Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Molecular Biology, Pathobiology, and Genetics

Patterns of EGFR, HER2, TP53, and KRAS Mutations of p14arf Expression in Non–Small Cell Lung Cancers in Relation to Smoking History

Mounia Mounawar, Anush Mukeria, Florence Le Calvez, Rayjean J. Hung, Helene Renard, Alexis Cortot, Claire Bollart, David Zaridze, Paul Brennan, Paolo Boffetta, Elisabeth Brambilla and Pierre Hainaut
Mounia Mounawar
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Anush Mukeria
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Florence Le Calvez
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rayjean J. Hung
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Helene Renard
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alexis Cortot
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Claire Bollart
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
David Zaridze
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Paul Brennan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Paolo Boffetta
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Elisabeth Brambilla
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Pierre Hainaut
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-06-4229 Published June 2007
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Mutations in the tyrosine kinase domain of the epidermal growth factor receptor EGFR are common in non–small cell lung cancer (NSCLC) of never smokers, whereas HER2 mutations are rare. We have analyzed EGFR and HER2 mutations and the expression of the two products of the CDKN2A gene (p14arf and p16INK4a) in 116 NSCLC that have been previously analyzed for TP53 and KRAS mutations in relation to smoking history of patients. EGFR mutations were detected in 20 of 116 (17%) tumors, whereas five (4.3%) tumors contained HER2 mutations. No tumor contained both mutations. Of tumors with EGFR or HER2 mutation, 72% were adenocarcinomas, 68% were from never smokers, and 32% were from former smokers. EGFR but not HER2 mutations were mutually exclusive with KRAS mutation. Among never smokers, 11 of 16 tumors with EGFR mutation also had TP53 mutation, in contrast with two of 17 tumors without EGFR mutation (P = 0.0008). Expression of p14arf, but not p16ink4a, was more frequently down-regulated in never smokers (62.5%) than ever smokers (35%; P = 0.008). All tumors with EGFR or HER2 mutations and wild-type TP53 showed down-regulation of p14arf expression. These observations suggest that functional inactivation of the p14arf/p53 connection is required in tumors with EGFR or HER2 mutations, consistent with the notion that these proteins are part of a fail-safe mechanism protecting cells against untimely or excessive mitotic signals. [Cancer Res 2007;67(12):5667–72]

  • EGFR
  • TP53
  • p14arf
  • Mutations
  • Lung Cancer

Introduction

Lung cancer accounts for over a million annual deaths worldwide, ∼90% attributable to smoking. Non–small cell lung cancer (NSCLC) represents 75% of primary lung cancers and includes squamous cell carcinomas (SCC), adenocarcinomas (ADC), and large-cell carcinomas (LCC). Irrespective of histologic type, common genetic alterations in NSCLC are mutations in TP53, defects in the CDKN2a/RB pathway, loss of alleles on chromosome 3p encompassing FHIT, SEMA3B, and RASSF1A, and aberrant promoter methylation in O6MGMT, CDKN2a, DAPK, TIMP-3, or RASSF1A. Mutations at codon 12 in KRAS are found in 30% to 40% of ADC but are rare in other histologies ( 1). In NSCLC of smokers, there is a characteristic pattern of TP53 mutations ( 2, 3), compatible with the effects of DNA damage by tobacco carcinogens, such as polycyclic aromatic hydrocarbons. Mutations in EGFR, encoding the epidermal growth factor receptor, have emerged as a frequent molecular alteration in NSCLC of never smokers. These mutations have attracted considerable clinical interest due to their association with tumor sensitivity to the antiproliferative effects of small-molecule tyrosine kinase (TK) inhibitors, erlotinib and gefinitib ( 4– 6). Most of described EGFR mutations (85%) fall into two categories: point mutations in exon 21 (L858R; 40%) and in-frame deletions of two to nine residues in exon 19, encompassing residues of a conserved LREA motif (residues 748–751; 45%; ref. 7). Other mutations include rare insertions in exon 20, a minor hotspot at exon 18 (5%) and scattered missense mutations in exons 18 to 21. There is structural and biochemical evidence that the L858R mutation and the short deletions in exon 19 modify the geometry of the ATP binding cleft in the TK of EGFR ( 7), resulting in a hyperactive form of the receptor. EGFR mutations are inversely correlated with tobacco consumption and are reportedly frequent in ADC of women of Asian descent ( 8). R1-C1: one sentence mutations in EGFR may coexist with mutations in TP53 ( 9, 10) but are mutually exclusive of mutations in KRAS, in agreement with the notion that KRAS is a downstream effector of EGFR signaling. Moreover, HER2 (a member of the ERBB family to which EGFR also belongs) is dysregulated in many cancers. The most common alteration of HER2 is overexpression with amplification, which is frequent in breast and ovarian cancers and is associated with poor prognosis ( 11). HER2 can dimerize with other members of the ERBB family. The strongest and the most powerful heterodimer formed is EGFR/HER2 ( 12). Recent studies have reported that mutations in the TK domain of HER2 are detected in lung cancers ( 13).

