Purpose:KRAS mutations are found in ∼25% of lung adenocarcinomas in Western countries and, as a group, have been strongly associated with cigarette smoking. These mutations are predictive of poor prognosis in resected disease as well as resistance to treatment with erlotinib or gefitinib.

Experimental Design: We determined the frequency and type of KRAS codon 12 and 13 mutations and characterized their association with cigarette smoking history in patients with lung adenocarcinomas.

Results:KRAS mutational analysis was done on 482 lung adenocarcinomas, 81 (17%) of which were obtained from patients who had never smoked cigarettes. KRAS mutations were found in 15% (12 of 81; 95% confidence intervals, 8-24%) of tumors from never smokers. Similarly, 22% (69 of 316; 95% confidence intervals, 17-27%) of tumors from former smokers, and 25% (21 of 85; 95% confidence intervals, 16-35%) of tumors from current smokers had KRAS mutations. The frequency of KRAS mutation was not associated with age, gender, or smoking history. The number of pack years of cigarette smoking did not predict an increased likelihood of KRAS mutations. Never smokers were significantly more likely than former or current smokers to have a transition mutation (G→A) rather than the transversion mutations known to be smoking-related (G→T or G→C; P < 0.0001).

Conclusions: Based on our data, KRAS mutations are not rare among never smokers with lung adenocarcinoma and such patients have a distinct KRAS mutation profile. The etiologic and biological heterogeneity of KRAS mutant lung adenocarcinomas is worthy of further study.

Translational Relevance

Mutations in the KRAS oncogene are found in ∼25% of lung adenocarcinomas in Western countries. Studies have linked KRAS mutations with poor prognosis in non–small cell lung cancer as well as resistance to treatment with erlotinib or gefitinib. These mutations have been reported to be strongly associated with cigarette smoking. However, previous studies which explored the association of smoking with KRAS mutation did not include large numbers of patients who never smoked cigarettes. We report that KRAS mutations are found in 15% of lung adenocarcinomas from patients who never smoked cigarettes compared with 22% in patients with a history of smoking cigarettes, a statistically insignificant difference in this study. Furthermore, the frequency of KRAS mutation was not associated with age, gender, or smoking history, making it difficult to predict which tumors have KRAS mutations by any clinical characteristics. Based on these data, we believe that molecular testing for KRAS mutations is necessary to identify this subgroup of patients with a different response to some treatments.

Since the identification of somatic epidermal growth factor receptor (EGFR) mutations, there has been heightened interest in the molecular basis of lung cancer in patients who never smoked cigarettes (13). Somatic mutations in EGFR have been identified in ∼15% of all patients with lung adenocarcinoma, with the proportion increasing to 50% in patients who never smoked cigarettes. There is an inverse relationship between cigarette smoking history and frequency of EGFR mutations, with the frequency of EGFR mutations decreasing significantly among patients who smoked more than 15 pack years (4). Such refined understanding of the relationship between smoking history and presence of EGFR mutations has allowed the design of clinical trials which use smoking history to enrich the number of patients with somatic EGFR mutations (57).

In contrast to EGFR mutations, KRAS mutations were initially identified in patients with lung adenocarcinoma who had a history of heavy cigarette smoking and were thought to be uncommon in patients without a history of smoking cigarettes (8). These mutations are found in ∼25% of lung adenocarcinomas in western countries but are less common in Asian populations (9, 10). KRAS mutations have been associated with poor prognosis in resected non–small cell lung cancer (NSCLC; refs. 1113), lack of survival benefit from adjuvant chemotherapy (14), and resistance to erlotinib or gefitinib (15). More than 95% of KRAS mutations in lung cancer occur in codons 12 and 13. In both KRAS and TP53, transversions (substituting a pyrimidine for a purine or purine for a pyrimidine) are more common than transitions (substituting purine for purine or pyrimidine for pyrimidine) identifying a molecular signature for the carcinogenic effects of cigarette smoke (16, 17). A detailed analysis of KRAS mutations in relation to smoking history has not been done. Using a cohort of patients with lung adenocarcinoma, we sought to determine the frequency and type of KRAS mutations in a large series of patients with known smoking histories.

Tumor specimens were obtained from an institutional tumor bank of patients who had undergone NSCLC resections between 2002 and 2007, as well as from patients with metastatic NSCLC who had KRAS mutation testing done as part of clinical trials or during routine clinical practice. Because KRAS mutations are rare in squamous tumors, only specimens with a histologic diagnosis of lung adenocarcinoma were included. All tumor specimens used for KRAS sequencing had >50% tumor. Specimens were microdissected as needed. This retrospective review was done under a waiver of authorization approved by the Memorial Sloan-Kettering Cancer Center Institutional Review Board. Standard direct sequencing was used to identify KRAS codon 12 and 13 mutations in tumors (15).

