Breast cancers are newly diagnosed in more than 200,000 American women annually. Breast cancer mortality is generally due to the occurrence of metastasis. The cyclooxygenase-2 enzyme (COX-2) is highly expressed in breast cancer and contributes to metastatic success. Selective COX-2 inhibitors limit breast tumor growth and metastasis, however, recently identified cardiac toxicities associated with global COX-2 inhibition prompted us to take a novel approach to target the COX-2 pathway. The chief COX-2 product in tumors is prostaglandin E2 (PGE2) which mediates biological effects by binding to each of four EP receptors (EP1-4). We have demonstrated that two of these receptors, EP1 and EP4, play opposing roles in modulating breast tumor metastasis. Pharmacologic antagonism or genetic silencing of EP4 reduces metastatic potential;antagonism of EP1 increases dissemination. We hypothesize that EP1 acts as a metastasis suppressor and that expression is decreased in women likely to develop disseminated disease. Furthermore, that EP1 controls metastasis by a mechanism dependent on Natural Killer cells. We will carry out the following Specific Objectives to test these hypotheses: Using a unique collection of early stage breast cancer specimens for which longterm survival is known we will (1) determine the relationship of EP1 expression to behavior in clinical breast cancer. Using a preclinical model, we will (2) determine the functional significance of EP1 and determine the contribution of Natural Killer cells to EP1 function. EP1 and other G-protein coupled receptors may mediate cellular effects in the nucleus as well as the plasma membrane. We will (3) identify the cellular location and function of EP1. Since all EP receptors bind the same ligand, altering the function of one receptor may adversely affect other EP receptor functions. We will (4) identify strategies to restore the balance of the protective (EP1) and the pro-metastatic (EP4) receptor. Potential Impact on Veterans Health Care: Breast cancer incidence increases with age and since the mean age of Veterans is 58, breast cancer will impact a significant number of Veterans in the coming years. The current Veteran population is approximately 26 million;of these, nearly 200,000 female Veterans will be diagnosed with breast cancer. Therefore, reduction in mortality from metastatic breast cancer would have a significant social and economic impact on Veterans Health Care. These studies will establish a rationale for modulating EP1 expression and function to achieve this goal.

Public Health Relevance

Relevance of the proposed work to the VA patient care mission. Cancers of the breast are newly diagnosed in nearly 200,000 Americans each year. Breast cancer has an enormous societal and health care impact and, due to the aging population, will become increasingly common. Given that the mean age of Veterans is 58, breast cancer will impact a significant number of Veterans in the next few years. The current Veteran population in the U.S. and Puerto Rico is approximately 26 million;of these, nearly 200,000 female Veterans will be diagnosed with breast cancer during their lifetimes. Reduction in mortality from this disease is an important goal. Breast cancer mortality is generally due to the occurrence of local and distant spread or metastasis. Our laboratory has identified several determinants of breast cancer metastasis. It is well established that the cyclooxygenase-2 (COX-2) enzyme is highly expressed and is associated with a poor prognosis in breast, colon, prostate and other malignancies. Recent concerns regarding the cardiac toxicity of COX-2 inhibitors prompted us to examine other approaches to therapeutically target the COX-2 pathway. We reasoned that preventing downstream signaling through the COX-2 product PGE2 would also be beneficial;however that hypothesis was only partially confirmed. We showed that breast tumors express all four known PGE receptors (EP1-4) and that antagonizing EP4 is beneficial whereas antagonism of EP1 actually promotes metastasis. Thus, the EP1 receptor appears to function as a metastasis suppressor. We will examine these relationships further in order to identify how to restore the balance between protective and anti-protective EP receptor function to reduce mortality from metastatic breast cancer.

Agency
National Institute of Health (NIH)
Institute
Veterans Affairs (VA)
Type
Non-HHS Research Projects (I01)
Project #
1I01BX000169-01
Application #
7682641
Study Section
Oncology A (ONCA)
Project Start
2009-04-01
Project End
2013-03-31
Budget Start
2009-04-01
Budget End
2010-03-31
Support Year
1
Fiscal Year
2009
Total Cost
Indirect Cost
Name
Baltimore VA Medical Center
Department
Type
DUNS #
796532609
City
Baltimore
State
MD
Country
United States
Zip Code
21201
Kochel, Tyler J; Goloubeva, Olga G; Fulton, Amy M (2016) Upregulation of Cyclooxygenase-2/Prostaglandin E2 (COX-2/PGE2) Pathway Member Multiple Drug Resistance-Associated Protein 4 (MRP4) and Downregulation of Prostaglandin Transporter (PGT) and 15-Prostaglandin Dehydrogenase (15-PGDH) in Triple-Negative Breast Breast Cancer (Auckl) 10:61-70
Bhooshan, Neha; Staats, Paul N; Fulton, Amy M et al. (2016) Prostaglandin E Receptor EP4 expression, survival and pattern of recurrence in locally advanced NSCLC. Lung Cancer 101:88-91
Clark, David J; Fondrie, William E; Liao, Zhongping et al. (2015) Redefining the Breast Cancer Exosome Proteome by Tandem Mass Tag Quantitative Proteomics and Multivariate Cluster Analysis. Anal Chem 87:10462-9
Kochel, Tyler J; Fulton, Amy M (2015) Multiple drug resistance-associated protein 4 (MRP4), prostaglandin transporter (PGT), and 15-hydroxyprostaglandin dehydrogenase (15-PGDH) as determinants of PGE2 levels in cancer. Prostaglandins Other Lipid Mediat 116-117:99-103
Kundu, Namita; Ma, Xinrong; Kochel, Tyler et al. (2014) Prostaglandin E receptor EP4 is a therapeutic target in breast cancer cells with stem-like properties. Breast Cancer Res Treat 143:19-31
Ma, Xinrong; Holt, Dawn; Kundu, Namita et al. (2013) A prostaglandin E (PGE) receptor EP4 antagonist protects natural killer cells from PGE2-mediated immunosuppression and inhibits breast cancer metastasis. Oncoimmunology 2:e22647
Holt, Dawn M; Ma, Xinrong; Kundu, Namita et al. (2012) Modulation of host natural killer cell functions in breast cancer via prostaglandin E2 receptors EP2 and EP4. J Immunother 35:179-88
Kundu, Namita; Campbell, Patricia; Hampton, Brian et al. (2012) Antimetastatic activity isolated from Colocasia esculenta (taro). Anticancer Drugs 23:200-11
Ma, Xinrong; Kundu, Namita; Collin, Peter D et al. (2012) Frondoside A inhibits breast cancer metastasis and antagonizes prostaglandin E receptors EP4 and EP2. Breast Cancer Res Treat 132:1001-8
Reader, Jocelyn; Holt, Dawn; Fulton, Amy (2011) Prostaglandin E2 EP receptors as therapeutic targets in breast cancer. Cancer Metastasis Rev 30:449-63

Showing the most recent 10 out of 13 publications