Purpose:

Activating missense mutations of KRAS are the most frequent oncogenic driver events in lung adenocarcinoma (LUAD). However, KRAS isoforms are highly heterogeneous, and data on the potential isoform-dependent therapeutic vulnerabilities are still lacking.

Experimental Design:

We developed an isogenic cell-based platform to compare the oncogenic properties and specific therapeutic actionability of KRAS-mutant isoforms. In parallel, we analyzed clinicopathologic and genomic data from 3,560 patients with non–small cell lung cancer (NSCLC) to survey allele-specific features associated with oncogenic KRAS mutations.

Results:

In isogenic cell lines expressing different mutant KRAS isoforms, we identified isoform-specific biochemical, biological, and oncogenic properties both in vitro and in vivo. These exclusive features correlated with different therapeutic responses to MEK inhibitors, with KRAS G12C and Q61H mutants being more sensitive compared with other isoforms. In vivo, combined KRAS G12C and MEK inhibition was more effective than either drug alone. Among patients with NSCLCs that underwent comprehensive tumor genomic profiling, STK11 and ATM mutations were significantly enriched among tumors harboring KRAS G12C, G12A, and G12V mutations. KEAP1 mutation was significantly enriched among KRAS G12C and KRAS G13X LUADs. KRAS G13X-mutated tumors had the highest frequency of concurrent STK11 and KEAP1 mutations. Transcriptomic profiling revealed unique patterns of gene expression in each KRAS isoform, compared with KRAS wild-type tumors.

Conclusions:

This study demonstrates that KRAS isoforms are highly heterogeneous in terms of concurrent genomic alterations and gene-expression profiles, and that stratification based on KRAS alleles should be considered in the design of future clinical trials.

This article is featured in Highlights of This Issue, p. 1475

Translational Relevance

KRAS is the most commonly mutated driver oncogene in solid tumors, including non–small cell lung cancer (NSCLC), and KRAS mutations have traditionally been associated with poor prognosis and treatment resistance. Comprehensive genomic, biological, and clinicopathologic analysis of KRAS-mutant variants in preclinical models and samples from patients with non-squamous NSCLC revealed isoform-dependent therapeutic vulnerabilities of these tumors. Our results suggest that the use of specific genetically defined in vitro tools together with the development of large patient-derived data sets facilitates the identification of potentially actionable vulnerabilities for KRAS-mutant NSCLC.

KRAS is one of the most commonly mutated oncogenes in human cancer, with selectively high frequency in tumors of the pancreas, colon, and lung. In non–small cell lung cancer (NSCLC), KRAS mutations are detected in about 30% of patients and are associated with poor prognosis (1, 2). Several strategies have been investigated over the last decades to target KRAS mutation in lung cancer, including the inhibition of downstream effectors such as MEK (3, 4). However, none of these strategies were effective in clinical trials, and the optimal first-line therapy for advanced KRAS-mutant lung adenocarcinoma (LUAD) still consists of PD-(L)1 blockade alone or in combination with platinum-based chemotherapy (5). More recently, alternative strategies targeting KRAS-mutant lung cancer have been investigated in preclinical models and are in various stages of clinical development (1, 6, 7).

To date, the greatest success in targeting KRAS mutations is represented by the development of isoform-specific direct KRAS inhibitors. Recent data from early-phase clinical trials of KRAS G12C direct inhibitors sotorasib and adagrasib have shown encouraging activity in KRAS G12C-mutant NSCLCs, with responses occurring in ∼30%–40% of patients (8–10). Based on the results of the CodeBreaK 100 study and KRYSTAL-1, sotorasib and adagrasib were granted accelerated FDA approval and breakthrough therapy designation (BTD), respectively, for patients with NSCLC who have received prior systemic therapy. Nevertheless, an urgent need remains for better stratification and effective therapeutic strategies to improve outcomes for KRAS-mutant cancer patients.

Although RAS mutations were discovered over 30 years ago (11), the detailed understanding of the biological properties of oncogenic KRAS is still far to be complete. New aspects of KRAS biology have been recently described, including the requirement of dimerization for oncogenic signaling (12), lipid-sorting specificity into the membrane (13), and tissue-specific comutation networks (14).

One intriguing aspect of KRAS biology is the potential impact of distinct KRAS-activating mutations on downstream signaling and drug sensitivity (15, 16). Until very recently, RAS proteins with alterations in codon 12, 13, or 61 have been considered oncogenic equivalents; however, emerging data suggest functional differences for each RAS mutation in colon cancer (17), melanoma (18), and NSCLC, where different mutant KRAS proteins are known to be heterogeneous in terms of clinical outcome (19–20). Recently, studies in isogenic cell lines demonstrated that each individual mutation generates a distinct oncogenic and network response (14, 15, 21–23). In terms of translational impact, the study of differences among specific KRAS mutants by means of more informative experimental approaches will be fundamental to discover new therapeutic strategies for individual KRAS mutations.

Here, we developed an isogenic system where a series of mutant KRAS isoforms commonly found in lung cancer were directly compared to highlight distinct GTP-loading levels, differential drug sensitivity to MEK inhibitors, and unique growth properties in vivo. To gather evidence on whether these differences also translated in different clinical outcomes, we investigated the impact of the different KRAS isoforms on survival in a large cohort of patients with non-squamous NSCLC. Together, these results suggest that genomic coalterations and residue-specific properties of KRAS mutations have important biological and clinical implications.

Generation of KRaslox KRASMUT cells

KRaslox KRASMUT cells were generated as previously described (12). Briefly, KRASMUT retroviral constructs were created by point mutagenesis from pBABE HA-tagged KRASWT plasmid (a gift from Channing Der, Addgene plasmid #75282). Retroviruses were generated by cotransfection of pBABE plasmids together with pAmpho plasmid into 293T cells using FuGENE HD Transfection Reagent (Promega). The retroviruses were transduced into HRas−/−; NRas−/−; KRaslox/lox MEFs (24) followed by puromycin selection (1 μg/mL). Selected cells were then cultured in the presence of 4-hydroxytamoxifen (4OHT; Sigma, 600 nmol/L) for at least 10 days in order to achieve complete deletion of endogenous KRas alleles. Cells were not cultured longer than 3 months after thawing from frozen stocks and were routinely checked for Mycoplasma as determined by the Mycoplasma Plus PCR Primer Set (Agilent).

Growth assays in vitro

Cells (1×103) were seeded in 96-well plates in 150 μL complete medium. The following day, cells were PBS washed, and 150 μL of medium with different concentrations of FBS was added to the plates. Cells were then incubated in the IncuCyte Zoom for real-time imaging, with three fields imaged per well under 10× magnification every two hours for a total of 60–96 hours. Data were analyzed using the IncuCyte Confluence version 1.5 software, which quantified cell-surface area coverage as confluence values. IncuCyte experiments were performed in triplicate. A single representative growth curve is shown for each condition. For hypoxic conditions, cells (1×103) were seeded in 96-well plates in 150 μL complete medium. The following day, cells were PBS washed, and 150 μL of medium with different concentrations of FBS was added to the plates that were incubated in 1% O2 incubator. Cell viability was assessed by CellTiter-Glo 2.0 (Promega) at the indicated time points.

Analysis of MEK/ERK inhibitor sensitivities

Data from a large-scale pharmacogenomics study, the Cancer Genome Project (CGP, data set version 2020), were accessible from http://www.cancerrxgene.org. A total of 310 lung, pancreas, and colon cancer cell lines and their drug-sensitivity values to MEK/ERK inhibitors in the CGP data set were used for analysis. In these data, the natural logarithm of the IC50 value represents the drug-sensitivity value. Cell lines were grouped according to KRAS mutational subtypes, and pairwise comparisons were performed using the Mann–Whitney U test.

Drug-response assays

Cells (1 × 103) were seeded in 96-well plates. The following day, cells were treated with drugs using a ten-point dose titration scheme. After 72 hours, cell viability was assessed using the colorimetric MTS assay. Absolute inhibitory concentration values were calculated using four-parameter logistic curve fitting. All experimental points were a result of three to six replicates, and all experiments were repeated at least three times. The data were graphically displayed using GraphPad Prism 5 for Windows (GraphPad Software). Each point (mean ± standard deviation) represents growth of treated cells compared with untreated cells. The curves were fitted using a nonlinear regression model with a sigmoidal dose response. For synergy distribution assays, cells were seeded at 1,500 cells per well in a 384-well plate (Corning). The following day, cells were treated with selumetinib and SHP099, varying concentration from 0.25 to 15 μmol/L and from 0.5 to 15 μmol/L, respectively. After 72 hours, cell viability was measured by CellTiter-Glo 2D (Promega). Four replicates of each mutant were analyzed implementing LOEWE model (Combenefit software); experiments were repeated three times.

Ras-GTP pulldown

Cells were grown in 0.1% FBS for 24 hours, stimulated with EFG (Thermo Fisher Cat#PHG0311) 50 ng/mL for 5 minutes, and Ras-GTP levels were assessed by Active Ras Detection Kit (Cell Signaling, cat. #8821) using Raf-RBD fused to GST to bind active (GTP-bound) Ras. Protein lysates (500 μg) were incubated with 30 μL glutathione resin and GST protein binding domains for one hour at 4°C to capture active small GTPases according to the manufacturer's protocol. After washing, the bound GTPase was recovered by eluting the GST-fusion protein from the glutathione resin. The purified GTPase was detected by Western blot using mouse monoclonal anti-KRAS (F234) (Santa Cruz Biotech, cat. #sc-30).

3D organoid culture and growth assay

After trypsinization, cells from the plastic culture were resuspended in ice-cold Matrigel (Corning) and incubated on prewarmed 6-cm plate to solidify. Organoids were cultured in Renaissance Essential Tumor Medium (Cellaria) with B-27 supplement (Thermo Fisher Scientific) and passaged over three times before performing any experiments. For the growth assay, organoids in Matrigel were dissociated into single cells using TrypLE Express (Invitrogen) at 37°C. One thousand cells were plated into each well of 384-well ultra-low attachment microplates (Corning) with the media and 10% Matrigel. On the next day, cells were treated with dose titration of trametinib or selumetinib for three days (from 1 nmol/L to 10 μmol/L). Each dose had more than three replicates. The viability assay was tested using CellTiter-Glo 3D (Promega) according to the manufacturer's protocol. The statistical significance was assessed using ANOVA and Tukey posttest for multiple comparison.

In vivo assays

Crl:NU-Foxn1nu mice (females, 8 weeks old) were purchased from Charles River. For lung colonization assays, KRaslox KRASMUT cells (1×106) were injected in the tail vein as single-cell suspension in 200 μL of sterile PBS. Mice were sacrificed synchronously at a single time point of one month, and lungs were collected in formalin. For drug treatment assays, KRaslox KRASMUT cells (2×106) were injected subcutaneously in a 1:1 mix of serum-free DMEM and Matrigel (phenol red–free; BD Biosciences) in both flanks of recipient mice. Once a palpable tumor formed, mice were randomly assigned to either selumetinib, sotorasib, or vehicle treatment, and measurements were taken daily using calipers. Selumetinib and sotorasib, resuspended in 2% hydroxypropyl methyl cellulose (HPMC), 1% Tween 80, were administered daily by oral gavage at a dose of 50 mg/kg. For IVIS longitudinal monitoring, KRaslox KRASMUT cells were transduced with pFUGW-Pol2-ffLuc2-eGFP (Addgene plasmid #71394) and FACS-sorted for GFP expression. KRaslox KRASMUT-luciferase cells (2 × 106) were injected in the tail vein of 8-week-old NCr nude mice (Taconic Biosciences) as single-cell suspension in 200 μL of sterile PBS and were monitored using an IVIS imaging system (PerkinElmer). All care and treatment of experimental animals were in strict accordance with Good Animal Practice as defined by the US Office of Laboratory Animal Welfare and approved by the Dana-Farber Cancer Institute Institutional Animal Care and Use Committee and by the Italian Health Minister (authorization n° 1227/2020-PR).

Study population

Clinicopathologic and genomic data from patients with NSCLC whose tumors underwent targeted next-generation sequencing at the Dana-Farber Cancer Institute who had provided written informed consent to institutional review board–approved protocols DF/HCC#02-180, #11-104, #13–364, and/or #17-000 were included in this study. This study was conducted in agreement with the Declaration of Helsinki.

Tumor mutational burden assessment and PD-L1 assessment

Tumor mutational burden (TMB), defined as the number of somatic, coding, base substitution, and indel mutations per megabase (Mb) of genome examined, was calculated from the DFCI OncoPanel next-generation sequencing platform. PD-L1 expression on tissue was assessed by IHC using an anti–PD-L1 rabbit monoclonal antibody (clone E1L3N, Cell Signaling Technology).

Clinical outcomes

Overall survival (OS) was defined as the time from the date of initial diagnosis to death. Patients who were still alive at the data cutoff were censored at the date of last contact. OS was compared between KRAS-mutated and KRAS wild-type cases, as well as across KRAS isoforms.

Co-occurrence and mutual exclusivity analysis

Fisher exact test P values and conditional odds ratios were used to assess co-occurrence and mutual exclusivity for genes with at least 2% frequency in the relevant groups. Positive odds ratios represented tendency to co-occurrence and negative odds ratios represented tendency to mutual exclusivity. Multiple comparison correction was performed using the Benjamini–Hochberg procedure using the qvalue package in R.

Gene-expression analysis from the TCGA

Gene-expression data were downloaded from the Firehose website (TCGA Firehose Legacy version), whereas somatic mutation data were downloaded from the cBioPortal website (cbioportal.org). The RSEM V2 values were used to represent gene expression and genes with counts fewer than 10 were filtered out. Gene-expression profiles were analyzed according to KRAS mutation status. Median expression within each group was used to estimate expression fold change (FC) to minimize the possible impact of outlier samples. Gene differential expression analyses across KRAS allele subgroups were conducted using R package DESeq2. P values were corrected for multiple hypothesis testing via false discovery rate (FDR) adjustment. FC threshold of an absolute value greater than 1.5 and FDR-adjusted P value threshold less than 0.1 were utilized to identify differentially expressed genes. Pathway enrichment analyses were conducted separately for upregulated and downregulated genes using R package Reactome.

Statistical analysis

We summarized continuous and categorical variables using percentages and medians. To test for differences between continuous variables we used the Wilcoxon-rank sum test and Kruskal–Wallis, when appropriate. The Fisher exact test was used to compare associations between categorical variables. Estimate event–time distributions were examined using the Kaplan–Meier methodology, and the log-rank test was used to test for differences in event-time distributions. Hazard ratios were estimated using the Cox regression models, as previously described (25). To investigate MEK/ERK inhibitor sensitivities for different KRAS-mutant isoforms across cancers, we grouped cancer cell lines based on their KRAS mutation status into the different groups. Mann–Whitney U test was used to compare drug sensitivities between two groups. All P values were two-sided with confidence intervals set at the 95% level. P < 0.05 were defined as significant. Multiple comparison correction was performed using the Benjamini–Hochberg procedure using the qvalue package in R. All statistical analyses were performed using R version 3.6.3.

Data availability statement

The institutional clinicopathologic and genomic data that support the findings of our study are available upon reasonable request from the corresponding author. RNA-seq data used in this study are publicly available from the TCGA (accession number: phs000178). Genomic data from the AACR project GENIE (Genomics Evidence Neoplasia Information Exchange) are publicly available (phs001337).

Generation of KRasloxKRASMUT cell line panel

Among KRAS-mutant lung cancer, codon 12 mutations predominate, accounting for nearly 90% of all KRAS mutations, followed by mutations in codons 13 and 61 (14). One question that arises from somatic genetic analysis is whether different KRAS mutations determine the clinical aspects of a given cancer. To investigate the role of individual KRAS isoforms on the biology of mutant KRAS avoiding context-dependent differences associated with different genetic backgrounds, we developed an isogenic system generated from HRas−/−; NRas−/−; KRaslox/lox mouse embryonic fibroblasts (MEF; ref. 24). In these cells, KRaslox/lox allele excision is controlled by a resident 4OHT-dependent CREERT2 recombinase. We transduced HRas−/−; NRas−/−; KRaslox/lox MEFs with retroviruses encoding for different human HA-tagged KRAS mutants, including the most frequent mutant isoforms detected in human lung adenocarcinoma (G12C, G12D, G12V, G12A, G13D, and Q61H; ref. 12).