In this retrospective study, we have analyzed EGFR (exons 18–21) and HER2 (exons 19 and 20) mutations in 116 NSCLC of well-characterized current, former, and never smokers, analyzed previously for KRAS and TP53 mutations ( 2). In parallel, we have assessed by immunohistochemistry the expression of p14arf and p16ink4a, the products of the CDKN2a locus ( 10, 14). p16ink4a is a cyclin kinase inhibitor that controls cyclin-dependent kinase 4/cyclin D1 complexes during progression into the G1 phase of the cell cycle. p14arf exerts complex functions in cell cycle, apoptosis, and DNA repair ( 15). Its expression is regulated by E2F, a transcription factor activated as end product of the growth signaling cascade initiated by EGFR ( 16, 17). p14arf complexes with and neutralizes Mdm2, the main controller of wild-type (WT) p53 protein stability, thus allowing p53 to escape degradation and to exert antiproliferative effects. We reasoned that p14arf represents an important connection between the EGFR and/or HER2 signaling pathways and the p53-dependent suppressive machinery. Here, we show that mutations in EGFR as well as HER2 are associated with TP53 mutation and/or with deficiency of p14arf expression in NSCLC of never and former smokers. This observation indicates that inactivation of the p14arf/p53 connection is required for the development of tumors containing EGFR or HER2 mutations.

Materials and Methods

Study subjects and tumors. The settings of the study have been described previously ( 2). The study is based on the INCO Central Europe Health Study, a hospital-based case-control study on lung cancer involving 600 cases and 600 controls. Each participant answered a questionnaire on lifestyle (residence, past health, direct and indirect exposure to tobacco, diet, alcohol consumption, sexual history, and occupational exposure to selected cancer risk factors). NSCLC from a total of 116 subjects from the Moscow area were selected into three groups: 33 never smokers (less than 100 cigarettes smoked in a lifetime; with or without exposure to involuntary smoking from the spouse or at workplace), 38 former smokers (smoking cessation for 2 years or more before diagnosis; average cumulative tobacco smoking, 27.6 pack-years), and 45 current smokers. The latter category was composed of heavy smokers (over 35 pack-years; average consumption, 48.8 pack-years). The characteristics of the patients and their cancers are summarized in Table 1 .

View this table:
  • View inline
  • View popup
Table 1.

Gender, smoking status, and histology of patients and lung tumors

Mutation analysis. DNA was extracted from areas of fresh formalin-fixed, paraffin-embedded tumor sections as selected by the pathologist and analyzed for TP53 (exons 4–10) and KRAS mutations (codon 12) as described elsewhere ( 2). EGFR mutations were detected using PCR-based direct sequencing of the four exons of the TK domain (exons 18–21) using primers and annealing conditions as described by Pao et al. ( 6). HER2 was amplified using the following sense and antisense primers for exons 19 and 20: 5′-GGATCCAGCCCACGCTCTT-3′ (19 forward) and 5′-CTGCAGCCATGGGGTCCTT-3′ (19 reverse) and 5′-CCATACCCTCTCAGCGTA-3′ (20 forward) and 5′-GCTCCGGAGAGACCTGCAA-3′ (20 reverse). Amplifications were done by using a touchdown protocol from 65°C to 62°C for exon 19 and from 61°C to 58°C for exon 20.

