Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Res ; 68(4): 1170-9, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18281493

ABSTRACT

Malignant solid tumors remain a significant clinical challenge, necessitating innovative therapeutic approaches. Oncolytic viral therapy is a nonmutagenic, biological anticancer therapeutic shown to be effective against human cancer in early studies. Because matrix metalloproteinases (MMP) play important roles in the pathogenesis and progression of cancer, we sought to determine if "arming" an oncolytic herpes simplex virus (oHSV) with an MMP-antagonizing transgene would increase virus-mediated antitumor efficacy. We generated oHSVs that express human tissue inhibitor of metalloproteinases 3 (TIMP3) or firefly luciferase and designated them rQT3 and rQLuc, respectively. We evaluated the antitumor efficacy of these viruses against neuroblastoma and malignant peripheral nerve sheath tumor (MPNST) xenografts. Relative to rQLuc, rQT3-infected primary human MPNST and neuroblastoma cells exhibited equivalent virus replication but increased cytotoxicity and reduced MMP activity. In vivo, rQT3-treated tumors showed delayed tumor growth, increased peak levels of infectious virus, immature collagen extracellular matrix, and reduced tumor vascular density. Remarkably, rQT3 treatment reduced circulating endothelial progenitors, suggesting virus-mediated antivasculogenesis. We conclude that rQT3 enhanced antitumor efficacy through multiple mechanisms, including direct cytotoxicity, elevated virus titer, and reduced tumor neovascularization. These findings support the further development of combined TIMP-3 and oncolytic virotherapy for cancer.


Subject(s)
Genetic Therapy/methods , Nerve Sheath Neoplasms/therapy , Neuroblastoma/therapy , Oncolytic Virotherapy/methods , Simplexvirus/physiology , Tissue Inhibitor of Metalloproteinase-3/genetics , Animals , Chlorocebus aethiops , Combined Modality Therapy , Female , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/virology , Humans , Luciferases, Firefly/genetics , Mice , Mice, Nude , Nerve Sheath Neoplasms/blood , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/virology , Neuroblastoma/blood , Neuroblastoma/genetics , Neuroblastoma/virology , Simplexvirus/genetics , Vero Cells , Xenograft Model Antitumor Assays
2.
Mol Ther ; 15(2): 279-86, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17235305

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs), driven in part by hyperactive Ras and epidermal growth factor receptor (EGFR) signaling, are often incurable. Testing of therapeutics for MPNST has been hampered by lack of adequate xenograft models. We previously documented that human MPNST cells are permissive for lytic infection by oncolytic herpes simplex viruses (oHSV). Herein we developed and characterized a xenograft model of human MPNST and evaluated the antitumor effects of oHSV mutants (G207 and hrR3) and the EGFR inhibitor, erlotinib. Additive cytotoxicity of these agents was found in human MPNST cell lines, suggesting that EGFR signaling is not critical for virus replication. Mice bearing human MPNST tumors treated with G207 or hrR3 by intraperitoneal or intratumoral injection showed tumor-selective virus biodistribution, virus replication, and reduced tumor burden. oHSV injection demonstrated more dramatic antitumor activity than erlotinib. Combination therapies showed a trend toward an increased antiproliferative effect. Both oHSV and erlotinib were antiangiogenic as measured by proangiogenic gene expression, effect on endothelial cells and xenograft vessel density. Overall, oHSVs showed highly potent antitumor effects against MPNST xenografts, an effect not diminished by EGFR inhibition. Our data suggest that inclusion of MPNSTs in clinical trials of oHSV is warranted.


Subject(s)
Neovascularization, Pathologic/prevention & control , Nerve Sheath Neoplasms/therapy , Quinazolines/pharmacology , Simplexvirus/genetics , Xenograft Model Antitumor Assays , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Genetic Therapy/methods , Humans , Immunoblotting , In Situ Hybridization , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Oncolytic Virotherapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Simplexvirus/metabolism , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL