HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Targeting the c-Met pathway potentiates glioblastoma responses to gamma-radiation.

AbstractPURPOSE:
Resistance to current cytotoxic therapies limits the treatment of most solid malignancies. This results, in part, from the overactivation of receptor tyrosine kinases and their downstream pathways in tumor cells and their associated vasculature. In this report, we ask if targeting the multifunctional mitogenic, cytoprotective, and angiogenic scatter factor/hepatocyte growth factor (SF/HGF)/c-Met pathway potentiates antitumor responses to gamma-radiation.
EXPERIMENTAL DESIGN:
Endogenous expression of SF/HGF and c-Met was targeted in U87 MG human malignant glioma cells and xenografts using chimeric U1/ribozymes. The effects of U1/ribozymes +/- gamma-radiation on glioma cell proliferation, apoptosis, xenograft growth, and animal survival were examined.
RESULTS:
U1/ribozymes knocked down SF/HGF and c-Met mRNA and protein levels, sensitized cells to gamma-radiation (P < 0.005), and enhanced radiation-induced caspase-dependent cytotoxicity in vitro (P < 0.005). Intravenous U1/ribozyme therapy as liposome/DNA complexes or radiation alone modestly and transiently inhibited the growth of s.c. U87 xenografts. Combining the therapies caused tumor regression and a 40% tumor cure rate. In animals bearing intracranial xenografts, long-term survival was 0% in response to radiation, 20% in response to intratumoral adenoviral-based U1/ribozyme delivery, and 80% (P < 0.0005) in response to combining U1/ribozymes with radiation. This apparent synergistic antitumor response was associated with a approximately 70% decrease in cell proliferation (P < 0.001) and a approximately 14- to 40-fold increase in apoptosis (P < 0.0001) within xenografts.
CONCLUSIONS:
Targeting the SF/HGF/c-Met pathway markedly potentiates the anti-glioma response to gamma-radiation. Clinical trials using novel SF/HGF/c-Met pathway inhibitors in glioma and other malignancies associated with c-Met activation should ultimate include concurrent radiation and potentially other cytotoxic therapeutics.
AuthorsBachchu Lal, Shuli Xia, Roger Abounader, John Laterra
JournalClinical cancer research : an official journal of the American Association for Cancer Research (Clin Cancer Res) Vol. 11 Issue 12 Pg. 4479-86 (Jun 15 2005) ISSN: 1078-0432 [Print] United States
PMID15958633 (Publication Type: Journal Article, Research Support, N.I.H., Extramural, Research Support, U.S. Gov't, P.H.S.)
Chemical References
  • Ki-67 Antigen
  • RNA, Catalytic
  • Hepatocyte Growth Factor
  • Proto-Oncogene Proteins c-met
Topics
  • Animals
  • Apoptosis (genetics, radiation effects)
  • Blotting, Northern
  • Cell Line, Tumor
  • Gamma Rays
  • Gene Expression Regulation, Neoplastic
  • Genetic Vectors (genetics)
  • Glioblastoma (genetics, pathology, radiotherapy)
  • Hepatocyte Growth Factor (genetics, metabolism)
  • Humans
  • Immunoblotting
  • Immunohistochemistry
  • In Situ Nick-End Labeling
  • Ki-67 Antigen (analysis)
  • Mice
  • Mice, Nude
  • Proto-Oncogene Proteins c-met (genetics, metabolism)
  • RNA, Catalytic (genetics, metabolism)
  • Xenograft Model Antitumor Assays (methods)

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: