Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition

KRAS GTPases are activated in one-third of cancers, and KRAS(G12C) is one of the most common activating alterations in lung adenocarcinoma 1 , 2 . KRAS(G12C) inhibitors 3 , 4 are in phase-I clinical trials and early data show partial responses in nearly half of patients with lung cancer. How cancer cells bypass inhibition to prevent maximal response to therapy is not understood. Because KRAS(G12C) cycles between an active and inactive conformation 4 – 6 , and the inhibitors bind only to the latter, we tested whether isogenic cell populations respond in a non-uniform manner by studying the effect of treatment at a single-cell resolution. Here we report that, shortly after treatment, some cancer cells are sequestered in a quiescent state with low KRAS activity, whereas others bypass this effect to resume proliferation. This rapid divergent response occurs because some quiescent cells produce new KRAS(G12C) in response to suppressed mitogen-activated protein kinase output. New KRAS(G12C) is maintained in its active, drug-insensitive state by epidermal growth factor receptor and aurora kinase signalling. Cells without these adaptive changes—or cells in which these changes are pharmacologically inhibited—remain sensitive to drug treatment, because new KRAS(G12C) is either not available or exists in its inactive, drug-sensitive state. The direct targeting of KRAS oncoproteins has been a longstanding objective in precision oncology. Our study uncovers a flexible non-uniform fitness mechanism that enables groups of cells within a population to rapidly bypass the effect of treatment. This adaptive process must be overcome if we are to achieve complete and durable responses in the clinic. Populations of KRAS(G12C)-mutant cancer cells can rapidly bypass the effects of treatment with KRAS(G12C) inhibitors because a subset of cells escapes drug-induced quiescence by producing new KRAS(G12C) that is maintained in its active, drug-insensitive state.

[1]  Sourav Bandyopadhyay,et al.  Aurora kinase A drives the evolution of resistance to third generation EGFR inhibitors in lung cancer , 2018, Nature Medicine.

[2]  Jun S. Liu,et al.  MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens , 2014, Genome Biology.

[3]  D. Bar-Sagi,et al.  RAS oncogenes: weaving a tumorigenic web , 2011, Nature Reviews Cancer.

[4]  Fabian J. Theis,et al.  Diffusion maps for high-dimensional single-cell analysis of differentiation data , 2015, Bioinform..

[5]  N. Rosen,et al.  Tumor adaptation and resistance to RAF inhibitors , 2013, Nature Medicine.

[6]  Kendall R. Sanson,et al.  Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities , 2018, Nature Communications.

[7]  Davis J. McCarthy,et al.  A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor , 2016, F1000Research.

[8]  M. Barbacid,et al.  Genetic analysis of Ras signalling pathways in cell proliferation, migration and survival , 2010, The EMBO journal.

[9]  S. Dudoit,et al.  A general and flexible method for signal extraction from single-cell RNA-seq data , 2018, Nature Communications.

[10]  Huimin Bian,et al.  Aurora A Kinase Inhibition Is Synthetic Lethal with Loss of the RB1 Tumor Suppressor Gene. , 2018, Cancer discovery.

[11]  James T. Webber,et al.  Kinome rewiring reveals AURKA limits PI3K-pathway inhibitor efficacy in breast cancer , 2018, Nature Chemical Biology.

[12]  Christof Fellmann,et al.  Disruption of CRAF-mediated MEK activation is required for effective MEK inhibition in KRAS mutant tumors. , 2014, Cancer cell.

[13]  Russell B. Fletcher,et al.  Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics , 2017, BMC Genomics.

[14]  A. Oudenaarden,et al.  Design and Analysis of Single-Cell Sequencing Experiments , 2015, Cell.

[15]  Allon M. Klein,et al.  Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells , 2015, Cell.

[16]  M. Berger,et al.  An approach to suppress the evolution of resistance in BRAFV600E-mutant cancer , 2017, Nature Medicine.

[17]  Evan Z. Macosko,et al.  Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets , 2015, Cell.

[18]  Neal Rosen,et al.  Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism , 2016, Science.

[19]  Di Wu,et al.  ROAST: rotation gene set tests for complex microarray experiments , 2010, Bioinform..

