101
|
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; 12:1101-10. [PMID: 16140040 DOI: 10.1016/j.ymthe.2005.07.533] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 07/12/2005] [Accepted: 07/13/2005] [Indexed: 11/28/2022] Open
Abstract
Oncolytic viruses that selectively replicate in cancer cells have been described for over 50 years. Despite the observation by several groups that multimutated herpes simplex type 1 vectors are oncolytic in a variety of murine tumor models, the oncolytic potential of ICP0 null mutants has not been described. This study characterizes a novel second-generation oncolytic herpes simplex type 1 vector null for the ICP0 gene. We tested three mutant viruses and found that all were selectively cytotoxic in a variety of human and murine tumor cells in vitro. Furthermore, we provide evidence of a mechanistic link between ICP0's function in interferon signaling pathways and the observed oncolytic capacity of ICP0 mutants. Using an immunocompetent murine model of breast adenocarcinoma we demonstrate that the ICP0 mutant KM100 completely eradicates tumors in approximately 80% of treated animals and significantly increases survival. Our data suggest that active viral replication is necessary for effective tumor regression. In addition, we characterized the potential of KM100 as an anti-tumor vaccine since cured mice were found to elicit a robust anti-tumor immune response and were refractory to subsequent tumor growth upon rechallenge.
Collapse
Affiliation(s)
- Jeff L Hummel
- Institute for Molecular Medicine and Health, Centre for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, MDCL 5026, 1200 Main Street West, Hamilton, Ontario, Canada L8N 3Z5
| | | | | |
Collapse
|
102
|
Fukuhara H, Ino Y, Kuroda T, Martuza RL, Todo T. 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; 65:10663-8. [PMID: 16322208 DOI: 10.1158/0008-5472.can-05-2534] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Conditionally replicating herpes simplex virus type 1 (HSV-1) vectors are promising therapeutic agents for cancer. Certain antitumor functions may be added to oncolytic activities of recombinant HSV-1 vectors by inserting transgenes into the viral genome. Because conventional homologous recombination techniques had required time-consuming processes to create "armed" oncolytic HSV-1 vectors, we established an innovative construction system using bacterial artificial chromosome and two recombinase systems (Cre/loxP and FLPe/FRT). Using G47Delta, a safe and efficacious oncolytic HSV-1 with triple gene mutations, as the backbone, this system allowed a rapid generation of multiple vectors with desired transgenes inserted in the deleted ICP6 locus. Four oncolytic HSV-1 vectors, expressing murine interleukin 18 (mIL-18), soluble murine B7-1 [B7-1-immunoglobulin (B7-1-Ig)], both, or none, were created simultaneously within 3 months. In vitro, all newly created recombinant vectors exhibited virus yields and cytopathic effects similar to the parental G47Delta. In two immunocompetent mouse tumor models, TRAMP-C2 prostate cancer and Neuro2a neuroblastoma, the vector expressing both mIL-18 and B7-1-Ig showed a significant enhancement of antitumor efficacy via T-cell-mediated immune responses. The results show that "arming" with multiple transgenes can improve the efficacy of oncolytic HSV-1 vectors. The use of our system may facilitate the development and testing of various armed oncolytic HSV-1 vectors.
Collapse
Affiliation(s)
- Hiroshi Fukuhara
- Molecular Neurosurgery Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | |
Collapse
|
103
|
Abstract
It has been 9 years since the beginning of the first clinical trial in which an oncolytic virus was administered to cancer patients. Since then, oncolytic viruses from five different species have been taken to phase I and II clinical trials in over 300 cancer patients. While additional studies will be required to ascertain if the efficacy of any of these agents is high enough to warrant adding them to the existing therapeutic regimen, it has been reassuring that DNA viruses engineered to achieve tumor selectivity and RNA viruses with relative inherent natural tumor selectivity have proven reasonably safe at the wide range of doses that were tested. Here, we review the biology and clinical results of these five species of viruses and discuss lessons learned and challenges for the future.
Collapse
Affiliation(s)
- Manish Aghi
- Department of Neurosurgery, Massachusetts General Hospital, White Building Room 502, 02114, USA.
| | | |
Collapse
|
104
|
Suter SE, Chein MB, von Messling V, Yip B, Cattaneo R, Vernau W, Madewell BR, London CA. In vitro canine distemper virus infection of canine lymphoid cells: a prelude to oncolytic therapy for lymphoma. Clin Cancer Res 2005; 11:1579-87. [PMID: 15746063 DOI: 10.1158/1078-0432.ccr-04-1944] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Measles virus (MV) causes the regression of human lymphoma xenografts. The purpose of this study was to determine if canine lymphoid cells could be infected in vitro with MV or canine distemper virus (CDV, the canine Morbillivirus equivalent of MV) and determine if in vitro viral infection leads to apoptotic cell death. EXPERIMENTAL DESIGN Reverse transcriptase-PCR was used to examine the expression of both signal lymphocyte activation molecule (CD150) and membrane cofactor molecule (CD46) mRNA. An attenuated CDV expressing enhanced green fluorescent protein was used to infect canine cells in vitro. Both flow cytometry and reverse transcriptase-PCR was used to document CDV infection. Cell death was examined using a propidium iodide staining assay and Annexin V binding. RESULTS Canine lymphoid cell lines and neoplastic B and T lymphocytes collected from dogs with spontaneous lymphoma expressed the Morbillivirus receptor CD150 mRNA. In contrast, only neoplastic lymphocytes expressed detectable levels of CD46 mRNA. Although MV did not infect canine cells, CDV efficiently infected between 40% and 70% of all three canine lymphoid lines tested. More importantly, CDV infected 50% to 90% of neoplastic lymphocytes isolated from dogs with both B and T cell lymphoma. Apoptosis of CDV-infected cell lines was documented. CONCLUSIONS Attenuated CDV may be a useful treatment for canine lymphoma. As such, dogs with lymphoma may represent a biologically relevant large animal model to investigate the feasibility, safety, and efficacy of Morbillivirus therapy in a clinical setting with findings that may have direct applicability in the treatment of human non-Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Steven E Suter
- Department of Surgical and Radiological Science, School of Veterinary Medicine, University of California, 2112 Tupper Hall, One Shields Avenue, Davis, CA 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|
105
|
Currier MA, Adams LC, Mahller YY, Cripe TP. Widespread intratumoral virus distribution with fractionated injection enables local control of large human rhabdomyosarcoma xenografts by oncolytic herpes simplex viruses. Cancer Gene Ther 2005; 12:407-16. [PMID: 15665822 DOI: 10.1038/sj.cgt.7700799] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Novel methods of local control for sarcomas are needed. We investigated the antitumor effect of two related herpes simplex virus (HSV) mutants, NV1020 and NV1066, on human rhabdomyosracoma cells and xenografts. Cell death correlated with virus replication and apoptosis in cultured cells and tumors. Complete regression was seen in all tumors <250 mm(3) following a single injection, yet only half of tumors >250 mm(3) showed a complete response. Fractionation of the virus dose into five injection sites did not increase transduction efficiency, transgene expression, or virus production, but did yield more widespread intratumoral distribution. Despite the same total dose of virus, improved control of large tumors was seen using fractionated injections as all large tumors (500-700 mm(3)) had durable, complete regression. Our data suggest that oncolytic HSVs may be useful for local control of bulky rhabdomyosarcoma tumors and that fractionated virus administration results in a more widespread virus infection and better tumor control. Therefore, strategies to maximize intratumoral virus distribution at initial delivery should be sought.
Collapse
Affiliation(s)
- Mark A Currier
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | |
Collapse
|
106
|
Benencia F, Courrèges MC, Conejo-García JR, Mohamed-Hadley A, Zhang L, Buckanovich RJ, Carroll R, Fraser N, Coukos G. HSV oncolytic therapy upregulates interferon-inducible chemokines and recruits immune effector cells in ovarian cancer. Mol Ther 2005; 12:789-802. [PMID: 15925544 DOI: 10.1016/j.ymthe.2005.03.026] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2004] [Revised: 03/18/2005] [Accepted: 03/19/2005] [Indexed: 11/28/2022] Open
Abstract
Cooperation between oncolytic herpes simplex virus (HSV) and host effector immune mechanisms has been previously described. In the present study, we investigated the mechanism underlying such cooperation in a murine syngeneic model of ovarian carcinoma. Therapeutic administration of HSV-1716, a replication-restricted mutant, resulted in significant reduction of tumor growth and a significant survival advantage. Intratumoral injection of HSV-1716 induced expression of IFN-gamma, MIG, and IP-10 in the tumor. This was accompanied by a significant increase in the number of tumor-associated NK and CD8+ T cells expressing CXCR3 and CD25. Ascites from HSV-1716-treated animals efficiently induced in vitro migration of NK and CD8+ T cells, which was dependent on the presence of MIG and IP-10. Murine monocytes and dendritic cells (DCs) were responsible for the production of MIG and IP-10 upon HSV-1716 infection. In monocytes, this was partially abrogated by neutralizing antibodies against IFN-alpha and -beta, thus indicating a role for type-1 IFNs in the reported effect. Human ovarian carcinomas showed high numbers of monocytes and DCs. Upon HSV-1716 infection, human monocyte-derived DCs produced large amounts of IFN-gamma and upregulated MIG and IP-10 expression. These results indicate that HSV-1716 induces an inflammatory response that may facilitate antitumor immune response upon oncolytic therapy.
