Squamous cell carcinoma (SCC) is the second most common subtype of lung cancer and is responsible for over 40,000 deaths each year in the United States alone. SCC has a survival rate of 15% and unlike the other major subtype of lung cancer, there are currently no approved targeted therapies for SCC. Recently, The Cancer Genome Atlas sequenced 178 human SCCs and identified numerous genetic alterations that may serve as valuable therapeutic targets. Because SCC is highly associated with smoking, the genetic mutation rate is one of the highest of all tumor types, making it difficult to distinguish driver from passenger mutations. Clearly, a major unmet need for the treatment of SCC is the identification of new therapeutic targets and treatment strategies. The transcription factor Sox2 is one of the most commonly altered genes in SCC, amplified in 23% and overexpressed in 60-90% of human SCCs. However, Sox2's function and key target genes in lung SCC are unclear. In addition, there are currently no targeted therapies that inhibit this transcription facor. We recently developed a novel mouse model of SCC driven by Sox2 expression that highly resembles the human disease at the level of histopathology, biomarker expression and pathway activation. This model will be a useful tool to identify novel therapeutic targets and biomarkers, elucidate mechanisms of tumor progression, identify the cell(s) of origin, and test novel therapies and drug combinations in SCC. The objective of this study is to use this novel mouse model and human lung cancer cells to identify pathways critical for Sox2-driven SCC and to elucidate the cell(s) of origin for SCC in vivo. We hypothesize that mouse tumors (since not exposed to carcinogen) will have fewer genetic alterations than the human disease and thereby serve as a biological filter to identify important pathways driven by or cooperating with Sox2. Based on preliminary in vitro and in vivo data, we predict that Sox2 drives targetable pathways that are critical for tumor initiation and/or maintenance. Given that Sox2-driven SCCs express markers of basal cells, we predict that Sox2 either arises in basal cells or transforms other lung cell types to a basal cell identity. To test these hypotheses, we will: 1) use next-generation sequencing to identify conserved genetic changes between mouse and human SCC, 2) test the impact of inhibiting key pathways in Sox2-expressing human lung cancer cells, and 3) use our bicistronic lentiviral system with cell type-specific promoters to determine which lung cell type functions as the cell(s) of origin for Sox2-driven SCC in vivo. This approach is innovative because we will employ a novel mouse model of SCC that highly recapitulates the human disease. This research is significant because lung cancer is the leading cause of cancer death in the United States and there are currently no targeted therapies approved for SCC. A better understanding of the critical pathways and cellular origins of SCC will impact the treatment and survival of patients with this intractable disease.

Public Health Relevance

Squamous cell lung cancer is a devastating subtype of lung cancer for which there are limited treatment options. This study will employ a novel mouse model of the disease to identify and validate new therapeutic targets and elucidate the cellular origins of the disease in vivo. This work is relevant to public health and the NIH mission because it will impact the development of new therapies to improve the quality of life and survival of lung cancer patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA187457-03
Application #
9066602
Study Section
Molecular Oncogenesis Study Section (MONC)
Program Officer
Kondapaka, Sudhir B
Project Start
2014-07-01
Project End
2019-05-31
Budget Start
2016-06-01
Budget End
2017-05-31
Support Year
3
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Utah
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
009095365
City
Salt Lake City
State
UT
Country
United States
Zip Code
84112
Mollaoglu, Gurkan; Jones, Alex; Wait, Sarah J et al. (2018) The Lineage-Defining Transcription Factors SOX2 and NKX2-1 Determine Lung Cancer Cell Fate and Shape the Tumor Immune Microenvironment. Immunity 49:764-779.e9
Huang, Fang; Ni, Min; Chalishazar, Milind D et al. (2018) Inosine Monophosphate Dehydrogenase Dependence in a Subset of Small Cell Lung Cancers. Cell Metab 28:369-382.e5
Mollaoglu, Gurkan; Guthrie, Matthew R; Böhm, Stefanie et al. (2017) MYC Drives Progression of Small Cell Lung Cancer to a Variant Neuroendocrine Subtype with Vulnerability to Aurora Kinase Inhibition. Cancer Cell 31:270-285
Brägelmann, Johannes; Böhm, Stefanie; Guthrie, Matthew R et al. (2017) Family matters: How MYC family oncogenes impact small cell lung cancer. Cell Cycle 16:1489-1498
Cardnell, Robert J; Li, Lerong; Sen, Triparna et al. (2017) Protein expression of TTF1 and cMYC define distinct molecular subgroups of small cell lung cancer with unique vulnerabilities to aurora kinase inhibition, DLL3 targeting, and other targeted therapies. Oncotarget 8:73419-73432
Terry, M R; Arya, R; Mukhopadhyay, A et al. (2015) Caspase-2 impacts lung tumorigenesis and chemotherapy response in vivo. Cell Death Differ 22:719-30
Mukhopadhyay, Anandaroop; Berrett, Kristofer C; Kc, Ushma et al. (2014) Sox2 cooperates with Lkb1 loss in a mouse model of squamous cell lung cancer. Cell Rep 8:40-9