Increased apoptosis and reduced replication efficiency of the E3 region-modified dl309 adenovirus in cancer cells. Virus Res.145,112–120 (2009).
    • 44  Kasuya H, Nishiyama Y, Nomoto S et al.: Suitability of a US3-inactivated HSV mutant (L1BR1) as an oncolytic virus for pancreatic cancer therapy. Cancer Gene Ther.14,533–542 (2007).
    • 45  Aghi M, Rabkin SD, Martuza RL: Angiogenic response caused by oncolytic herpes simplex virus-induced reduced thrombospondin expression can be prevented by specific viral mutations or by administering a thrombospondin-derived peptide. Cancer Res.67,440–444 (2007).
    • 46  Kurozumi K, Hardcastle J, Thakur R et al.: Effect of tumor microenvironment modulation on the efficacy of oncolytic virus therapy. J. Natl Cancer Inst.99,1768–1781 (2007).
    • 47  Kaur B, Cripe TP, Chiocca EA: “Buy one get one free”: armed viruses for the treatment of cancer cells and their microenvironment. Curr. Gene Ther.9,341–355 (2009).
    • 48  Toyoizumi T, Mick R, Abbas AE, Kang EH, Kaiser LR, Molnar-Kimber KL: Combined therapy with chemotherapeutic agents and herpes simplex virus type 1 ICP34.5 mutant (HSV-1716) in human non-small cell lung cancer. Hum. Gene Ther.10,3013–3029 (1999).
    • 49  Mohr I: To replicate or not to replicate: achieving selective oncolytic virus replication in cancer cells through translational control. Oncogene24,7697–7709 (2005).
    • 50  Petrowsky H, Roberts GD, Kooby DA et al.: Functional interaction between fluorodeoxyuridine-induced cellular alterations and replication of a ribonucleotide reductase-negative herpes simplex virus. J. Virol.75,7050–7058 (2001).▪▪ First demonstration that chemotherapy-induced upregulation of cellular homologs of viral genes mutated in oHSV enhances their replication and cytotoxicity.
    • 51  Nakano K, Todo T, Zhao γ et al.: Enhanced efficacy of conditionally replicating herpes simplex virus (G207) combined with 5-fluorouracil and surgical resection in peritoneal cancer dissemination models. J. Gene Med.7,638–648 (2005).
    • 52  Eisenberg DP, Adusumilli PS, Hendershott KJ et al.: 5-fluorouracil and gemcitabine potentiate the efficacy of oncolytic herpes viral gene therapy in the treatment of pancreatic cancer. J. Gastrointest. Surg.9,1068–1077 (2005).
    • 53  Bennett JJ, Adusumilli P, Petrowsky H et al.: Up-regulation of GADD34 mediates the synergistic anticancer activity of mitomycin C and a γ134.5 deleted oncolytic herpes virus (G207). FASEB J.18,1001–1003 (2004).
    • 54  Mullerad M, Bochner BH, Adusumilli PS et al.: Herpes simplex virus based gene therapy enhances the efficacy of mitomycin C for the treatment of human bladder transitional cell carcinoma. J. Urol.174,741–746 (2005).
    • 55  Hadjipanayis CG, Fellows-Mayle W, Deluca NA: Therapeutic efficacy of a herpes simplex virus with radiation or temozolomide for intracranial glioblastoma after convection-enhanced delivery. Mol. Ther.16,1783–1788 (2008).
    • 56  Stahel RA, Felley-Bosco E, Opitz I, Weder W: Malignant pleural mesothelioma. Future Oncol.5(3),391–402 (2009).
    • 57  Adusumilli PS, Chan MK, Chun YS et al.: Cisplatin-induced GADD34 upregulation potentiates oncolytic viral therapy in the treatment of malignant pleural mesothelioma. Cancer Biol. Ther.5,48–53 (2006).
    • 58  Lin SF, Gao SP, Price DL et al.: Synergy of a herpes oncolytic virus and paclitaxel for anaplastic thyroid cancer. Clin. Cancer Res.14,1519–1528 (2008).▪ Demonstrated that synergy is due to oHSV enhancing paclitaxel activity and cellular sensitivity.
    • 59  Passer BJ, Castelo-Branco P, Buhrman JS, Varghese S, Rabkin SD, Martuza RL: Oncolytic herpes simplex virus vectors and taxanes synergize to promote killing of prostate cancer cells. Cancer Gene Ther.16,551–560 (2009).
    • 60  Shimoyama S, Goshima F, Teshigahara O et al.: Enhanced efficacy of herpes simplex virus mutant HF10 combined with paclitaxel in peritoneal cancer dissemination models. Hepatogastroenterology54,1038–1042 (2007).
    • 61  Cinatl J Jr, Cinatl J, Michaelis M et al.: Potent oncolytic activity of multimutated herpes simplex virus G207 in combination with vincristine against human rhabdomyosarcoma. Cancer Res.63,1508–1514 (2003).
    • 62  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.10,385–393 (1999).
    • 63  Lee KC, Hamstra DA, Bhojani MS, Khan AP, Ross BD, Rehemtulla A: Noninvasive molecular imaging sheds light on the synergy between 5-fluorouracil and TRAIL/Apo2L for cancer therapy. Clin. Cancer Res.13,1839–1846 (2007).
    • 64  Nakamura M, Nagano H, Sakon M et al.: Role of the Fas/FasL pathway in combination therapy with interferon-a and fluorouracil against hepatocellular carcinoma in vitro. J. Hepatol.46,77–88 (2007).
    • 65  Qiu S, Ruan H, Pei Z et al.: Combination of targeting gene-virotherapy with 5-FU enhances antitumor efficacy in malignant colorectal carcinoma. J. Interferon Cytokine Res.24,219–230 (2004).
    • 66  Mace AT, Harrow SJ, Ganly I, Brown SM: Cytotoxic effects of the oncolytic herpes simplex virus HSV1716 alone and in combination with cisplatin in head and neck squamous cell carcinoma. Acta Otolaryngol.127,880–887 (2007).
    • 67  Carew JS, Giles FJ, Nawrocki ST: Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett.269,7–17 (2008).
    • 68  Marks PA, Xu WS: Histone deacetylase inhibitors: potential in cancer therapy. J. Cell. Biochem.107,600–608 (2009).
    • 69  Otsuki A, Patel A, Kasai K et al.: Histone deacetylase inhibitors augment antitumor efficacy of herpes-based oncolytic viruses. Mol. Ther.16,1546–1555 (2008).▪ Highlights how inhibiting interferon responses with histone deacetylase inhibitors can enhance oHSV efficacy.
    • 70  Nguyen TL, Abdelbary H, Arguello M et al.: Chemical targeting of the innate antiviral response by histone deacetylase inhibitors renders refractory cancers sensitive to viral oncolysis. Proc. Natl Acad. Sci. USA105,14981–14986 (2008).
    • 71  Hoti N, Chowdhury W, Hsieh JT, Sachs MD, Lupold SE, Rodriguez R: Valproic acid, a histone deacetylase inhibitor, is an antagonist for oncolytic adenoviral gene therapy. Mol. Ther.14,768–778 (2006).
    • 72  Liu TC, Castelo-Branco P, Rabkin SD, Martuza RL: Trichostatin A and oncolytic HSV combination therapy shows enhanced antitumoral and anti-angiogenic effects. Mol. Ther.16,1041–1047 (2008).
    • 73  Katsura T, Iwai S, Ota Y, Shimizu H, Ikuta K, Yura Y: The effects of trichostatin A on the oncolytic ability of herpes simplex virus for oral squamous cell carcinoma cells. Cancer Gene Ther.16,237–245 (2009).
    • 74  Liu P, Cheng H, Roberts TM, Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov.8,627–644 (2009).
    • 75  Benetti L, Roizman B: Protein kinase B/Akt is present in activated form throughout the entire replicative cycle of ΔU(S)3 mutant virus but only at early times after infection with wild-type herpes simplex virus 1. J. Virol.80,3341–3348 (2006).
