Approximately 35% of follicular thyroid carcinomas harbor a chromosomal translocation that fuses paired box gene 8 (PAX8) with the peroxisome proliferator-activated receptor gamma gene (PPARG), resulting in production of a PAX8-PPARG fusion protein denoted PPFP. PPFP contains the full sequence of the nuclear receptor PPARG1, and hence PPFP binds to PPARGresponsive genes and to PPARG ligands. We have created the first transgenic mouse model of this cancer. The cancer is locally invasive and forms lung metastases. Treatment with the PPARG agonist pioglitazone (Pio) shrinks the thyroid almost to control size and eliminates metastatic disease. Most remarkably, this therapeutic response is characterized by a trans differentiation-type process whereby the remaining thyroid cells develop large lipid droplets and express a wide array of PPARG- inducible adipocyte genes. Since PPARG is the master regulator of adipogenesis, these results indicate that, in the presence of Pio, PPFP is strongly PPARG-like. We postulate that the anti-tumor action of Pio in PPFP cancers is tied to this adipocyte trans differentiation-like effect;i.e., the more the thyroi cancer cells acquire a mature adipocyte phenotype, the less they retain of their malignant phenotype.
In Aim 1, we will evaluate the oncogenic action of PPFP in the mouse model. There is evidence that, in the absence of Pio, PPFP can inhibit PPARG induction of some target genes, and that inhibition of endogenous PPARG may underlie the oncogenic nature of PPFP. Therefore, we will test whether the genetic deletion of PPARG mimics the expression of PPFP in terms of the development of thyroid cancer. We also will assess whether both the PAX8 and PPARG DNA binding domains within PPFP are important by studying mice in which PPFP has appropriate mutations. Analyses of histology, gene expression and DNA binding will provide insight into the genes regulated by PPFP that contribute to the development and progression of thyroid cancer. We will use a non-adipogenic PPARg ligand to test whether the adipogenic nature of the Pio response is critical to its therapeutic effect. We also will test whether arsenic trioxide is therapeutic, especially in combination with Pio. This hypothesis derives from the observation that PPFP + Pio strongly induces AQP7, a channel protein through which arsenic enters cells.
In Aim 2, we will perform a phase II clinical trial to determine whether Pio is therapeutic in patients with metastatic PPFP thyroid cancer not treatable by standard therapies. The primary endpoint will be a decrease in the size of metastases. A secondary endpoint will be measurement of lipid content of metastases that do not completely resolve, based upon the observation that treatment of our mouse model of this cancer with Pio results in an adipogenic response in the surviving thyroid cells. Other secondary endpoints include changes in serum thyroglobulin and testing the ability of Pio to induce radioiodine uptake in the cancer, followed b radioiodine therapy if indicated. Overall, these studies will help elucidate mechanisms through which PPFP contributes to thyroid cancer and will help identify novel therapies both in the transgenic mouse model and in patients.

Public Health Relevance

Follicular thyroid cancers contain a protein called PPFP that is not found in normal cells and that underlies development of this cancer. These studies will investigate the mechanisms through which PPFP contributes to thyroid cancer, and will test novel therapies both in mice and patients with this disease.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA166033-01A1
Application #
8451143
Study Section
Special Emphasis Panel (ZRG1-EMNR-H (02))
Program Officer
Timmer, William C
Project Start
2013-04-01
Project End
2018-03-31
Budget Start
2013-04-01
Budget End
2014-03-31
Support Year
1
Fiscal Year
2013
Total Cost
$504,330
Indirect Cost
$173,491
Name
University of Michigan Ann Arbor
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Yu, Jingcheng; Koenig, Ronald J (2018) Thyroid-Specific PPAR? Deletion Is Benign in the Mouse. Endocrinology 159:1463-1468
Giordano, Thomas J; Haugen, Bryan R; Sherman, Steven I et al. (2018) Pioglitazone Therapy of PAX8-PPAR? Fusion Protein Thyroid Carcinoma. J Clin Endocrinol Metab 103:1277-1281
Zhang, Yanxiao; Yu, Jingcheng; Grachtchouk, Vladimir et al. (2017) Genomic binding of PAX8-PPARG fusion protein regulates cancer-related pathways and alters the immune landscape of thyroid cancer. Oncotarget 8:5761-5773
Xu, Bin; O'Donnell, Michael; O'Donnell, Jeffrey et al. (2016) Adipogenic Differentiation of Thyroid Cancer Cells Through the Pax8-PPAR? Fusion Protein Is Regulated by Thyroid Transcription Factor 1 (TTF-1). J Biol Chem 291:19274-86
Raman, Priyadarshini; Grachtchouk, Vladimir; Lyons Jr, Robert H et al. (2015) Identification of the Genomic Insertion Site of the Thyroid Peroxidase Promoter-Cre Recombinase Transgene Using a Novel, Efficient, Next-Generation DNA Sequencing Method. Thyroid 25:1162-6
Zhang, Yanxiao; Yu, Jingcheng; Lee, Chee et al. (2015) Genomic binding and regulation of gene expression by the thyroid carcinoma-associated PAX8-PPARG fusion protein. Oncotarget 6:40418-32
Vu-Phan, Dang; Koenig, Ronald J (2014) Genetics and epigenetics of sporadic thyroid cancer. Mol Cell Endocrinol 386:55-66
Raman, Priyadarshini; Koenig, Ronald J (2014) Pax-8-PPAR-? fusion protein in thyroid carcinoma. Nat Rev Endocrinol 10:616-23
Vu-Phan, Dang; Grachtchouk, Vladimir; Yu, Jingcheng et al. (2013) The thyroid cancer PAX8-PPARG fusion protein activates Wnt/TCF-responsive cells that have a transformed phenotype. Endocr Relat Cancer 20:725-39