4.6 Article

Integrated phosphoproteomics and transcriptional classifiers reveal hidden RAS signaling dynamics in multiple myeloma

Journal

BLOOD ADVANCES
Volume 3, Issue 21, Pages 3214-3227

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/bloodadvances.2019000303

Keywords

-

Categories

Funding

  1. Damon Runyon Cancer Research Foundation Dale Frey Breakthrough Award [DFS 14-15]
  2. National Institutes of Health (National Cancer Institute) [K08CA184116, R01CA226851, P30 CA138292]
  3. National Institutes of Health (National Institute of General Medical Sciences) [DP2OD022552]
  4. UCSF Stephen and Nancy Grand Multiple Myeloma Translational Initiative
  5. MMRF Answer Fund
  6. American Cancer Society postdoctoral fellowship [PF-17-109-1-TBG]
  7. National Institutes of Health (National Human Genome Research Institute) [T32 HG000046]
  8. Gordon and Betty Moore Foundation [GBMF 4552]

Ask authors/readers for more resources

A major driver of multiple myeloma (MM) is thought to be aberrant signaling, yet no kinase inhibitors have proven successful in the clinic. Here, we employed an integrated, systems approach combining phosphoproteomic and transcriptome analysis to dissect cellular signaling in MM to inform precision medicine strategies. Unbiased phosphoproteomics initially revealed differential activation of kinases across MM cell lines and that sensitivity to mammalian target of rapamycin (mTOR) inhibition may be particularly dependent on mTOR kinase baseline activity. We further noted differential activity of immediate downstream effectors of Ras as a function of cell line genotype. We extended these observations to patient transcriptome data in the Multiple Myeloma Research Foundation CoMMpass study. A machine-learning-based classifier identified surprisingly divergent transcriptional outputs between NRAS- and KRAS-mutated tumors. Genetic dependency and gene expression analysis revealed mutated Ras as a selective vulnerability, but not other MAPK pathway genes. Transcriptional analysis further suggested that aberrant MAPK pathway activation is only present in a fraction of RAS-mutated vs wild-type RAS patients. These high-MAPK patients, enriched for NRAS Q61 mutations, have inferior outcomes, whereas RAS mutations overall carry no survival impact. We further developed an interactive software tool to relate pharmacologic and genetic kinase dependencies in myeloma. Collectively, these predictive models identify vulnerable signaling signatures and highlight surprising differences in functional signaling patterns between NRAS and KRAS mutants invisible to the genomic landscape. These results will lead to improved m stratification of MM patients in precision medicine trials while also revealing unexplored modes of Ras biology in MM.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available