Primary breast tumors can shed millions of cells into the blood stream, known as circulating tumor cells (CTCs), at early stages of tumor progression. CTCs are able to survive in the circulation and arrest at distant sites to form metastatic disease. The presence of CTCs in a patient with breast cancer is associated with poor prognosis and increased risk of metastasis. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) has anti-tumor activity in breast cancer and is currently being evaluated in clinical trials as a target for breast cancer therapy. However, the role of AMPK in breast cancer metastasis is largely unknown. Most studies have focused on the function of AMPK as a cellular metabolic regulator, but AMPK has numerous other roles including maintaining epithelial cell polarity and regulating cytoskeletal proteins. Disseminated epithelial tumor cells use tubulin-based structures known as microtentacles to adhere to blood vessel walls in order to survive in the circulation and arrest in the capillaries of distant organs. There is a higher frequency of microtentacles in metastatic breast cancer cells, and microtentacles are enriched with a stable, detyrosinated form of a- tubulin. AMPK can phosphorylate the microtubule plus-end binding protein, clip-170, which results in decreased microtubule stability in normal epithelial cells. In this study, we will establish the role of AMPK in stabilizing microtubules in breast cancer. Furthermore, we will investigate the effects of clip-170 phosphorylation on microtubule stability and the metastatic efficiency of detached breast tumor cells. This study will test the hypothesis that AMPK activation decreases microtubule stability, resulting in decreased CTC attachment. We will test this hypothesis in the following specific aims: 1) Determine the extent of AMPK regulation of microtubule stability and microtentacle formation. 2) Define how AMPK phosphorylation of clip- 170 affects CTC attachment and metastasis. Current chemotherapeutics used in breast cancer therapy that target microtubules can enhance microtentacle formation and increase CTC re- attachment. Clip-170 provides a novel therapeutic target for microtubule stability that may reduce the metastatic success of CTCs or act as a biomarker for AMPK therapy. Completion of this study will help determine the therapeutic potential of AMPK-directed drugs to decrease the attachment of CTCs to blood vessels. Therefore, we can take advantage of the existing and developing pharmacological AMPK activators to decrease CTC metastasis and increase survival of patients with breast cancer.

Public Health Relevance

The presence of tumor cells in the blood, known as circulating tumor cells, is associated with an increased risk of developing distant spread of disease or metastasis, which is the main cause of death in patients with breast cancer. However, there are currently no treatments to specifically target circulating tumor cells to prevent metastatic disease. The goal of this study is to better understand the structural changes in circulating tumor cells that allow them to successfully metastasize in order develop new therapies to decrease metastasis and increase survival of patients with breast cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Individual Predoctoral NRSA for M.D./Ph.D. Fellowships (ADAMHA) (F30)
Project #
5F30CA196075-03
Application #
9330781
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Damico, Mark W
Project Start
2015-04-01
Project End
2019-03-31
Budget Start
2017-04-01
Budget End
2018-03-31
Support Year
3
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Maryland Baltimore
Department
Physiology
Type
Schools of Medicine
DUNS #
188435911
City
Baltimore
State
MD
Country
United States
Zip Code
21201
Ory, Eleanor C; Chen, Desu; Chakrabarti, Kristi R et al. (2017) Extracting microtentacle dynamics of tumor cells in a non-adherent environment. Oncotarget 8:111567-111580
Briggs, Joseph W; Ren, Ling; Chakrabarti, Kristi R et al. (2017) Activation of the unfolded protein response in sarcoma cells treated with rapamycin or temsirolimus. PLoS One 12:e0185089
Ory, Eleanor C; Bhandary, Lekhana; Boggs, Amanda E et al. (2017) Analysis of microtubule growth dynamics arising from altered actin network structure and contractility in breast tumor cells. Phys Biol 14:026005
Chakrabarti, Kristi R; Andorko, James I; Whipple, Rebecca A et al. (2016) Lipid tethering of breast tumor cells enables real-time imaging of free-floating cell dynamics and drug response. Oncotarget 7:10486-97
Chakrabarti, Kristi R; Hessler, Lindsay; Bhandary, Lekhana et al. (2015) Molecular Pathways: New Signaling Considerations When Targeting Cytoskeletal Balance to Reduce Tumor Growth. Clin Cancer Res 21:5209-5214
Thompson, Keyata N; Whipple, Rebecca A; Yoon, Jennifer R et al. (2015) The combinatorial activation of the PI3K and Ras/MAPK pathways is sufficient for aggressive tumor formation, while individual pathway activation supports cell persistence. Oncotarget 6:35231-46
Chakrabarti, Kristi R; Whipple, Rebecca A; Boggs, Amanda E et al. (2015) Pharmacologic regulation of AMPK in breast cancer affects cytoskeletal properties involved with microtentacle formation and re-attachment. Oncotarget 6:36292-307
Bhandary, Lekhana; Whipple, Rebecca A; Vitolo, Michele I et al. (2015) ROCK inhibition promotes microtentacles that enhance reattachment of breast cancer cells. Oncotarget 6:6251-66