Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-19T03:28:00.165Z Has data issue: false hasContentIssue false

76 - Herpesviruses as therapeutic agents

from Part VIII - Herpes as therapeutic agents

Published online by Cambridge University Press:  24 December 2009

Frank Tufaro
Affiliation:
Allera Health Products, Inc., 360 Central Avenue, Suite 1560, St. Petersburg, FL 33701, USA
James M. Markert
Affiliation:
University of British Columbia, British Columbia, Canada; Department of Surgery, Brain Tumor Research Laboratories, The University of Alabama at Birmingham, Birmingham, AL, USA
Ann Arvin
Affiliation:
Stanford University, California
Gabriella Campadelli-Fiume
Affiliation:
Università degli Studi, Bologna, Italy
Edward Mocarski
Affiliation:
Emory University, Atlanta
Patrick S. Moore
Affiliation:
University of Pittsburgh
Bernard Roizman
Affiliation:
University of Chicago
Richard Whitley
Affiliation:
University of Alabama, Birmingham
Koichi Yamanishi
Affiliation:
University of Osaka, Japan
Get access

Summary

Introduction

After more than a decade of intensive research and development efforts, the translation of promising viral-based gene therapies from the research lab to the clinic is both promising and unexpectedly challenging. Many of the same properties that make viral vectors attractive candidates to deliver genes for therapeutic purposes also impede the path to successful clinical development.

Vectors for clinical use must be manufactured in relatively high yields such that hundreds of thousands or even millions of “doses” can be generated in a safe and cost-effective manner. Moreover, the resulting vector must exhibit genetic as well as structural stability, withstand storage at various temperatures for up to several years, and cause little or no toxicity in animals, and ultimately in humans.

Herpes simplex viruses (HSV), while widespread in nature, have not been tested in human clinical studies as often as several other commonly used vectors, such as adenovirus, adeno-associated viruses (AAV), and retroviruses. In many ways however, HSV is emerging as a viable therapeutic platform and several clinical studies are either ongoing or planned for the very near future. The reason for this increased focus on HSV is due in part attributable to the unique properties that make HSV a stable and potentially potent vector for controlled gene delivery. In addition, the increasing experience with replication-competent vectors in human clinical studies has made it more familiar with clinicians.

Properties of therapeutic HSV vectors

Therapeutic HSV can be characterized as replication-competent or replication-defective.

Type
Chapter
Information
Human Herpesviruses
Biology, Therapy, and Immunoprophylaxis
, pp. 1341 - 1352
Publisher: Cambridge University Press
Print publication year: 2007

