4.8 Article

Diverse alterations associated with resistance to KRAS(G12C) inhibition

Journal

NATURE
Volume 599, Issue 7886, Pages 679-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41586-021-04065-2

Keywords

-

Funding

  1. NIH/NCI [1R01CA23074501, 1R01CA23026701A1, P50 CA247749 01]
  2. Pew Charitable Trusts
  3. Damon Runyon Cancer Research Foundation
  4. American Lung Association
  5. Charles H. Revson Senior Fellowship in Biomedical Science [21-31]
  6. NRSA F30 program [1F30CA232549-01]
  7. MSKCC Pilot Center for Precision Disease Modeling program [U54 OD020355]
  8. LUNGevity foundation
  9. Breast Cancer Research Foundation
  10. Gerald Leigh Charitable Trust
  11. Cycle for Survival
  12. Conquer Cancer from the ASCO Foundation
  13. institutional K30 grant (CTSA) [UL1TR00457]
  14. Medical Scientist Training Program grant [T32GM007739]
  15. MSKCC Support Grant-Core Grant program [P30 CA008748]
  16. NCI Cancer Center Support Grant [P30 CA08748]
  17. Amgen Inc.
  18. Marie-Josee and Henry R. Kravis Center for Molecular Oncology

Ask authors/readers for more resources

Resistance to KRAS(G12C) inhibitors demonstrates a heterogeneous pattern with multiple subclonal events emerging during treatment. Co-targeting of ERK signaling intermediates can enhance the antiproliferative effect of G12C inhibitor treatment in models with acquired RAS or BRAF mutations.
Inactive state-selective KRAS(G12C) inhibitors(1-8) demonstrate a 30-40% response rate and result in approximately 6-month median progression-free survival in patients with lung cancer(9). The genetic basis for resistance to these first-in-class mutant GTPase inhibitors remains under investigation. Here we evaluated matched pre-treatment and post-treatment specimens from 43 patients treated with the KRAS(G12C) inhibitor sotorasib. Multiple treatment-emergent alterations were observed across 27 patients, including alterations in KRAS, NRAS, BRAF, EGFR, FGFR2, MYC and other genes. In preclinical patient-derived xenograft and cell line models, resistance to KRAS(G12C) inhibition was associated with low allele frequency hotspot mutations in KRAS(G12V or G13D), NRAS(Q61K or G13R), MRAS(Q71R) and/or BRAF(G596R), mirroring observations in patients. Single-cell sequencing in an isogenic lineage identified secondary RAS and/or BRAF mutations in the same cells as KRAS(G12C), where they bypassed inhibition without affecting target inactivation. Genetic or pharmacological targeting of ERK signalling intermediates enhanced the antiproliferative effect of G12C inhibitor treatment in models with acquired RAS or BRAF mutations. Our study thus suggests a heterogenous pattern of resistance with multiple subclonal events emerging during G12C inhibitor treatment. A subset of patients in our cohort acquired oncogenic KRAS, NRAS or BRAF mutations, and resistance in this setting may be delayed by co-targeting of ERK signalling intermediates. These findings merit broader evaluation in prospective clinical trials.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available