Aliquots of PCR products were examined by electrophoresis on 2% agarose gel containing ethidium bromide. PCR products were treated with 2 μL ExoSAP-IT (Amersham Biosciences) at 37°C for 15 min followed by inactivation at 80°C for 15 min and direct sequencing using Applied Biosystems Prism dye terminator cycle sequencing method (Perkin-Elmer) on ABI Prism 3100 Genetic Analyzer (Applied Biosystems).

Immunochemistry. Deparaffinized tissue sections were labeled with specific antibodies against p16ink4a and p14arf. The monoclonal antibody p16INK4a (16P07) (Neomarkers) was used at 1:200 dilution after 60-min Tris-citrate (pH 6) retrieving treatment on a Ventana automated system. The secondary antibody was biotinylated antibody (goat anti-rabbit/goat anti-mouse) followed by streptavidin-peroxidase complex (View kit, Ventana Systems). Detection was done using 3,3′-diaminobenzidine with H2O2 (View kit). Results were expressed as expression scores combining the percentage of positive cells for the marker and the staining intensity (1–3 on an arbitrary, semiquantitative scale). Cases were considered as positive when the expression score was >60. This value corresponds to the minimal score of normal pulmonary cells ( 18). p14arf primary antibody was a polyclonal rabbit antibody (Neomarkers) used at 1:50 after 25-min treatment in citrate (pH 6). As described previously, the secondary antibody was a biotin retrieving labeled donkey anti-rabbit (1:1,250; Jackson ImmunoResearch) followed by detecting the streptavidin-biotin/horseradish peroxidase complex (DAKO). Normal lymphocytes, stromal cells, and normal epithelial cells (bronchial cells and alveolar cells) were considered as positive controls. p14arf was considered as down-regulated in tumors when the expression score was <60.

Statistical analysis. Relative risks and 95% confidence intervals (95% CI) were calculated after adjustment for sex, age, and education, based on unconditional multivariate logistic regression model using the SAS System for Windows (release 9.1), as were pooled t tests for independent samples with equal variances, Satterthwaite t test for samples with unequal variances (when folded F tests P < 0.05), Pearson χ2 analyses, Fisher's exact tests, and ΰ statistics.

Results

EGFR and HER2 mutations in relation to histology, gender, and smoking history. A total of 23 EGFR mutations (exons 18–21) were found in 20 of 116 (17%) NSCLC cases (see detailed results in Supplementary Table). Of these mutated cases, 15 (75%) were ADC, 3 (15%) SCC, and 2 (10%) adeno-squamous (ADC-SCC) cases. Mutations were of the following types: deletions of variable size in exon 19, all encompassing codons 746 to 750 (14 mutations), missense mutations in exon 18 (2 mutations, I706T and S720P), exon 20 (2 mutations, S768I), and exon 21 (4 mutations, L858R, and 1 mutation L858Q). Three cases contained two mutations: in two cases, short deletions in exon 19 coexisted with a missense mutation (L858R or I706T); in the other case, two missense mutations were detected (L858R and S768I).

Mutations were inversely correlated with tobacco consumption, with no mutation in 45 current smokers, 4 in 38 (10%) former smokers, and 16 in 33 (48%) never smokers [odds ratio (OR), 0.05; 95% CI, 0.02–0.18]. In former smokers, 2 of the tumors with mutations were SCC and 2 were ADC. In never smokers, 13 of the tumors with mutations were ADC, 2 were ADC-SCC, and 1 was a SCC ( Table 2 ).

View this table:
  • View inline
  • View popup
Table 2.

Distribution of EGFR mutations in relation to smoking status, gender, and tumor histology

Of patients with EGFR mutations, 15 were women and 5 were men. Thus, EGFR mutations were detected in 48% (15 of 31) of the women included in the study and in 6% (5 of 85) of men. EGFR mutations were found in 50% of ADC in never smokers, irrespective of gender (2 of 4 males and 11 of 21 females).

On the other hand, only 5 (4.3%) mutations were found in HER2 [4 (10.5%) in former smokers and 1 (3%) in never smokers]. Of the 5 mutated cases, 3 were ADC, 1 was SCC, and 1 was ADC-SCC. Four of the five mutations were detected in former smokers, including 2 silent (G787G and L823L) and the 2 missense (E744G and R745D) mutations. The mutation T791I was found in a tumor of never smoker that harbored a KRAS mutation. All HER2 mutations were detected in men and the three missense mutations were associated with mutant TP53 and/or low expression of p14arf (see details in Supplementary Table).