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Advani, S. J., Sibley, G., Song, P. Y.et al. (1998). Enhancement of replication of genetically engineered herpes simplex virus by ionizing radiation: a new paradign for destruction of therapeutically intractable tumors. Gene Ther., 5, 160–165.CrossRefGoogle Scholar
Blank, S. V., Rubin, S. C., Coukos, G., Amin, K. M., Albelda, S. M., and Molnar-Kimber, K. L. (1999). Replication-selective herpes simplex virus type 1 mutant therapy of cervical cancer is enhanced by low-dose radiation. Hum. Gene Ther., 13, 627–639.CrossRefGoogle Scholar
Bradley, J. D., Kataoka, Y., Advani, S.et al. (1999). Ionizing radiation improves survival in mice bearing intracranial high-grade gliomas injected with genetically modified herpes simplex virus. Clin. Cancer Res., 5, 1517–1522.Google ScholarPubMed
Chahlavi, A., Todo, T., Martuza, R. L., and Rabkin, S. D. (1999). Replication-competent herpes simpelx virus vector G207 and cisplatin combination therapy for head and neck squamous cell carcinoma. Neoplasia, 1, 162–169.CrossRefGoogle ScholarPubMed
Chattopadhyay, M., Wolfe, D., Huang, S.et al. (2002). In vivo gene therapy for pyridoxine-induced neuropathy by herpes simplex virus-mediated gene transfer of neurotrophin-3. Ann. Neurol., 51, 19–27.CrossRefGoogle ScholarPubMed
Chattopadhyay, M., Goss, J., Lacomis, D.et al. (2003). Protective effect of HSV-mediated gene transfer of nerve growth factor in pyridoxine neuropathy demonstrates functional activity of trkA receptors in large sensory neurons of adult animals. Eur. J. Neurosci., 17, 732–740.CrossRefGoogle ScholarPubMed
Chou, J. and Roizman, B. (1992). The gamma 1(34.5) gene of herpes simplex virus 1 precludes neuroblastoma cells from triggering total shutoff of protein synthesis characteristic of programed cell death in neuronal cells. Proc. Natl Acad. Sci. USA, 89, 3266–3270.CrossRefGoogle ScholarPubMed
Chou, J., Kern, E. R., Whitley, R. J., and Roizman, B. (1990). Mapping of herpes simplex virus-1 neurovirulence to gamma 134.5, a gene nonessential for growth in culture. Science, 250, 1262–1266.CrossRefGoogle ScholarPubMed
Coen, D. M., Goldstein, D. J., and Weller, S. K. (1989) Herpes simplex virus ribonucleotide reductase mutants are hypersensitive to acyclovir. Antimicrob. Agents Chemother., 33, 1395–1399 (Erratum appears in Antimicrob. Agents Chemother., 33, 1827).CrossRefGoogle ScholarPubMed
DeLuca, N. A., McCarthy, A. M., and Schaffer, P. A. (1985). Isolation and characterization of deletion mutants of herpes simplex virus type 1 in the gene encoding immediate-early regulatory protein ICP4. J. Virol., 56, 558–570.Google ScholarPubMed
Farassati, F., Yang, A. D., and Lee, P. W. (2001). Oncogenes in Ras signalling pathway dictate host-cell permissiveness to herpes simplex virus 1. Nat. Cell. Biol., 3, 745–750.CrossRefGoogle ScholarPubMed
Fong, Y., Kermeny, N., Jarnagin, W. et al. (2002). Phase I study of a replication-competent herpes simplex oncolytic virus for treatment of hepatic colorectal metastases. ASCO Annual Meeting. Orlando, FL, May 18–21.
Fraefel, C., Song, S., Lim, F.et al. (1996). Helper virus-free transfer of herpes simplex virus type 1 plasmid vectors into neural cells. J. Virol., 70, 7190–7197.Google ScholarPubMed
Goins, W. F., Lee, K. A., Cavalcoli, J. D.et al. (1999). Herpes simplex virus type 1 vector-mediated expression of nerve growth factor protects dorsal root ganglion neurons from peroxide toxicity. J. Virol., 73, 519–532.Google ScholarPubMed