Patient smoking history was obtained by review of a patient-completed smoking questionnaire and the medical record. The prospectively administered questionnaire contained the following questions: Have you smoked more than 100 cigarettes in your life? Are you currently smoking? How many years have you been a regular smoker; and on average, how many cigarettes did you smoke per day? The smoking questionnaire was administered at the time of the first evaluation by a thoracic surgeon or medical oncologist at this institution. Never smokers had smoked <100 cigarettes. Former smokers had previously smoked cigarettes but quit smoking more than 1 year prior to diagnosis of lung cancer. Pack years of smoking was defined as [(average number of cigarettes per day / 20) × years smoking].

In 482 lung adenocarcinomas, KRAS mutations in codons 12 or 13 were found in 21% (102 of 482; 95% confidence intervals, 18-25%). Patients whose tumors harbored KRAS mutations were not significantly different from patients with KRAS wild-type tumors with regard to gender, age, or prior smoking history (Table 1). KRAS mutations were identified in 15% (12 of 81; 95% confidence intervals, 8-24%) of never smokers, 22% (69 of 316; 95% confidence intervals, 17-27%) of former smokers, and 25% (21 of 85; 95% confidence intervals, 16-35%) of current smokers (Fig. 1). No tumor with a KRAS mutation had a mutation in EGFR. There were no significant differences in frequency of KRAS mutations by category of smoking history (Mantel-Haenszel χ2P = 0.12). Next, we examined the frequency of KRAS mutation by pack years of cigarette smoking (Table 2), and found no trend (Mantel-Haenszel χ2P = 0.19). To determine whether there was a cutpoint for pack years of cigarette smoking above which KRAS mutations were more frequent, a receiver operating characteristic curve was generated. The area under the receiver operating characteristic curve was 0.56 (data not shown) suggesting no value in using pack years of cigarette smoking to predict KRAS mutational status.

Table 1.

KRAS codon 12 and 13 mutations and clinical characteristics

AllMutant KRASWild-type KRASP
Total 482 102 (21%) 380 (79%)  
Men 197 (41%) 40 (39%) 157 (41%) 0.73* 
Women 285 (59%) 62 (61%) 223 (59%)  
Never smokers 81 (17%) 12 (12%) 69 (18%) 0.14* 
Former/current smokers 401 (83%) 90 (88%) 311 (82%)  
Age, median (range) 68 (30-89) 68 (33-85) 67 (30-89) 0.98 
AllMutant KRASWild-type KRASP
Total 482 102 (21%) 380 (79%)  
Men 197 (41%) 40 (39%) 157 (41%) 0.73* 
Women 285 (59%) 62 (61%) 223 (59%)  
Never smokers 81 (17%) 12 (12%) 69 (18%) 0.14* 
Former/current smokers 401 (83%) 90 (88%) 311 (82%)  
Age, median (range) 68 (30-89) 68 (33-85) 67 (30-89) 0.98 
*

Fisher's exact test.

Wilcoxon rank sum test.

Fig. 1.

Frequency of KRAS mutation by smoking history. Mantel-Haenszel χ2 test for trend was used to calculate P value.

Fig. 1.

Frequency of KRAS mutation by smoking history. Mantel-Haenszel χ2 test for trend was used to calculate P value.

Close modal
Table 2.

Frequency of KRAS mutation by smoking history in pack years

MutantWild-typeTotalFrequency (%)95% Confidence intervals (%)
Never smokers 12 69 81 15 8-23 
<5 py 25 28 11 2-28 
6-10 py 25 28 11 2-28 
11-15 py 13 19 32 13-57 
16-25 py 10 45 55 18 9-31 
26-50 py 40 106 146 27 20-35 
51-75 py 16 51 67 24 14-36 
>75 py 12 46 58 21 11-33 
Total 102 380 482 21  
MutantWild-typeTotalFrequency (%)95% Confidence intervals (%)
Never smokers 12 69 81 15 8-23 
<5 py 25 28 11 2-28 
6-10 py 25 28 11 2-28 
11-15 py 13 19 32 13-57 
16-25 py 10 45 55 18 9-31 
26-50 py 40 106 146 27 20-35 
51-75 py 16 51 67 24 14-36 
>75 py 12 46 58 21 11-33 
Total 102 380 482 21  

Abbreviation: py, pack years.

