Europe PMC KRAS-Mutant Non-Small Cell Lung Cancer.">KRAS-Mutant Non-Small Cell Lung Cancer.">
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Purpose:KRAS mutations occur in approximately 25% of patients with non-small cell lung cancer (NSCLC). Despite the uniform presence of KRAS mutations, patients with KRAS-mutant NSCLC can have a heterogeneous clinical course. As the pattern of co-occurring mutations may describe different biological subsets of patients with KRAS-mutant lung adenocarcinoma, we explored the effects of co-occurring mutations on patient outcomes and response to therapy.Experimental Design: We identified patients with advanced KRAS-mutant NSCLC and evaluated the most common co-occurring genomic alterations. Multivariate analyses were performed incorporating the most frequent co-mutations and clinical characteristics to evaluate association with overall survival as well as response to platinum-pemetrexed chemotherapy and immune checkpoint inhibitors.Results: Among 330 patients with advanced KRAS-mutant lung cancers, the most frequent co-mutations were found in TP53 (42%), STK11 (29%), and KEAP1/NFE2L2 (27%). In a multivariate analysis, there was a significantly shorter survival in patients with co-mutations in KEAP1/NFE2L2 [HR, 1.96; 95% confidence interval (CI), 1.33-2.92; P ≤ 0.001]. STK11 (HR, 1.3; P = 0.22) and TP53 (HR 1.11, P = 0.58) co-mutation statuses were not associated with survival. Co-mutation in KEAP1/NFE2L2 was also associated with shorter duration of initial chemotherapy (HR, 1.64; 95% CI, 1.04-2.59; P = 0.03) and shorter overall survival from initiation of immune therapy (HR, 3.54; 95% CI, 1.55-8.11; P = 0.003).Conclusions: Among people with KRAS-mutant advanced NSCLC, TP53, STK11, and KEAP1/NFE2L2 are the most commonly co-occurring somatic genomic alterations. Co-mutation of KRAS and KEAP1/ NFE2L2 is an independent prognostic factor, predicting shorter survival, duration of response to initial platinum-based chemotherapy, and survival from the start of immune therapy. Clin Cancer Res; 24(2); 334-40. ©2017 AACR.

Free full text 


Logo of nihpaLink to Publisher's site
Clin Cancer Res. Author manuscript; available in PMC 2019 Jan 15.
Published in final edited form as:
PMCID: PMC5771996
NIHMSID: NIHMS917304
PMID: 29089357

Effects of co-occurring genomic alterations on outcomes in patients with KRAS-mutant non-small cell lung cancer

Abstract

Background

KRAS mutations occur in approximately 25% of patients with non-small cell lung cancer (NSCLC). Despite the uniform presence of KRAS mutations, patients with KRAS-mutant NSCLC can have a heterogeneous clinical course. Since the pattern of co-occurring mutations may describe different biological subsets of patients with KRAS-mutant lung adenocarcinoma, we explored the effects of co-occurring mutations on patient outcomes and response to therapy.

Methods

We identified patients with advanced KRAS-mutant NSCLC and evaluated the most common co-occurring genomic alterations. Multivariate analyses were performed incorporating the most frequent co-mutations and clinical characteristics to evaluate association with overall survival as well as response to platinum-pemetrexed chemotherapy and immune checkpoint inhibitors.

Results

Among 330 patients with advanced KRAS-mutant lung cancers, the most frequent co-mutations were found in TP53 (42%), STK11 (29%), and KEAP1/NFE2L2 (27%). In a multivariate analysis, there was a significantly shorter survival in patients with co-mutations in KEAP1/NFE2L2 (HR 1.96, 95%CI 1.33–2.92, p=<0.001). STK11 (HR1.3, p=0.22) and TP53 (HR 1.11, p= 0.58) co-mutation status were not associated with survival. Co-mutation in KEAP1/NFE2L2 was also associated with shorter duration of initial chemotherapy (HR 1.64, 95% CI 1.04–2.59, p=0.03) and shorter overall survival from initiation of immune therapy (HR 3.54, 95% CI 1.55–8.11, p=0.003).

Conclusions

Among people with KRAS-mutant advanced NSCLC, TP53, STK11, and KEAP1/NFE2L2 are the most commonly co-occurring somatic genomic alterations. Co-mutation of KRAS and KEAP1/ NFE2L2 is an independent prognostic factor, predicting shorter survival, duration of response to initial platinum based chemotherapy, and survival from start of immune therapy.

