Lung cancer is the leading cause of cancer deaths in both men and women, with the majority of cases being classified as non-small cell lung cancer (NSCLC). Since at time of diagnosis, the majority of lung cancers are already advanced, new strategies are required to target tumor progression and metastasis. Peroxisome proliferator-activated receptor-? (PPAR?), a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors, plays a critical role in adipocyte activation, but has also been implicated in a variety of cancers. During the previous funding period we demonstrated that activation of PPAR? has pleiotropic effects on human NSCLC cells, including promotion of a differentiated phenotype and inhibition of transformed growth and invasiveness. This receptor is the target of thiazolidinediones (TZDs), such as rosiglitazone and pioglitazone. Recent retrospective studies have suggested that these agents specifically reduce the risk of developing lung cancer in patients, making PPAR? an attractive target for treatment of lung cancer. Cancer progression and metastasis require complex interactions between tumor cells and the surrounding tumor microenvironment (TME). We have developed an immunocompetent mouse model for lung cancer cells progression, in which mouse lung cancer cells are injected into the lungs of syngeneic mice. These cells form a primary tumor which metastasizes to the other lobes of the lung, lymph nodes, and distant organs including liver and brain. This model allows manipulation of specific genes in either tumor cells or the TME. Using this model we have unexpectedly shown that in contrast to effects on tumor cells, systemic administration of pioglitazone accelerates tumor progression and promotes metastases. We hypothesize that these disparate effects of PPAR? activation are a result of opposing effects in tumor cells vs. the TME, with activation in tumor cells inhibiting progression and activation in cells of the TME contributing to progression. Our preliminary data indicate that pioglitazone affects the distribution of tumor-associated macrophages and may promote tumor angiogenesis. This project will use in vivo models and in vitro studies to define the contribution of PPAR? in each cell type to tumor progression and metastasis.
Three specific aims are proposed.
Aim 1 will examine the specific role of PPAR? activation in tumor cells on cancer progression, and define effector pathways in human and murine NSCLC cells.
Aim 2 will assess the role of PPAR? in macrophages using a targeted knockout strategy. Studies will use in vitro systems to define cross-talk between cancer cells and macrophages.
Aim 3 will employ a similar targeted knockout strategy to assess the role of PPAR? in endothelial cell on tumor progression in the setting of pioglitazone. Since large numbers of patients are treated with TZDs, defining the role of these agents and PPAR? on cancer progression and metastasis is of critical importance.

Public Health Relevance

Studies indicate that activators of PPAR? can inhibit initiation of lung cancer but have conflicting roles on cancer progression and metastasis. This project will define the role for this protein both in tumor cells, macrophages, and endothelial cells using both in vivo and in vitro approaches. Understanding the cell-specific mechanisms whereby PPAR? regulates cancer progression and metastasis is critical in light of the wide-spread use of these agents, and will lead to identification of novel targets and new therapeutic approaches.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA108610-07
Application #
8206507
Study Section
Tumor Microenvironment Study Section (TME)
Program Officer
Sathyamoorthy, Neeraja
Project Start
2004-08-05
Project End
2015-11-30
Budget Start
2011-12-01
Budget End
2012-11-30
Support Year
7
Fiscal Year
2012
Total Cost
$297,398
Indirect Cost
$103,020
Name
University of Colorado Denver
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
041096314
City
Aurora
State
CO
Country
United States
Zip Code
80045
Li, Howard Y; McSharry, Maria; Walker, Deandra et al. (2018) Targeted overexpression of prostacyclin synthase inhibits lung tumor progression by recruiting CD4+ T lymphocytes in tumors that express MHC class II. Oncoimmunology 7:e1423182
Kwak, Jeff W; Laskowski, Jennifer; Li, Howard Y et al. (2018) Complement Activation via a C3a Receptor Pathway Alters CD4+ T Lymphocytes and Mediates Lung Cancer Progression. Cancer Res 78:143-156
Li, Howard Y; McSharry, Maria; Bullock, Bonnie et al. (2017) The Tumor Microenvironment Regulates Sensitivity of Murine Lung Tumors to PD-1/PD-L1 Antibody Blockade. Cancer Immunol Res 5:767-777
Poczobutt, Joanna M; De, Subhajyoti; Yadav, Vinod K et al. (2016) Expression Profiling of Macrophages Reveals Multiple Populations with Distinct Biological Roles in an Immunocompetent Orthotopic Model of Lung Cancer. J Immunol 196:2847-59
Poczobutt, Joanna M; Nguyen, Teresa T; Hanson, Dwight et al. (2016) Deletion of 5-Lipoxygenase in the Tumor Microenvironment Promotes Lung Cancer Progression and Metastasis through Regulating T Cell Recruitment. J Immunol 196:891-901
Fernandez-Boyanapalli, Ruby F; Frasch, S Courtney; Thomas, Stacey M et al. (2015) Pioglitazone restores phagocyte mitochondrial oxidants and bactericidal capacity in chronic granulomatous disease. J Allergy Clin Immunol 135:517-527.e12
Marek, Lindsay A; Hinz, Trista K; von Mässenhausen, Anne et al. (2014) Nonamplified FGFR1 is a growth driver in malignant pleural mesothelioma. Mol Cancer Res 12:1460-9
Wang, Chu-An; Drasin, David; Pham, Catherine et al. (2014) Homeoprotein Six2 promotes breast cancer metastasis via transcriptional and epigenetic control of E-cadherin expression. Cancer Res 74:7357-70
Campbell, Eric L; Bruyninckx, Walter J; Kelly, Caleb J et al. (2014) Transmigrating neutrophils shape the mucosal microenvironment through localized oxygen depletion to influence resolution of inflammation. Immunity 40:66-77
Ostriker, Allison; Horita, Henrick N; Poczobutt, Joanna et al. (2014) Vascular smooth muscle cell-derived transforming growth factor-? promotes maturation of activated, neointima lesion-like macrophages. Arterioscler Thromb Vasc Biol 34:877-86

Showing the most recent 10 out of 27 publications