[20]  David A. Scott,et al.  Potent and Selective Covalent Quinazoline Inhibitors of KRAS G12C. , 2017, Cell chemical biology.

[21]  S. Chandarlapaty,et al.  Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. , 2012, Cancer cell.

[22]  R. Bernards,et al.  Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR , 2012, Nature.

[23]  A. Balmain,et al.  A model for RAS mutation patterns in cancers: finding the sweet spot , 2018, Nature Reviews Cancer.

[24]  Mari Mino-Kenudson,et al.  EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib. , 2012, Cancer discovery.

[25]  H. Aburatani,et al.  A novel cell-cycle-indicator, mVenus-p27K−, identifies quiescent cells and visualizes G0–G1 transition , 2014, Scientific Reports.

[26]  Yi Liu,et al.  Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State. , 2016, Cancer discovery.

[27]  John C. Hunter,et al.  Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations , 2015, Molecular Cancer Research.

[28]  Charlotte Soneson,et al.  Observation weights unlock bulk RNA-seq tools for zero inflation and single-cell applications , 2018, Genome Biology.

[29]  Sandrine Dudoit,et al.  Normalizing single-cell RNA sequencing data: challenges and opportunities , 2017, Nature Methods.

[30]  Aaron T. L. Lun,et al.  Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R , 2017, Bioinform..

[31]  Y. Saeys,et al.  Computational methods for trajectory inference from single‐cell transcriptomics , 2016, European journal of immunology.

[32]  J. Minna,et al.  Knockdown of Oncogenic KRAS in Non–Small Cell Lung Cancers Suppresses Tumor Growth and Sensitizes Tumor Cells to Targeted Therapy , 2011, Molecular Cancer Therapeutics.

[33]  R. Satija,et al.  Integrative single-cell analysis , 2019, Nature Reviews Genetics.

[34]  P. Zarrinkar,et al.  Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor , 2018, Cell.

[35]  Frank McCormick,et al.  RAS Proteins and Their Regulators in Human Disease , 2017, Cell.

[36]  Ambrose J. Carr,et al.  Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment , 2018, Cell.

[37]  Stephen S. Taylor,et al.  Aurora-kinase inhibitors as anticancer agents , 2004, Nature Reviews Cancer.

[38]  Raphael Gottardo,et al.  Orchestrating high-throughput genomic analysis with Bioconductor , 2015, Nature Methods.

[39]  Kaori Sasai,et al.  Phosphorylation by aurora kinase A induces Mdm2-mediated destabilization and inhibition of p53 , 2004, Nature Genetics.

[40]  Cole Trapnell,et al.  The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells , 2014, Nature Biotechnology.

[41]  Phillip W Iversen,et al.  Aurora-A kinase inhibition is synthetic lethal with loss of the RB1 tumor suppressor gene. , 2019 .

[42]  C. Der,et al.  Aurora-A Phosphorylates, Activates, and Relocalizes the Small GTPase RalA , 2009, Molecular and Cellular Biology.

[43]  Kevin R. Moon,et al.  Recovering Gene Interactions from Single-Cell Data Using Data Diffusion , 2018, Cell.

[44]  Tom H. Cheung,et al.  Molecular regulation of stem cell quiescence , 2013, Nature Reviews Molecular Cell Biology.

[45]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[46]  Alessandro Laio,et al.  Clustering by fast search and find of density peaks , 2014, Science.

[47]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[48]  H. Fu,et al.  Aurora kinase A interacts with H-Ras and potentiates Ras-MAPK signaling , 2017, Oncotarget.

[49]  J. Schlessinger,et al.  Cell Signaling by Receptor Tyrosine Kinases , 2000, Cell.

[50]  Kevan M. Shokat,et al.  K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions , 2013, Nature.

[51]  S. Dudoit,et al.  Normalization of RNA-seq data using factor analysis of control genes or samples , 2014, Nature Biotechnology.

[52]  J. Desai,et al.  Phase 1 study evaluating the safety, tolerability, pharmacokinetics (PK), and efficacy of AMG 510, a novel small molecule KRASG12C inhibitor, in advanced solid tumors. , 2019, Journal of Clinical Oncology.

[53]  Krister Wennerberg,et al.  Methods for High-Throughput Drug Combination Screening and Synergy Scoring , 2016, bioRxiv.