Patterns of EGFR, HER2, KRAS, and TP53 mutations according to smoking history. Figure 1 shows the prevalence of EGFR mutations according to smoking history, combined with the prevalence of TP53 and KRAS mutations as reported previously ( 2). None of the cases was found to contain mutations in all three genes ( Table 3 ). In never smokers, TP53 mutations were detected in 11 of 16 (69%) cases with EGFR mutation but in only 2 of 17 cases without EGFR mutation. Only one never smoker (N11) had a mutation in KRAS, without TP53 or EGFR mutation but with HER2 mutation. In former smokers, EGFR mutations were found in 10% (4 of 38) of the cases, KRAS mutations in 24%, and TP53 in 59%. Of the four patients with EGFR mutations, none had a TP53 or a KRAS mutation. In contrast, two of the eight cases with KRAS mutation also had a TP53 mutation. In current smokers, EGFR mutations were absent, and KRAS and TP53 mutations were found in, respectively, 16% and 84% of the cases. All but one case with KRAS mutation contained a TP53 mutation. Overall, these results show that although TP53 mutation load was particularly high in current smokers, TP53 mutations were also detected in never smokers in relation to the presence of EGFR mutation. In contrast, mutations in KRAS were relatively rare in never smokers (6%) but more common in current (24%) and former smokers (21%). These differences were mostly due to differences in mutation patterns for ADC: KRAS mutations were detected twice as frequently in ADC of former smokers (38%) than in ADC of never or current smokers (19% in each group).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Prevalence of TP53, KRAS, and EGFR mutations in relation to smoking history. Percentage of tumors with mutation in EGFR (exons 18–21), TP53 (exons 4–9), and KRAS (codon 12), in relation to smoking status (never smokers, n = 33; former smokers, n = 38; and current smokers, n = 45). TP53 and KRAS mutation data are from Le Calvez et al. ( 2).

View this table:
  • View inline
  • View popup
Table 3.

Alterations of TP53 and KRAS in relation to EGFR mutations

Expression of p14arf and p16ink4 in relation to TP53, KRAS, and EGFR/HER2 mutation patterns. The observation that TP53 mutation is more common in never smokers with than without EGFR mutation led us to formulate the hypothesis that inactivation of p53 function may be required for carcinogenesis driven by mutant EGFR. In vitro studies in human diploid fibroblasts have shown that activation of growth signaling cascades resulted in increased expression of p14arf, leading to accumulation of p53 and growth suppression ( 16). This model is consistent with the fact that TP53 mutation is uncommon in knockout mice lacking p14arf expression ( 19). We thus analyzed by immunohistochemistry the expression of p14arf and of p16ink4a, the two product of CDKN2a, a locus often altered by deletion, mutation, or hypermethylation in lung cancer (see Introduction). Results were expressed as an expression score combining staining intensity and distribution in tumor cells (see methods). Figure 2 shows an example of staining patterns in tumors with low (expression score < 60) and normal (expression score ≥ 60) p14arf or p16ink4a expression. Results are presented in Table 4 . Loss of p14arf expression seemed to be more common in never smokers (63%) than in former smokers (47%) or current smokers (25%). This trend was not observed with p16ink4a, the loss of expression of which was marginally more common in current smokers (68%) and former smokers (61%) than in never smokers (53%). Among the 20 tumors containing EGFR mutations, 17 (85%) showed low p14arf expression. Figure 3 shows the interrelation between TP53 status and p14arf expression in tumors containing EGFR mutations. All the tumors with WT TP53 showed deficiency of p14arf expression. Thus, overall, the p14arf/p53 pathway was altered in all tumors containing mutant EGFR, either by mutation of TP53 (3 cases), by loss of expression of p14arf (9 cases), or by both (8 cases). Despite the low number of HER2 alterations, a similar concordance between mutation and p14arf expression was observed with HER2. All cases with missense HER2 mutation were deficient for p14arf expression, two of them also harboring a TP53 mutation (see Supplementary Table).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Immunohistochemical detection of p14arf an p16INK4a in lung tumors. Results were expressed as expression scores combining the percentage of cells positive for each marker and the staining intensity (1–3 on an arbitrary, semiquantitative scale). Representative examples of staining with different scores in SCC (D) and ADC (A–C). p14arf staining in ADC (A and B). Expression scores, 120 (A) and 20 (B). p16ink4a staining in ADC (C) and SCC (D). Expression scores, 80 (C) and 0 (D). Note the presence of positive nontumor cells in the stroma of the specimen in (D). Bars, 10 μm.

