Bladder cancer kills 12,000 Americans each year, but few research projects are targeted to this disease. For most of these patients, the cause of death is attributable to metastatic spread, commonly to the lungs. Hence, a fuller understanding of the molecular mechanisms driving the dissemination and growth of bladder cancer to the lungs is likely to present new therapeutic opportunities. The Ras family small GTPases RalA and RalB have been shown to play important roles in human tumor formation and progression. Specifically, RalB is elevated in bladder cancer and promotes migration and metastasis. Genome-wide analysis of RalB dependent changes in gene expression revealed that RalB regulates the expression of CD24, a GPI-linked glycoprotein, necessary for the growth of many human cancer cell lines. CD24 is also a biomarker of metastasis in bladder cancer. RalB induction of CD24 expression involves the Ral effector RalBP1, which is also elevated in bladder cancer, and the zinc finger transcription factor RREB1. Our Guiding Hypothesis is that a novel RalB ? RalBP1 ? RREB1 ? CD24 signaling pathway that was discovered by this project, promotes bladder cancer metastasis. To test this hypothesis we propose the following Specific Aims:
In Aim 1, the mechanistic contributions of RalB and RalBP1 to bladder cancer lung metastasis will be dissected using mutants of both that are impaired in specific functions and effector interactions. Since RalA does not promote migration or metastasis, we will use RalA/RalB chimeras to identify RalB sequences necessary for metastasis and subsequently proteins that specifically bind to RalB via those regions. Successful completion of this aim should result in new targets for drug discovery.
Aim 2 will determine how RalB and RalBP1 regulate RREB1 activity and evaluate the requirement for RREB1 regulation in CD24 expression. Since genome-wide profiling of RalB dependent gene expression led to the discovery of the metastasis biomarker CD24, we will use a human bladder cancer tissue repository and advanced computational tools to generate a gene signature of RalB expression. Given the importance of RalB in bladder cancer progression, the ability of this signature to predict the development of metastatic disease in patients will be evaluated. Eventually, this signature may be of prognostic value and an avenue to select patients for individualized RalB targeted therapy.
In Aim 3 we will evaluate the requirement for CD24 in bladder cancer metastasis. An existing CD24 knockout mouse will be used to study the role of CD24 in a chemical carcinogenesis model of bladder cancer formation, invasion, and metastasis. In a preclinical study, we will also test whether anti-CD24 immunotherapy can block the growth of established human bladder cancer lung metastases. These mechanistic studies of a novel signaling pathway will provide molecular information on how Ral GTPases promote bladder cancer lung metastasis, thus providing both a framework for future patient risk stratification and opportunities for novel therapies of metastatic disease. PROJECT NARRATIVE Bladder cancer kills 12,000 Americans each year, but few research projects are targeted to this disease. For most of these patients, the cause of death is attributable to metastatic spread, commonly to the lungs. The goal of this project is to understand the mechanisms that underlie lung metastasis in human bladder cancer and use this knowledge to predict and treat this condition in patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA075115-15
Application #
8278059
Study Section
Tumor Progression and Metastasis Study Section (TPM)
Program Officer
Woodhouse, Elizabeth
Project Start
1997-09-01
Project End
2014-04-30
Budget Start
2012-05-01
Budget End
2014-04-30
Support Year
15
Fiscal Year
2012
Total Cost
$340,613
Indirect Cost
$117,990
Name
University of Colorado Denver
Department
Surgery
Type
Schools of Medicine
DUNS #
041096314
City
Aurora
State
CO
Country
United States
Zip Code
80045
Fantini, Damiano; Glaser, Alexander P; Rimar, Kalen J et al. (2018) A Carcinogen-induced mouse model recapitulates the molecular alterations of human muscle invasive bladder cancer. Oncogene 37:1911-1925
Yan, Chao; Theodorescu, Dan (2018) RAL GTPases: Biology and Potential as Therapeutic Targets in Cancer. Pharmacol Rev 70:1-11
Duex, Jason E; Swain, Kalin E; Dancik, Garrett M et al. (2018) Functional Impact of Chromatin Remodeling Gene Mutations and Predictive Signature for Therapeutic Response in Bladder Cancer. Mol Cancer Res 16:69-77
Duex, Jason E; Owens, Charles; Chauca-Diaz, Ana et al. (2017) Nuclear CD24 Drives Tumor Growth and Is Predictive of Poor Patient Prognosis. Cancer Res 77:4858-4867
Nickerson, M L; Witte, N; Im, K M et al. (2017) Molecular analysis of urothelial cancer cell lines for modeling tumor biology and drug response. Oncogene 36:35-46
Yan, Chao; Theodorescu, Dan; Miller, Bettina et al. (2016) Synthesis of novel Ral inhibitors: An in vitro and in vivo study. Bioorg Med Chem Lett 26:5815-5818
Agarwal, Neeraj; Dancik, Garrett M; Goodspeed, Andrew et al. (2016) GON4L Drives Cancer Growth through a YY1-Androgen Receptor-CD24 Axis. Cancer Res 76:5175-85
Jones, Robert T; Felsenstein, Kenneth M; Theodorescu, Dan (2016) Pharmacogenomics: Biomarker-Directed Therapy for Bladder Cancer. Urol Clin North Am 43:77-86
Duberow, David P; Brait, Mariana; Hoque, Mohammad O et al. (2016) High-performance detection of somatic D-loop mutation in urothelial cell carcinoma patients by polymorphism ratio sequencing. J Mol Med (Berl) 94:1015-24
Frantzi, Maria; van Kessel, Kim E; Zwarthoff, Ellen C et al. (2016) Development and Validation of Urine-based Peptide Biomarker Panels for Detecting Bladder Cancer in a Multi-center Study. Clin Cancer Res 22:4077-86

Showing the most recent 10 out of 66 publications