Activation of an invasive program is a critical event in the multi-step process of tumor metastasis driven by the activation of tyrosine kinases and ligand-stimulated chemokine receptors. We now show that the Abl family of nonreceptor tyrosine kinases, Abl (Abl1) and Arg (Abl2), which are activated downstream of multiple receptor tyrosine kinases (RTKs), are also activated by chemokine receptors and play a critical role in the regulation of cancer cell invasion. We show that Abl kinases are required for epithelial cancer cell invasion and matrix degradation and identify a novel signaling pathway that links the Abl kinases to the regulation of the matrix metalloproteinase MT1-MMP in breast cancer cells. Moreover, we found that endogenous Abl kinases are hyperactivated in a subset of breast cancer cell lines that are negative for Her2 and hormone receptors. Further, expression of activated Abl kinases elicits a striking disruption of epithelial cell polarity which is associated with cancer progression. Based on these findings, we hypothesize that Abl kinases are required for metastasis of a subset of epithelial tumors through regulation of invasive programs and thus combination therapies that target inhibition of Abl kinases might be exploited for the treatment of a subset of invasive breast tumors. To this end we propose the following aims: 1) Identify the phosphoproteomic signature for activated Abl kinases in human breast cancer cells. Knowledge of the molecular signature induced by activated Abl kinases in breast tumors will allow for identification of patients that might benefit from targeted therapy with approved and novel Abl kinase inhibitors;2) Elucidate the mechanisms employed by Abl kinases to regulate breast cancer cell invasion;and 3) Define the role of Abl kinases in mammary tumor progression and metastasis using mouse models. Together these aims will uncover Abl-dependent signaling networks that regulate invasive programs that drive cancer tumor progression and metastasis. The long-term goal of these studies is to develop novel therapeutic approaches with greater specificity and reduced toxicity than those provided by current therapies for the treatment of specific tumor subtypes. Inhibition of the Abl kinases is expected to simultaneously block multiple signaling pathways required for tumor invasion that converge on the activation of these unique kinases.

Public Health Relevance

Disruption of cell polarity and activation of an invasive program are critical events in the multi-step process of tumor metastasis. We have identified the Abl family of nonreceptor tyrosine kinases as critical regulators of cell polarity and cancer cell invasion. We found that Abl kinases, which are activated downstream of multiple RTKs, are also activated downstream of chemokine receptors in breast cancer cells and are required for cancer cell invasion and matrix degradation. Results from the proposed studies have wide-ranging implications for the development of novel therapeutic approaches with greater specificity and reduced toxicity than current therapies for the treatment of invasive solid tumors. Inhibition of the Abl kinases will result in the simultaneous block of multiple signaling pathways required for the invasion of epithelial tumors driven by the activation of RTKs and/or chemokines that converge on the activation of these unique kinases.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA155160-03
Application #
8458615
Study Section
Tumor Progression and Metastasis Study Section (TPM)
Program Officer
Snyderwine, Elizabeth G
Project Start
2011-07-21
Project End
2016-04-30
Budget Start
2013-05-01
Budget End
2014-04-30
Support Year
3
Fiscal Year
2013
Total Cost
$302,232
Indirect Cost
$107,182
Name
Duke University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
044387793
City
Durham
State
NC
Country
United States
Zip Code
27705
Wang, Jun; Rouse, Clay; Jasper, Jeff S et al. (2016) ABL kinases promote breast cancer osteolytic metastasis by modulating tumor-bone interactions through TAZ and STAT5 signaling. Sci Signal 9:ra12
Khatri, Aaditya; Wang, Jun; Pendergast, Ann Marie (2016) Multifunctional Abl kinases in health and disease. J Cell Sci 129:9-16
Matsumoto, Yoshinori; La Rose, Jose; Kent, Oliver A et al. (2016) Reciprocal stabilization of ABL and TAZ regulates osteoblastogenesis through transcription factor RUNX2. J Clin Invest 126:4482-4496
Li, Ran; Knight, Jennifer F; Park, Morag et al. (2015) Abl Kinases Regulate HGF/Met Signaling Required for Epithelial Cell Scattering, Tubulogenesis and Motility. PLoS One 10:e0124960
Wang, Jun; Pendergast, Ann Marie (2015) The Emerging Role of ABL Kinases in Solid Tumors. Trends Cancer 1:110-123
Sourbier, Carole; Ricketts, Christopher J; Matsumoto, Shingo et al. (2014) Targeting ABL1-mediated oxidative stress adaptation in fumarate hydratase-deficient cancer. Cancer Cell 26:840-850
Chislock, Elizabeth M; Pendergast, Ann Marie (2013) Abl family kinases regulate endothelial barrier function in vitro and in mice. PLoS One 8:e85231
Chislock, Elizabeth M; Ring, Colleen; Pendergast, Ann Marie (2013) Abl kinases are required for vascular function, Tie2 expression, and angiopoietin-1-mediated survival. Proc Natl Acad Sci U S A 110:12432-7
Greuber, Emileigh K; Smith-Pearson, Pameeka; Wang, Jun et al. (2013) Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nat Rev Cancer 13:559-71
Chislock, Elizabeth M; Pendergast, Ann Marie (2013) Tie2 (to) Abl: Signaling to endothelial cell survival. Cell Cycle 12:3709-10

Showing the most recent 10 out of 13 publications