Much remains to be learned about how RAS isoforms differ functionally from each other, and about the impact of specific mutations on each RAS protein. NRAS-mutant melanoma represents both a critical unmet need in terms of efficacious therapeutic options and also an outstanding opportunity to elucidate these functional differences. Although KRAS is the predominant RAS isoform mutated in cancers overall, NRAS is the predominant RAS isoform mutated in malignant melanoma, and whereas mutations at codon 61 are rare in KRAS, they predominate in NRAS. Project 2 has recently demonstrated that Nras Q61R but not G12D could drive melanoma formation in Ink4a-deficient mice. These provocative findings indicate that NRAS Q61R and G12D must activate distinct effectors, and/or activate effectors in a distinct manner. Given the preferential occurrence of NRAS Q61 mutations in melanoma, we propose studies to elucidate the signaling mechanisms that distinguish the roles of Q61- versus G12-mutant NRAS in driving these cancers. We hypothesize that there are structural, biochemical and biological properties distinct from those of the more common G12-mutant KRAS proteins found in lung, colorectal and pancreatic carcinomas. We have also identified an unexpected requirement for both KRAS and HRAS WT forms in NRAS-mutant melanomas. We will investigate effector signaling mechanisms driven by different NRAS mutants, define the requirements for WT isoforms, and leverage both candidate- and unbiased methodologies to identify targets that can lead to novel combination therapies for effective treatment of NRAS-driven melanomas. To address our goals, we propose three aims.
In Aim 1, we will elucidate known and unknown effector signaling pathways downstream of cellular NRAS mutated at codon 61 versus codon 12.
This Aim will be performed in close collaboration with Project 2, which will focus on structural and biochemical differences in the same panel of NRAS mutants. Projects 1 and 4 will also examine some of the cellular and tumorigenic phenotypes of the same mutations in different RAS isoforms.
In Aim 2, we will characterize the requirement for WT RAS isoforms, and determine functional differences in effectors and signaling networks, as well as in transformed growth properties.
In Aim 3, we will determine whether the same mechanisms that overcome melanoma addiction to mutant NRAS also overcome dependency on WT RAS isoforms. To do this, we will first interrogate YAP, known to be capable of rescuing addiction to mutant KRAS in carcinomas, and we will next perform a novel functional genetic screen to identify non-YAP mechanisms in an unbiased manner. Collectively, our studies will elucidate a better understanding of the neglected NRAS isoform, characterize functional distinctions among different NRAS mutations, determine relationships between WT and mutant NRAS-driven effector signaling pathways and networks, and identify new directions for novel therapeutic options in NRAS-driven melanoma.

Public Health Relevance

Malignant cutaneous melanoma is a very aggressive skin cancer once it has metastasized, and its incidence is rising. About a quarter of these melanomas are driven by the NRAS oncogene, which is mutated in ways that are different from other NRAS-mutant cancers such as certain leukemias, and also different from other RAS types such as KRAS that drives pancreatic cancer. Our goal is to understand specifically how the melanoma- associated NRAS proteins function so that we can create effective therapies against them.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA203657-03
Application #
9488434
Study Section
Special Emphasis Panel (ZCA1)
Project Start
Project End
Budget Start
2018-06-01
Budget End
2019-05-31
Support Year
3
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of North Carolina Chapel Hill
Department
Type
DUNS #
608195277
City
Chapel Hill
State
NC
Country
United States
Zip Code
27599
Fu, Jingjing; Dang, Yunkun; Counter, Christopher et al. (2018) Codon usage regulates human KRAS expression at both transcriptional and translational levels. J Biol Chem 293:17929-17940
Adhikari, Hema; Counter, Christopher M (2018) Interrogating the protein interactomes of RAS isoforms identifies PIP5K1A as a KRAS-specific vulnerability. Nat Commun 9:3646
Waters, Andrew M; Der, Channing J (2018) KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb Perspect Med 8:
Vaseva, Angelina V; Blake, Devon R; Gilbert, Thomas S K et al. (2018) KRAS Suppression-Induced Degradation of MYC Is Antagonized by a MEK5-ERK5 Compensatory Mechanism. Cancer Cell 34:807-822.e7
Papke, Bjoern; Der, Channing J (2017) Drugging RAS: Know the enemy. Science 355:1158-1163
Bryant, Kirsten L; Der, Channing J (2017) Mutant RAS Calms Stressed-Out Cancer Cells. Dev Cell 40:120-122
Waters, Andrew M; Ozkan-Dagliyan, Irem; Vaseva, Angelina V et al. (2017) Evaluation of the selectivity and sensitivity of isoform- and mutation-specific RAS antibodies. Sci Signal 10:
Ali, Moiez; Kaltenbrun, Erin; Anderson, Grace R et al. (2017) Codon bias imposes a targetable limitation on KRAS-driven therapeutic resistance. Nat Commun 8:15617
Yin, Guowei; Kistler, Samantha; George, Samuel D et al. (2017) A KRAS GTPase K104Q Mutant Retains Downstream Signaling by Offsetting Defects in Regulation. J Biol Chem 292:4446-4456
Huynh, Minh V; Campbell, Sharon L (2016) Getting a Handle on RAS-targeted Therapies: Cysteine Directed Inhibitors. Mini Rev Med Chem 16:383-90

Showing the most recent 10 out of 11 publications