View this table:
  • View inline
  • View popup
Table 4.

Expression of p14arf and p16Ink4a in lung cancers according to smoking status

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Concordance TP53, EGFR mutations and p14arf expression in never smokers. A, percentage of tumors with down-regulated (gray) or normal (white) p14arf expression among tumors with WT or mutant EGFR. B, among tumors with mutant EGFR, proportion of cases with down-regulated p14arf expression (gray), TP53 mutation (white), or both (shaded).

Discussion

Somatic mutations in the TK domain of EGFR have emerged recently as common alterations in ADC of never smokers ( 8), whereas mutations in HER2 are not frequent ( 13, 20, 21). EGFR mutation prevalence is inversely correlated with tobacco consumption ( 22, 23). Furthermore, EGFR and KRAS mutations exhibit a mutually exclusive distribution in ADC, suggesting that they belong to redundant oncogenic pathways. EGFR/HER2 and KRAS mutations have been reported to be mutually exclusive ( 24); however, thus far, little is known on whether these mutations may coexist with TP53 mutations. TP53 has been reported as mutated in ADC of never smokers with prevalences between 15% and 50% according to different studies ( 2). In a study by Kosaka et al. ( 10), TP53 mutations were found at similar prevalences in tumors with or without EGFR or KRAS mutations, suggesting that mutation in TP53 occur independently of mutation in EGFR or KRAS.

In the present study, conducted on a series of 116 patients with well-defined smoking history, we found that HER2 mutations are detectable but rare in lung cancer (4.3%), whereas EGFR mutations are more common in (a) tumors of never smokers (16 of 33, 48%) as opposed to former smokers (4 of 38, 10%) or current smokers (0%) and (b) ADC (15 of 20 tumors, 75%) as opposed to ADC-SCC (2 of 20, 10%) and to SCC (3 of 20, 15%). EGFR mutations are mutually exclusive with KRAS but not TP53 mutations. On the other hand, HER2 mutations are mutually exclusive with EGFR mutations but can coexist with KRAS or TP53 mutations. The overall type and distribution of EGFR mutations is compatible with previous reports: mutation in exon 19 represented 61% of the mutations and mutations in exons 20 and 21 represented, respectively, 9% and 22%.

It has been suggested that the prevalence of EGFR mutations was particularly high in ADC of never smoking women ( 25). In the present study, cases harboring EGFR mutations were equally distributed among male and female never smokers (50% in each group). This observation suggests that the high prevalence of EGFR mutations in females is mostly an effect of the overrepresentation of women in the “never smoker” category, compatible with patterns of tobacco consumption among genders.

The CDKN2a locus encodes two distinct suppressor proteins, p16Ink4a and p14arf, that are often down-regulated in lung cancers by several mechanisms, including promoter methylation, loss of heterozygosity, point mutations, and loss of expression ( 26). How alterations in these genes correlate with EGFR mutation in lung cancer is not clearly understood. In a recent study, Toyooka et al. ( 27) have shown that the probability of having EGFR mutation was lower among tumors with methylated CDKN2a gene. The patterns of alterations of p14arf in lung cancers are much less well described as those of p16ink4a. Eymin et al. ( 28) have shown that loss of p14arf expression tended to correlate with high expression of Mdm2 in primary lung cancers, suggesting a role for Mdm2/p14arf in modulating the activity of p53. Nicholson et al. ( 29) have detected homozygous deletions, but not intragenic mutations of p14arf in 19% of NSCLC, equally distributed among TP53 mutated and WT tumors. CDKN2a promoter methylation has been described in lung cancer cell lines ( 30), in DNA from bronchial lavages ( 31), and in primary SCC ( 32), but how methylation correlates with expression is poorly documented. To determine whether down-regulation of p16ink4a and p14arf correlates with the mutation status of EGFR, HER2, KRAS, and TP53 in lung cancers, we have analyzed their expression by immunohistochemistry, a method that captures inactivation of these products by several mechanisms. In agreement with previous reports, p16ink4a was frequently down-regulated in lung cancers, with a nonsignificant tendency for more frequent loss of expression in current smokers. In contrast, p14arf was significantly more frequently down-regulated in never smokers than in current smokers. Among patients with EGFR mutations, 85% showed down-regulation of p14arf expression, whereas the 15% of the cases that retained p14arf expression harbored a mutation in TP53. Furthermore, the three tumors carrying missense mutation of HER2 were down-regulated for p14arf, and two of them were mutant for TP53. These results suggest that mutation in EGFR/HER2 requires the functional inactivation of the p14arf/p53 connection, either by down-regulation of p14arf, by mutation of TP53, or by both mechanisms. This interpretation is consistent with a model in which hyperproliferative signals generated by mutant EGFR may induce p14arf, resulting in the activation of p53 as part of a fail-safe mechanism to counter mitotic signaling. The fact that some cancers show both TP53 mutation and low p14arf expression could reflect a selection for the loss of one or both the INK4 genes that flank the ARF locus. Alternatively, mutation of TP53 could reflect the fact that loss of p14arf might not completely abrogate p53 function, perhaps due to its participation to other signaling pathways ( 33– 35). Further dissection of these mechanisms will require the analysis of the status of other partners in the p53/p14arf connection, in particular mdm2.

According to this model, only cells with a functional deficiency in the p14arf/p53 connection may bypass growth suppression and clonally expand to cause cancer. This hypothesis requires that EGFR mutation is an early event in lung carcinogenesis, which may occur in normal lung, and that mutation in TP53 and/or inactivation of p14arf is the rate-limiting event that permits clonal expansion. It is intriguing to note that a concordance with p14arf/p53 inactivation was not observed for KRAS mutations. Indeed, constitutive activation of KRAS is expected to induce an oncogenic stress that activates the p14arf/p53 pathway, leading to the same type of selection pressure for inactivating this pathway as with EGFR mutation. One possible explanation is that these mutations trend to arise in distinct etiologic context, perhaps as the consequence of distinct mechanisms of carcinogenesis. Of the 16 cases with KRAS mutation in the present series, only 1 was a never smoker, whereas 8 and 7 were former or current smokers, respectively. In line with this hypothesis, Toyooka et al. ( 27) have shown that there were differences in the evolvement of epigenetic alterations between EGFR- and KRAS-mediated tumorigenesis. Further studies are needed to determine whether such differences in genetic and epigenetic alterations may predict different tumor behavior and response to therapy.

Acknowledgments

Grant support: European Commission INCO-Copernicus program IC15-CT96-0313 and the French “Programme National d'Expertise Spécialisée” on Lung Cancer of the French Institut National du Cancer. M. Mounawar is a recipient of a IARC Special Training Award. A. Cortot is supported by a fellowship of the Association pour la Recherche sur le Cancer.

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.

We thank Ghyslaine Martel-Planche for technical assistance and Stephanie Villar for assistance with automated sequencing. The authors of this article are partners of European Cancer Risk, Nutrition and Individual Susceptibility, a network of excellence operating within the European Union 6th Framework Program, Priority 5: “Food Quality and Safety” (contract no. 513943).

Footnotes

  • Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

  • Received November 16, 2006.
  • Revision received March 8, 2007.
  • Accepted April 5, 2007.
  • ©2007 American Association for Cancer Research.

References

  1. ↵
    Kohno T, Yokota J. How many tumor suppressor genes are involved in human lung carcinogenesis? Carcinogenesis 1999; 20: 1403–10.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Le Calvez F, Mukeria A, Hunt JD, et al. TP53 and KRAS mutation load and types in lung cancers in relation to tobacco smoke: distinct patterns in never, former, and current smokers. Cancer Res 2005; 65: 5076–83.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    Pfeifer GP, Denissenko MF, Olivier M, Tretyakova N, Hecht SS, Hainaut P. Tobacco smoke carcinogens, DNA damage, and p53 mutations in smoking-associated cancers. Oncogene 2002; 21: 7435–51.
    OpenUrlCrossRefPubMed
  4. ↵
    Kobayashi S, Ji H, Yuza Y, et al. An alternative inhibitor overcomes resistance caused by a mutation of the epidermal growth factor receptor. Cancer Res 2005; 65: 7096–101.
    OpenUrlAbstract/FREE Full Text
  5. 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.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    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.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    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.
    OpenUrlCrossRefPubMed
  8. ↵
    Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst 2005; 97: 339–46.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Conde E, Angulo B, Tang M, et al. Molecular context of the EGFR mutations: evidence for the activation of mTOR/S6K signaling. Clin Cancer Res 2006; 12: 710–7.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    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.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Harari D, Yarden Y. Molecular mechanisms underlying ErbB2/HER2 action in breast cancer. Oncogene 2000; 19: 6102–14.
    OpenUrlCrossRefPubMed
  12. ↵
    Cohen EE, Lingen MW, Martin LE, et al. Response of some head and neck cancers to epidermal growth factor receptor tyrosine kinase inhibitors may be linked to mutation of ERBB2 rather than EGFR. Clin Cancer Res 2005; 11: 8105–8.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Shigematsu H, Takahashi T, Nomura M, et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res 2005; 65: 1642–6.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Gallucci M, Guadagni F, Marzano R, et al. Status of the p53, p16, RB1, and HER-2 genes and chromosomes 3, 7, 9, and 17 in advanced bladder cancer: correlation with adjacent mucosa and pathological parameters. J Clin Pathol 2005; 58: 367–71.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Sarkar-Agrawal P, Vergilis I, Sharpless NE, DePinho RA, Runger TM. Impaired processing of DNA photoproducts and ultraviolet hypermutability with loss of p16INK4a or p19ARF. J Natl Cancer Inst 2004; 96: 1790–3.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Lomazzi M, Moroni MC, Jensen MR, Frittoli E, Helin K. Suppression of the p53- or pRB-mediated G1 checkpoint is required for E2F-induced S-phase entry. Nat Genet 2002; 31: 190–4.
    OpenUrlCrossRefPubMed
  17. ↵
    Aslanian A, Iaquinta PJ, Verona R, Lees JA. Repression of the Arf tumor suppressor by E2F3 is required for normal cell cycle kinetics. Genes Dev 2004; 18: 1413–22.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Salon C, Eymin B, Micheau O, et al. E2F1 induces apoptosis and sensitizes human lung adenocarcinoma cells to death-receptor-mediated apoptosis through specific downregulation of c-FLIP(short). Cell Death Differ 2006; 13: 260–72.
    OpenUrlCrossRefPubMed
  19. ↵
    Kamijo T, Zindy F, Roussel MF, et al. Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 1997; 91: 649–59.
    OpenUrlCrossRefPubMed
  20. ↵
    Sasaki H, Shimizu S, Endo K, et al. EGFR and erbB2 mutation status in Japanese lung cancer patients. Int J Cancer 2006; 118: 180–4.
    OpenUrlCrossRefPubMed
  21. ↵
    Senior K. ERBB2 kinase mutations in lung-cancer tumours. Lancet Oncol 2004; 5: 647.
    OpenUrlPubMed
  22. ↵
    Haneda H, Sasaki H, Lindeman N, et al. A correlation between EGFR gene mutation status and bronchioloalveolar carcinoma features in Japanese patients with adenocarcinoma. Jpn J Clin Oncol 2006; 36: 69–75.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Sugio K, Uramoto H, Ono K, et al. Mutations within the tyrosine kinase domain of EGFR gene specifically occur in lung adenocarcinoma patients with a low exposure of tobacco smoking. Br J Cancer 2006; 94: 896–903.