To determine whether the type of KRAS mutation identified in never smokers correlated with the previously described dominance of transversions in smoking-associated cancers, we compared the type of KRAS mutation found in never smokers to those found in former or current smokers (Table 3). Never smokers were significantly more likely (Fisher's exact test P < 0.0001) to have a transition mutation. The ratio of transition/transversion for never smokers was 11:1 as compared with 17:73 for former or current smokers.

Table 3.

KRAS mutation type as a function of smoking history

KRAS
Former/currentNeverTotal
MutationNucleotide
G12A GGT→GC13 13 
G12C GGT→TGT 38 38 
G12V GGT→GT20 21 
G13C GGC→TGC 
     
G13D GGC→GA
G12D GGT→GA15 10 25 
G12S GGT→AGT 
Total  90 12  
KRAS
Former/currentNeverTotal
MutationNucleotide
G12A GGT→GC13 13 
G12C GGT→TGT 38 38 
G12V GGT→GT20 21 
G13C GGC→TGC 
     
G13D GGC→GA
G12D GGT→GA15 10 25 
G12S GGT→AGT 
Total  90 12  

In these patients with lung adenocarcinoma, we find that KRAS mutations are not rare in never smokers. This is a striking finding given the widespread perception that cigarette smoking and KRAS mutations are invariably linked (reviewed in ref. 16). The association between cigarette smoking and KRAS mutations has been inferred from a number of series that included a relatively small numbers of patients who never smoked cigarettes. For example, Nelson et al. examined tumors from 365 patients with NSCLC, of which only 22 were never smokers (18). Among the patients in that series in which KRAS mutational analysis was done, there were only 16 never smokers, none of whom had KRAS mutations. However, another series which included some never smokers did identify KRAS mutation in 14% (3 of 21) of never smokers (19). A difference between our series and previous series is the method of collection of smoking history. We determined smoking history using prospectively collected smoking questionnaires completed by patients with a diagnosis of lung cancer. These patients completed a detailed questionnaire which included the age of onset of smoking, the average number of cigarettes per day, the number of years in which they smoked cigarettes, and the time that the patient quit smoking cigarettes. The characteristics of patients included in this analysis are similar to the patient population seen at our institution with regard to age, gender, and smoking history.

The KRAS mutations observed in these never smokers, in addition to being more frequent than previously reported, are more likely to be transitions, unlike the transversions more common in patients with a history of cigarette smoking. In both KRAS and TP53, transversions (substituting a pyrimidine for a purine, or purine for a pyrimidine) are more common than transitions (substituting purine for purine, or pyrimidine for pyrimidine; refs. 16, 17). The etiology of G→T transversions in tumors from patients with lung cancer is thought to be related to exposure to polycyclic aromatic hydrocarbons found in cigarette smoke (20). In the case of TP53, investigators have recently noted that TP53 G→T transversions were distinctly uncommon in lung adenocarcinomas with EGFR mutations, a mutation more commonly seen in never smokers (21).

Because patients without smoking history represent ∼15% of patients with lung cancer, it is critical that any analysis seeking to examine the biology of these tumors examine a relatively large number of patients with NSCLC (22, 23). Relatively little is understood about the biology and epidemiology of lung cancer in never smokers. A number of possible causative factors have been suggested including exposure to environmental tobacco smoke or radon, as well as genetic and hormonal abnormalities (reviewed in refs. 24, 25). The distinct profile of KRAS mutations observed here in never smokers further suggests that such cancers are only rarely caused by environmental tobacco exposure. Whether this etiologic heterogeneity within KRAS mutant lung adenocarcinomas is associated with differences in cooperating genetic lesions and overall biological behavior warrants further investigation. Finally, because tumors from never smokers may have KRAS mutations, and such mutations have been associated with resistance to erlotinib and gefitinib (15), molecular analysis of NSCLC specimens for KRAS mutations may improve a clinician's ability to predict response and resistance to therapy with erlotinib or gefitinib.

No potential conflicts of interest were disclosed.

Grant support: Supported in part by NIH P01 CA129243 (M.G.K., W.P., and M.L.).

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.

Note: V.A. Miller and M. Ladanyi contributed equally to this work.

1
Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib.
N Engl J Med
2004
;
350
:
2129
–39.
2
Paez JG, Janne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy.
Science
2004
;
304
:
1497
–500.
3
Pao W, Miller V, Zakowski M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib.
Proc Natl Acad Sci U S A
2004
;
101
:
13306
–11.
4
Pham D, Kris MG, Riely GJ, et al. Use of cigarette-smoking history to estimate the likelihood of mutations in epidermal growth factor receptor gene exons 19 and 21 in lung adenocarcinomas.
J Clin Oncol
2006
;
24
:
1700
–4.
5
Cappuzzo F, Ligorio C, Janne PA, et al. Prospective study of gefitinib in epidermal growth factor receptor fluorescence in situ hybridization-positive/phospho-Akt-positive or never smoker patients with advanced non-small-cell lung cancer: the ONCOBELL trial.
J Clin Oncol
2007
;
25
:
2248
–55.
6
Kris MG, Pao W, Zakowski M, et al. Prospective trial with preoperative gefitinib to correlate lung cancer response with EGFR exon 19 and 21 mutations and to select patients for adjuvant therapy. 2006 ASCO Annual Meeting; 2006; Atlanta (GA): Am Soc Clin Oncol; 2006. p. 369s.
7
Sequist LV, Martins RG, Spigel D, et al. Fine-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations.
J Clin Oncol
2008
;
26
:
2442
–9.
8
Rodenhuis S, van de Wetering ML, Mooi WJ, et al. Mutational activation of the K-ras oncogene: a possible pathogenetic factor in adenocarcinoma of the lung.
N Engl J Med
1987
;
317
:
929
–35.
9
Buttitta F, Barassi F, Fresu G, et al. Mutational analysis of the HER2 gene in lung tumors from Caucasian patients: mutations are mainly present in adenocarcinomas with bronchioloalveolar features.
Int J Cancer
2006
;
119
:
2586
–91.
10
Suzuki M, Shigematsu H, Iizasa T, et al. Exclusive mutation in epidermal growth factor receptor gene, HER-2, and KRAS, and synchronous methylation of nonsmall cell lung cancer.
Cancer
2006
;
106
:
2200
–7.
11
Graziano SL, Gamble GP, Newman NB, et al. Prognostic significance of K-ras codon 12 mutations in patients with resected stage I and II non-small-cell lung cancer.
J Clin Oncol
1999
;
17
:
668
–75.
12
Slebos R, Kibbelaar R, Dalesio O. K-RAS oncogene activation as a prognostic marker in adenocarcinoma of the lung.
N Engl J Med
1990
;
323
:
561
–5.
13
Rosell R, Molina F, Moreno I, et al. Mutated K-ras gene analysis in a randomized trial of preoperative chemotherapy plus surgery versus surgery in stage IIIA non-small cell lung cancer.
Lung Cancer
1995
;
12
Suppl 1:
S59
–70.
14
Winton T, Livingston R, Johnson D, et al. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer.
N Engl J Med
2005
;
352
:
2589
–97.
15
Pao W, Wang TY, Riely GJ, et al. KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib.
PLoS Med
2005
;
2
:
e17
.
16
Ahrendt SA, Decker PA, Alawi EA, et al. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung.
Cancer
2001
;
92
:
1525
–30.
17
Hainaut P, Pfeifer GP. Patterns of p53 G to T transversions in lung cancer reflect the primary mutagenic signature of DNA-damage by tobacco smoke.
Carcinogenesis
2001
;
22
:
367
–74.
18
Nelson HH, Christiani DC, Mark EJ, Wiencke JK, Wain JC, Kelsey KT. Implications and prognostic value of K-ras mutation for early-stage lung cancer in women.
J Natl Cancer Inst
1999
;
91
:
2032
–8.
19
Tsao MS, Aviel-Ronen S, Ding K, et al. Prognostic and predictive importance of p53 and RAS for adjuvant chemotherapy in non small-cell lung cancer.
J Clin Oncol
2007
;
25
:
5240
–7.
20
Hecht SS. Tobacco smoke carcinogens and lung cancer.
J Natl Cancer Inst
1999
;
91
:
1194
–210.
21
Kosaka T, Yatabe Y, Endoh H, Kuwano H, Takahashi T, Mitsudomi T. Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications.
Cancer Res
2004
;
64
:
8919
–23.
22
Parkin DM, Pisani P, Lopez AD, Masuyer E. At least one in seven cases of cancer is caused by smoking. Global estimates for 1985.
Int J Cancer
1994
;
59
:
494
–504.
23
Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002.
CA Cancer J Clin
2005
;
55
:
74
–108.
24
Subramanian J, Govindan R. Lung cancer in never smokers: a review.
J Clin Oncol
2007
;
25
:
561
–70.
25
Sun S, Schiller JH, Gazdar AF. Lung cancer in never smokers—a different disease.
Nat Rev Cancer
2007
;
7
:
778
–90.