Significant discoveries at UT Southwestern have set the foundation for the first molecular genetic and biological classification of sporadic renal cell carcinoma of clear -cell type (ccRCC), the most common type. Leveraging this innovation, we seek to assess the prognostic and predictive value of a mutated gene we have discovered, build the first immunocompetent animal model of ccRCC reproducing the genetics of human tumors, and identify novel targets for drug development. We discovered that the tumor suppre ssor gene BAP1 is mutated in 15% of sporadic ccRCC and that BAP1-deficient tumors tend not to have mutations in a second tumor suppressor gene PBRM1 (mutated in 50% of ccRCC). We developed and validated immunohistochemistry tests and found that whereas BAP1-deficient tumors tend to be of high grade, PBRM1- deficient tumors are typically of low grade. In patients with localized disease, BAP1 loss was associated with significantly worse prognosis (HR for RCC-specific death, 3.1; 95% CI, 2.3-4.1; p=6.77?10-14). By leveraging samples from the International mRCC Database Consortium, a consortium that has already introduced changes in clinical practice, we seek to determine whether BAP1 loss leads to more aggressive tumors even in the metastatic setting. Based on our previous discovery that BAP1 loss is associated with mTORC1 activation, we will also test whether BAP1 is a biomarker of responsiveness to mTORC1 inhibitors clinically. Notably, both BAP1 and PBRM1 genes are located in the proximity of VHL (which is inactivated in >80% of ccRCC) in a region on chromosome 3p that is deleted in the vast majority of ccRCC. This may explain a longstanding paradox ? why do humans with VHL germline mutations develop kidney cancer, but not Vhl+/- mice? We found that in mice, Bap1 and Pbrm1 are on different chromosomes than Vhl. Thus, LOH of the Vhl region in the mouse would still leave two copies of Bap1 and Pbrm1. We predicted that combined inactivation of Vhl and Bap1 (or Pbrm1) would cause ccRCC in the mouse but have been prevented from fully testing this hypothesis due to renal failure and perinatal lethality of mice with conditional loss of Bap1 in the kidney. However, conditional loss of Vhl is tolerated up to a year of age and VhlF/F;BapF/+ mice develop microscopic RCC. To bypass perinatal lethality and obtain macroscopic tumors, we will inactivate Vhl and Bap1 in the adult nephron. Finally, BAP1 is lost in tumors and consequently is not a suitable target for therapy. BAP1 is a deubiquitinase, and to develop the next generation of therapies, we will identify BAP1 substrates implicated in renal cancer. If successful, these aims will identify patients most likely to respond to mTORC1 inhibitors, help stratify patients with metastatic disease, lead to the first faithful immunocompetent animal model of ccRCC, and, following the path from gene discovery to drug development of Project 1, pave the way for the next generation of targeted therapies.

Public Health Relevance

Metastatic kidney cancer is incurable. However, the discovery of mutated genes and an understanding of how they trigger tumor development have led to a new set of drugs and a doubling of life expectancy. We have discovered a new gene mutated in an aggressive form of kidney cancer and seek to (1) determine its impact on the prognosis and treatment of patients with metastases, (2) disrupt it in the mouse to generate the first animal model resembling kidney cancer in humans, and (3) understand how it causes cancer so we can identify targets for the next generation of drugs.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Specialized Center (P50)
Project #
1P50CA196516-01A1
Application #
9071071
Study Section
Special Emphasis Panel (ZCA1)
Project Start
2016-08-01
Project End
2021-07-31
Budget Start
2016-08-01
Budget End
2017-07-31
Support Year
1
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Texas Sw Medical Center Dallas
Department
Type
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Kay, Fernando U; Canvasser, Noah E; Xi, Yin et al. (2018) Diagnostic Performance and Interreader Agreement of a Standardized MR Imaging Approach in the Prediction of Small Renal Mass Histology. Radiology 287:543-553
Chen, Xi; Zhou, Zhiguo; Hannan, Raquibul et al. (2018) Reliable gene mutation prediction in clear cell renal cell carcinoma through multi-classifier multi-objective radiogenomics model. Phys Med Biol 63:215008
Puertollano, Rosa; Ferguson, Shawn M; Brugarolas, James et al. (2018) The complex relationship between TFEB transcription factor phosphorylation and subcellular localization. EMBO J 37:
Chen, Kenneth S; Stroup, Emily K; Budhipramono, Albert et al. (2018) Mutations in microRNA processing genes in Wilms tumors derepress the IGF2 regulator PLAG1. Genes Dev 32:996-1007
O'Kelly, Devin; Zhou, Heling; Mason, Ralph P (2018) Tomographic breathing detection: a method to noninvasively assess in situ respiratory dynamics. J Biomed Opt 23:1-6
Wang, Xinzeng; Pirasteh, Ali; Brugarolas, James et al. (2018) Whole-body MRI for metastatic cancer detection using T2 -weighted imaging with fat and fluid suppression. Magn Reson Med 80:1402-1415
Courtney, Kevin D; Infante, Jeffrey R; Lam, Elaine T et al. (2018) Phase I Dose-Escalation Trial of PT2385, a First-in-Class Hypoxia-Inducible Factor-2? Antagonist in Patients With Previously Treated Advanced Clear Cell Renal Cell Carcinoma. J Clin Oncol 36:867-874
Krajewski, Katherine M; Pedrosa, Ivan (2018) Imaging Advances in the Management of Kidney Cancer. J Clin Oncol :JCO2018791236
Carbone, Michele; Amelio, Ivano; Affar, El Bachir et al. (2018) Consensus report of the 8 and 9th Weinman Symposia on Gene x Environment Interaction in carcinogenesis: novel opportunities for precision medicine. Cell Death Differ 25:1885-1904
Kay, Fernando U; Pedrosa, Ivan (2018) Imaging of Solid Renal Masses. Urol Clin North Am 45:311-330

Showing the most recent 10 out of 28 publications