Goldstein, D. J. and , Weller S. K. (1988). Factor(s) present in herpes simplex virus type 1-infected cells can compensate for the loss of the large subunit of the viral ribonucleotide reductase: characterization of an ICP6 deletion mutant. Virology, 166, 41–51.CrossRefGoogle ScholarPubMed
Goss, J. R., Mata, M., Goins, W. F., Wu, H. H., Glorioso, J. C., and Fink, D. J. (2001). Antinociceptive effect of a genomic herpes simplex virus-based vector expressing human proenkephalin in rat dorsal root ganglion. Gene Ther., 8, 551–556.CrossRefGoogle ScholarPubMed
Goss, J. R., Harley, C. F., Mata, M.et al. (2002a). Herpes vector-mediated expression of proenkephalin reduces bone cancer pain. Ann. Neurol., 52, 662–665.CrossRefGoogle Scholar
Goss, J. R., Goins, W. F., Lacomis, D., Mata, M., Glorioso, J. C., and Fink, D. J. (2002b). Herpes simplex-mediated gene transfer of nerve growth factor protects against peripheral neuropathy in streptozotocin-induced diabetes in the mouse. Diabetes, 51, 2227–2232.CrossRefGoogle Scholar
Grandi, P., Spear, M., Breakefield, X. O., and Wang, S. (2004). Targeting HSV amplicon vectors. Methods, 33, 179–186.CrossRefGoogle ScholarPubMed
Hao, S., Mata, M., Goins, W., Glorioso, J. C., and Fink, D. J. (2003). Transgene-mediated enkephalin release enhances the effect of morphine and evades tolerance to produce a sustained antiallodynic effect in neuropathic pain. Pain, 102, 135–142.CrossRefGoogle ScholarPubMed
Harland, J., Papanastassiou, V., and Brown, S. M. (2002). HSV1716 persistence in primary human glioma cells in vitro. Gene Ther., 9, 1194–1198.CrossRefGoogle ScholarPubMed
Harrow, S., Papanastassiou, V., Harland, J.et al. (2004). HSV 1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther., 11, 1648–1658.CrossRefGoogle Scholar
He, B., Gross, M., and Roizman, B. (1997). The gamma(1)34.5 protein of herpes simplex virus 1 complexes with protein phosphatase 1alpha to dephosphorylate the alpha subunit of the eukaryotic translation initiation factor 2 and preclude the shutoff of protein synthesis by double-stranded RNA-activated protein kinase. Proc. Natl Acad. Sci. USA, 94, 843–848.CrossRefGoogle ScholarPubMed
Ho, I. A., Hui, K. M., and Lam, P. Y. (2004). Glioma-specific and cell cycle-regulated herpes simplex virus type 1 amplicon viral vector. Hum. Gene Ther., 15, 495–508.CrossRefGoogle ScholarPubMed
Hunter, L. D., Martuza, R. L., Feigenbaum, F.et al. (1999). Attenuated, replication-competent herpes simplex virus type 1 mutant G207: safety evaluation of intracerebral injection in nonhuman primates. J. Virol., 73, 6319–6326.Google ScholarPubMed
Jacobs, A. H., Winkeler, A., Hartung, M.et al. (2003). Improved herpes simplex virus type 1 amplicon vectors for proportional coexpression of positron emission tomography marker and therapeutic genes. Hum. Gene Ther., 14, 277–297.CrossRefGoogle ScholarPubMed
Krisky, D. M., Wolfe, D., Goins, W. F., et al. (1998). Deletion of multiple immediate-early genes from herpes simplex virus reduces cytotoxicity and permits long-term gene expression in neurons. Gene Ther., 5, 1593–1603.CrossRefGoogle ScholarPubMed
Lilley, C. E., Groutsi, F., Han, Z.et al. (2001). Multiple immediate-early gene-deficient herpes simplex virus vectors allowing efficient gene delivery to neurons in culture and widespread gene delivery to the central nervous system in vivo. J. Virol., 75, 4343–4356.CrossRefGoogle ScholarPubMed
McCarthy, A. M., McMahan, L., and Schaffer, P. A. (1989). Herpes simplex virus type 1 ICP27 deletion mutants exhibit altered patterns of transcription and are DNA deficient. J. Virol., 63, 18–27.Google ScholarPubMed
MacKie, R. M., Stewart, B., and Brown, S. M. (2001). Intralesional injection of herpes simplex virus 1716 in metastatic melanoma. Lancet, 357, 525–526.CrossRefGoogle ScholarPubMed