Introduction

Somatic KRAS mutations are identified in 25% of patients with non-small cell lung cancers (NSCLC). Patients with these mutations have shorter survival compared to patients with EGFR-mutant NSCLC or KRAS wild type tumors.1 Since there is significant clinical heterogeneity in patients with KRAS-mutant NSCLC, defining clinically relevant subsets of KRAS-mutant NSCLC is important. In small cohorts, investigators have found specific KRAS point mutations (such as G12V and C12R), were associated with poorer outcomes.2 However in a large retrospective analysis of nearly 700 patients with metastatic disease, no apparent differences in outcome based on KRAS mutation subtype were identified.3

In patients with lung cancer, the predictive utility of KRAS mutations as a marker of response to both targeted therapy and standard cytotoxic chemotherapy has been of particular interest. The presence of a KRAS mutation suggests lack of response to EGFR tyrosine kinase inhibitors,4,5 likely because EGFR and KRAS mutations only rarely occur together. Patients with KRAS codon 13 mutations appear to have poorer outcomes with adjuvant cisplatin-based chemotherapy following resection of early stage disease,6 but in the metastatic setting KRAS mutations do not appear to independently predict response or resistance to chemotherapy treatments.7,8

While the identification of subsets of NSCLC with oncogenic drivers has transformed the treatment of this disease, these advances have thus far largely been limited to patients with mutations in EGFR9 or oncogenic fusions involving ALK10, RET11, or ROS112 kinases. There has been progress in the development of compounds that selectively target KRAS G12C, but efforts to specifically target mutant KRAS in the clinic have thus far been largely unsuccessful. Clinical testing of agents targeting downstream pathways, such as MEK and PI3K-AKT, in patients with KRAS-mutant tumors has yielded relatively low response rates.13,14 This may be due to the fact that there is a significant molecular diversity in KRAS-mutant tumors compared to other known driver events and it is these underlying mechanisms that drive divergent biologic and clinical behavior.15 However, even KRAS G12C mutant cell lines exhibit a range of responses to pharmacologic inhibition of KRAS G12C, a finding that further highlights the molecular diversity of KRAS-mutant lung cancers.16

We hypothesized that broad next-generation sequencing may allow an in-depth description of clinically heterogenous group of patients and offer prognostic and predictive markers based on the presence of co-occurring mutations in patients with KRAS-mutant NSCLC. To evaluate this hypothesis, we investigated the effect of commonly co-occurring genomic alterations, clinical characteristics on survival and treatment response in patients with advanced KRAS-mutant NSCLC.

Methods

Patients

Consecutive patients with metastatic or recurrent lung cancers found to have a KRAS mutation by next-generation sequencing were included in the analysis. A medical record search was used to identify individuals seen at Memorial Sloan Kettering with a primary tumor diagnosis of lung cancer by ICD-O code who had also undergone hybridization capture-based next-generation sequencing (NGS) testing from January of 2014 to October 2016. The list was then manually reviewed to exclude patients who did not have metastatic or recurrent disease, or a tumor diagnosis of primary lung cancer. Data collection was approved by the MSKCC Institutional Review Board/Privacy Board. Clinical characteristics and treatment course were collected for all patients. Overall survival was defined as the time from date of diagnosis of advanced disease (stage IV or recurrent cancer) until date of death or last follow up.

Genotype Analysis

Tumor and germline DNA were processed to generate bar-coded libraries and subjected to exon capture using custom-designed probes. Matched normal DNA was analyzed simultaneously to identify and filter out germline SNPs. Genomic analysis was performed using the MSK-IMPACT assay17, a clinical test approved by the New York State Department of Health designed to detect mutations, copy-number alterations, and select fusions involving 341 (version 1), 410 (version 2), or 468 (version 3) cancer-associated genes. Genomic analysis was performed using assay version 1 (341 genes) for 66 samples, version 2 (410 genes) for 250 samples, and version 3 (468 genes) for 14 samples. Normalized mutation burden was calculated as the absolute mutation burden (number of non-synonymous mutations per sample) divided by the genomic coverage for that sample (0.98Mb for version 1, 1.06Mb for version 2, and 1.22Mb for version 3).

Statistical Methods

Survival following diagnosis of stage IV lung cancer and treatment duration was estimated using Kaplan-Meier methodology. Patients were followed until death; patients alive at the end of the study were censored at the time of last available follow up. Univariate group comparisons were performed using log-rank tests. A multivariable Cox proportional hazards model was used to assess the independent effect of co-occurring mutations (STK11, KEAP1/NFE2L2, and TP53), adjusting for age, gender, performance status, and smoking history. Tumor mutational burden normalized by the size of the coding region (MB) captured by sequencing was evaluated as a continuous variable.

Results

Clinical Characteristics

We identified 550 patients with lung cancer and KRAS mutations on NGS testing between January of 2014 and October 2016. Of that cohort of patients, 330 had metastatic or recurrent lung cancer. Seventy-two percent of patients had metastatic disease at the time of initial diagnosis (n=240) while 28% (n=90) had recurrent disease. The predominant histology was adenocarcinoma (n=298, 90%). Patient demographics and histology are noted in Table 1. The most common KRAS mutation observed was G12C (44%) (Figure 1A).

An external file that holds a picture, illustration, etc.
Object name is nihms917304f1a.jpg
An external file that holds a picture, illustration, etc.
Object name is nihms917304f1b.jpg

KRAS genotype analysis. Figure 1A: Type of KRAS Codons: KRAS point mutations in dataset. Mutations occurring in less than 1% of patients grouped into “other” category. Figure 1B: Distribution of three most frequently co-occurring mutations as depicted in a proportional Venn diagram.

Table 1

Baseline patient characteristics

CharacteristicN=330 (%)
Age at diagnosis (stage IV)
  Median (range)61 (45–80)

Sex
  Women195 (59)
  Men135 (41)

KPS (%)
  ≥80241 (73)
  <8072 (22)
  Not recorded17 (5)

Smoking History Category
  Current37 (11)
  Former271 (82)
  Never22 (7)

Smoker History, pack-years
  Median Pack-year30
  Range0–135
  Unknown2

Pathology
  Adenocarcinoma294 (89)
  Squamous11 (3)
  Adenosquamous4 (1)
  Other21 (6)

Co-occurring Mutations

Along with KRAS, 377 different genes were mutated in this group of patients. The median number of co-occurring mutations per tumor was 8 (range 0–58). The most frequent mutations were found in TP53 (41%), STK11 (28%), KEAP1 (24%), RBM10 (16%), and PTPRD (15%) (Table 2). Given that genomic alteration in NFE2L2 elicit similar effects as alterations in KEAP1 in their effects on the Nrf2 pathway, patients with genomic alterations in either gene were grouped.18 An additional 3% of patients (n=9) were found to have mutations in NFE2L2 and therefore 27% of patients were grouped as having either KEAP1 or NFE2L2 mutations concurrent with KRAS. The three most frequently co-occurring genomic alterations (TP53, STK11, and KEAP1) were selected for further statistical analysis. The distributions of the 3 most frequent co-occurring mutations (TP53, STK11, and KEAP1/NFE2L2) are depicted in a proportional Venn diagram in Figure 1B.

Table 2

Most Frequent Co-Occurring Mutations Among patients with KRAS-mutant NSCLC

MutationFrequency (n)
TP5342% (138)
STK1129% (95)
KEAP1/NFE2L227% (93)
RBM1016% (52)
PTPRD15% (50)
SMARCA414% (46)
ATM13% (42)
FAT110% (32)
ARID1A9% (31)
PTPRT9% (31)

Co-occurring Mutations and Survival

The median follow-up among the 177 patients alive at the data cutoff of January 2017 was 12 months (range 1–114). The median overall survival (mOS) for all patients with KRAS-mutant advanced lung cancers in this cohort was 17 months (95% CI: 14–25). Patients with and without concurrent mutation in TP53 had a similar overall survival (HR 0.9, 95% CI 0.6–1.2, p=0.5). Patients with a concurrent mutation in STK11 (KRAS/STK11) were found to have a shorter overall survival (OS) (HR 1.7, 95% CI 1.1–2.4, p=0.002, Figure 2A). In addition, patients with concurrent mutation in KEAP1 or NFE2L2 (KRAS/KEAP1/NFE2L2) were also found to have a shorter OS (HR 2.1, 95% CI 1.4–3.1, p<.0001, Figure 2B). A multivariable Cox proportional hazards model was used to assess the independent effect of the most frequently identified co-occurring mutations (TP53, STK11, KEAP1/NFE2L2), adjusting for age, gender, performance status, and smoking history. After adjustment for these clinical variables, only KEAP1/NFE2L2 was independently associated with shorter OS (HR 1.96, 95% CI 1.3-3-2.92, p<0.001, Table 3)

An external file that holds a picture, illustration, etc.
Object name is nihms917304f2a.jpg
An external file that holds a picture, illustration, etc.
Object name is nihms917304f2b.jpg
An external file that holds a picture, illustration, etc.
Object name is nihms917304f2c.jpg

