Genetic modification of neurons to express bevacizumab for local anti-angiogenesis treatment of glioblastoma.

TitleGenetic modification of neurons to express bevacizumab for local anti-angiogenesis treatment of glioblastoma.
Publication TypeJournal Article
Year of Publication2015
AuthorsHicks MJ, Funato K, Wang L, Aronowitz E, Dyke JP, Ballon DJ, Havlicek DF, Frenk EZ, De BP, Chiuchiolo MJ, Sondhi D, Hackett NR, Kaminsky SM, Tabar V, Crystal RG
JournalCancer Gene Ther
Volume22
Issue1
Pagination1-8
Date Published2015 Jan
ISSN1476-5500
KeywordsAnimals, Antibodies, Monoclonal, Humanized, Bevacizumab, Brain, Dependovirus, Disease Models, Animal, Female, Gene Expression, Genetic Vectors, Glioblastoma, Humans, Magnetic Resonance Imaging, Mice, Neovascularization, Pathologic, Neurons, Transduction, Genetic, Tumor Burden, Xenograft Model Antitumor Assays
Abstract

The median survival of glioblastoma multiforme (GBM) is approximately 1 year. Following surgical removal, systemic therapies are limited by the blood-brain barrier. To circumvent this, we developed a method to modify neurons with the genetic sequence for therapeutic monoclonal antibodies using adeno-associated virus (AAV) gene transfer vectors, directing persistent, local expression in the tumor milieu. The human U87MG GBM cell line or patient-derived early passage GBM cells were administered to the striatum of NOD/SCID immunodeficient mice. AAVrh.10BevMab, an AAVrh.10-based vector coding for bevacizumab (Avastin), an anti-human vascular endothelial growth factor (VEGF) monoclonal antibody, was delivered to the area of the GBM xenograft. Localized expression of bevacizumab was demonstrated by quantitative PCR, ELISA and western blotting. Immunohistochemistry showed that bevacizumab was expressed in neurons. Concurrent administration of AAVrh.10BevMab with the U87MG tumor reduced tumor blood vessel density and tumor volume, and increased survival. Administration of AAVrh.10BevMab 1 week after U87MG xenograft reduced growth and increased survival. Studies with patient-derived early passage GBM primary cells showed a reduction in primary tumor burden with an increased survival. These data support the strategy of AAV-mediated central nervous system gene therapy to treat GBM, overcoming the blood-brain barrier through local, persistent delivery of an anti-angiogenesis monoclonal antibody.

DOI10.1038/cgt.2014.58
Alternate JournalCancer Gene Ther
PubMed ID25501993
PubMed Central IDPMC4293257
Grant ListT32 HL094284 / HL / NHLBI NIH HHS / United States
T32HL094284 / HL / NHLBI NIH HHS / United States
Related Institute: 
MRI Research Institute (MRIRI)

Weill Cornell Medicine
Department of Radiology
525 East 68th Street New York, NY 10065