In all cell lines, the presence of the respective KRAS mutation was confirmed by Sanger sequencing (data not shown). Cells were treated with 4OHT to abolish the expression of endogenous wild-type KRas, thus allowing the characterization of specific oncogenic KRAS mutations in isogenic cell lines without the interference of H- or NRas isoforms or the endogenous wild-type KRas (herein referred to KRaslox KRASMUT system; Fig. 1A). Interestingly, all KRAS mutants showed increased expression upon deletion of the endogenous wild-type KRas allele. Therefore, we utilized only 4OHT-treated cells in order to eliminate any bias imposed by the wild-type allele. In the full-serum condition, phosphorylation of ERK, MEK, SRC, and AKT was slightly increased in all KRAS mutants compared with wild-type control cells (Fig. 1B). In contrast, STAT3 and EGFR showed marked differences between mutants in terms of both phosphorylation and expression levels (Fig. 1B). Remarkably, KRASGTP levels upon stimulation with EGF showed profound variability among mutants, with KRAS G12D and G12V being already saturated in untreated condition in contrast with the other mutants that are still responsive to mitogenic stimulation (Fig. 1C).

Figure 1.

A, Schematic representation of the KRaslox KRASMUT system (top). HRas−/−; NRas−/−; KRaslox/lox MEFs were infected with 5-MOI of retroviruses encoding for different human HA-tagged KRAS mutants, selected with puromycin (1 μg/mL), treated with 4OHT (600 nmol/L) for at least 10 days, and probed by Western blot with the indicated antibodies (bottom). B, KRaslox KRASMUT cells were treated with 4OHT for 10 days and probed by Western blot with the indicated antibodies. Results are representative of one of three similar experiments. C, KRas-GTP levels and activation of downstream pMEK signaling in KRaslox KRASMUT cells in 0.1% FBS medium upon stimulation with EGF (50 ng/mL). Results are representative of one of three similar experiments. D, Growth rates of KRaslox KRASMUT cells in 10% or 0.5% FBS medium in 2D conditions as assessed by IncuCyte measurements (left, P < 0.0001 by unpaired Student t test). Results are representative of one of three similar experiments. Growth of KRaslox KRASMUT organoids was monitored by CTG assay. ANOVA analysis followed by Tukey multiple comparisons posttest was used for statistical analysis (P < 0.0001). E, KRaslox KRASMUT cells (1 × 106) were injected into the tail vein of nude mice (n = 3 per isoform). Animals were sacrificed after one month, and lung colonization was checked by serial sections stained with hematoxylin and eosin. The number of animals with evidence of lung colonization is indicated in the table. Scale bar, 100 μm.

Figure 1.

A, Schematic representation of the KRaslox KRASMUT system (top). HRas−/−; NRas−/−; KRaslox/lox MEFs were infected with 5-MOI of retroviruses encoding for different human HA-tagged KRAS mutants, selected with puromycin (1 μg/mL), treated with 4OHT (600 nmol/L) for at least 10 days, and probed by Western blot with the indicated antibodies (bottom). B, KRaslox KRASMUT cells were treated with 4OHT for 10 days and probed by Western blot with the indicated antibodies. Results are representative of one of three similar experiments. C, KRas-GTP levels and activation of downstream pMEK signaling in KRaslox KRASMUT cells in 0.1% FBS medium upon stimulation with EGF (50 ng/mL). Results are representative of one of three similar experiments. D, Growth rates of KRaslox KRASMUT cells in 10% or 0.5% FBS medium in 2D conditions as assessed by IncuCyte measurements (left, P < 0.0001 by unpaired Student t test). Results are representative of one of three similar experiments. Growth of KRaslox KRASMUT organoids was monitored by CTG assay. ANOVA analysis followed by Tukey multiple comparisons posttest was used for statistical analysis (P < 0.0001). E, KRaslox KRASMUT cells (1 × 106) were injected into the tail vein of nude mice (n = 3 per isoform). Animals were sacrificed after one month, and lung colonization was checked by serial sections stained with hematoxylin and eosin. The number of animals with evidence of lung colonization is indicated in the table. Scale bar, 100 μm.

Close modal

Growth properties of different KRAS mutants in vitro and in vivo

Next, we aimed at characterizing the proliferation kinetics of KRaslox KRASMUT cells both in vitro and in vivo. We tested growth properties of different KRAS mutants by IncuCyte in full-serum and starving conditions. All mutants displayed a strong proliferative advantage over wild-type KRAS controls in full-serum; nevertheless, no significant differences were observed among mutants (Fig. 1D). Under starving conditions, however, a clear stratification was evident, with G12D, and G12V to a lesser extent, showing the best fitness over the other KRAS mutants (Fig. 1D). Of note, G12D and G12V are the mutant isoforms displaying higher GTP-bound fraction in limited serum conditions (Fig. 1C). Similar results were obtained in hypoxic conditions, with G12D mutant cells showing improved fitness compared with the other KRAS mutants upon concomitant starvation (Supplementary Fig. S1A). In 3D conditions, all KRAS-mutant isoforms showed comparable growth kinetics with the exception of Q61H, which was significantly less proliferative (Fig. 1D). The advantage of G12D versus the other mutants was also maintained in tumor growth assays in vivo. We injected KRaslox KRASMUT cells expressing different mutants into the tail vein of nude mice and euthanized the animals after one month in order to check for colonization property into the lungs. At histologic evaluation, G12D-mutant cells displayed tumor nodule formation into the lungs, whereas all the other mutants did not (Fig. 1E). Using the same mutant cells expressing luciferase gene, we noninvasively monitored tumor growth in the mice. Consistent with Fig. 1E and in vitro results, G12D and G12V mutants were established in the mouse lung much quicker than other mutants (Supplementary Fig. S1B and S1C). These results confirmed an in vivo advantage for KRASG12D mutants (26), although further assessment at longer time points should be performed in order to detect delayed tumor onset in mutants other than G12D.

Sensitivity to MEK inhibitors and G12C inhibitors in vitro and in vivo

Due to the prominent role of the MAPK pathway in KRAS signaling, MEK inhibitors were thought to be able to achieve disease control in KRAS-mutant tumors. However, phase III clinical trials demonstrated no overall benefit compared with chemotherapy (4). Retrospective analysis in different subpopulations of KRAS codon mutations showed that patients with KRAS G12C or G12V receiving selumetinib + docetaxel had improved OS compared with other KRAS mutations, suggesting that the type of KRAS mutations may affect MEK inhibitor sensitivity in LUAD (20). We next surveyed the CGP pharmacogenomic data set (27) to identify correlations between sensitivity to MEK inhibitors and KRAS-mutant isoforms in a broad panel of lung, pancreas, and colon cancer cell lines. We could not observe any obvious trend for increased sensitivity for specific isoforms other than G12R, likely due to the limited representation of small number of cell lines (Supplementary Fig. S2). Considering that cell line heterogeneity poses challenging limits to the characterization of KRAS isoform as a single variable to predict MEK inhibitor sensitivity, we tested a panel of MAPK inhibitors in our isogenic KRaslox KRASMUT system. GSK1120212 (trametinib) showed similar antiproliferative effects in all KRAS mutants, whereas selumetinib and the other MAPK inhibitors exhibited up to 10-fold IC50 variability in different KRAS isoforms, being G12C and Q61H among the most sensitive (Fig. 2A; Supplementary Fig. S3; refs. 28, 29). The same trend was observed in 3D culture (Fig. 2B). Western blot assessment of the MAPK pathway confirmed dose-dependent reduction of pERK in selumetinib-treated KRaslox KRASMUT cells, with the exception of G12A and G13D mutants (Fig. 2C). To validate these findings into an in vivo setting, we implanted KRaslox KRASMUT cells into nude mice and followed tumor growth in the presence or absence of selumetinib treatment (50 mg/kg daily). In agreement with our findings in 2D and 3D conditions, we could stratify the outcome as tumor regression (G12C and Q61H), stable disease (G12D, G12V, and G13D), and no response (G12A) based on tumor volume FC on treatment (Fig. 2D).

