Malignant progression and poor survival in glioma and many other solid malignancies are linked with over-expression of the transmembraneous receptor tyrosine kinase c-Met and its secreted ligand hepatocyte growth factor (HGF). HGF and c-Met are increasingly seen as promising therapeutic targets due to these clinical associations and to their multifunctional autocrine and paracrine tumor promoting activities that include the direct stimulation of tumor cell growth, tumor cell resistance to cytotoxic therapy, tumor cell migration/invasion, and tumor angiogenesis. We have shown that the neutralizing anti- HGF monoclonal antibody (L2G7) specifically and potently blocks HGF functions and inhibits the growth of HGF-expressing orthotopic human glioma xenografts, including primary glioma xenografts, and prolonged animal survival. L2G7 inhibits glioma Akt and MAP-kinase activity, tumor angiogenesis, proliferation index, and increases tumor apoptosis index. Numerous questions must be answered in order to optimally develop anti-HGF mAb therapy for malignant brain tumors. Unanswered questions include -What is the best route of L2G7 delivery? What are the benefits of combining L2G7 with standard cytotoxic modalities? Can combination therapies be optimized based on the dynamics of HGF:c-Met pathway inhibition? Can we identify subsets of gliomas that are more or less likely to respond to L2G7 therapy? This research will use innovative xenograft model systems, and an array of complementary molecular, biological, and imaging tumor response endpoints to answer these and related questions.
Our specific aims are: (1) To determine the relative efficacy of direct intratumoral convection enhanced delivery vs. systemic delivery of anti-HGF HuL2G7 in glioma xenografts, (2) To determine if HuL2G7 enhances the efficacy of radiation and/or chemotherapy in glioma xenografts, and (3) To identify clinically translatable molecular markers for predicting glioma sensitivity/resistance to HGF:c-Met pathway inhibition. Positive results will elucidate mechanisms of anti- HGF therapeutics, dosing schemes, and molecular/biological response criteria necessary for optimally developing anti-HGF L2G7 and potentially other HGF:c-Met pathway inhibitors .for CNS cancer. Findings will also contribute substantially to the application of other antibody-based therapeutics to brain cancer. Hepatocyte growth factor (HGF) is a promising therapeutic target for the most common primary CNS malignancies such as glioblastoma and medulloblastoma and brain metastases. This research will examine how to best use a neutralizing anti-HGF monoclonal antibody to inhibit experimental brain tumor growth. Histological, biochemical, and genetic markers for predicting tumor sensitivity and assessing anti-tumor responses to HGF inhibition will also be determined. Results will impact on how HGF inhibitors are used in brain tumor patients. ? ? ?

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA129192-01
Application #
7299276
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Yovandich, Jason L
Project Start
2007-08-01
Project End
2011-07-30
Budget Start
2007-08-01
Budget End
2008-07-31
Support Year
1
Fiscal Year
2007
Total Cost
$305,829
Indirect Cost
Name
Hugo W. Moser Research Institute Kennedy Krieger
Department
Type
DUNS #
155342439
City
Baltimore
State
MD
Country
United States
Zip Code
21205
Goodwin, C Rory; Woodard, Crystal L; Zhou, Xin et al. (2016) Microarray-Based Phospho-Proteomic Profiling of Complex Biological Systems. Transl Oncol 9:124-129
Woodard, Crystal L; Goodwin, C Rory; Wan, Jun et al. (2013) Profiling the dynamics of a human phosphorylome reveals new components in HGF/c-Met signaling. PLoS One 8:e72671
Rath, Prakash; Lal, Bachchu; Ajala, Olutobi et al. (2013) In Vivo c-Met Pathway Inhibition Depletes Human Glioma Xenografts of Tumor-Propagating Stem-Like Cells. Transl Oncol 6:104-11
Li, Yunqing; Laterra, John (2012) Cancer stem cells: distinct entities or dynamically regulated phenotypes? Cancer Res 72:576-80
Li, Yunqing; Li, Angela; Glas, Martin et al. (2011) c-Met signaling induces a reprogramming network and supports the glioblastoma stem-like phenotype. Proc Natl Acad Sci U S A 108:9951-6
Goodwin, C Rory; Lal, Bachchu; Ho, Sandra et al. (2011) PTEN reconstitution alters glioma responses to c-Met pathway inhibition. Anticancer Drugs 22:905-12
Goodwin, C Rory; Laterra, John (2010) Neuro-oncology: unmasking the multiforme in glioblastoma. Nat Rev Neurol 6:304-5
Goodwin, C Rory; Lal, Bachchu; Zhou, Xin et al. (2010) Cyr61 mediates hepatocyte growth factor-dependent tumor cell growth, migration, and Akt activation. Cancer Res 70:2932-41
Coon, Valerie; Laukert, Tamara; Pedone, Carolyn A et al. (2010) Molecular therapy targeting Sonic hedgehog and hepatocyte growth factor signaling in a mouse model of medulloblastoma. Mol Cancer Ther 9:2627-36
Lal, Bachchu; Goodwin, C Rory; Sang, Yingying et al. (2009) EGFRvIII and c-Met pathway inhibitors synergize against PTEN-null/EGFRvIII+ glioblastoma xenografts. Mol Cancer Ther 8:1751-60

Showing the most recent 10 out of 12 publications