Marconi, P., Krisky, D., Oligino, T.et al. (1996). Replication-defective herpes simplex virus vectors for gene transfer in vivo. Proc. Natl Acad. Sci. USA, 93, 11319–11320.CrossRefGoogle ScholarPubMed
Markert, J. M., Coen, D. M., Malick, A., Mineta, T., and Martuza, R. L. (1992). Expanded spectrum of viral therapy in the treatment of nervous system tumors. J. Neurosurg. 77, 590–594.CrossRefGoogle ScholarPubMed
Markert, J. M., Malick, A., Coen, D. M., and Martuza, R. L. (1993). Reduction and elimination of encephalitis in an experimental glioma therapy model with attenuated herpes simplex mutants that retain susceptibility to acyclovir. Neurosurgery, 32, 597–603.CrossRefGoogle Scholar
Markert, J. M., Gillespie, G. Y., Weichselbaum, R. R., Roizman, B., and Whitley, R. J. (2000a). Genetically engineered HSV in the treatment of glioma: a review. Rev. Med. Virol., 10, 17–30.3.0.CO;2-G>CrossRefGoogle Scholar
Markert, J. M., Medlock, M. D., Rabkin, S. D.et al. (2000b). Conditionally replicating herpes simplex, virus., mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther., 7, 867–874.CrossRefGoogle Scholar
Marshall, K. R., Lachmann, R. H., Efstathiou, S., Rinaldi, A., and Preston, C. M. (2000). Long-term transgene expression in mice infected with a herpes simplex virus type 1 mutant severely impaired for immediate-early gene expression. J. Virol., 74, 956–964.CrossRefGoogle ScholarPubMed
Martuza, R. L., Malick, A., Markert, J. M., Ruffner, K. L., and Coen, D. M. (1991). Experimental therapy of human glioma by means of a genetically engineered virus mutant. Science, 252, 854–856.CrossRefGoogle ScholarPubMed
Meignier, B. (1991). Genetically engineered attenuated herpes simplex viruses. Rev. Infect. Dis., 13 Suppl 11, S895–S897.CrossRefGoogle ScholarPubMed
Meignier, B., Longnecker, R., and Roizman, B. (1988). In vivo behavior of genetically engineered herpes simplex viruses R7017 and R7020: construction and evaluation in rodents. J. Infect. Dis., 158, 602–614.CrossRefGoogle ScholarPubMed
Meignier, B., Martin, B., Whitley, R. J., and Roizman, B. (1990). In vivo behavior of genetically engineered herpes simplex viruses R7017 and R7020. II. Studies in immunocompetent and immunosuppressed owl monkeys (Aotus trivirgatus). J. Infect. Dis., 162, 313–321.CrossRefGoogle Scholar
Miller, C. G. and Fraser, N. W. (2000). Role of the immune response during neuro-attenuated herpes simplex virus-mediated tumor destruction in a murine intracranial melanoma model. Cancer Res., 60, 5714–5722.Google Scholar
Miller, C. G., Krummenacher, C., Eisenberg, R. J., Cohen, G. H., and Fraser, N. W. (2001). Development of a syngenic murine B16 cell line-derived melanoma susceptible to destruction by neuroattenuated HSV-1. Mol. Ther., 3, 160–168.CrossRefGoogle ScholarPubMed
Mineta, T., Rabkin, S. D., and Martuza, R. L. (1994). Treatment of malignant gliomas using ganciclovir-hypersensitive, ribonucleotide reductase-deficient herpes simplex viral mutant. Cancer Res., 54, 3963–3966.Google ScholarPubMed
Mineta, T., Rabkin, S. D., Yazaki, T., Hunter, W. D., and Martuza, R. L. (1995). Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat. Med., 1, 938–943.CrossRefGoogle ScholarPubMed
Moriuchi, S., Oligino, T., Krisky, D.et al. (1998). 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 Resi., 58, 5731–5737.Google ScholarPubMed
Palmer, J. A., Branston, R. H., Lilley, C. E., et al. (2000). Development and optimization of herpes simplex virus vectors for multiple long-term gene delivery to the peripheral nervous system. J. Virol., 74, 5604–5618.CrossRefGoogle ScholarPubMed