Collapse
Affiliation(s)
- Fabian Benencia
- Abramson Family Cancer Research Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Everts B, van der Poel HG. Replication-selective oncolytic viruses in the treatment of cancer. Cancer Gene Ther 2005; 12:141-61. [PMID: 15472714 DOI: 10.1038/sj.cgt.7700771] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the search for novel strategies, oncolytic virotherapy has recently emerged as a viable approach to specifically kill tumor cells. Unlike conventional gene therapy, it uses replication competent viruses that are able to spread through tumor tissue by virtue of viral replication and concomitant cell lysis. Recent advances in molecular biology have allowed the design of several genetically modified viruses, such as adenovirus and herpes simplex virus that specifically replicate in, and kill tumor cells. On the other hand, viruses with intrinsic oncolytic capacity are also being evaluated for therapeutic purposes. In this review, an overview is given of the general mechanisms and genetic modifications by which these viruses achieve tumor cell-specific replication and antitumor efficacy. However, although generally the oncolytic efficacy of these approaches has been demonstrated in preclinical studies the therapeutic efficacy in clinical trails is still not optimal. Therefore, strategies are evaluated that could further enhance the oncolytic potential of conditionally replicating viruses. In this respect, the use of tumor-selective viruses in conjunction with other standard therapies seems most promising. However, still several hurdles regarding clinical limitations and safety issues should be overcome before this mode of therapy can become of clinical relevance.
Collapse
Affiliation(s)
- Bart Everts
- Department of Biomedical sciences, University of Amsterdam, Kruislaan 318, 1098 SM, Amsterdam, The Netherlands
| | | |
Collapse
|
108
|
Cinatl J, Michaelis M, Driever PH, Cinatl J, Hrabeta J, Suhan T, Doerr HW, Vogel JU. Multimutated herpes simplex virus g207 is a potent inhibitor of angiogenesis. Neoplasia 2005; 6:725-35. [PMID: 15720798 PMCID: PMC1531676 DOI: 10.1593/neo.04265] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The mode of the antitumoral activity of multimutated oncolytic herpes simplex virus type 1 G207 has not been fully elucidated yet. Because the antitumoral activity of many drugs involves the inhibition of tumor blood vessel formation, we determined if G207 had an influence on angiogenesis. Monolayers of human umbilical vein endothelial cells and human dermal microvascular endothelial cells, but not human dermal fibroblasts, bronchial epithelial cells, and retinal glial cells, were highly sensitive to the replicative and cytotoxic effects of G207. Moreover, G207 infection caused the destruction of endothelial cell tubes in vitro. In the in vivo Matrigel plug assay in mice, G207 suppressed the formation of perfused vessels. Intratumoral treatment of established human rhabdomyosarcoma xenografts with G207 led to the destruction of tumor vessels and tumor regression. Ultrastructural investigations revealed the presence of viral particles in both tumor and endothelial cells of G207-treated xenografts, but not in adjacent normal tissues. These findings show that G207 may suppress tumor growth, in part, due to inhibition of angiogenesis.
Collapse
Affiliation(s)
- Jindrich Cinatl
- Institute of Medical Virology, Center of Hygiene, Paul-Ehrlich Str. 40, Frankfurt am Main D-60596, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
109
|
Abstract
Molecular research has vastly advanced our understanding of the mechanism of cancer growth and spread. Targeted approaches utilizing molecular science have yielded provocative results in the treatment of cancer. Oncolytic viruses genetically programmed to replicate within cancer cells and directly induce toxic effect via cell lysis or apoptosis are currently being explored in the clinic. Safety has been confirmed and despite variable efficacy results several dramatic responses have been observed with some oncolytic viruses. This review summarizes results of clinical trials with oncolytic viruses in cancer.
Collapse
Affiliation(s)
- Eugene Lin
- Mary Crowley Medical Research Center, Dallas, Texas, USA
| | | |
Collapse
|
110
|
Liu R, Varghese S, Rabkin SD. Oncolytic Herpes Simplex Virus Vector Therapy of Breast Cancer in C3(1)/SV40 T-antigen Transgenic Mice. Cancer Res 2005; 65:1532-40. [PMID: 15735042 DOI: 10.1158/0008-5472.can-04-3353] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oncolytic herpes simplex virus vectors are a promising strategy for cancer therapy, as direct cytotoxic agents, inducers of antitumor immune responses, and as expressers of anticancer genes. Progress is dependent upon representative preclinical models to evaluate therapy. In this study, two families of oncolytic herpes simplex virus vectors (G207 and NV1020 series) that have been in clinical trials were examined for the treatment of breast cancer, using the C3(1)/T-Ag transgenic mouse model. Female mice spontaneously develop mammary carcinomas, and the C3(1)/T-Ag-derived tumor cell line M6c forms implantable tumors. Both in vitro and in vivo, G47Delta, derived from G207 by deletion of ICP47 and the US11 promoter, was more efficacious than G207. Whereas NV1023, derived from NV1020 by deletion of ICP47 and insertion of LacZ, was as cytotoxic to M6c cells in vitro as G47Delta, it did not inhibit the growth of s.c. M6c tumors but did extend the survival of intracerebral tumor bearing mice. In contrast, NV1042, NV1023 expressing interleukin 12, inhibited s.c. M6c tumor growth to a similar extent as G47Delta, but was less effective than NV1023 in intracerebral tumors. In the spontaneously arising mammary tumor model, when only the first arising tumor per mouse was treated, G47Delta inhibited the growth of a subset of tumors, and when all tumors were treated, G47Delta significantly delayed tumor progression. When the first mammary tumor was treated and the remaining mammary glands removed, NV1042 was more efficacious than G47Delta at inhibiting the growth and progression of injected tumors.
Collapse
Affiliation(s)
- Renbin Liu
- Molecular Neurosurgery Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | |
Collapse
|
111
|
Wong RJ, Chan MK, Yu Z, Ghossein RA, Ngai I, Adusumilli PS, Stiles BM, Shah JP, Singh B, Fong Y. Angiogenesis inhibition by an oncolytic herpes virus expressing interleukin 12. Clin Cancer Res 2005; 10:4509-16. [PMID: 15240543 DOI: 10.1158/1078-0432.ccr-04-0081] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Oncolytic herpes simplex viruses (HSVs) may have significant antitumor effects resulting from the direct lysis of cancer cells. HSVs may also be used to express inserted transgenes to exploit additional therapeutic strategies. The ability of an interleukin (IL)-12-expressing HSV to treat squamous cell carcinoma (SCC) by inhibition of tumor angiogenesis is investigated in this study. EXPERIMENTAL DESIGN A replication-competent, attenuated, oncolytic HSV carrying the murine IL-12 gene (NV1042), its non-cytokine-carrying analog (NV1023), or saline was used to treat established murine SCC flank tumors by intratumoral injection. The expression of secondary antiangiogenic mediators was measured. Angiogenesis inhibition was assessed by in vivo Matrigel plug assays, flank tumor subdermal vascularity, and in vitro endothelial cell tubule formation assay. RESULTS Intratumoral injections of NV1042 (2 x 10(7) plaque-forming units) into murine SCC VII flank tumors resulted in smaller tumor volumes as compared with NV1023 or saline. IL-12 and IFN-gamma expression in tumors was 440 and 2.2 pg/mg, respectively, at 24 h after NV1042 injection, but both IL-12 and IFN-gamma were undetectable (<0.2 pg/mg) after NV1023 or saline injections. Expression of two antiangiogenesis mediators, monokine induced by IFN-gamma and IFN-inducible protein 10, was elevated after NV1042 treatment. Matrigel plug assays of NV1042-transfected SCC VII tumor cells demonstrated significantly decreased hemoglobin content and microvessel density as compared with NV1023 and PBS. Excised murine flank tumors treated with NV1042 had decreased subdermal vascularity as compared with NV1023 and PBS. Both splenocytes and IL-12 expression by NV1042 were required for in vitro inhibition of endothelial tubule formation. CONCLUSIONS IL-12 expression by an oncolytic herpes virus enhances therapy of SCC through antiangiogenic mechanisms. Strategies combining HSV oncolysis with angiogenesis inhibition merit further investigation for potential clinical application.
