? Project 7 Use of metformin, a medicine with a 40-year track record of safety and efficacy for the treatment of diabetes and other non-malignant conditions, has been associated with improved cancer survival in observational studies. Our groups at the University of Chicago (U of C) and the Mayo Clinic (Mayo) have independently shown that ovarian cancer (OvCa) patients with diabetes who are treated with metformin have an improved prognosis. Our preclinical studies, using OvCa cell lines and mouse models, showed that metformin induces metabolic alterations in OvCa cells, inhibits interactions between cancer cells and stromal cells in the tumor microenvironment, and slows growth of both genetically engineered and xenograft models of OvCa in vivo. In view of these findings, the primary hypothesis underlying this application is that metformin can be used as a cancer therapeutic in patients with OvCa.
In Aim 1, we will evaluate the effect of metformin on cells in the tumor microenvironment and on functional aspects of metabolism (glycolysis, glycogen storage, lipid homeostasis, insulin signaling).
In Aim 2, we will use patient-derived xenograft models (Avatars) to evaluate the effect of metformin alone and in combination with chemotherapy on tumor burden. To assess the impact of diabetes on the action of metformin in OvCa, this experiment will be performed in both normoglycemic and obese/hyperglycemic mice. To better understand the metformin mechanism of action in OvCa, we will utilize proteomic profiling to identify the key oncogenic signaling pathways affected by metformin treatment in the patient-derived xenografts.
In Aim 3, we will examine pharmacodynamic readouts and explore possible predictive biomarkers of metformin's effect with chemotherapy using biospecimens from our ongoing randomized phase II trial of chemotherapy metformin. As part of the SPORE, prospectively collected serum and tissue samples, including paired samples collected before and after neo-adjuvant chemotherapy metformin, will be used to study the effect of metformin on protein expression using shotgun proteomics followed by label-free quantification. The goal of Aim 3 is to (a) identify proteins and pathways that are altered with metformin treatment to guide research aimed at understanding the mechanism of metformin action in patients and (b) assess whether a proteomic signature in the tumor at the time of diagnosis predicts a beneficial effect of metformin treatment. Collectively, these studies will not only elucidate the mechanistic basis for metformin's effects on cancer metabolism and the microenvironment, but also provide the first prospective assessment of whether adding metformin to chemotherapy improves progression-free survival in OvCa patients.

Public Health Relevance

The cure rate for ovarian cancer has not changed in decades; thus, new ideas for therapies for ovarian cancer are much-needed. It is increasingly recognized that ovarian cancer cells develop unique metabolic adaptations that allow them to survive in the face of chemotherapy. This project team studies the metabolism of ovarian cancer and has shown how metformin, an already available metabolic agent, can block the pro-tumorigenic metabolic adaptations of ovarian cancer cells.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
2P50CA136393-06A1
Application #
8932130
Study Section
Special Emphasis Panel (ZCA1-RPRB-C (M1))
Project Start
2008-10-01
Project End
2020-08-31
Budget Start
2015-09-11
Budget End
2016-08-31
Support Year
6
Fiscal Year
2015
Total Cost
$337,376
Indirect Cost
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Li, Lei; Liu, Tongzheng; Li, Yunhui et al. (2018) The deubiquitinase USP9X promotes tumor cell survival and confers chemoresistance through YAP1 stabilization. Oncogene 37:2422-2431
Lu, Yingchang; Beeghly-Fadiel, Alicia; Wu, Lang et al. (2018) A Transcriptome-Wide Association Study Among 97,898 Women to Identify Candidate Susceptibility Genes for Epithelial Ovarian Cancer Risk. Cancer Res 78:5419-5430
Wu, Dongyan; Yang, Haitao; Winham, Stacey J et al. (2018) Mediation analysis of alcohol consumption, DNA methylation, and epithelial ovarian cancer. J Hum Genet 63:339-348
Painter, Jodie N; O'Mara, Tracy A; Morris, Andrew P et al. (2018) Genetic overlap between endometriosis and endometrial cancer: evidence from cross-disease genetic correlation and GWAS meta-analyses. Cancer Med 7:1978-1987
Wahner Hendrickson, Andrea E; Menefee, Michael E; Hartmann, Lynn C et al. (2018) A Phase I Clinical Trial of the Poly(ADP-ribose) Polymerase Inhibitor Veliparib and Weekly Topotecan in Patients with Solid Tumors. Clin Cancer Res 24:744-752
Natanzon, Yanina; Goode, Ellen L; Cunningham, Julie M (2018) Epigenetics in ovarian cancer. Semin Cancer Biol 51:160-169
Knijnenburg, Theo A; Wang, Linghua; Zimmermann, Michael T et al. (2018) Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep 23:239-254.e6
Jung, DeokBeom; Khurana, Ashwani; Roy, Debarshi et al. (2018) Quinacrine upregulates p21/p27 independent of p53 through autophagy-mediated downregulation of p62-Skp2 axis in ovarian cancer. Sci Rep 8:2487
Liu, Gang; Mukherjee, Bhramar; Lee, Seunggeun et al. (2018) Robust Tests for Additive Gene-Environment Interaction in Case-Control Studies Using Gene-Environment Independence. Am J Epidemiol 187:366-377
Ong, Jue-Sheng; Hwang, Liang-Dar; Cuellar-Partida, Gabriel et al. (2018) Assessment of moderate coffee consumption and risk of epithelial ovarian cancer: a Mendelian randomization study. Int J Epidemiol 47:450-459

Showing the most recent 10 out of 294 publications