Papanastassiou, V., Rampling, R., Fraser, M.et al. (2002). 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., 9, 398–406.CrossRefGoogle ScholarPubMed
Rampling, R., Cruickshank, G., Papanastassiou, V.et al. (2000). Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. [see comment]. Gene Ther., 7, 859–866.CrossRefGoogle Scholar
Randazzo, B. P., Bhat, M. G., Kesari, S., Fraser, N. W., and Brown, S. M. (1997). Treatment of experimental subcutaneous human melanoma with a replication-restricted herpes simplex virus mutant. J. Invest. Dermatol., 108, 933–937.CrossRefGoogle ScholarPubMed
Samaniego, L. A., Webb, A. L., and DeLuca, N. A. (1995). Functional interactions between herpes simplex virus immediate-early proteins during infection: gene expression as a consequence of ICP27 and different domains of ICP4. J. Virol., 69, 5705–5715.Google ScholarPubMed
Samaniego, L. A., Wu, N., and DeLuca, N. A. (1997). The herpes simplex virus immediate-early protein ICP0 affects transcription from the viral genome and infected-cell survival in the absence of ICP4 and ICP27. J. Virol., 71, 4614–4625.Google ScholarPubMed
Samaniego, L. A., Neiderhiser, L., and DeLuca, N. A. (1998). Persistence and expression of the herpes simplex virus genome in the absence of immediate-early proteins. J. Virol., 72, 3307–3320.Google ScholarPubMed
Sanson, M., Marcaud, V., Robin, E., Valery, C., Sturtz, F., and Zalc, B. (2002). Connexin 43-mediated bystander effect in two rat glioma cell models. Cancer Gene Ther., 9, 149–155.CrossRefGoogle ScholarPubMed
Schmeisser, F., Donohue, M., and Weir, J. P. (2002). Tetracycline-regulated gene expression in replication-incompetent herpes simplex virus vectors. Hum. Gene Ther., 13, 2113–2124.CrossRefGoogle ScholarPubMed
Shah, A. C., Benos, D., Gillespie, G. Y., and Markert, J. M.Oncolytic viruses: clinical applications as vectors for the treatment of malignant gliomas. J. Neurooncol., 65, 203–226.CrossRef
Spaete, R. R. and Frenkel, N. (1982). The herpes simplex virus amplicon: a new eucaryotic defective-virus cloning-amplifying vector. Cell, 30, 295–304.CrossRefGoogle ScholarPubMed
Toda, M., Rabkin, S. D., Kojima, H., and Martuza, R. L. (1999). Herpes simplex virus as an in situ cancer vaccine for the induction of specific anti-tumor immunity. Hum. Gene Ther., 10, 385–393.CrossRefGoogle Scholar
Valyi-Nagy, T., Fareed, M. U., O'Keefe, J. S.et al. (1994). The herpes simplex virus type 1 strain 17+ gamma 34.5 deletion mutant 1716 is avirulent in SCID mice. J. Gen. Virol., 75, 2059–2063.CrossRefGoogle ScholarPubMed
Wade-Martins, R., Smith, E. R., Tyminski, E., Chiocca, E. A., and Saeki, Y. (2001). An infectious transfer and expression system for genomic DNA loci in human and mouse cells. Nat. Biotechnol., 19, 1067–1070.CrossRefGoogle ScholarPubMed
Wang, Y., Camp, S. M., Niwano, M.et al. (2002). Herpes simplex virus type 1/adeno-associated virus rep(+) hybrid amplicon vector improves the stability of transgene expression in human cells by site-specific integration. J. Virol., 76, 7150–7162.CrossRefGoogle ScholarPubMed
Wu, N., Watkins, S. C., Schaffer, P. A., and DeLuca, N. A. (1996). Prolonged gene expression and cell survival after infection by a herpes simplex virus mutant defective in the immediate-early genes encoding ICP4, ICP27, and ICP22. J. Virol., 70, 6358–6369.Google ScholarPubMed
Yamada, M., Oligino, T., Mata, M., Goss, J., Glorioso, J., and Fink, D. J. (1999). Herpes simplex virus vector-mediated expression of Bcl-2 prevents 6-hydroxydopamine-induced degeneration of neurons in the substantia nigra in vivo. Proc. Natl Acad. Sci. USA, 96, 4078–4083.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×