Collapse
Affiliation(s)
- Richard J Wong
- Head and Neck Service, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Abstract
Most cases of cancer, when detected at an advanced stage, cannot be cured with conventional therapeutic modalities. Therefore, novel targeted approaches such as gene therapy are needed. Nevertheless, while the safety record of gene therapy for cancer has been excellent with more than a thousand patients treated without mortality related to the therapy, clinical efficacy has so far been limited. Moreover, it has become evident that clinical efficacy is partly determined by efficacy of gene delivery. Most adenoviruses used for gene therapy have been based on serotype 5 (Ad5). Unfortunately, recent data suggest that the primary receptor, the coxsackie-adenovirus receptor (CAR) expression in tumors may be highly variable resulting in resistance to adenovirus infection. Consequently, various strategies have been evaluated to modify adenovirus tropism in order to circumvent CAR deficiency, including retargeting complexes or genetic capsid modifications. To further improve tumor penetration and local amplification of the anti-tumor effect, selectively oncolytic agents, e.g. conditionally replicating adenoviruses (CRAds), have been constructed. Infection of tumor cells results in replication, oncolysis, and subsequent release of the virus progeny. Normal tissue is spared due to lack of replication. This review will focus on a discussion of various modifications of adenovirus to achieve efficient anti-tumor effect, and special emphasis will be placed on CRAds in multimodality treatments.
Collapse
Affiliation(s)
- Anna Kanerva
- Cancer Gene Therapy Group, Rational Drug Design, Biomedicum Helsinki, University of Helsinki, Finland
| | | |
Collapse
|
113
|
Nakamori M, Fu X, Rousseau R, Chen SY, Zhang X. Destruction of nonimmunogenic mammary tumor cells by a fusogenic oncolytic herpes simplex virus induces potent antitumor immunity. Mol Ther 2004; 9:658-65. [PMID: 15120326 DOI: 10.1016/j.ymthe.2004.02.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Accepted: 02/27/2004] [Indexed: 10/26/2022] Open
Abstract
In principle, destruction of tumor cells in vivo by oncolytic agents would release the entire repertoire of tumor antigens in their natural forms, leading to effective antitumor immunity. This goal has been elusive despite extensive testing of numerous strategies. We developed a doubly fusogenic oncolytic herpes simplex virus (Synco-2D) that kills tumor cells by a unique dual mechanism combining direct cytolysis with syncytial formation induced by cell membrane fusion. A single intratumor injection of Synco-2D induced strong antitumor immunity against an otherwise nonimmunogenic murine mammary tumor growing in immune-competent mice. CD8+ T cells were the primary mediators of immunity, contributing to the destruction of both primary and metastatic tumors. We conclude that the fusogenic capacity of Synco-2D enables it to elicit antitumor immunity exceeding that induced by more conventional oncolytic viruses.
Collapse
Affiliation(s)
- Mikihito Nakamori
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
114
|
Delman KA, Zager JS, Bhargava A, Petrowsky H, Malhotra S, Ebright MI, Bennett JJ, Gusani NJ, Kooby DA, Roberts GD, Fong Y. Effect of murine liver cell proliferation on herpes viral behavior: implications for oncolytic viral therapy. Hepatology 2004; 39:1525-32. [PMID: 15185293 DOI: 10.1002/hep.20198] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Replication-competent herpes simplex oncolytic viruses are promising anticancer agents that partly target increased DNA synthesis in tumor cells. Investigators have proposed that these DNA viruses may be combined with liver resection to enhance killing of liver malignancies. Whether or not the cellular alterations associated with hepatic regeneration affect the efficacy and toxicity of these promising anticancer agents is unknown. This study examined the behavior of two oncolytic viruses, NV1020 and G207, during liver regeneration. When delivered during the peak of liver regeneration, replication and appearance of both G207 and NV1020 in hepatic tissue are enhanced as demonstrated by histochemical staining for the marker gene lac Z, immunohistochemical staining, and quantitative polymerase chain reaction. This increased appearance of virus in liver tissue correlates with increases in cellular ribonucleotide reductase activity and DNA synthesis and is also associated with increased viral binding. However, increased viral presence is transient, and viral detection declines to baseline within 7 days. When these viruses were delivered to animals even as early as 7 days after hepatectomy, there proved to be no measurable viral replication in any organ and no increased morbidity or mortality. In conclusion, the early stages of hepatic regeneration after resection provide an environment suitable for viral replication. Administration of replication-competent herpes simplex virus during the peak of hepatocyte regeneration (24-48 hours) permits viral productivity in tissue that otherwise does not support viral growth. The increase in hepatotoxicity after hepatectomy is short-lived and can be predicted by peak hepatocyte DNA synthesis.
Collapse
Affiliation(s)
- Keith A Delman
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Kirn D. Oncolytic virotherapy as a novel treatment platform for cancer. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2004:89-105. [PMID: 12894453 DOI: 10.1007/978-3-662-05352-2_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
116
|
Jacobs AH, Voges J, Kracht LW, Dittmar C, Winkeler A, Thomas A, Wienhard K, Herholz K, Heiss WD. Imaging in gene therapy of patients with glioma. J Neurooncol 2004; 65:291-305. [PMID: 14682379 DOI: 10.1023/b:neon.0000003658.51816.3f] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Over 10 years ago, the first successful gene therapy paradigms for experimental brain tumors models have been conducted, and they were thought to revolutionize the treatment of patients with gliomas. Application of gene therapy has been quickly forced into clinical trials, the first patients being enrolled in 1994, with overall results being disappointing. However, single patients seemed to benefit from gene therapy showing long-term treatment response, and most of these patients bearing small glioblastomas. Whereas the gene therapy itself has been performed with high sophistication, limited attention has been paid on technologies, which (i) allow an identification of viable target tissue in heterogenous glioma tissue and which (ii) enable an assessment of successful vector administration and vector-mediated gene expression in vivo. However, these measures are a prerequisite for the development of successful gene therapy in the clinical application. As biological treatment strategies such as gene and cell-based therapies hold promise to selectively correct disease pathogenesis, successful clinical implementation of these treatment strategies rely on the establishment of molecular imaging technology allowing the non-invasive assessment of endogenous and exogenous gene expression in vivo. Imaging endogenous gene expression will allow the characterization and identification of target tissue for gene therapy. Imaging exogenously introduced cells and genes will allow the determination of the 'tissue dose' of transduced cell function and vector-mediated gene expression, which in turn can be correlated to the induced therapeutic effect. Only these combined strategies of non-invasive imaging of gene expression in vivo will enable the establishment of safe and efficient vector administration and gene therapy protocols for clinical application. Here, we review some aspects of imaging in gene therapy trials for glioblastoma, and we present a 'proof-of-principle' 2nd-generation gene therapy protocol integrating molecular imaging technology for the establishment of efficient gene therapy in clinical application.
Collapse
Affiliation(s)
- A H Jacobs
- Max Planck-Institute for Neurological Research, Center of Molecular Medicine (ZMMK), Department of Neurology, University of Cologne, Cologne, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Abstract
Oncolytic viruses have been considered as a potential form of cancer treatment throughout the last century because of their ability to lyse and destroy tumor cells both in tissue culture and in animal models of cancer. However, it is only during the past decade that new molecular technologies have become available and understanding of genetic and molecular components of these viruses has increased to the point that they can be manipulated and made safe for use in treatment in humans. Thus there has been a revival of the concepts of conditionally replication-competent viruses and suicide gene therapy to supplement currently existing cancer therapies. While a wide variety of viruses have been closely studied for this purpose, herpes simplex virus type-1 (HSV-1) has received particularly close attention. The inherent cytotoxicity of this virus, if harnessed and made to be selective in the context of a tumor microenvironment, makes this an ideal candidate for further development. Furthermore, its large genome size, ability to infect cells with a high degree of efficiency, and the presence of an inherent viral-specific thymidine kinase gene add to its potential capabilities. This review explores work performed in this field and its potential for application in the treatment of cancers in humans.
Collapse
Affiliation(s)
- Emil Lou
- Department of Microbiology and Immunology, SUNY Upstate Medical University, College of Medicine, Syracuse, NY 13210, USA.
| |
Collapse
|
118
|
Lou E. Oncolytic viral therapy and immunotherapy of malignant brain tumors: two potential new approaches of translational research. Ann Med 2004; 36:2-8. [PMID: 15000342 DOI: 10.1080/07853890310016315] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Brain tumors arise at a rate of nearly 5/100,000 in the general population, with over 17,000 U.S. residents being diagnosed each year. Approximately 60% of all brain tumors are gliomas, which are derived from interstitial tissue of the brain, such as astrocytic or ependymal tissue, or oligodendrocytes. The traditional protocols for treatment of malignant gliomas include diagnostic surgery, followed by regimens of radio- and chemotherapies. In the case of chemotherapy, the treatment protocols have remained nearly unchanged for over 30 years despite high mortality rates, and with little to no improvement in outcome. New advances in the fields of molecular biology and immunology have resulted in new possibilities for treating malignant gliomas by targeting cellular and molecular mechanisms of tumor cells, and stand in contrast to traditional forms of treatment. In the field of gene therapy, the possibility of using oncolytic viruses, such as HSV-1, for glioma therapy--specifically, of high grade astrocytomas--is being explored, and trials have begun using a replication-selective mutant strain known as G207. An increased understanding of the role of the cytokine TGF-beta2 has led to developments of anti-sense immunotherapy targeting this factor. The two examples mentioned here are discussed in this review and cited as possible improvements in the treatment of high grade astrocytomas.