    OpenUrlCrossRefPubMed
  24. ↵
    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.
    OpenUrlCrossRefPubMed
  25. ↵
    Thomas SK, Fossella FV, Liu D, et al. Asian ethnicity as a predictor of response in patients with non-small-cell lung cancer treated with gefitinib on an expanded access program. Clin Lung Cancer 2006; 7: 326–31.
    OpenUrlPubMed
  26. ↵
    Sharpless NE. INK4a/ARF: a multifunctional tumor suppressor locus. Mutat Res 2005; 576: 22–38.
    OpenUrlPubMed
  27. ↵
    Toyooka S, Tokumo M, Shigematsu H, et al. Mutational and epigenetic evidence for independent pathways for lung adenocarcinomas arising in smokers and never smokers. Cancer Res 2006; 66: 1371–5.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Eymin B, Gazzeri S, Brambilla C, Brambilla E. Mdm2 overexpression and p14(ARF) inactivation are two mutually exclusive events in primary human lung tumors. Oncogene 2002; 21: 2750–61.
    OpenUrlCrossRefPubMed
  29. ↵
    Nicholson SA, Okby NT, Khan MA, et al. Alterations of p14ARF, p53, and p73 genes involved in the E2F-1-mediated apoptotic pathways in non-small cell lung carcinoma. Cancer Res 2001; 61: 5636–43.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Park MJ, Shimizu K, Nakano T, et al. Pathogenetic and biologic significance of TP14ARF alterations in nonsmall cell lung carcinoma. Cancer Genet Cytogenet 2003; 141: 5–13.
    OpenUrlCrossRefPubMed
  31. ↵
    Topaloglu O, Hoque MO, Tokumaru Y, et al. Detection of promoter hypermethylation of multiple genes in the tumor and bronchoalveolar lavage of patients with lung cancer. Clin Cancer Res 2004; 10: 2284–8.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Furonaka O, Takeshima Y, Awaya H, Ishida H, Kohno N, Inai K. Aberrant methylation of p14(ARF), p15(INK4b), and p16(INK4a) genes and location of the primary site in pulmonary squamous cell carcinoma. Pathol Int 2004; 54: 549–55.
    OpenUrlCrossRefPubMed
  33. ↵
    Sherr CJ. Divorcing ARF and p53: an unsettled case. Nat Rev Cancer 2006; 6: 663–73.
    OpenUrlCrossRefPubMed
  34. Efeyan A, Garcia-Cao I, Herranz D, Velasco-Miguel S, Serrano M. Tumour biology: policing of oncogene activity by p53. Nature 2006; 443: 159.
    OpenUrlCrossRefPubMed
  35. ↵
    Eymin B, Claverie P, Salon C, et al. p14ARF activates a Tip60-dependent and p53-independent ATM/ATR/CHK pathway in response to genotoxic stress. Mol Cell Biol 2006; 26: 4339–50.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Cancer Research: 67 (12)
June 2007
Volume 67, Issue 12
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Patterns of EGFR, HER2, TP53, and KRAS Mutations of p14arf Expression in Non–Small Cell Lung Cancers in Relation to Smoking History
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Patterns of EGFR, HER2, TP53, and KRAS Mutations of p14arf Expression in Non–Small Cell Lung Cancers in Relation to Smoking History
Mounia Mounawar, Anush Mukeria, Florence Le Calvez, Rayjean J. Hung, Helene Renard, Alexis Cortot, Claire Bollart, David Zaridze, Paul Brennan, Paolo Boffetta, Elisabeth Brambilla and Pierre Hainaut
Cancer Res June 15 2007 (67) (12) 5667-5672; DOI: 10.1158/0008-5472.CAN-06-4229

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Patterns of EGFR, HER2, TP53, and KRAS Mutations of p14arf Expression in Non–Small Cell Lung Cancers in Relation to Smoking History
Mounia Mounawar, Anush Mukeria, Florence Le Calvez, Rayjean J. Hung, Helene Renard, Alexis Cortot, Claire Bollart, David Zaridze, Paul Brennan, Paolo Boffetta, Elisabeth Brambilla and Pierre Hainaut
Cancer Res June 15 2007 (67) (12) 5667-5672; DOI: 10.1158/0008-5472.CAN-06-4229
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • HSF1-Mediated Aneuploidy in p53-Defective Cells
  • Mechanisms of Invasive Urothelial Tumorigenesis
  • Alternate mRNA 3′ Processing Characterizes Distinct Tumors
Show more Molecular Biology, Pathobiology, and Genetics
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement