This application proposes the fourth renewal of the UNC Breast Cancer SPORE. Originally funded in 1992, the UNC SPORE has used significant institutional investment and the distinctive SPORE elements: funding flexibility, interdisciplinary collaboration, bidirectional translational research, developmental programs, and interSPORE partners to build an outstanding program in translational breast cancer research. The UNC SPORE'S combination of population-based research, molecular genetics, breast cancer biology, health disparities research, tissue-acquiring clinical trials, database development, and expertise in bioinformatics and biostatistics was made possible with long-term SPORE support. Interaction between disciplines has resulted in substantial productivity, as measured by important findings published in high impact journals, career advancement for junior investigators, developmental research leading to grants and new SPORE projects, multiple funded interSPORE collaborations, and innovative approaches to breast cancer etiology, classification, prognosis, and therapy. Our projects are conceptually linked by studies of breast cancer molecular intrinsic subtypes, particularly the triple negative breast cancers, basal-like cancer and a newly-defined subtype, Claudin low. We feature a nation leading population science study of breast cancer and minority disparities, the Carolina Breast Cancer Study (CBCS), entering its 20th year of SPORE funding. Our molecular genetics translational group is developing new technology for intrinsic subtyping with which to analyze clinically-annotated samples from CBCS and local, national, and international trials. Novel translational studies of gene expression, tumor microevironment and a systems approach to the kinome will utilize both preclinical models and human tumors. The projects are entitled: 1) Carolina Breast Cancer Study: Genomic and epidemiologic determinants of breast cancer minority disparities. 2) Targeting the infiltrating immune cells in Claudin-low tumors. 3) Development and validation of predictive markers for triple negative breast cancers. 4) Stromal response subtypes in breast cancer risk and progression 5) Quantitative proteomic analysis of the human kinome in Claudin-low and basal-like breast cancer Five of the co-Project leaders are products of the SPORE career development program. Two of the projects resulted from our SPORE Developmental Research Program. The SPORE is supported by exceptional infrastructure and institutional commitment from the UNC Lineberger Comprehensive Cancer Center and three SPORE-funded cores: Pathology, Genomics, Biostatistics & Bioinformatics, and Administration.

Public Health Relevance

The UNC Breast Cancer SPORE contributes to the clinical application of molecular subtyping to breast cancer discovery, prognostication, conduct of clinical trials, and understanding of minority disparities in breast cancer. Our projects continue to explore the effect of intrinsic subtype on the predisposition, progression, and therapeutic resistance of difficult-to-treat breast cancers. UNO SPORE's technology development, population-based studies, and analysis of clinical trials will have substantial translational impact.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
5P50CA058223-22
Application #
8919987
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Rohan, Joyann
Project Start
1997-08-05
Project End
2016-08-31
Budget Start
2015-09-01
Budget End
2016-08-31
Support Year
22
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of North Carolina Chapel Hill
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
608195277
City
Chapel Hill
State
NC
Country
United States
Zip Code
27599
Williams, Michelle M; Lee, Linus; Werfel, Thomas et al. (2018) Intrinsic apoptotic pathway activation increases response to anti-estrogens in luminal breast cancers. Cell Death Dis 9:21
Allott, Emma H; Geradts, Joseph; Cohen, Stephanie M et al. (2018) Frequency of breast cancer subtypes among African American women in the AMBER consortium. Breast Cancer Res 20:12
Matsunuma, Ryoichi; Chan, Doug W; Kim, Beom-Jun et al. (2018) DPYSL3 modulates mitosis, migration, and epithelial-to-mesenchymal transition in claudin-low breast cancer. Proc Natl Acad Sci U S A 115:E11978-E11987
Panda, Anshuman; de Cubas, Aguirre A; Stein, Mark et al. (2018) Endogenous retrovirus expression is associated with response to immune checkpoint blockade in clear cell renal cell carcinoma. JCI Insight 3:
Sharma, Priyanka; López-Tarruella, Sara; García-Saenz, José Angel et al. (2018) Pathological Response and Survival in Triple-Negative Breast Cancer Following Neoadjuvant Carboplatin plus Docetaxel. Clin Cancer Res 24:5820-5829
Siegel, Marni B; He, Xiaping; Hoadley, Katherine A et al. (2018) Integrated RNA and DNA sequencing reveals early drivers of metastatic breast cancer. J Clin Invest 128:1371-1383
Kumar, Sunil; Lindsay, Daniel; Chen, Q Brent et al. (2018) Tracking plasma DNA mutation dynamics in estrogen receptor positive metastatic breast cancer with dPCR-SEQ. NPJ Breast Cancer 4:39
Smith, Christof C; Beckermann, Kathryn E; Bortone, Dante S et al. (2018) Endogenous retroviral signatures predict immunotherapy response in clear cell renal cell carcinoma. J Clin Invest 128:4804-4820
Wheeler, Stephanie B; Spencer, Jennifer C; Pinheiro, Laura C et al. (2018) Financial Impact of Breast Cancer in Black Versus White Women. J Clin Oncol 36:1695-1701
Hong, Chi-Chen; Sucheston-Campbell, Lara E; Liu, Song et al. (2018) Genetic Variants in Immune-Related Pathways and Breast Cancer Risk in African American Women in the AMBER Consortium. Cancer Epidemiol Biomarkers Prev 27:321-330

Showing the most recent 10 out of 598 publications