Collapse
Affiliation(s)
- Emil Lou
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
119
|
Hu JCC, Coffin RS. Oncolytic herpes simplex virus for tumor therapy. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 55:165-84. [PMID: 12968536 DOI: 10.1016/s0074-7742(03)01007-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Affiliation(s)
- Jennifer C C Hu
- Cancer Cell Biology, Hammersmith Hospital Campus, Imperial College School of Medicine, London W12 0NN, United Kingdom
| | | |
Collapse
|
120
|
Abstract
OBJECTIVE Gene therapy is a rapidly evolving novel treatment for human disease. This review discusses the latest development in gene transfer technology and its potential use in the female reproductive tract. METHODS A comprehensive search using the MEDLINE database was performed to review current, innovative trends in gene transfer technology. In addition, articles on reproductive tract gene transfer were reviewed. CONCLUSION(S) Recent developments, such as the Human Genome Project, have generated great interest in the genetic basis of human health and disease. Gene therapy is a rapidly evolving field that uses gene transfer to treat disease. Ongoing research in the field focuses on improving vector technology to enable efficient in vivo gene transfer. Although multiple techniques for gene transfer have been described, no single technique can be used in all instances. The human female reproductive tract is easily accessible and can be readily transfected. In vivo gene transfer has resulted in successful alteration of implantation rates and has demonstrated potential for use in treatment of ovarian cancer.
Collapse
Affiliation(s)
- Gaurang S Daftary
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | |
Collapse
|
121
|
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 2003; 10:983-90. [PMID: 12756419 DOI: 10.1038/sj.gt.3302038] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Several clinical trials have or are being performed testing the safety and efficacy of different strains of oncolytic viruses (OV) for malignant cancers. OVs represent either naturally occurring or genetically engineered strains of viruses that exhibit relatively selective replication in tumor cells. Several types of OV have been derived from herpes simplex virus 1 (HSV1). Tumor oncolysis depends on the processes of initial OV infection of tumor, followed by subsequent propagation of OV within the tumor itself. The role of the immune responses in these processes has not been extensively studied. On the contrary, effects of the immune response on the processes of wild-type HSV1 infection and propagation in the central nervous system have been studied and described in detail. The first line of defense against a wild-type HSV1 infection in both naive and immunized individuals is provided by innate humoral (complement, cytokines, chemokines) and cellular (macrophages, neutrophils, NK cells, gammadelta T cells, and interferon-producing cells) responses. These orchestrate the lysis of virions and virus-infected cells as well as provide a link to effective adaptive immunity. The role of innate defenses in curtailing the oncolytic effect of genetically engineered HSV has only recently been studied, but several of the same host responses appear to be operative in limiting anticancer effects by the replicating virus. The importance of this knowledge lies in finding avenues to modulate such initial innate responses, in order to allow for increased oncolysis of tumors while minimizing host toxicity.
Collapse
Affiliation(s)
- H Wakimoto
- Molecular Neuro-Oncology Laboratories, Neurosurgery Service, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | |
Collapse
|
122
|
Miller CG, Fraser NW. Requirement of an integrated immune response for successful neuroattenuated HSV-1 therapy in an intracranial metastatic melanoma model. Mol Ther 2003; 7:741-7. [PMID: 12788647 PMCID: PMC2661757 DOI: 10.1016/s1525-0016(03)00120-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neuroattenuated herpes simplex virus ICP34.5 mutants slow progression of preformed tumors and lead to complete regression of some tumors. Although this was previously thought to be due to viral lysis of infected tumor cells, it is now understood that there is an immune component to tumor destruction. We have previously shown that no difference in survival is seen in lymphocyte-depleted mice after viral or mock therapy of syngeneic intracranial melanomas. We have also demonstrated the presence of a wide spectrum of immune cells following viral therapy, including larger percentages of CD4+ T cells and macrophages. In this paper, the contribution of the immune system to tumor destruction has been further delineated. Viral therapy of intracranial melanoma induces a tumor-specific cytotoxic and proliferative T cell response. However, there is no increase following viral therapy in either serum tumor antibody levels or viral-neutralizing antibodies. Thus specific T cell responses appear to mediate viral-elicited prolongation in survival. These data suggest that designing new viruses capable of augmenting T cell responses may induce stronger tumor destruction upon viral therapy.
Collapse
MESH Headings
- Animals
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- CD4 Antigens/genetics
- CD4 Antigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8 Antigens/genetics
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Division/physiology
- DNA-Binding Proteins/deficiency
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Female
- Herpes Simplex/genetics
- Herpes Simplex/immunology
- Herpes Simplex/virology
- Herpes Simplex Virus Vaccines/administration & dosage
- Herpes Simplex Virus Vaccines/immunology
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/physiology
- Injections, Intraperitoneal
- Killer Cells, Natural/immunology
- Macrophages/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Transplantation
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, Attenuated/administration & dosage
Collapse
Affiliation(s)
| | - Nigel W. Fraser
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104-6076, USA
| |
Collapse
|
123
|
Solly SK, Trajcevski S, Frisén C, Holzer GW, Nelson E, Clerc B, Abordo-Adesida E, Castro M, Lowenstein P, Klatzmann D. Replicative retroviral vectors for cancer gene therapy. Cancer Gene Ther 2003; 10:30-9. [PMID: 12489026 DOI: 10.1038/sj.cgt.7700521] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2002] [Indexed: 11/08/2022]
Abstract
Poor efficiency of gene transfer into cancer cells constitutes the major bottleneck of current cancer gene therapy. We reasoned that because tumors are masses of rapidly dividing cells, they would be most efficiently transduced with vector systems allowing transgene propagation. We thus designed two replicative retrovirus-derived vector systems: one inherently replicative vector, and one defective vector propagated by a helper retrovirus. In vitro, both systems achieved very efficient transgene propagation. In immunocompetent mice, replicative vectors transduced >85% tumor cells, whereas defective vectors transduced <1% under similar conditions. It is noteworthy that viral propagation could be efficiently blocked by azido-thymidine, in vitro and in vivo. In a model of established brain tumors treated with suicide genes, replicative retroviral vectors (RRVs) were approximately 1000 times more efficient than defective adenoviral vectors. These results demonstrate the advantage and potential of RRVs and strongly support their development for cancer gene therapy.
Collapse
Affiliation(s)
- Sounkary K Solly
- Biologie et Thérapeutique des Pathologies Immunitaires, CNRS/UPMC UMR 7087, Hôpital Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Rainov NG, Ren H. Oncolytic viruses for treatment of malignant brain tumours. ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 88:113-23. [PMID: 14531569 DOI: 10.1007/978-3-7091-6090-9_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Wild type viruses have been known for decades for their capability to destroy malignant tumour cells upon infection and intracellular replication. Genetic engineering of such viruses was, however, only recently done in an attempt to improve their utility as biological anticancer agents. Wild type or recombinant viruses able to selectively destroy tumour cells while sparing normal tissue are known as oncolytic viruses. Most oncolytic viruses currently investigated in clinical trials are derived from adenovirus (AV) or herpes simplex virus type I (HSVI). More than 300 patients with solid tumours were now treated in clinical trials with oncolytic viruses, and in most cases virus was administered directly into the tumour mass. About 10% of the above patients had recurrent malignant glioma. Total intratumoral doses of up to 2 x 10(12) virus particles were well tolerated, and in general no severe side effects resulted from the clinical use of oncolytic AV and HSVI, either in the brain or in the rest of the body. Encouraging anti-tumoral activity was demonstrated in some types of tumours treated locally with oncolytic viruses, and systemic chemotherapy was found to potentiate the anti-tumour effect of virus mediated oncolysis. In malignant glioma, standard gene therapy approaches employing non-replicating virus vectors failed to demonstrate significant benefit in clinical studies. Therapy with oncolytic viruses seems to hold more promise in early clinical trials than gene therapy with non-replicating virus vectors. However, further major advancements in virus designs, application modalities, and understanding of the interactions of the host's immune system with the virus are clearly needed before oncolytic virus therapy of malignant brain tumours can be introduced to clinical practice.