Figure 2.

A, Comparison of IC50 values to the MEK inhibitor selumetinib between KRaslox KRASMUT cells expressing different KRAS isoforms. Error bars, mean ± SD. Results are representative of one of three similar experiments. The table indicates cell lines ranked by IC50 values (lowest to highest). B, Log GR50 values for selumetinib and trametinib are shown in the box plot. The numbers and vertical lines through each box indicate the median. C, KRaslox KRASMUT cells were seeded and treated with the indicated concentrations of selumetinib for 12 hours and then probed by Western blot with the indicated antibodies. Results are representative of one of three similar experiments. D, KRaslox KRASMUT cells (2 × 106) were injected subcutaneously with Matrigel (1:1) into nude mice. Mice with palpable tumors were randomized into two cohorts (n = 6) and treated with vehicle or selumetinib (50 mg/kg) daily by oral gavage for one week. Tumor size was measured every day with a caliper. Mean FC in tumor volume relative to initial tumor volume is shown. Error bars, mean ± SD.

Figure 2.

A, Comparison of IC50 values to the MEK inhibitor selumetinib between KRaslox KRASMUT cells expressing different KRAS isoforms. Error bars, mean ± SD. Results are representative of one of three similar experiments. The table indicates cell lines ranked by IC50 values (lowest to highest). B, Log GR50 values for selumetinib and trametinib are shown in the box plot. The numbers and vertical lines through each box indicate the median. C, KRaslox KRASMUT cells were seeded and treated with the indicated concentrations of selumetinib for 12 hours and then probed by Western blot with the indicated antibodies. Results are representative of one of three similar experiments. D, KRaslox KRASMUT cells (2 × 106) were injected subcutaneously with Matrigel (1:1) into nude mice. Mice with palpable tumors were randomized into two cohorts (n = 6) and treated with vehicle or selumetinib (50 mg/kg) daily by oral gavage for one week. Tumor size was measured every day with a caliper. Mean FC in tumor volume relative to initial tumor volume is shown. Error bars, mean ± SD.

Close modal

Considering that preclinical in vivo studies demonstrated that in KRAS-mutant LUAD, combined SHP2/MEK inhibition is therapeutically effective (30, 31), we tested the same combination in KRaslox KRASMUT cells. Interestingly, we observed that combined SHP2/MEK blockade was more effective in G12C, G12D, and G12V mutants compared with the other mutants, in agreement with previous findings (refs. 31, 32; Supplementary Fig. S4).

Given the very recent FDA approval of the selective G12C inhibitor sotorasib (CodeBreaK 100, NCT03600883), we aimed at assessing the efficacy of G12C inhibition in our isoform-specific KRASG12C system, both in vitro and in vivo. As expected, drug–response curves with sotorasib demonstrated a clear selectivity for G12C-mutant cells compared with the other KRAS-mutant isoforms (Fig. 3A). To determine how G12C inhibition compared with MEK inhibition in this model, we next implanted KRaslox KRASG12C cells into nude mice and randomized these mice to receive either vehicle, selumetinib (50 mg/kg daily), sotorasib (50 mg/kg daily), or selumetinib + sotorasib. Interestingly, selumetinib exerted better therapeutic response in comparison with sotorasib, possibly likely due to the delay in implementing feedback mechanisms for MEK inhibitors as compared with selective G12C inhibitors as single agent. Importantly, combined treatment with selumetinib + sotorasib resulted in complete growth abrogation and disease control in mice treated with this combination (Fig. 3BE). This preliminary finding further supports the hypothesis that combined KRAS G12C and MEK inhibition may improve clinical outcomes in patients with KRAS-mutant NSCLC over G12C inhibition alone. A phase Ib clinical trial of sotorasib in combination with other anticancer agents, including MEK inhibitors is currently ongoing (NCT04185883).

Figure 3.

A, Comparison of IC50 values to the selective G12C inhibitor sotorasib between KRaslox KRASMUT cells expressing different KRAS isoforms. Error bars, mean ± SD. Results are representative of one of three similar experiments. B, KRaslox KRASG12C cells (2 × 106) were injected subcutaneously with Matrigel (1:1) into nude mice. Mice with palpable tumors were randomized into four cohorts and treated with vehicle, selumetinib (50 mg/kg), sotorasib (50 mg/kg), or combo treatment daily by oral gavage. Tumor size was measured every day with a caliper. Tumor volume change relative to initial tumor volume is shown for individual tumors. C, Bar graph showing average tumor volume FC to baseline at endpoint. Error bars, mean ± SD. D, Waterfall plot showing individual tumor volume change from baseline (%) at endpoint. E, Macroscopic photographs after 10 days of treatment and tumor volume tracking of KRaslox KRASG12C cells.

Figure 3.

A, Comparison of IC50 values to the selective G12C inhibitor sotorasib between KRaslox KRASMUT cells expressing different KRAS isoforms. Error bars, mean ± SD. Results are representative of one of three similar experiments. B, KRaslox KRASG12C cells (2 × 106) were injected subcutaneously with Matrigel (1:1) into nude mice. Mice with palpable tumors were randomized into four cohorts and treated with vehicle, selumetinib (50 mg/kg), sotorasib (50 mg/kg), or combo treatment daily by oral gavage. Tumor size was measured every day with a caliper. Tumor volume change relative to initial tumor volume is shown for individual tumors. C, Bar graph showing average tumor volume FC to baseline at endpoint. Error bars, mean ± SD. D, Waterfall plot showing individual tumor volume change from baseline (%) at endpoint. E, Macroscopic photographs after 10 days of treatment and tumor volume tracking of KRaslox KRASG12C cells.

Close modal

Clinicopathologic and genomic correlation of KRAS variants in patients with non-squamous NSCLC

In order to translate our findings based on genetically defined cell lines to a clinically relevant context, a total of 3,560 patients with NSCLCs who underwent successful comprehensive tumor genomic profiling at the Dana-Farber Cancer Institute were identified. Of these, 3,126 (87.8%) were non-squamous NSCLC and were included in the final analysis (Supplementary Fig. S5A). The median age of the 3,126 patients with non-squamous NSCLC was 66 (range, 18–99), 61.2% were female, 18.6% were current smokers, 58.4% were former smokers, and 18.6% were never smokers. KRAS mutation (KRASMUT) occurred in 1,131 cases (36.2%), whereas the remaining 1,995 cases (63.8%) had a KRAS wild-type (KRASWT) genotype (Supplementary Table S1). The most common KRAS mutations identified occurred at codon 12 (990/1131, 87.5%), of which the KRAS G12C was the most frequent (476/990, 47.2%; Supplementary Fig. S5B).

We first investigated differences in clinicopathologic and genomic factors according to KRAS mutation status. Compared with KRASWT cases, patients with KRASMUT tumors were more likely to be older (P < 0.001), females (P < 0.0001), current/former smokers (P < 0.0001), and to have adenocarcinoma histology (P < 0.01), consistent with previous reports (33, 34). Baseline clinicopathologic features of patients with KRASWT versus KRASMUT are shown in Supplementary Table S2. We next focused on comutations in preselected genes of interest that define major subsets of NSCLC, including TP53, STK11, and KEAP1. Of note, although mutations in TP53 were more likely to occur in KRASWT tumors (P < 0.0001), STK11 and KEAP1 were significantly enriched in KRAS-mutated tumors (P < 0.0001; Supplementary Fig. S5C). We also examined whether PD-L1 expression and TMB distributions differed between KRASMUT and KRASWT NSq-NSCLCs. We noted that both median PD-L1 expression (10 vs. 5%, P < 0.001) and median TMB (9.8 vs. 8.4 mut/Mb, P < 0.0001) were significantly higher in KRASMUT versus KRASWT tumors (Supplementary Fig. S5D).

Next, we examined the impact of KRAS mutation on survival among patients with NSq-NSCLC. Although we found no significant impact of KRAS mutation on OS in all comers (HR: 1.07, P = 0.21), as well as among patients with stage I–III NSq-NSCLC (HR: 1.08, P = 0.39), we observed a significant deleterious impact of KRAS mutation among patients with stage IV tumors (16.2 vs. 25.5 months, HR:1.37, P < 0.0001; Fig. 4AC).