    • 76  Liu TC, Wakimoto H, Martuza RL, Rabkin SD: Herpes simplex virus Us3(-) mutant as oncolytic strategy and synergizes with phosphatidylinositol 3-kinase-Akt targeting molecular therapeutics. Clin. Cancer Res.13,5897–5902 (2007).▪ oHSV could sensitize cancer cells to molecular targeted drugs and thereby enhance efficacy.
    • 77  Sarinella F, Calistri A, Sette P, Palu G, Parolin C: Oncolysis of pancreatic tumour cells by a γ34.5-deleted HSV-1 does not rely upon Ras-activation, but on the PI 3-kinase pathway. Gene Ther.13,1080–1087 (2006).
    • 78  Mahller YY, Vaikunth SS, Currier MA et al.: Oncolytic HSV and erlotinib inhibit tumor growth and angiogenesis in a novel malignant peripheral nerve sheath tumor xenograft model. Mol. Ther.15,279–286 (2007).
    • 79  Palumbo A, Facon T, Sonneveld P et al.: Thalidomide for treatment of multiple myeloma: 10 years later. Blood111,3968–3977 (2008).
    • 80  Israyelyan A, Shannon EJ, Baghian A, Kearney MT, Kousoulas KG: Thalidomide suppressed the growth of 4T1 cells into solid tumors in Balb/c mice in a combination therapy with the oncolytic fusogenic HSV-1 OncdSyn. Cancer Chemother. Pharmacol.64,1201–1210 (2009).
    • 81  Portsmouth D, Hlavaty J, Renner M: Suicide genes for cancer therapy. Mol. Aspects Med.28,4–41 (2007).
    • 82  Chase M, Chung RY, Chiocca EA: An oncolytic viral mutant that delivers the CYP2B1 transgene and augments cyclophosphamide chemotherapy. Nat. Biotechnol.16,444–448 (1998).
    • 83  Braidwood L, Dunn PD, Hardy S, Evans TR, Brown SM: Antitumor activity of a selectively replication competent herpes simplex virus (HSV) with enzyme prodrug therapy. Anticancer Res.29,2159–2166 (2009).
    • 84  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.7,939–946 (2000).
    • 85  Luo C, Mori I, Goshima F et al.: Replication-competent, oncolytic herpes simplex virus type 1 mutants induce a bystander effect following ganciclovir treatment. J. Gene Med.9,875–883 (2007).
    • 86  Guffey MB, Parker JN, Luckett WS Jr et al.: Engineered herpes simplex virus expressing bacterial cytosine deaminase for experimental therapy of brain tumors. Cancer Gene Ther.14,45–56 (2007).
    • 87  Tyminski E, Leroy S, Terada K et al.: Brain tumor oncolysis with replication-conditional herpes simplex virus type 1 expressing the prodrug-activating genes, CYP2B1 and secreted human intestinal carboxylesterase, in combination with cyclophosphamide and irinotecan. Cancer Res.65,6850–6857 (2005).▪ Example of the power of oHSV to deliver multiple prodrug-activating genes.
    • 88  Ishida D, Nawa A, Tanino T et al.: Enhanced cytotoxicity with a novel system combining the paclitaxel-2´-ethylcarbonate prodrug and an HSV amplicon with an attenuated replication-competent virus, HF10 as a helper virus. Cancer Lett.288(1),17–27 (2009).
    • 89  Fulci G, Breymann L, Gianni D et al.: Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proc. Natl Acad. Sci. USA103,12873–12878 (2006).
    • 90  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.67,7850–7855 (2007).
    • 91  Pawlik TM, Nakamura H, Mullen JT et al.: Prodrug bioactivation and oncolysis of diffuse liver metastases by a herpes simplex virus 1 mutant that expresses the CYP2B1 transgene. Cancer95,1171–1181 (2002).
    • 92  Currier MA, Gillespie RA, Sawtell NM et al.: Efficacy and safety of the oncolytic herpes simplex virus rRp450 alone and combined with cyclophosphamide. Mol. Ther.16,879–885 (2008).
    • 93  Orvedahl A, Alexander D, Talloczy Z et al.: HSV-1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe1,23–35 (2007).