Collapse
Affiliation(s)
- N G Rainov
- Department of Neurological Science, University of Liverpool, UK.
| | | |
Collapse
|
125
|
Tanaka M, Kagawa H, Yamanashi Y, Sata T, Kawaguchi Y. Construction of an excisable bacterial artificial chromosome containing a full-length infectious clone of herpes simplex virus type 1: viruses reconstituted from the clone exhibit wild-type properties in vitro and in vivo. J Virol 2003; 77:1382-91. [PMID: 12502854 PMCID: PMC140785 DOI: 10.1128/jvi.77.2.1382-1391.2003] [Citation(s) in RCA: 244] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In recent years, several laboratories have reported on the cloning of herpes simplex virus type 1 (HSV-1) genomes as bacterial artificial chromosomes (BACs) in Escherichia coli and on procedures to manipulate these genomes by using the bacterial recombination machinery. However, the HSV-BACs reported so far are either replication incompetent or infectious, with a deletion of one or more viral genes due to the BAC vector insertion. For use as a multipurpose clone in research on HSV-1, we attempted to generate infectious HSV-BACs containing the full genome of HSV-1 without any loss of viral genes. Our results were as follows. (i) E. coli (YEbac102) harboring the full-length HSV-1 genome (pYEbac102) in which a BAC flanked by loxP sites was inserted into the intergenic region between U(L)3 and U(L)4 was constructed. (ii) pYEbac102 was an infectious molecular clone, given that its transfection into rabbit skin cells resulted in production of infectious virus (YK304). (iii) The BAC vector sequence was almost perfectly excisable from the genome of the reconstituted virus YK304 by coinfection of Vero cells with YK304 and a recombinant adenovirus, AxCANCre, expressing Cre recombinase. (iv) As far as was examined, the reconstituted viruses from pYEbac102 could not be phenotypically differentiated from wild-type viruses in vitro and in vivo. Thus, the viruses grew as well in Vero cells as did the wild-type virus and exhibited wild-type virulence in mice on intracerebral inoculation. (v) The infectious molecular clone pYEbac102 is in fact useful for mutagenesis of the HSV-1 genome by bacterial genetics, and a recombinant virus carrying amino acid substitutions in both copies of the alpha0 gene was generated. pYEbac102 will have multiple applications to the rapid generation of genetically engineered HSV-1 recombinants in basic research into HSV-1 and in the development of HSV vectors in human therapy.
Collapse
Affiliation(s)
- Michiko Tanaka
- Department of Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | | | | | | | | |
Collapse
|
126
|
Abstract
Although the cytotoxic effects of viruses are usually viewed in terms of pathogenicity, it is possible to harness this activity for therapeutic purposes. Viral genomes are highly versatile, and can be modified to direct their cytotoxicity towards cancer cells. These viruses are known as oncolytic viruses. How are viruses engineered to become tumour specific, and can they be used to safely treat cancer in humans?
Collapse
Affiliation(s)
- E Antonio Chiocca
- Molecular Neuro-Oncology Laboratories, Neurosurgery Service, Massachusetts General Hospital, East Building, 13th Street, Bldg 149, Charlestown, Massachusetts 02129, USA. chioccahelix.mgh.harvard.edu
| |
Collapse
|
127
|
Abstract
There is reason to believe that the unfolding revolution in molecular biology and translational research will allow selective targeting of tumor cells, and radically change the way general practitioners and pediatric oncologists treat and follow children with cancer. This article highlights some of the most promising approaches being tested in the field. By learning about the underlying biology, the remaining hurdles, the projected timeline, and the possible impact of new therapies on the practice of pediatric oncology, health care professionals and patients should be better prepared for the future of pediatric oncology.
Collapse
Affiliation(s)
- Robert J Arceci
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans Street, Room 2M51, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|
128
|
Varghese S, Rabkin SD. Oncolytic herpes simplex virus vectors for cancer virotherapy. Cancer Gene Ther 2002; 9:967-78. [PMID: 12522436 DOI: 10.1038/sj.cgt.7700537] [Citation(s) in RCA: 175] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2002] [Indexed: 12/29/2022]
Abstract
Oncolytic herpes simplex virus type 1 (HSV-1) vectors are emerging as an effective and powerful therapeutic approach for cancer. Replication-competent HSV-1 vectors with mutations in genes that affect viral replication, neuropathogenicity, and immune evasiveness have been developed and tested for their safety and efficacy in a variety of mouse models. Evidence to-date following administration into the brain attests to their safety, an important observation in light of the neuropathogenicity of the virus. Phase I clinical traits of three vectors, G207, 1716, and NV1020, are either ongoing or completed, with no adverse events attributed to the virus. These and other HSV-1 vectors are effective against a myriad of solid tumors in mice, including glioma, melanoma, breast, prostate, colon, ovarian, and pancreatic cancer. Enhancement of activity was observed when HSV-1 vectors were used in combination with traditional therapies such as radiotherapy and chemotherapy, providing an attractive strategy to pursue in the clinic. Oncolytic HSV-1 vectors expressing "suicide" genes (thymidine kinase, cytosine deaminase, rat cytochrome P450) or immunostimulatory genes (IL-12, GM-CSF, etc.) have been constructed to maximize tumor destruction through multimodal therapeutic mechanisms. Further advances in virus delivery and tumor specificity should improve the likelihood for successful translation to the clinic.
Collapse
Affiliation(s)
- Susan Varghese
- Molecular Neurosurgery Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | |
Collapse
|
129
|
Vile R, Ando D, Kirn D. The oncolytic virotherapy treatment platform for cancer: unique biological and biosafety points to consider. Cancer Gene Ther 2002; 9:1062-7. [PMID: 12522445 DOI: 10.1038/sj.cgt.7700548] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2002] [Indexed: 11/09/2022]
Abstract
The field of replication-selective oncolytic viruses (virotherapy) has exploded over the last 10 years. As with many novel therapeutic approaches, initial overexuberance has been tempered by clinical trial results with first-generation agents. Although a number of significant hurdles to this approach have now been identified, novel solutions have been proposed and improvements are being made at a furious rate. This article seeks to initiate a discussion of these hurdles, approaches to overcome them, and unique safety and regulatory issues to consider.
Collapse
Affiliation(s)
- Richard Vile
- Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
130
|
Abstract
The grave outlook for malignant glioma patients in spite of improvements to current modalities has ushered in new approaches to therapy. Viruses have emerged on the scene and gained attention for their ability to play essentially two roles: first, as vectors for therapeutic gene delivery and second, as engineered infectious agents capable of selectively lysing tumor cells. To date, clinical brain tumor trials using viruses for gene delivery have employed retroviral or adenoviral vectors to introduce ganciclovir susceptibility to tumors in the form of the HSV1-TK gene. Clinical oncolytic studies, on the other hand, have evaluated a conditionally replicating HSV as an antineoplastic agent. Despite some promise afforded by these trials, further studies are warranted; the investigation of additional viruses to play these roles is inevitable and is now precedented.
Collapse
Affiliation(s)
- Peter E Fecci
- Departments of Neurosurgery and Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
131
|
Abstract
An increasing number of oncolytic virus vectors has been developed lately for cancer therapy. Herpes simplex virus type 1 (HSV-1) vectors are particularly useful, because they can be genetically engineered to replicate and spread highly selectively in tumor cells and can also express multiple foreign transgenes. These vectors can manifest cytopathic effect in a wide variety of tumor types without damaging normal tissues, provide amplified gene delivery within the tumor, and induce specific antitumor immunity. Multiple recombinant HSV-1 vectors have been tested in patients with brain tumors and other cancers, which showed the feasibility of administering replication-competent HSV-1 vectors safely in human organs including the brain. Different approaches are currently undertaken to improve the efficacy of oncolytic HSV-1 therapy which include development of new generation vectors via further genetic engineering of existing safe vectors, combination with immune gene therapy, and combination with conventional therapies. Oncolytic virus therapy is a promising therapeutic modality that awaits establishing as an important treatment option for cancer patients in the near future.
Collapse
Affiliation(s)
- Tomoki Todo
- Molecular Neurosurgery Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA.
| |
Collapse
|
132
|
Bharatan NS, Currier MA, Cripe TP. Differential susceptibility of pediatric sarcoma cells to oncolysis by conditionally replication-competent herpes simplex viruses. J Pediatr Hematol Oncol 2002; 24:447-53. [PMID: 12218591 DOI: 10.1097/00043426-200208000-00008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Attenuated viruses derived from herpes simplex virus (HSV) type 1 that kill tumor cells (oncolysis) are currently in clinical trials for selected cancers, primarily carcinomas and gliomas. The authors sought to determine if pediatric sarcoma cells are also sensitive to HSV-mediated oncolysis. MATERIALS AND METHODS The authors tested a panel of ten cell lines derived from rhabdomyosarcoma, osteosarcoma, Ewing sarcoma, and a secondary malignant fibrous histiocytoma for survival after exposure to attenuated HSV vectors. The viruses used included NV1020, haploid for the neurovirulence gene, and G207, deleted for both and ribonucleotide reductase but expressing the beta-galactosidase reporter gene. G207 transduction was determined by measuring beta-galactosidase expression. RESULTS Sarcoma cells differed in their sensitivity to viral oncolysis but were relatively consistent by histologic type. Rhabdomyosarcoma and malignant fibrous histiocytoma cells were most sensitive while osteosarcoma cells were intermediately sensitive to oncolysis by both HSV recombinants. Although Ewing sarcoma cells showed efficient viral entry and gene transfer, these cells were the least susceptible to oncolysis by HSV. CONCLUSIONS Conditionally replication-competent HSV-derived vectors may be useful for the treatment of rhabdomyosarcoma and osteosarcoma, but may not be as efficacious for treating Ewing sarcoma until the mechanism of resistance is defined and circumvented.