Figure 4.

OS in (A) all comers with non-squamous NSCLC, and (B) stage I–III, and (C) stage IV disease according to KRAS mutation status. OS in (D) all comers with non-squamous NSCLC, and (E) stage I–III, and (F) stage IV disease according to KRAS alleles.

Figure 4.

OS in (A) all comers with non-squamous NSCLC, and (B) stage I–III, and (C) stage IV disease according to KRAS mutation status. OS in (D) all comers with non-squamous NSCLC, and (E) stage I–III, and (F) stage IV disease according to KRAS alleles.

Close modal

Although we identified a detrimental impact of KRAS mutation among patients with advanced NSq-NSCLC, this difference likely reflects the use of highly effective targeted therapies among patients with KRAS wild-type tumors harboring EGFR mutations and ALK fusions.

Because we have shown that different KRAS mutation have different in vitro and in vivo growth properties, and differential responses to MEK inhibitors, we lastly investigated whether different KRAS mutation affected survival in patients with NSq-NSCLC. The clinicopathologic characteristics of patients according to KRAS alleles are shown in Supplementary Table S3. Consistent with previous reports (35), we found a significantly higher proportion of never smokers among patients with KRAS G12D mutation (21.8%) as opposed to the other KRAS variants (<10%). Although there was no difference in terms of enrichment in TP53 mutation across the KRAS variants, the rates of concurrent STK11 and KEAP1 mutations were highest among KRAS codon 13 mutations (Supplementary Fig. S5C). Pairwise comparisons between KRAS alleles in terms of TP53, STK11, and KEAP1 comutation patterns are shown in Supplementary Fig. S6A. When we examined PD-L1 expression and TMB distributions in this across the different KRAS alleles, we found that the median TMB differed significantly across the seven groups, with KRAS G12D-mutated tumors having the lowest number of non-synonymous mutations/megabase (Supplementary Fig. S5D). By contrast, no difference in PD-L1 expression was observed among these groups (Supplementary Fig. S5D). Pairwise comparisons between KRAS alleles in terms of PD-L1 and TMB distributions are shown in Supplementary Fig. S6B.

We lastly analyzed the impact of KRAS alleles on survival in patients with non-squamous NSCLC. We found no significant difference across KRAS mutation subtype in all comers, as well as among those with stage I–III or stage IV disease (Fig. 4DF).

KRAS variants have different comutation patterns and gene-expression profiles in patients with NSCLC

Although we did not find differences in survival in patients with NSCLC with different KRAS alleles, we identified several differences in clinicopathologic and genomic factors among NSCLC harboring different KRAS alleles in the DFCI cohort. Therefore, we hypothesized that one of the reasons by which our findings in isogenic models did not translate into different clinical outcomes could be the biological heterogeneity of KRAS-mutant LUADs. To further dissect this heterogeneity in patients with KRAS-mutant LUADs, we interrogated a large publicly available cohort of 12,931 LUADs (GENIE v.9.0) that underwent tumor genomic profiling to comprehensively determine differences in comutation patterns across KRAS variants. After QC removing duplicate samples and gene annotation, 10,663 unique samples were included in the final analysis. KRAS mutation was detected in 3,843 of cases (36%). In this cohort, the most common genomic alterations included TP53 (33%), STK11 (16%), ATM (11%), KEAP1 (11%), and RBM10 (7%; Fig. 5A). When compared with KRAS wild-type cases, however, KRAS-mutant LUADs were found to be significantly enriched in STK11, KEAP1, ATM, RBM10 mutations, whereas TP53, EGFR, BRAF, and ERBB2 were less likely to co-occur with any KRAS mutation (Supplementary Fig. S7A and S7B).

Figure 5.

A, Oncoprint plot of KRAS-mutant LUADs in the GENIE v9.0 data set. B, Volcano plots showing comutation patterns in KRAS G12C, G12D, G12V, G12A, G13X, and G61X compared with KRAS wild-type LUADs. Gene ontology analysis showing (C) downregulated and (D) upregulated pathways in KRAS G12C, G12D, G12V, G12A, G13X, and G61X compared with KRAS wild-type LUADs.

Figure 5.

A, Oncoprint plot of KRAS-mutant LUADs in the GENIE v9.0 data set. B, Volcano plots showing comutation patterns in KRAS G12C, G12D, G12V, G12A, G13X, and G61X compared with KRAS wild-type LUADs. Gene ontology analysis showing (C) downregulated and (D) upregulated pathways in KRAS G12C, G12D, G12V, G12A, G13X, and G61X compared with KRAS wild-type LUADs.

Close modal

We next focused on the individual KRAS variants and compared the comutation patterns of each KRAS variant with KRAS wild-type LUADs as reference. We identified that all KRAS variants tended to be mutually exclusive with EGFR mutation and were more likely to lack concurrent TP53 mutations (Fig. 5B). Of note, among LUADs with codon 12 mutations, STK11 and ATM mutations was significantly enriched among tumors harboring KRAS G12C, G12A, and G12V mutations, whereas such enrichment was not detected in LUADs with KRAS G12D (Fig. 5B). Additionally, we also found that KEAP1 mutation was significantly enriched among KRAS G12C and KRAS G13X LUADs. Importantly, the highest frequency of concurrent STK11 and KEAP1 mutation was observed in tumors with KRAS codon 13 mutations (Supplementary Fig. S7C). Pairwise comparisons in concurrent and mutually exclusive mutations between each KRAS variant and the remaining KRAS alleles are reported in Supplementary Fig. S8A and S8B.

Having shown that the KRAS-mutant LUADs are highly heterogeneous in terms of concurrent genomic alterations, we lastly examined whether tumors with different KRAS mutations also had different gene-expression profiles. To address this question, we performed a gene ontology analysis of differentially expressed genes in each KRAS variant versus KRAS wild-type tumors in the TCGA LUAD cohort. We found that several pathways were uniquely upregulated and downregulated in specific KRAS isoform (Fig. 5C and D). Interestingly, KRAS G12D-mutant LUADs showed a significant downregulation of pathways involved in chromosome maintenance, DNA double break repair, and cellular senescence (Fig. 5C), whereas KRAS G13X LUADs had a significant downregulation of pathways involved in innate and adaptative immunity, including PD-1, CD28, INFγ, and IL2 signaling, which is consistent with the high rate of concurrent STK11 and/or KEAP1 (36, 37) mutations we identified in this group of LUADs. A full list of differentially regulated pathways in all KRAS isoforms is reported in Supplementary Table S4.

Activating mutations in the KRAS oncogene are known to trigger and sustain the development of tumors with different histology, being particularly frequent in lung, colon, and pancreatic cancers (1). The comparison between predicted and observed frequency of different mutant KRAS isoforms suggests the existence of tissue-specific mechanisms of biological selection (14); however, the connection between KRAS-mutant isoforms and therapeutic vulnerabilities has not been fully addressed yet. Very recently, the development of two specific inhibitors of the KRAS-G12C allele, sotorasib and adagrasib, which were granted accelerated FDA approval and BTD, respectively, in patients with previously treated advanced NSCLC (10, 38–41) highlighted the need to identify new isoform-specific therapeutic strategies. Here we demonstrate that KRAS isoforms are characterized by different oncogenic properties and therapeutic sensitivities in isogenic models, and that KRAS-mutant LUADs are highly heterogeneous in terms of clinicopathologic, genomic, and transcriptomic features.

Studies have been inconsistent in reporting whether KRAS isoforms differ in terms of oncogenic properties, preferred signaling pathways, genomics, and clinical outcomes in patients with LUAD (1). Previously, NSCLC patient samples and cell lines harboring KRAS G12A have been shown to primarily rely on phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) signaling, whereas those with mutant KRAS G12C or mutant KRAS G12V are more likely to activate Ral signaling and decrease growth factor-dependent Akt activation (19). Similarly, in KRAS-overexpressing clones from human cell lines harboring the three most common KRAS mutations in NSCLC (G12C, G12V, and G12D), the expression of specific KRAS isoforms induced a different sensitivity pattern to cytotoxic agents, such as cisplatin, pemetrexed, and taxanes (42). Nonetheless, subsequent retrospective analysis yielded highly variable results in terms of clinical outcomes to either chemotherapy or PD-(L)1 inhibition in patients with advanced NSCLC, as well as among those with resected tumors (19, 42, 43). However, data on KRAS comutation pattern have been underreported in these studies; therefore, dissecting the biological differences among KRAS isoforms and their impact on clinical outcomes in patients with LUAD remains challenging.