    • 94  Fong Y, Kim T, Bhargava A et al.: A herpes oncolytic virus can be delivered via the vasculature to produce biologic changes in human colorectal cancer. Mol. Ther.17,389–394 (2009).
    • 95  Huang C, Li M, Chen C, Yao Q: Small interfering RNA therapy in cancer: mechanism, potential targets, and clinical applications. Expert Opin. Ther. Targets12,637–645 (2008).
    • 96  Croce CM: Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet.10,704–714 (2009).
    • 97  Diehn M, Cho RW, Clarke MF: Therapeutic implications of the cancer stem cell hypothesis. Semin. Radiat. Oncol.19,78–86 (2009).
    • 98  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.69,3472–3481 (2009).
    • 99  Mahller YY, Williams JP, Baird WH et al.: Neuroblastoma cell lines contain pluripotent tumor initiating cells that are susceptible to a targeted oncolytic virus. PLoS One4,E4235 (2009).
    • 100  Friedman GK, Langford CP, Coleman JM et al.: Engineered herpes simplex viruses efficiently infect and kill CD133+ human glioma xenograft cells that express CD111. J. Neurooncol.95,199–209 (2009).
    • 101  Tesniere A, Panaretakis T, Kepp O et al.: Molecular characteristics of immunogenic cancer cell death. Cell Death Differ.15,3–12 (2008).
    • 102  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.72,631–639 (1991).
    • 103  Liu BL, Robinson M, Han ZQ et al.: ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther.10,292–303 (2003).
    • 104  Wong RJ, Kim SH, Joe JK, Shah JP, Johnson PA, Fong Y: Effective treatment of head and neck squamous cell carcinoma by an oncolytic herpes simplex virus. J. Am. Coll. Surg.193,12–21 (2001).
    • 105  Takakuwa H, Goshima F, Nozawa N et al.: Oncolytic viral therapy using a spontaneously generated herpes simplex virus type 1 variant for disseminated peritoneal tumor in immunocompetent mice. Arch. Virol.148,813–825 (2003).
    • 106  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. USA98,6396–6401 (2001).
    • 107  Chou J, Kern ER, Whitley RJ, Roizman B: Mapping of herpes simplex virus-1 neurovirulence to γ34.5, a gene nonessential for growth in culture. Science250,1262–1266 (1990).
    • 108  Goldstein DJ, Weller SK: Herpes simplex virus type 1-induced ribonucleotide reductase activity is dispensible for virus growth and DNA synthesis: isolation and characterization of an ICP6 LacZ insertion mutant. J. Virol.62,196–205 (1988).
    • 109  Kambara H, Okano H, Chiocca EA, Saeki Y: An oncolytic HSV-1 mutant expressing ICP34.5 under control of a nestin promoter increases survival of animals even when symptomatic from a brain tumor. Cancer Res.65,2832–2839 (2005).
    • 110  Wong RJ, Joe JK, Kim SH, Shah JP, Horsburgh B, Fong Y: Oncolytic herpesvirus effectively treats murine squamous cell carcinoma and spreads by natural lymphatics to treat sites of lymphatic metastases. Hum. Gene Ther.13,1213–1223 (2002).
    • 111  Purves FC, Longnecker RM, Leader DP, Roizman B: Herpes simplex virus 1 protein kinase is encoded by open reading frame US3 which is not essential for virus growth in cell culture. J. Virol.61,2896–2901 (1987).
    • 112  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.57,1502–1509 (1997).
    • 113  Israyelyan A, Chouljenko VN, Baghian A, David AT, Kearney MT, Kousoulas KG: Herpes simplex virus type-1(HSV-1) oncolytic and highly fusogenic mutants carrying the NV1020 genomic deletion effectively inhibit primary and metastatic tumors in mice. Virol. J.5,68 (2008).
    • 114  Nagano S, Perentes JY, Jain RK, Boucher Y: Cancer cell death enhances the penetration and efficacy of oncolytic herpes simplex virus in tumors. Cancer Res.68,3795–3802 (2008).
    • 201  National Cancer Institute: clinical trials www.cancer.gov/clinicaltrials