Collapse
Affiliation(s)
- Neeti S Bharatan
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | | | |
Collapse
|
133
|
Abstract
The concept of using replicating viruses as anticancer agents is not a new one, but the ability to genetically modify these viruses into increasingly potent and tumor-specific vectors is a recent phenomenon. As more is learned about the functions of viral gene products in controlling the mammalian cell cycle and in disabling cellular defense mechanisms, specific viral functions can be augmented or eliminated to enhance antineoplastic efficacy. In this article, general mechanisms by which oncolytic viruses achieve their antitumor efficacy and specificity are reviewed. The paradoxical roles of the immune response are addressed with respect to oncolytic viral therapy, as it, on one hand, impedes the spread of viral infection, and on the other, augments tumor cell destruction through the recruitment of T cells "vaccinated" against tumor antigens. The most commonly used oncolytic viruses are each reviewed in turn, including adenoviruses, herpes simplex viruses, vaccinia viruses, reoviruses, and Newcastle disease viruses. Special attention is focused on the unique biology of each of these viruses as well as the status of several of these mutants in clinical trials.
Collapse
Affiliation(s)
- John T Mullen
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114-2696, USA
| | | |
Collapse
|
134
|
Jacobs A, Heiss WD. Towards non-invasive imaging of HSV-1 vector-mediated gene expression by positron emission tomography. Vet Microbiol 2002; 86:27-36. [PMID: 11888687 DOI: 10.1016/s0378-1135(01)00488-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The overall goals of the broad and growing field of molecular medicine is to identify fundamental errors of disease and to develop corrections of them on the molecular level. At the same time, real-time imaging of gene expression in vivo aims towards a detailed analysis of both endogenous and exogenous gene expression in animal models of disease and in the clinical setting. Non-invasive imaging of endogenous gene expression may reveal insight into the molecular basis of disease pathogenesis and the extent of treatment response. When exogenous genes are introduced, e.g. by herpes simplex virus type 1 (HSV-1)-based vectors, to ameliorate a genetic defect or to add an additional gene function to cells, imaging techniques may reveal the assessment of the location, magnitude and duration of therapeutic gene expression and its correlation to the therapeutic effect. Here, we review the main approaches of non-invasive imaging techniques of gene expression in vivo with special reference to HSV-1 vector-mediated gene expression.
Collapse
Affiliation(s)
- A Jacobs
- Department of Neurology, University of Cologne, Max-Planck-Institute for Neurological Research, Center of Molecular Medicine, Cologne, Germany.
| | | |
Collapse
|
135
|
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. [PMID: 11960286 DOI: 10.1038/sj.cgt.7700446] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Here we developed an effective therapeutic approach using a replication-conditional mutant of herpes simplex virus (HSV), G207, for the treatment of metastatic tumors in the immunologically privileged central nervous system. An experimental model of brain metastasis was developed using BALB/c mice that harbored both intracranial (i.c.) and subcutaneous (s.c.) mouse CT26 colon adenocarcinoma tumors. Intratumoral injections of G207 into s.c. tumors elicited cytotoxic T-cell responses not only to HSV but also to a tumor antigen; however, only a limited antitumor effect was observed on metastatic brain tumors. To improve this antitumor effect, G207 was also injected into the brain tumor. After intratumoral injections of G207 into both i.c. and s.c. CT26 tumors, a significant antitumor effect was observed in the metastatic brain tumors. This therapeutic efficacy was absent in athymic mice, indicating that the antitumor effect could be mediated by T cells. Cytotoxic T-cell responses to HSV and the tumor antigen were induced by injections of G207 into i.c. and s.c. CT26 tumors. These results suggest that HSV-infected brain tumors may be efficiently eliminated by the induced anti-HSV T cells as well as by antitumor T cells. Therefore, this strategy of immuno-viral therapy, involving direct viral oncolytic activities and inducing antitumor and antiviral immune responses, may be useful for the treatment of tumors in the immunologically privileged central nervous system.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Antineoplastic Combined Chemotherapy Protocols
- Brain Neoplasms/immunology
- Brain Neoplasms/secondary
- Brain Neoplasms/therapy
- Cancer Vaccines/therapeutic use
- Cytotoxicity, Immunologic
- Female
- Genetic Therapy/methods
- Genetic Vectors
- Humans
- Immunotherapy/methods
- Injections, Intralesional
- Injections, Intraventricular
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Simplexvirus/physiology
- Spleen/physiology
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Vaccination
- Viral Vaccines/therapeutic use
- Virus Replication/genetics
Collapse
Affiliation(s)
- Masahiro Toda
- Division of Cellular Signaling, Institute for Advanced Medical Research, Shinju-ku, Tokyo 160-8582, Japan.
| | | | | | | | | |
Collapse
|
136
|
Ali SA, Lynam J, McLean CS, Entwisle C, Loudon P, Rojas JM, McArdle SEB, Li G, Mian S, Rees RC. Tumor regression induced by intratumor therapy with a disabled infectious single cycle (DISC) herpes simplex virus (HSV) vector, DISC/HSV/murine granulocyte-macrophage colony-stimulating factor, correlates with antigen-specific adaptive immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3512-9. [PMID: 11907113 DOI: 10.4049/jimmunol.168.7.3512] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Direct intratumor injection of a disabled infectious single cycle HSV-2 virus encoding the murine GM-CSF gene (DISC/mGM-CSF) into established murine colon carcinoma CT26 tumors induced a significant delay in tumor growth and complete tumor regression in up to 70% of animals. Pre-existing immunity to HSV did not reduce the therapeutic efficacy of DISC/mGM-CSF, and, when administered in combination with syngeneic dendritic cells, further decreased tumor growth and increased the incidence of complete tumor regression. Direct intratumor injection of DISC/mGM-CSF also inhibited the growth of CT26 tumor cells implanted on the contralateral flank or seeded into the lungs following i.v. injection of tumor cells (experimental lung metastasis). Proliferation of splenocytes in response to Con A was impaired in progressor and tumor-bearer, but not regressor, mice. A potent tumor-specific CTL response was generated from splenocytes of all mice with regressing, but not progressing tumors following in vitro peptide stimulation; this response was specific for the gp70 AH-1 peptide SPSYVYHQF and correlated with IFN-gamma, but not IL-4 cytokine production. Depletion of CD8(+) T cells from regressor splenocytes before in vitro stimulation with the relevant peptide abolished their cytolytic activity, while depletion of CD4(+) T cells only partially inhibited CTL generation. Tumor regression induced by DISC/mGM-CSF virus immunotherapy provides a unique model for evaluating the immune mechanism(s) involved in tumor rejection, upon which tumor immunotherapy regimes may be based.
Collapse
Affiliation(s)
- Selman A Ali
- Department of Life Sciences, Nottingham Trent University, Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Endo T, Toda M, Watanabe M, Iizuka Y, Kubota T, Kitajima M, Kawakami Y. 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. [PMID: 11857031 DOI: 10.1038/sj.cgt.7700407] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2001] [Indexed: 11/09/2022]
Abstract
In this study, we investigated the therapeutic efficacy of a replication-conditional mutant HSV, G207, for the treatment of liver metastasis of colon carcinoma. Three liver metastasis models in syngeneic BALB/c mice were developed: (i) splenic injection, (ii) splenic and subcutaneous (s.c.) injection, and (iii) orthotopic implantation of CT26 colon carcinoma. In the splenic injection model, G207 was injected into the established splenic tumor on day 7. In the splenic and s.c. injection model, G207 were injected into the established s.c. tumor on days 5 and 8. In the orthotopic implantation model, a piece of CT26 tumor tissue was transplanted onto the wall of the cecum and G207 was injected in the established cecum tumor on day 7. On day 21 or 28, animals were sacrificed and liver metastases were evaluated. In all three models in immunocompetent mice, liver metastases were significantly reduced by intratumoral inoculation with G207 compared to the control. In athymic mice, however, there was no significant therapeutic effect of intratumoral inoculation with G207 on liver metastases. Tumor-specific cytotoxic T-lymphocyte responses were induced in mice treated with G207 in the orthotopic implantation model. These results suggest that intratumoral inoculation of G207, as an in situ cancer vaccine, can be an effective approach against liver metastasis of colon cancer and the efficacy involves tumor-specific T-cell responses.
Collapse
Affiliation(s)
- Takashi Endo
- Department of Surgery, Institute for Advanced Medical Research, Keio University, School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
138
|
Abstract
There is a clear need for new, selective, cancer treatments that do not cause the cross-resistance which occurs with currently available chemotherapeutic agents. Gene therapy is a promising approach, but to date, it has shown limited effectiveness in clinical trials because of insufficient gene transduction. Many investigators are now revisiting the 'old' idea of using tumour-specific, replication-selective viruses or bacteria to treat cancer. These agents can be directly oncolytic, but can also be used to simultaneously express therapeutic genes in target cells or induce tumour-specific, cell-mediated immunity. We discuss the promise of this rapidly evolving field and examine the potential barriers to its success.