To study the oncogenic properties of KRAS isoforms in a genetic-controlled system with the same genomic background, we generated isogenic models to dissect potential isoform-dependent therapeutic vulnerabilities. It has already been demonstrated that specific KRAS isoforms exhibit diverse biochemical and biological features in vitro and in vivo (19, 21, 25, 44, 45). However, the therapeutic impact of different KRAS alleles on treatment outcome has had limited investigated so far (29, 46, 47). In our KRaslox KRASMUT platform expressing the most frequent KRAS-mutant alleles (G12C, G12D, G12V, G12A, G13D, and Q61H), we observed that G12D-mutant cells show a significant growth advantage over the other mutants both in vitro and in vivo and are characterized, together with G12V, by the highest levels of GTP-bound fraction even in the absence of upstream EGF stimulation.

To determine whether these differences translated in differential clinical outcomes, we analyzed a large cohort of patients with LUAD. Although we did not identify significant differences in survival in patients with either early-stage or advanced LUADs according to the different KRAS variants, we found that each KRAS isoform was associated with distinct clinicopathologic, genomic, and transcriptomic features, which may have affected our ability to identify differences in outcomes. These data are in agreement with current knowledge confirming no significant differences in survival in LUAD patients according to specific KRAS-mutant alleles. Still, KRAS isoform specificity could be used as a stratification criterion for better tailored therapies based on exclusive vulnerabilities. For instance, KRAS G12D-mutated LUADs were more likely to develop in patients with no history of tobacco use. Consistently, tumors with KRAS G12D mutation also had the lowest TMB among KRAS isoforms, were less likely to harbor concurrent mutations in STK11 or KEAP1, and had a marked downregulation of pathways involved in cellular senescence and chromosome maintenance. In contrast, the highest rate of concurrent mutations in STK11 and KEAP1 was identified among KRAS G13X-mutated tumors. Concurrent mutations in STK11 and KEAP1 define unique subsets of KRAS-mutant LUADs, with distinct immunophenotype and transcriptomic profile, and are generally characterized by significantly worse survival with both chemotherapy and PD-(L)1 blockade (48, 49). Consistently, we identified that G13X LUADs had a significant downregulation of pathways involved in innate and adaptative immunity, such as PD-1 signaling, TCR signaling, and INFγ signaling, among others. It should be highlighted that gene-expression profiling was performed in NSCLC samples from the TCGA, rather than in our isogenic models. Therefore, these results should be interpreted in the context of the different genomic backgrounds which characterize each KRAS allele. Overall, our results support the hypothesis that in addition to the allele-specific and -intrinsic differences that we identified in our isogenic, each KRAS allele has a different spectrum of comutations and transcriptomic profiles in patient samples, which may not be dependent uniquely from KRAS alleles, but still have great biological relevance. Together, these results may have direct and immediate implications for the design and interpretation of panKRAS or multi/KRAS-targeted therapies, as each individual allele has unique oncogenic properties and features.

To further dissect this question, we also investigated differential response to targeted therapy in KRaslox KRASMUT cells. As a proof of concept we chose MEK inhibitors, which showed promising results in a phase II clinical trial but failed to provide any significant benefit in addition to chemotherapy in KRAS-mutant LUAD in a phase III trial (4). Interestingly, retrospective analysis of the phase II trial showed a trend for KRAS G12C and G12V patients toward greater improvement in OS, progression-free survival, and overall response rate compared with other KRAS mutations (20). Indeed, we observed a consistently increased sensitivity of G12C and Q61H mutants with a panel of MEK inhibitors in vitro, which was maintained in a preclinical setting in vivo where G12C and Q61H implants showed greater tumor volume reduction compared with other mutants. This effect may partly rely on increased affinity to CRAF for some isoforms (29, 50). Also, we validated the convenience of our preclinical tool in surveying combination therapies that can be easily tested in high-throughput screening, such as G12C/MEK inhibitors or SHP2/MEK inhibitors (30, 31).

Our study has limitations inherent to the artificial generation of our isogenic model and the retrospective assessment of clinical outcomes. However, we believe that our model does isolate the biological contribution of each isoform and allow the exploration of drug sensitivity in the absence of potential confounding factors. This is of particular importance, given that our results indicate that each KRAS isoform is associated with unique comutation patterns and transcriptomic profiles that may affect drug sensitivity independently from KRAS-intrinsic activity. Our results suggest that the use of specific genetically defined in vitro tools together with the development of large patient-derived data sets will facilitate the identification of potentially actionable vulnerabilities for KRAS-mutant NSCLC.

In summary, this study indicates that KRAS isoforms are biologically and clinically heterogeneous, and that such heterogeneity should be considered to optimize drug development strategies for KRAS-mutant NSCLC.

A.J. Redig reports grants from NCI, Conquer Cancer Foundation, A Breath of Hope Foundation, and Harvard Catalyst/KL2 during the conduct of the study. During the time that A.J. Redig was employed by Dana-Farber (ending in September 2017), A.J. Redig had financial consulting relationships that did not fund any of the work included in this paper but would have been present during the time when contributions to this manuscript were conducted; these relationships are now 36 months from publication and are itemized as follows: (i) Boehringer Ingelheim: personal fee; (ii) Medtronic: personal fee, nonfinancial support; (iii) Roche: personal fee, nonfinancial support; (iv) Ariad: personal fee; and (v) AstraZeneca: personal fee, nonfinancial support, grant funding. A.J. Redig also reports employment by Takeda Pharmaceuticals (November 2017–February 2020) and Blackstone Life Sciences (August 2020–present). P.C. Gokhale reports grants from Marengo Therapeutics, Epizyme Inc, Daiichi Sankyo, Foghorn Therapeutics, 28-7 Therapeutics, and Moderna outside the submitted work. M.M. Awad reports grants and personal fees from Genentech, Bristol Myers Squibb, and AstraZeneca; grants from Lilly; and personal fees from Merck, Maverick, Blueprint Medicine, Syndax, Ariad, Nektar, Gritstone, ArcherDx, Mirati, NextCure, Novartis, EMD Serono, NovaRx, and Iovance outside the submitted work. P.A. Jänne reports grants and personal fees from AstraZeneca, Boehringer Ingelheim, Eli Lilly, Daiichi Sankyo, and Takeda Oncology; personal fees from Pfizer, Roche/Genentech, Chugai, SFJ Pharmaceuticals, Voronoi, Biocartis, Novartis, Sanofi Oncology, Mirati Therapeutics, Transcenta, Silicon Therapeutics, Syndax, Nuvalent, Bayer, Esai, Allorion Therapeutics, Accutar Biotech, and AbbVie; and grants from Revolution Medicines and PUMA outside the submitted work. In addition, P.A. Jänne has a patent for EGFR mutations issued and licensed to LabCorp. C. Ambrogio reports other support from Revolution Medicines, Verastem, and Boehringer Ingelheim outside the submitted work. No disclosures were reported by the other authors.

B. Ricciuti: Conceptualization, validation, investigation, methodology, writing–original draft. J. Son: Validation, investigation, methodology, writing–original draft. J.J. Okoro: Validation, investigation, methodology. A. Mira: Validation, investigation, methodology. E. Patrucco: Validation, investigation, methodology. Y. Eum: Validation, investigation, methodology. X. Wang: Validation, investigation, methodology. R. Paranal: Validation, investigation, methodology. H. Wang: Formal analysis. M. Lin: Validation, investigation, methodology. H.M. Haikala: Validation, investigation, methodology. J. Li: Validation, investigation, methodology. Y. Xu: Validation, investigation, methodology. J.V. Alessi: Validation, investigation, methodology. C. Chhoeu: Methodology. A.J. Redig: Formal analysis. J. Köhler: Formal analysis. K.H. Dholakia: Methodology. Y. Chen: Methodology. E. Richard: Validation, investigation, methodology. M.-J. Nokin: Validation, investigation, methodology. D. Santamaria: Formal analysis. P.C. Gokhale: Formal analysis. M.M. Awad: Conceptualization, supervision, writing–original draft. P.A. Jänne: Conceptualization, supervision, writing–original draft. C. Ambrogio: Conceptualization, supervision, funding acquisition, validation, investigation, methodology, writing–original draft.