Associations of co-occurring genomic alterations and KRAS with overall survival from time Stage IV diagnosis A, STK11 B, KEAP1 or NFE2L2, C, TP53

Table 3

Multivariate Analysis of Overall Survival in patients with KRAS-mutant NSCLC

VariableHR (95% CI of HR)p-Value
TP53 (+ vs −)1.11 (0.77 to 1.58)0.581
STK11 (+ vs −)1.30 (0.86 to 1.97)0.216
KEAP1/NFE2L2 (+/−)1.96 (1.33 to 2.92)<0.001
Gender (male vs female)1.20 (0.86 to 1.68)0.284
Age at diagnosis1.03 (1.01 to 1.05)0.003
Smoking (former/current vs never)1.43 (0.86 to 1.68)0.402
KPS at diagnosis (KPS 80–100 vs <80)0.88 (0.60 to 1.29)0.514

Initial chemotherapy and co-mutational status

To evaluate the effects of co-mutations on outcomes after chemotherapy, we obtained treatment history of patients who received initial chemotherapy treatment with a platinum agent, pemetrexed, +/− bevacizumab following diagnosis of recurrent or metastatic NSCLC. In a univariate analysis the presence of a co-occurring mutation in KEAP1/ NFE2L2 was associated with a shorter duration of therapy (HR 1.6 95% CI 1.1–2.4, p=0.008, Supplemental Figure 1). The presence of either a STK11 or TP53 mutation was not associated with a difference in duration of platinum based therapy. A multivariable Cox proportional hazards model (adjusting for STK11, TP53, KEAP1/NFE2L2 as well as clinical characteristics of age, gender, performance status, and smoking history, and bevacizumab use), found KEAP1/NFE2L2 was associated with shorter treatment duration (HR 1.64, 95% CI 1.04–2.59, p=0.03, Supplemental Table 1).

Immunotherapy and co-mutation

To evaluate the effects of co-mutations on outcomes after immunotherapy, we obtained treatment history of 86 patients that underwent therapy with an immune checkpoint inhibitor as monotherapy (nivolumab or pembrolizumab). In a univariate analysis, neither the presence of TP53, SKT11, nor KEAP1/NFE2L2 was associated with a difference in duration of immune therapy (Supplemental Figure 2). These results were confirmed in a multivariate analysis adjusting for these three co-mutations as well as, age, gender, performance status, and line of therapy. Patients with a co-occurring mutation in KEAP1 or NFE2L2 were found to have a shorter overall survival from the start of immune checkpoint inhibitor in both a univariate (Figure 3) and multivariate analysis (HR 3.54, 95% CI 1.55–8.11, p=0.003, Supplemental Table 3) adjusting for co-mutations as well as, tumor mutation burden, age, gender, performance status, and line of therapy. The presence of a co-occurring mutation in neither STK11 nor TP53 was associated with a significant difference in mOS from start of therapy. Tumor mutation burden was associated with difference in overall survival from time of initiation of immunotherapy as patients with higher mutation burdens were found to have longer survival from start of treatment (HR 0.9, 95% CI 0.83–0.99, p=0.025, Supplemental Table 3).

An external file that holds a picture, illustration, etc.
Object name is nihms917304f3a.jpg
An external file that holds a picture, illustration, etc.
Object name is nihms917304f3b.jpg

Overall survival (from time of start of immune checkpoint inhibitor therapy) for treatment of Stage IV disease based on presence of A, KEAP1/NFE2L2 co-mutation and B, STK11 co-mutation

Discussion

We report the landscape of co-occurring genomic alterations in a series of 330 patients with advanced KRAS-mutant lung cancer, highlighting the most common events and identifying mutations in the KEAP1/NFE2L2 pathway as a significant, independent negative prognostic factor for patients with KRAS-mutant NSCLC. We go on to demonstrate that this association with poor overall survival is also associated with shorter duration of therapy with platinum-doublet chemotherapy and overall survival after immunotherapy. These data suggest that the observed clinical heterogeneity in patients with KRAS-mutant NSCLC is likely due in part to differences in co-occurring molecular events.

Concurrent mutations are common in patients with KRAS-mutant NSCLC, with the most common events in STK11 and TP53. Although TP53 mutations are common, our analysis concurs with that of others, reporting that concurrent TP53 mutation in KRAS-mutant NSCLC is not prognostic.19 Multiple prior analyses have reported STK11 mutations are more frequent in KRAS-mutant NSCLC as opposed to KRAS wild type tumors.20 Preclinical data has suggested that loss of STK11 leads to a more aggressive tumor phenotype.21 However there has been conflicting evidence whether STK11 mutations have prognostic or predictive implications.15,22 Our initial univariate analysis also demonstrated that concurrent STK11 mutations were associated with shorter overall survival in patients with advanced KRAS-mutant NSCLC. However, when a multivariable analysis was conducted adjusting for concurrent mutations in KEAP1/NFE2L2, TP53 and other clinical variables, concurrent mutation in STK11 was no longer associated with difference in overall survival.

The differences seen in the univariate analysis compared to multivariable analysis is likely due to significant overlap between patients with STK11 and KEAP1 concurrent mutations (also observed by others15). Our analysis suggests, however, that it is not the concurrent STK11 mutation that is associated with adverse outcomes in these patients, but rather the presence of concurrent KEAP1 or NFE2L2 mutation. This observation is limited as we evaluated only somatic mutations and not protein expression, and therefore it is possible that we did not capture tumors that have suppressed STK11 mRNA expression through a mechanism other than STK11 mutation which were observed in the subsets described by Skoulidis et al.15 These results highlight the importance of multivariable analyses, incorporating not just clinical features but also genomic features, in order to more accurately describe prognostic features in the genomically complicated landscape of KRAS-mutant NSCLC.

Mutations in the KEAP1/NFE2L2 pathway identify 27% of patients with KRAS-mutant lung cancer as having an independent negative prognostic factor. KEAP1 and NFE2L2 mutations have been best described in squamous cell lung cancer. Activation of the NRF2 pathway (through activation of NFE2L2 or inactivation of KEAP1) was found to be altered in 34% of squamous cell cancers of the lung when evaluated by TCGA investigators.23 Solis et al. also showed that increased nuclear expression of Nrf2 and decreased or absent expression of KEAP1 in 38% and 46% of patients with squamous cell carcinoma, respectively and less commonly in adenocarcinoma (18%).24 While low cytoplasmic KEAP1 expression was associated with worse overall survival in squamous cell carcinomas, no association was found between KEAP1 expression and outcomes in patients with adenocarcinoma. That analysis, similar to the TCGA analysis, is heavily weighted towards patients with early stage resected disease as opposed to advanced stage disease which is the focus of our report.

The poor prognosis of patients with concurrent KEAP1 or NFE2L2 mutations may be due in part to the observation that activation of the Nrf2 pathway may be associated with resistance to chemotherapy. Mutations in KEAP1 affect the repressive activity of KEAP1, stimulating nuclear accumulation of Nrf2, and induce constitutive expression of cytoprotective enzymes.25 Cell lines expressing lower levels of KEAP1 or KEAP1 mutant cells demonstrated greater resistance to cisplatin than cell lines with normal KEAP1.26 Concordant with this, we observed that patients with a concurrent KEAP1 or NFE2L2 mutation who were treated with a platinum/pemetrexed combination had more rapid disease progression (as suggested by shorter duration of therapy) compared to KEAP1 or NFE2L2 wild type patients. Our analysis was limited to patients with KRAS-mutant NSCLC, suggesting a cooperativity between these mutations in KEAP1 or NFE2L2 and KRAS. Moreover, the frequency of KEAP1/NFE2L2 in KRAS mutant NSCLC is much higher than seen in other lung cancer with other oncogenic drivers.

Treatment with immune check point inhibitors has been a significant advance in the treatment of NSCLC,27,28 particularly for patients without targetable oncogenic drivers. Despite these advances, single-agent response rates in unselected populations remain relatively low (10–30% in reported clinical trials). Various biomarkers have been reported that may potentially predict response to immune checkpoint inhibitors in solid tumors including PD-L1 expression29 and tumor mutation burden or neoantigen load30,31. We attempted to explore the impact of concurrent genomic alterations in patients who received single agent immune checkpoint inhibitors during the treatment of their disease. Our analysis of clinical benefit with immunotherapy was limited in size, since only a subset of the patients in our larger analysis received this therapy. Keeping these limitations in mind, our analysis suggests that patients with KRAS-mutant advanced NSCLC and a concurrent mutation in KEAP1 or NFE2L2, have significantly shorter overall survival from initiation of immune checkpoint therapy. No statistically significant differences in OS from start of therapy were observed based on concurrent STK11 or TP53 mutations. Formal response assessment was available on a subset of these patients (data not shown), and no significant difference in response was observed based on concurrent mutation in either KEAP1/NFE2L2, STK11, or TP53.