Collapse
Affiliation(s)
- Lynda K Hawkins
- Imperial College School of Medicine, Imperial Cancer Research Fund Programme for Viral and Genetic Therapy of Cancer, Hammersmith Hospital, London, UK
| | | | | |
Collapse
|
139
|
Abstract
Viral vectors have been widely used as gene delivery vehicles for both experimental and clinical investigations. Although these vectors are capable of achieving high gene transduction efficiency in vitro, one of the major limitations facing the therapeutic viral vectors is that the preexisting host anti-vector immunity can substantially reduce their transduction efficiency in vivo. This is especially of concern when the therapeutic remedy requires repeated systemic administration. Here we report the delivery of herpes simplex virus (HSV) derived vectors through liposome formulation. In these studies, we have prepared HSV vectors in three different forms for liposome formulation: purified viral DNA (obtained from a bacterial artificial chromosome containing an infectious HSV genome), HSV capsids, and intact viral particles. All three forms of HSV were readily transfected into cultured cells and infectious virus was efficiently generated. Furthermore, introduction of HSV vectors as DNA/liposome complexes improved in vivo transduction efficiency, by effectively evading the host anti-HSV immunity during systemic administration. We conclude that viral vectors such as HSV can be systemically delivered through liposome formulation for safe and repeated administration for gene transduction or oncolytic purposes.
Collapse
Affiliation(s)
- X Fu
- Center for Cell and Gene Therapy, Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
140
|
Peng KW, Ahmann GJ, Pham L, Greipp PR, Cattaneo R, Russell SJ. Systemic therapy of myeloma xenografts by an attenuated measles virus. Blood 2001; 98:2002-7. [PMID: 11567982 DOI: 10.1182/blood.v98.7.2002] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Conditionally replicating viruses are promising agents for the treatment of malignancy. Here it is shown that the live attenuated Edmonston-B vaccine strain of measles virus (MV-Edm) replicates selectively in human myeloma cells and has potent antitumor activity. In vitro, replication of MV-Edm was restricted in phytohemagglutinin (PHA)-stimulated peripheral blood lymphocytes (PBLs) but proceeded efficiently in a panel of 6 myeloma cell lines-ARH-77, RPMI 8226, JJN-3, MM1, KAS-6/1, and KMS-11-and in primary myeloma cells isolated by CD138 sorting from the bone marrow aspirates of 6 patients. MV-Edm infection induced potent cytopathic effects in these myeloma cells, resulting in the formation of multinucleated syncytia that eventually became nonviable. In contrast, syncytial formation in PHA-stimulated PBLs was minimal after MV-Edm infection. In vivo, MV-Edm was antitumorigenic and inhibited the establishment of myeloma cells as xenografts in immunocompromised mice. When injected directly into ARH-77 myeloma xenografts in the mice, MV-Edm caused complete regression of these xenografts. MV-Edm administered intravenously into the tail veins of mice also showed significant antineoplastic activity against established RPMI 8226 and ARH-77 xenografts. In particular, the ARH-77 myeloma xenografts were exquisitely sensitive to MV-Edm therapy, and tumors in all mice regressed completely. In light of its selectivity for myeloma cells and its potent antineoplastic activity against myeloma xenografts in vivo, MV-Edm merits further development for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- K W Peng
- Molecular Medicine Program and the Department of Hematology, Mayo Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
141
|
Jorgensen TJ, Katz S, Wittmack EK, Varghese S, Todo T, Rabkin SD, Martuza RL. 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. [PMID: 11687957 PMCID: PMC1506202 DOI: 10.1038/sj.neo.7900193] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2001] [Accepted: 05/15/2001] [Indexed: 11/09/2022] Open
Abstract
Viral gene therapy against malignant tumors holds great promise for tumors that are susceptible to the oncolytic activity of viruses. One advantage of oncolytic viral therapy is that it can potentially be combined with other therapies, such as radiotherapy, to obtain an enhanced tumor response. In the case of prostate cancer, herpes simplex virus-mediated therapies have been shown to be highly effective in animal models; however, studies of the efficacy of combined viral and radiation therapy have not yet been reported. In this study, we have combined G207, a multimutated HSV type 1 vector, with external beam radiation therapy of prostate tumors grown subcutaneously in mice. We examined both the human LNCaP tumor in athymic mice and the mouse transgenic TRAMP tumor in either athymic mice or its syngeneic host, C57BL/6 mice. Virus was delivered either intravenously, in the case of LNCaP, or intratumorally, in the case of TRAMP. We found that individually, either G207 or radiation was effective in delaying tumor growth in these models. However, delivering the treatments simultaneously did not produce an enhanced effect.
Collapse
Affiliation(s)
- T J Jorgensen
- Department of Radiation Medicine, Lombardi Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA.
| | | | | | | | | | | | | |
Collapse
|
142
|
Okada T, Shah M, Higginbotham JN, Li Q, Wildner O, Walbridge S, Oldfield E, Blaese RM, Ramsey WJ. AV.TK-mediated killing of subcutaneous tumors in situ results in effective immunization against established secondary intracranial tumor deposits. Gene Ther 2001; 8:1315-22. [PMID: 11571568 DOI: 10.1038/sj.gt.3301526] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2000] [Accepted: 06/11/2001] [Indexed: 11/09/2022]
Abstract
Gene transfer vectors expressing herpes simplex thymidine kinase (HSVtk), in addition to direct killing of tumor cells, often have an associated local "bystander effect" mediated by metabolic coupling of tumor cells. A systemic antitumor effect mediated by the immune system, termed the distant bystander effect, has also been reported. We have observed the development of cytotoxic T-lymphocyte (CTL) populations and long-lasting antitumor immunity following treatment of subcutaneous tumors with an adenoviral vector expressing HSVtk (AV.TK) and ganciclovir (GCV) in rat glioma model. This vaccination effect seen with AV.TK/GCV treatment of subcutaneous tumor could even abrogate or retard growth of previously established secondary intracranial tumors.
Collapse
Affiliation(s)
- T Okada
- Clinical Gene Therapy Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Kirn D, Martuza RL, Zwiebel J. Replication-selective virotherapy for cancer: Biological principles, risk management and future directions. Nat Med 2001; 7:781-7. [PMID: 11433341 DOI: 10.1038/89901] [Citation(s) in RCA: 375] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the search for novel cancer therapies that can be used in conjunction with existing treatments, one promising area of research is the use of viral vectors and whole viruses. This review describes the underlying biological principles and current status of the field, outlines approaches for improving clinical effectiveness and discusses the unique safety and regulatory issues surrounding viral therapies.
Collapse
Affiliation(s)
- D Kirn
- Viral and Genetic Therapy Programme, Imperial College School of Medicine and the Imperial Cancer Research Fund, Hammersmith Hospital, London, UK.
| | | | | |
Collapse
|
144
|
Abstract
The concept of using viruses as oncolytic agents has a long history. However, relatively new developments are the use of these viruses as gene delivery vehicles and the restriction of viral replication and lysis to tumour cells. The latter is attempted by the use of tumour-specific promoters, which transcriptionally target viral genes involved in replication, or by deletion of viral functions dispensable for replication in tumour cells but essential for productive infection of normal cells. In addition, retargeting of the viral tropism towards tumours by capsid modifications has been examined. Although much progress has been made in developing oncolytic vectors for clinical use, there is still a long way to go to determine which combinations of virus, gene therapy, surgery, radiation, and/or chemotherapy will provide improved therapy for the control and eradication of a variety of human cancers. First controlled clinical trials with an oncolytic adenovirus in combination with chemotherapy have shown encouraging antineoplastic activity. For future vector developments it will be crucial to achieve maximum vector distribution and transgene expression within tumours, to trigger a specific systemic immune effector response against treated and untreated lesions, and to modulate the immune system to avoid immune-mediated inactivation or destruction of the virus. In the context of replication-competent vectors, suicide genes might be used as fail-safe mechanism in the case of a runaway infection.
Collapse
Affiliation(s)
- O Wildner
- Humboldt-Universität zu Berlin, Labor für Gentherapie, Germany.
| |
Collapse
|
145
|
Varghese S, Newsome JT, Rabkin SD, McGeagh K, Mahoney D, Nielsen P, Todo T, Martuza RL. Preclinical safety evaluation of G207, a replication-competent herpes simplex virus type 1, inoculated intraprostatically in mice and nonhuman primates. Hum Gene Ther 2001; 12:999-1010. [PMID: 11387063 DOI: 10.1089/104303401750195944] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
G207, a replication-competent herpes simplex virus type 1 (HSV-1) virus, has been previously shown to be effective against human prostate cancer xenografts in mice. This study assesses its safety in the prostate of two animal models known for their sensitivity to HSV-1. BALB/c mice were injected intraprostatically with either HSV-1 G207 or strain F and observed for 5 months. None of the G207-injected animals exhibited any clinical signs of disease or died. However, 50% of strain F-injected mice displayed sluggish, hunched behavior and died by day 13. Histopathologically, the G207-injected prostates were normal whereas strain F-injected prostates showed epithelial flattening, sloughing, and stromal edema. Four Aotus nancymae monkeys were also injected with G207 intraprostatically and observed short term (up to 21 days) and long term (56 days). Safety was assessed on the basis of clinical observations, viral biodistribution, virus shedding, and histopathology. None of the injected monkeys displayed evidence of clinical disease, shedding of infectious virus, or spread of the virus into other organs. Except for minor histological changes unrelated to the study, no significant abnormalities were observed. These results demonstrate that G207 can be safely inoculated into the prostate and should be considered for human trials for the treatment of prostate cancer.