This work was funded by the Giovanni Armenise–Harvard Foundation, the Lung Cancer Research Foundation, the International Lung Cancer Foundation, the European Research Council under the European Union's Horizon 2020 research and innovation program (grant agreement No. 101001288) and AIRC under IG 2021 ID. 25737 project (to C. Ambrogio). B. Ricciuti's work was supported by the Conquer Cancer Foundation of ASCO YIA, and the IASLC Fellowship Award. C. Ambrogio is supported by the Zanon di Valgiurata family through Justus SS.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Salgia
R
,
Pharaon
R
,
Mambetsariev
I
,
Nam
A
,
Sattler
M
.
The improbable targeted therapy: KRAS as an emerging target in non-small cell lung cancer (NSCLC)
.
Cell Reports Med
2021
;
2
:
100186
.
2.
Judd
J
,
Abdel Karim
N
,
Khan
H
,
Naqash
AR
,
Baca
Y
,
Xiu
J
, et al
.
Characterization of KRAS mutation subtypes in non–small cell lung cancer
.
Mol Cancer Ther
2021
;
20
:
2577
84
.
3.
Köhler
J
,
Catalano
M
,
Ambrogio
C
.
Back to the bench? MEK and ERK inhibitors for the treatment of KRAS mutant lung adenocarcinoma
.
Curr Med Chem
2017
;
24
:
1
17
.
4.
Jänne
P
,
van den Heuvel
M
,
Barlesi
F
,
Cobo
M
,
Mazieres
J
,
Crinò
L
, et al
.
Selumetinib plus docetaxel compared with docetaxel alone and progression-free survival in patients with KRAS-mutant advanced non-small cell lung cancer: the SELECT-1 randomized clinical trial
.
JAMA
2017
;
317
:
1844
53
.
5.
Planchard
D
,
Popat
S
,
Kerr
K
,
Novello
S
,
Smit
EF
,
Faivre-Finn
C
, et al
.
Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
2018
;
29
:
iv192
237
.
6.
Ambrogio
C
,
López-Gómez
G
,
Falcone
M
,
Vidal
A
,
Nadal
E
,
Crosetto
N
, et al
.
Combined inhibition of DDR1 and Notch signaling is a therapeutic strategy for KRAS-driven lung adenocarcinoma
.
Nat Med
2016
;
22
:
270
7
.
7.
Manchado
E
,
Weissmueller
S
,
Morris
JP
,
Chen
C-C
,
Wullenkord
R
,
Lujambio
A
, et al
.
A combinatorial strategy for treating KRAS-mutant lung cancer
.
Nature
2016
;
534
:
647
51
.
8.
Awad
MM
,
Liu
S
,
Rybkin
II
,
Arbour
KC
,
Dilly
J
,
Zhu
VW
, et al
.
Acquired resistance to KRAS G12C inhibition in cancer
.
N Engl J Med
2021
;
384
:
2382
93
.
9.
Skoulidis
F
,
Li
BT
,
Dy
GK
,
Price
TJ
,
Falchook
GS
,
Wolf
J
, et al
.
Sotorasib for lung cancers with KRAS p.G12C mutation
.
N Engl J Med
2021
;
384
:
2371
81
.
10.
Riely
GJ
,
Ou
S-HI
,
Rybkin
I
,
Spira
A
,
Papadopoulos
K
,
Sabari
JK
, et al
.
99O_PR KRYSTAL-1: activity and preliminary pharmacodynamic (PD) analysis of adagrasib (MRTX849) in patients (Pts) with advanced non–small cell lung cancer (NSCLC) harboring KRASG12C mutation
.
J Thorac Oncol
2021
;
16
:
S751
S752
.
11.
Santos
E
,
Martin-Zanca
D
,
Reddy
EP
,
Pierotti
Ma
,
Della
Porta G
,
Barbacid
M
.
Malignant activation of a K-ras oncogene in lung carcinoma but not in normal tissue of the same patient
.
Science
1984
;
223
:
661
4
.
12.
Ambrogio
C
,
Köhler
J
,
Zhou
Z
,
Wang
H
,
Paranal
R
,
Li
J
, et al
.
KRAS dimerization impacts MEK inhibitor sensitivity and oncogenic activity of mutant KRAS
.
Cell
2018
;
172
:
857
68
.
13.
Zhou
Y
,
Prakash
P
,
Liang
H
,
Cho
KJ
,
Gorfe
AA
,
Hancock
JF
.
Lipid-sorting specificity encoded in K-Ras membrane anchor regulates signal output
.
Cell
2017
;
168
:
239
51
.
14.
Cook
J
,
Melloni
G
,
Gulhan
D
,
Park
P
,
Haigis
KM
.
The origins and genetic interactions of KRAS mutations are allele- and tissue-specific
.
Nat Commun
2021
;
12
:
1808
.
15.
Hood
FE
,
Klinger
B
,
Newlaczyl
AU
,
Sieber
A
,
Dorel
M
,
Oliver
SP
, et al
.
Isoform-specific Ras signaling is growth factor dependent
.
Mol Biol Cell
2019
;
30
:
1108
17
.
16.
Tang
R
,
Shuldiner
EG
,
Kelly
M
,
Murray
CW
,
Hebert
JD
,
Andrejka
L
, et al
.
Multiplexed identification of RAS paralog imbalance as a driver of lung cancer growth
.
bioRxiv
2021
.
Available from:
https://doi.org/10.1101.2021.07.08.451571.
17.
Tejpar
S
,
Celik
I
,
Schlichting
M
,
Sartorius
U
,
Bokemeyer
C
,
Van Cutsem
E
.
Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab
.
J Clin Oncol
2012
;
30
:
3570
7
.
18.
Burd
CE
,
Liu
W
,
Huynh
MV
,
Waqas
MA
,
Gillahan
JE
,
Clark
KS
, et al
.
Mutation-specific RAS oncogenicity explains NRAS codon 61 selection in melanoma
.
Cancer Discov
2014
;
4
:
1418
29
.
19.
Ihle
NT
,
Byers
LA
,
Kim
ES
,
Saintigny
P
,
Lee
JJ
,
Blumenschein
GR
, et al
.
Effect of KRAS oncogene substitutions on protein behavior: implications for signaling and clinical outcome
.
J Natl Cancer Inst
2012
;
104
:
228
39
.
20.
Jänne
Pa
,
Smith
I
,
McWalter
G
,
Mann
H
,
Dougherty
B
,
Walker
J
, et al
.
Impact of KRAS codon subtypes from a randomised phase II trial of selumetinib plus docetaxel in KRAS mutant advanced non-small-cell lung cancer
.
Br J Cancer
2015
;
113
:
199
203
.
21.
Hammond
DE
,
Mageean
CJ
,
Rusilowicz
EV
,
Wickenden
JA
,
Clague
MJ
,
Prior
IA
.
Differential reprogramming of isogenic colorectal cancer cells by distinct activating KRAS mutations
.
J Proteome Res
2015
;
14
:
1535
46
.
22.
Stolze
B
,
Reinhart
S
,
Bulllinger
L
,
Fröhling
S
,
Scholl
C
.
Comparative analysis of KRAS codon 12, 13, 18, 61, and 117 mutations using human MCF10A isogenic cell lines
.
Sci Rep
2015
;
5
:
8535
.
23.
Brubaker
DK
,
Paulo
JA
,
Sheth
S
,
Poulin
EJ
,
Popow
O
,
Joughin
BA
, et al
.
Proteogenomic network analysis of context-specific KRAS signaling in mouse-to-human cross-species translation
.
Cell Syst
2019
;
9
:
258
70
.
24.
Drosten
M
,
Dhawahir
A
,
Sum
EYMM
,
Urosevic
J
,
Lechuga
CG
,
Esteban
LM
, et al
.
Genetic analysis of Ras signalling pathways in cell proliferation, migration and survival
.
EMBO J
2010