It is possible that mutations in KEAP1/NFE2L2 are associated with other features that have been associated with outcomes after immunotherapy, such as PD-L1 expression status which was not available in this series of patients. PD-L1 expression has been reported in KRAS-mutant NSCLC and was more frequently observed in smokers,32 however relationship to concurrent mutation is thus far unknown. Skouldolis et al. described that the cluster of KRAS-mutant NSCLC with low STK11 expression demonstrated a lack of immune system engagement however concurrent mutations in KEAP1 were also common in this cluster.15 In a follow up analysis, they reported that patients with concurrent mutations in STK11 had lower objective response rates to immunotherapy agents,33 a finding we did not observe in our larger series of patients.

Higher nonsynonymous mutation burden in tumors has been associated with improved objective response, durable clinical benefit, and progression free survival in patients treated with the PD-1 antibody pembrolizumab.30 More recently, normalized mutation count calculated from results of routine next-generation sequencing was reported to be predictive of response to nivolumab in NSCLC.34,35 Our multivariable analysis did incorporate normalized tumor mutation count and demonstrated that tumor mutation burden was associated with longer overall survival from initiation of immune checkpoint inhibitor. However, presence of concurrent mutation in KEAP1 or NFE2L2 was associated with significantly shorter mOS independent of tumor mutation burden.

While the findings we have described are consistent with other reports analyzing the molecular characteristics of patients with KRAS-mutant lung cancer, there are some limitations to our analysis. All patients were identified based on molecular testing at a single institution. Although molecular analysis is offered routinely for all patients, some patients may have had inadequate biopsy specimens, precluding next-generation sequencing analysis. Patients received diverse treatments and therefore the analyses of initial platinum/pemetrexed chemotherapy and immunotherapy involve a much smaller subset of patients. Duration of therapy was used as a surrogate marker for progression free survival when describing treatment history which has potential pitfalls as patients may discontinue therapy for other reasons (e.g. toxicity) as opposed to progression.

While it has been evident that there is significant clinical heterogeneity in patients with KRAS-mutant NSCLC, it is now becoming clear that this clinical heterogeneity is like due to biologic heterogeneity. Skouldolis et al. have previously reported molecular stratification of KRAS-mutant lung adenocarcinomas using RNA sequencing expression data from a subset of lung adenocarcinomas in the TCGA. These results were further validated in other small cohorts, however this dataset focused on primarily early stage disease with only a subset of patients analyzed having Stage IV disease (and all of these from the BATTLE-2 clinical trial and therefore platinum refractory).15

Our analysis shows that routine next-generation sequencing can not only provide information regarding potential actionable mutations, but also suggest prognostic and predictive features of a patient’s cancer by exploring the presence of various co-occurring mutations. Considering any given mutation in the context of other mutations and using multivariate analyses is crucial in evaluating the significance of somatic genetic events. Prospective identification of patients with concurrent KEAP1 or NFE2L2 mutations in patients with KRAS-mutant NSCLC should be considered as a prognostic factor in clinical trials evaluating therapies for these patients. Our results indicate that patients with concurrent KRAS and KEAP1/NFE2L2 have a clinically distinct behavior and may require stratification in trials.

Statement of Translational Relevance

While KRAS mutations identify the largest group of patients with oncogene-driven non-small cell lung cancer (NSCLC), patients with KRAS-mutant NSCLC have a heterogenous clinical course. We used the results of next-generation sequencing of tumors from patients with KRAS-mutant NSCLC to describe the pattern of co-occurring mutations and explore clinical outcomes. In a multivariable analysis, we identified a molecular subtype of KRAS-mutant NSCLC, with co-mutations in KEAP1/NFE2L2 in which patients had a significantly shorter overall survival than other patients with KRAS-mutant NSCLC. Patients with concurrent mutations in KRAS and KEAP1 / NFE2L2 had a shorter duration of therapy with platinum-based chemotherapy than other patients with KRAS-mutant lung cancer. These clinical findings align with prior pre-clinical work showing that mutations in KEAP1 or NFE2L2 result in activation of the Nrf2 pathway inducing constitutive expression of cytoprotective enzymes thus conferring resistance to platinum agents. Mutations in KEAP1/NFE2L2 were also associated with decreased overall survival from start of immune checkpoint inhibitors, independent of tumor mutational burden. Since our results indicate that patients with KEAP1 or NFE2L2 mutations occurring in the context of KRAS-mutations have a worse clinical course than other patients with KRAS- mutant NSCLC, we recommend that this information be captured as part of clinical trials evaluating therapy for these patients.