Collapse
Affiliation(s)
- S Varghese
- Molecular Neurosurgery Laboratory, Department of Surgery (Neurosurgery), Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
146
|
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 U S A 2001; 98:6396-401. [PMID: 11353831 PMCID: PMC33479 DOI: 10.1073/pnas.101136398] [Citation(s) in RCA: 340] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oncolytic herpes simplex virus type 1 (HSV-1) vectors are promising therapeutic agents for cancer. Their efficacy depends on the extent of both intratumoral viral replication and induction of a host antitumor immune response. To enhance these properties while employing ample safeguards, two conditionally replicating HSV-1 vectors, termed G47Delta and R47Delta, have been constructed by deleting the alpha47 gene and the promoter region of US11 from gamma34.5-deficient HSV-1 vectors, G207 and R3616, respectively. Because the alpha47 gene product is responsible for inhibiting the transporter associated with antigen presentation (TAP), its absence led to increased MHC class I expression in infected human cells. Moreover, some G47Delta-infected human melanoma cells exhibited enhanced stimulation of matched antitumor T cell activity. The deletion also places the late US11 gene under control of the immediate-early alpha47 promoter, which suppresses the reduced growth properties of gamma34.5-deficient mutants. G47Delta and R47Delta showed enhanced viral growth in a variety of cell lines, leading to higher virus yields and enhanced cytopathic effect in tumor cells. G47Delta was significantly more efficacious in vivo than its parent G207 at inhibiting tumor growth in both immune-competent and immune-deficient animal models. Yet, when inoculated into the brains of HSV-1-sensitive A/J mice at 2 x 10(6) plaque forming units, G47Delta was as safe as G207. These results suggest that G47Delta may have enhanced antitumor activity in humans.
Collapse
Affiliation(s)
- T Todo
- Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| | | | | | | |
Collapse
|
147
|
Van Tendeloo VF, Van Broeckhoven C, Berneman ZN. Gene therapy: principles and applications to hematopoietic cells. Leukemia 2001; 15:523-44. [PMID: 11368355 DOI: 10.1038/sj.leu.2402085] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Ever since the development of technology allowing the transfer of new genes into eukaryotic cells, the hematopoietic system has been an obvious and desirable target for gene therapy. The last 10 years have witnessed an explosion of interest in this approach to treat human disease, both inherited and acquired, with the initiation of multiple clinical protocols. All gene therapy strategies have two essential technical requirements. These are: (1) the efficient introduction of the relevant genetic material into the target cell and (2) the expression of the transgene at therapeutic levels. Conceptual and technical hurdles involved with these requirements are still the objects of active research. To date, the most widely used and best understood vectors for gene transfer in hematopoietic cells are derived from retroviruses, although they suffer from several limitations. However, as gene transfer mechanisms become more efficient and long-term gene expression is enhanced, the variety of diseases that can be tackled by gene therapy will continue to expand. However, until the problem of delivery and subsequent expression is adequately resolved, gene therapy will not realize its full potential. The first part of this review gives an overview of the gene delivery technology available at present to transfer genetic sequences in human somatic cells. The relevance of the hematopoietic system to the development of gene therapy strategies as well as hematopoietic cell-based gene therapy is discussed in the second part.
Collapse
Affiliation(s)
- V F Van Tendeloo
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp University Hospital, Belgium
| | | | | |
Collapse
|
148
|
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 immunocompetent hamsters. Mol Ther 2001; 3:431-7. [PMID: 11319903 DOI: 10.1006/mthe.2001.0303] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Gallbladder cancer is an extremely difficult disease to cure once metastases occur. In this paper, we explored the potential of G207, an oncolytic, replication-competent herpes simplex virus type 1 mutant, as a new therapeutic means for gallbladder cancer. Gallbladder carcinoma cell lines (four human and one hamster) showed nearly total cell killing within 72 h of G207 infection at a m.o.i. of 0.25 to 2.5 in vitro. The susceptibility to G207 cytopathic activity correlated with the infection efficiency demonstrated by lacZ expression. Intraneoplastic inoculation of G207 (1 x 10(7) pfu) in immunocompetent hamsters bearing established subcutaneous KIGB-5 tumors caused a significant inhibition of tumor growth and prolongation of survival. Repeated inoculations (three times with 4-day intervals) were significantly more efficacious than a single inoculation. In hamsters with bilateral subcutaneous KIGB-5 tumors, inoculation of one tumor alone with G207 caused regression or growth reduction of uninoculated tumors as well as inoculated tumors. In athymic mice, however, the anti-tumor effect was largely reduced in inoculated tumors and completely abolished in remote tumors, suggesting large contribution of T-cell-mediated immune responses to both local and systemic anti-tumor effect of G207. These results indicate that G207 may be useful as a new strategy for gallbladder cancer treatment.
Collapse
Affiliation(s)
- K Nakano
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | | | | | | |
Collapse
|
149
|
Wong RJ, Patel SG, Kim S, DeMatteo RP, Malhotra S, Bennett JJ, St-Louis M, Shah JP, Johnson PA, Fong Y. Cytokine gene transfer enhances herpes oncolytic therapy in murine squamous cell carcinoma. Hum Gene Ther 2001; 12:253-65. [PMID: 11177562 DOI: 10.1089/10430340150218396] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Replication-competent, attenuated herpes simplex viruses (HSV) have been demonstrated to be effective oncolytic agents in a variety of malignant tumors. Cytokine gene transfer has also been used as immunomodulatory therapy for cancer. To test the utility of combining these two approaches, two oncolytic HSV vectors (NV1034 and NV1042) were designed to express the murine GM-CSF and murine IL-12 genes, respectively. These cytokine-carrying variants were compared with the analogous non-cytokine-carrying control virus (NV1023) in the treatment of murine SCC VII squamous cell carcinoma. All three viruses demonstrated similar infection efficiency, viral replication, and cytotoxicity in vitro. SCC VII cells infected by NV1034 and NV1042 effectively produced GM-CSF and IL-12, respectively. In an SCC VII subcutaneous flank tumor model in immunocompetent C3H/HeJ mice, intratumoral injection with each virus caused a significant reduction in tumor volume compared with saline injections. The NV1042-treated tumors showed a striking reduction in tumor volume compared with the NV1023- and NV1034-treated tumors. On subsequent rechallenge in the contralateral flank with SCC VII cells, 57% of animals treated with NV1042 failed to develop tumors, in comparison with 14% of animals treated with NV1023 or NV1034, and 0% of naive animals. The increased antitumor efficacy seen with NV1042 in comparison with NV1023 and NV1034 was abrogated by CD4(+) and CD8(+) lymphocyte depletion. NV1042 is a novel, attenuated, oncolytic herpesvirus that effectively expresses IL-12 and elicits a T lymphocyte-mediated antitumor immune response against murine squamous cell carcinoma. Such combined oncolytic and immunomodulatory strategies hold promise in the treatment of cancer.
Collapse
Affiliation(s)
- R J Wong
- Head and Neck Division, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
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. [PMID: 11313808 DOI: 10.1038/sj.gt.3301392] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2000] [Accepted: 11/16/2000] [Indexed: 11/08/2022]
Abstract
We have used syngeneic, established bilateral subcutaneous tumor models to examine the antitumor activity of herpes simplex virus (HSV) vectors, including the induction of an immune response against non-inoculated distant tumors. In such a model with CT26 murine colon adenocarcinoma, unilateral intratumoral inoculation of replication-deficient HSV-1 tsK inhibited the growth of both the inoculated and noninoculated established tumors. To enhance this limited antitumor immune response, we generated a defective HSV vector, dvIL12-tk encoding both interleukin-12 (IL-12) and HSV thymidine kinase (TK), with tsK as the helper virus. In a 'suicide gene' strategy, ganciclovir (GCV) treatment after intratumoral inoculation of dvlacZ-tk/tsK, encoding E. coli lacZ instead of IL-12, resulted in enhanced antitumor activity. Antitumor activity was also enhanced by local expression of IL-12 from dvIL12-tk/tsK. The combination of IL-12 cytokine therapy with GCV treatment was the most efficacious approach, with significantly greater inhibition of tumor growth than IL-12 or TK + GCV alone. These results illustrate the power of combining different cancer therapy approaches; 'suicide gene' therapy, cytokine therapy, and HSV vector infection. HSV vectors are particularly well suited to this because they can accommodate the insertion of large and multiple gene sequences.
Collapse
Affiliation(s)
- M Toda
- Department of Neurosurgery, Georgetown University Medical Center, Washington, DC, USA
| | | | | |
Collapse
|