;
29
:
1091
104
.
25.
Ricciuti
B
,
Jones
G
,
Severgnini
M
,
Alessi
JV
,
Recondo
G
,
Lawrence
M
, et al
.
Early plasma circulating tumor DNA (ctDNA) changes predict response to first-line pembrolizumab-based therapy in non–small cell lung cancer (NSCLC)
.
J Immunother Cancer
2021
;
9
:
e001504
.
26.
Winters
I
,
Chiou
S
,
Paulk
N
,
McFarland
C
,
Lalgudi
P
,
Ma
R
, et al
.
Multiplexed in vivo homology-directed repair and tumor barcoding enables parallel quantification of Kras variant oncogenicity
.
Nat Commun
2017
;
8
:
2053
.
27.
Garnett
MJ
,
Edelman
EJ
,
Heidorn
SJ
,
Greenman
CD
,
Dastur
A
,
Lau
KW
, et al
.
Systematic identification of genomic markers of drug sensitivity in cancer cells
.
Nature
2012
;
483
:
570
5
.
28.
Li
S
,
Liu
S
,
Deng
J
,
Akbay
E
,
Hai
J
,
Ambrogio
C
, et al
.
Assessing therapeutic efficacy of MEK inhibition in a KRAS G12C-driven mouse model of lung cancer
.
Clin Cancer Res
2018
;
24
:
4854
64
.
29.
Zhou
Z-W
,
Ambrogio
C
,
Bera
AK
,
Li
Q
,
Li
X-X
,
Li
L
, et al
.
KRASQ61H preferentially signals through MAPK in a RAF dimer-dependent manner in non-small cell lung cancer
.
Cancer Res
2020
;
80
:
3719
31
.
30.
Mainardi
S
,
Mulero-Sánchez
A
,
Prahallad
A
,
Germano
G
,
Bosma
A
,
Krimpenfort
P
, et al
.
SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo letter
.
Nat Med
2018
;
24
:
961
7
.
31.
Ruess
DA
,
Heynen
GJ
,
Ciecielski
KJ
,
Ai
J
,
Berninger
A
,
Kabacaoglu
D
, et al
.
Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase
.
Nat Med
2018
;
24
:
954
60
.
32.
Gebregiworgis
T
,
Kano
Y
,
St-Germain
J
,
Radulovich
N
,
Udaskin
M
,
Mentes
A
, et al
.
The Q61H mutation decouples KRAS from upstream regulation and renders cancer cells resistant to SHP2 inhibitors
.
Nat Commun
2021
;
12
:
6274
.
33.
El Osta
B
,
Behera
M
,
Kim
S
,
Berry
LD
,
Sica
G
,
Pillai
RN
, et al
.
Characteristics and outcomes of patients with metastatic KRAS-mutant lung adenocarcinomas: the lung cancer mutation consortium experience
.
J Thorac Oncol
2019
;
14
:
876
89
.
34.
Dogan
S
,
Shen
R
,
Ang
DC
,
Johnson
ML
,
D'Angelo
SP
,
Paik
PK
, et al
.
Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: Higher susceptibility of women to smoking-related KRAS-mutant cancers
.
Clin Cancer Res
2012
;
18
:
6169
77
.
35.
Riely
GJ
,
Kris
MG
,
Rosenbaum
D
,
Marks
J
,
Li
A
,
Chitale
DA
, et al
.
Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma
.
Clin Cancer Res
2008
;
14
:
5731
4
.
36.
Skoulidis
F
,
Goldberg
ME
,
Greenawalt
DM
,
Hellmann
MD
,
Awad
MM
,
Gainor
JF
, et al
.
STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma
.
Cancer Discov
2018
;
8
:
822
35
.
37.
Chen
X
,
Su
C
,
Ren
S
,
Zhou
C
,
Jiang
T
.
Pan-cancer analysis of KEAP1 mutations as biomarkers for immunotherapy outcomes
.
Ann Transl Med
2020
;
8
:
141
.
38.
Li
S
,
Balmain
A
,
Counter
CM
.
A model for RAS mutation patterns in cancers: finding the sweet spot
.
Nat Rev Cancer
2018
;
18
:
767
77
.
39.
Canon
J
,
Rex
K
,
Saiki
A
,
Mohr
C
,
Cooke
K
,
Bagal
D
, et al
.
The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity
.
Nature
2019
;
575
:
217
23
.
40.
Hallin
J
,
Engstrom
L
,
Hargis
L
,
Calinisan
A
,
Aranda
R
,
Briere
D
, et al
.
The KRAS G12C inhibitor, MRTX849, provides insight toward therapeutic susceptibility of KRAS mutant cancers in mouse models and patients
.
Cancer Discov
2020
;
10
:
54
71
.
41.
Hong
DS
,
Fakih
MG
,
Strickler
JH
,
Desai
J
,
Durm
GA
,
Shapiro
GI
, et al
.
KRAS-G12C inhibition with sotorasib in advanced solid tumors
.
N Engl J Med
2020
;
383
:
1207
17
.
42.
Garassino
M
,
Marabese
M
,
Rusconi
P
,
Rulli
E
,
Martelli
O
,
Farina
G
, et al
.
Different types of K-Ras mutations could affect drug sensitivity and tumour behaviour in non-small-cell lung cancer
.
Ann Oncol
2011
;
22
:
235
7
.
43.
Shepherd
FA
,
Domerg
C
,
Hainaut
P
,
Jänne
P
,
Pignon
J
,
Graziano
S
, et al
.
Pooled analysis of the prognostic and predictive effects of KRAS mutation status and KRAS mutation subtype in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapy
.
J Clin Oncol
2013
;
31
:
2173
81
.
44.
Hunter
JC
,
Manandhar
A
,
Carrasco
MA
,
Gurbani
D
,
Gondi
S
,
Westover
KD
.
Biochemical and structural analysis of common cancer-associated KRAS mutations
.
Mol Cancer Res
2015
;
13
:
1325
35
.
45.
Zafra
MP
,
Parsons
MJ
,
Kim
J
,
Alonso-Curbelo
D
,
Goswami
S
,
Schatoff
EM
, et al
.
An in vivo kras allelic series reveals distinct phenotypes of common oncogenic variants
.
Cancer Discov
2020
;
10
:
1654
71
.
46.
De Roock
W
,
Jonker
DJ
,
Di Nicolantonio
F
,
Sartore-Bianchi
A
,
Tu
D
,
Siena
S
, et al
.
Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab
.
JAMA
2010
;
304
:
1812
20
.
47.
Bournet
B
,
Muscari
F
,
Buscail
C
,
Assenat
E
,
Barthet
M
,
Hammel
P
, et al
.
KRAS G12D mutation subtype is a prognostic factor for advanced pancreatic adenocarcinoma
.
Clin Transl Gastroenterol
2016
;
7
:
e157
.
48.
Skoulidis
F
,
Byers
LA
,
Diao
L
,
Papadimitrakopoulou
VA
,
Tong
P
,
Izzo
J
, et al
.
Co-occurring genomic alterations define major subsets of KRAS-mutant lung adenocarcinoma with distinct biology, immune profiles, and therapeutic vulnerabilities
.
Cancer Discov
2015
;
5
:
861
78
.
49.
Ricciuti
B
,
Arbour
K
,
Lin
J
,
Vajdi
A
,
Vokes
N
,
Hong
L
, et al
.
Diminished efficacy of programmed death-(Ligand)1 inhibition in STK11- and KEAP1-mutant lung adenocarcinoma is affected by KRAS mutation status
.
J Thorac Oncol
2021;S1556–0864(21)03284–6.
50.
Mysore
VP
,
Zhou
ZW
,
Ambrogio
C
,
Li
L
,
Kapp
JN
,
Lu
C
, et al
.
A structural model of a Ras–Raf signalosome
.
Nat Struct Mol Biol
2021
;
28
:
847
57
.

Supplementary data