Supplementary Material

Acknowledgments

Funding Support: This study was funded by the US Department of Health and Human Services through the National Cancer Institute of the National Institutes of Health (P30 CA008748), the DallePezze Family Foundation, and a Stand Up To Cancer – American Cancer Society Lung Cancer Dream Team Translational Research Grant (Grant Number: SU2C-AACR-DT1715). Stand Up To Cancer is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the Scientific Partner of SU2C.

Conflict of Interest Statement: Dr. Riely’s institution receives clinical research support from Takeda, Pfizer, Novartis, and Infinity. He has been a compensated consultant for Genentech/Roche.

References

1. Johnson ML, Sima CS, Chaft J, et al. Association of KRAS and EGFR mutations with survival in patients with advanced lung adenocarcinomas. Cancer. 2013;119(2):356–362. 10.1002/cncr.27730. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
2. Villaruz LC, Socinski MA, Cunningham DE, et al. The prognostic and predictive value of KRAS oncogene substitutions in lung adenocarcinoma. Cancer. 2013;119(12):2268–2274. 10.1002/cncr.28039. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
3. Yu HA, Sima CS, Shen R, et al. Prognostic Impact of KRAS Mutation Subtypes in 677 Patients with Metastatic Lung Adenocarcinomas. J Thorac Oncol. 2015;10(3):431–437. 10.1097/JTO.0000000000000432. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
4. Linardou H, Dahabreh IJ, Kanaloupiti D, et al. Assessment of somatic k-RAS mutations as a mechanism associated with resistance to EGFR-targeted agents: a systematic review and meta-analysis of studies in advanced non-small-cell lung cancer and metastatic colorectal cancer. Lancet Oncol. 2008;9(10):962–972. 10.1016/S1470-2045(08)70206-7. [Abstract] [CrossRef] [Google Scholar]
5. Pao W, Wang TY, Riely GJ, et al. KRAS Mutations and Primary Resistance of Lung Adenocarcinomas to Gefitinib or Erlotinib. PLOS Med. 2005;2(1):e17. 10.1371/journal.pmed.0020017. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
6. Shepherd FA, Domerg C, Hainaut P, 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(17):2173–2181. 10.1200/JCO.2012.48.1390. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
7. Rodenhuis S, Boerrigter L, Top B, et al. Mutational activation of the K-ras oncogene and the effect of chemotherapy in advanced adenocarcinoma of the lung: a prospective study. J Clin Oncol. 1997;15(1):285–291. 10.1200/JCO.1997.15.1.285. [Abstract] [CrossRef] [Google Scholar]
8. Kalikaki A, Koutsopoulos A, Hatzidaki D, et al. Clinical outcome of patients with non-small cell lung cancer receiving front-line chemotherapy according to EGFR and K-RAS mutation status. Lung Cancer. 2010;69(1):110–115. 10.1016/j.lungcan.2009.09.010. [Abstract] [CrossRef] [Google Scholar]
9. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13(3):239–246. 10.1016/S1470-2045(11)70393-X. [Abstract] [CrossRef] [Google Scholar]
10. Camidge DR, Bang Y-J, Kwak EL, et al. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13(10):1011–1019. 10.1016/S1470-2045(12)70344-3. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
11. Drilon A, Wang L, Hasanovic A, et al. Response to Cabozantinib in Patients with RET Fusion-Positive Lung Adenocarcinomas. Cancer Discov. 2013;3(6):630–635. 10.1158/2159-8290.CD-13-0035. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
12. Shaw AT, Ou S-HI, Bang Y-J, et al. Crizotinib in ROS1-Rearranged Non–Small-Cell Lung Cancer. N Engl J Med. 2014;371(21):1963–1971. 10.1056/NEJMoa1406766. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
13. Jänne PA, Shaw AT, Pereira JR, et al. Selumetinib plus docetaxel for KRAS-mutant advanced non-small-cell lung cancer: a randomised, multicentre, placebo-controlled, phase 2 study. Lancet Oncol. 2013;14(1):38–47. 10.1016/S1470-2045(12)70489-8. [Abstract] [CrossRef] [Google Scholar]
14. Blumenschein GR, Smit EF, Planchard D, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC) Ann Oncol. 2015;26(5):894–901. 10.1093/annonc/mdv072. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
15. Skoulidis F, Byers LA, Diao L, 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(8):860–877. 10.1158/2159-8290.CD-14-1236. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
16. Lito P, Solomon M, Li L-S, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016;351(6273):604–608. 10.1126/science.aad6204. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
17. Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology. J Mol Diagn. 2015;17(3):251–264. 10.1016/j.jmoldx.2014.12.006. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
18. Goldstein LD, Lee J, Gnad F, et al. Recurrent Loss of NFE2L2 Exon 2 Is a Mechanism for Nrf2 Pathway Activation in Human Cancers. Cell Rep. 2016;16(10):2605–2617. 10.1016/j.celrep.2016.08.010. [Abstract] [CrossRef] [Google Scholar]
19. Shepherd FA, Lacas B, Le Teuff G, et al. Pooled Analysis of the Prognostic and Predictive Effects of TP53 Comutation Status Combined With KRAS or EGFR Mutation in Early-Stage Resected Non-Small-Cell Lung Cancer in Four Trials of Adjuvant Chemotherapy. J Clin Oncol Off J Am Soc Clin Oncol. 2017 Apr; 10.1200/JCO.2016.71.2893. JCO2016712893. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
20. Matsumoto S, Iwakawa R, Takahashi K, et al. Prevalence and specificity of LKB1 genetic alterations in lung cancers. Oncogene. 2007;26(40):5911–5918. 10.1038/sj.onc.1210418. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
21. Ji H, Ramsey MR, Hayes DN, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448(7155):807–810. 10.1038/nature06030. [Abstract] [CrossRef] [Google Scholar]
22. Facchinetti F, Bluthgen MV, Clain GT, et al. Abstract 2250: STK11 mutations in non-small cell lung cancer (NSCLC): descriptive analysis and prognostic significance. Cancer Res. 2016;76(14 Supplement):2250–2250. 10.1158/1538-7445.AM2016-2250. [CrossRef] [Google Scholar]
23. Network TCGAR. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519–525. 10.1038/nature11404. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
24. Solis LM, Behrens C, Dong W, et al. Nrf2 and Keap1 Abnormalities in Non–Small Cell Lung Carcinoma and Association with Clinicopathologic Features. Clin Cancer Res. 2010;16(14):3743–3753. 10.1158/1078-0432.CCR-09-3352. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
25. Padmanabhan B, Tong KI, Ohta T, et al. Structural Basis for Defects of Keap1 Activity Provoked by Its Point Mutations in Lung Cancer. Mol Cell. 2006;21(5):689–700. 10.1016/j.molcel.2006.01.013. [Abstract] [CrossRef] [Google Scholar]
26. Ohta T, Iijima K, Miyamoto M, et al. Loss of Keap1 Function Activates Nrf2 and Provides Advantages for Lung Cancer Cell Growth. Cancer Res. 2008;68(5):1303–1309. 10.1158/0008-5472.CAN-07-5003. [Abstract] [CrossRef] [Google Scholar]
27. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N Engl J Med. 2016;375(19):1823–1833. 10.1056/NEJMoa1606774. [Abstract] [CrossRef] [Google Scholar]
28. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non–Small-Cell Lung Cancer. N Engl J Med. 2015;373(17):1627–1639. 10.1056/NEJMoa1507643. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
29. Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res Off J Am Assoc Cancer Res. 2014;20(19):5064–5074. 10.1158/1078-0432.CCR-13-3271. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
30. Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science. 2015;348(6230):124–128. 10.1126/science.aaa1348. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
31. Snyder A, Makarov V, Merghoub T, et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N Engl J Med. 2014;371(23):2189–2199. 10.1056/NEJMoa1406498. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
32. Calles A, Liao X, Sholl LM, et al. Expression of PD-1 and Its Ligands, PD-L1 and PD-L2, in Smokers and Never Smokers with KRAS-Mutant Lung Cancer. J Thorac Oncol. 2015;10(12):1726–1735. 10.1097/JTO.0000000000000687. [Abstract] [CrossRef] [Google Scholar]
33. Skoulidis F, Elamin Y, Papadimitrakopoulou V, et al. MA04.07 Impact of Major Co-Mutations on the Immune Contexture and Response of KRAS-Mutant Lung Adenocarcinoma to Immunotherapy. J Thorac Oncol. 2017;12(1):S361–S362. 10.1016/j.jtho.2016.11.402. [CrossRef] [Google Scholar]
34. Carbone DP, Reck M, Paz-Ares L, et al. First-Line Nivolumab in Stage IV or Recurrent Non–Small-Cell Lung Cancer. N Engl J Med. 2017;376(25):2415–2426. 10.1056/NEJMoa1613493. [Europe PMC free article] [Abstract] [CrossRef] [Google Scholar]
35. Stephens P. Identification of novel modulators of response to immune checkpoint blockade in lung cancer patients through the marrying of clinical and genomic data; AACR; April 1–7th 2017; Washington, DC. Abstract SY40-02. [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/28191345
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/28191345

Article citations


Go to all (221) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

NCI NIH HHS (4)