Hostname: page-component-7c8c6479df-7qhmt Total loading time: 0 Render date: 2024-03-28T23:03:29.229Z Has data issue: false hasContentIssue false

Spectrum of somatic EGFR, KRAS, BRAF, PTEN mutations and TTF-1 expression in Brazilian lung cancer patients

Published online by Cambridge University Press:  05 March 2014

JULIANA G. CARNEIRO
Affiliation:
Faculdade de Ciências Médicas, Centro de Ensino Superior e Desenvolvimento, Campina Grande, 58411-020, Brasil Department of Surgery, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brasil
PATRICIA G. COUTO
Affiliation:
Department of Surgery, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brasil
LUCIANA BASTOS-RODRIGUES
Affiliation:
Department of Surgery, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brasil
MARIA APARECIDA C. BICALHO
Affiliation:
Department of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brasil
PAULA V. VIDIGAL
Affiliation:
Department of Pathology, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brasil
ALYNE VILHENA
Affiliation:
Hospital Julia Kubitscheck, Belo Horizonte, 30620-470, Brasil
NILSON F. AMARAL
Affiliation:
Hospital Julia Kubitscheck, Belo Horizonte, 30620-470, Brasil
ALLEN E. BALE
Affiliation:
Department of Genetics, Yale University School of Medicine, New Haven, CT 06520-8005, USA
EITAN FRIEDMAN
Affiliation:
The Susanne Levy Gertner Oncogenetics Unit, Chaim Sheba Medical Center, Tel-Hashomer, 52621, Israel
LUIZ DE MARCO*
Affiliation:
Department of Surgery, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Brasil
*
*Corresponding author: Av. Alfredo Balena 190, room 325, Belo Horizonte 30130-100, Brasil. Tel: +55 (31) 3409-9134. Fax: +55 (31) 3409-9134. E-mail: Ldemarco@ufmg.br
Rights & Permissions [Opens in a new window]

Summary

Lung cancer is the leading global cause of cancer-related mortality. Inter-individual variability in treatment response and prognosis has been associated with genetic polymorphisms in specific genes: EGFR, KRAS, BRAF, PTEN and TTF-1. Somatic mutations in EGFR and KRAS genes are reported at rates of 15–40% in non-small cell lung cancer (NSCLC) in ethnically diverse populations. BRAF and PTEN are commonly mutated genes in various cancer types, including NSCLC, with PTEN mutations exerting an effect on the therapeutic response of EGFR/AKT/PI3K pathway inhibitors. TTF-1 is expressed in approximately 80% of lung adenocarcinomas and its positivity correlates with higher prevalence of EGFR mutation in this cancer type. To determine molecular markers for lung cancer in Brazilian patients, the rate of the predominant EGFR, KRAS, BRAF and PTEN mutations, as well as TTF-1 expression, was assessed in 88 Brazilian NSCLC patients. EGFR exon 19 deletions (del746–750) were detected in 3/88 (3·4%) patients. Activating KRAS mutations in codons 12 and 61 were noted in five (5·7%) and two (2·3%) patients, respectively. None of the common somatic mutations were detected in either the BRAF or PTEN genes. TTF-1 was overexpressed in 40·7% of squamous-cell carcinoma (SCC). Our findings add to a growing body of data that highlights the genetic heterogeneity of the abnormal EGFR pathway in lung cancer among ethnically diverse populations.

Type
Research Papers
Copyright
Copyright © Cambridge University Press 2014 

1. Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide, stemming in part from the lack of effective early detection schemes impacting survival (Kadara et al., Reference Kadara, Kabbout and Wistuba2011). There is an urgent need for novel biomarkers that could be clinically applied as prognostic factors and somatic mutations are obvious candidates for being used as prognostic markers (Li et al., Reference Li, Kung, Mack and Gandara2013; Travis et al., Reference Travis, Brambilla and Riely2013).

Epidermal growth factor receptor (EGFR) plays an important role in cell proliferation and survival (Reungwetwattana et al., Reference Reungwetwattana, Weroha and Molina2012). The frequency of activating mutations of the EGFR gene in non-small cell lung cancer (NSCLC) varies according to ethnicity and is noted in 15% of NSCLC diagnosed in Caucasians, 40% in Asians and 33·3% in Latin Americans mostly of Spanish origin (Paez et al., Reference Paez, Jänne, Lee, Tracy, Greulich, Gabriel, Herman, Kaye, Lindeman, Boggon, Naoki, Sasaki, Fujii, Eck, Sellers, Johnson and Meyerson2004; Leidner et al., Reference Leidner, Fu, Clifford, Hamdan, Jin, Eisenberg, Boggon, Skokan, Franklin, Cappuzzo, Hirsch, Varella-Garcia and Halmos2009; Rosell et al., Reference Rosell, Moran, Queralt, Porta, Cardenal, Camps, Majem, Lopez-Vivanco, Isla, Provencio, Insa, Massuti, Gonzalez-Larriba, Paz-Ares, Bover, Garcia-Campelo, Moreno, Catot, Rolfo, Reguart, Palmero, Sánchez, Bastus, Mayo, Bertran-Alamillo, Molina, Sanchez and Taron2009; Arrieta et al., Reference Arrieta, Cardona, Bramuglia, Gallo, Campos-Parra, Serrano, Castro, Avilés, Amorin, Kirchuk, Cuello, Borbolla, Riemersma, Becerra and Rosell2011; Cote et al., Reference Cote, Haddad and Edwards2011).

Activating KRAS mutations that predominantly cluster to either codons 12 and 13, and rarely in codon 61 (Suda et al., Reference Suda, Tomizawa and Mitsudomi2010) are encountered with differing rates in NSCLC diagnosed in different ethnic groups: 30% in Caucasians, 10% in East Asians and 16·6% in Latin Americans (Hunt et al., Reference Hunt, Strimas, Eyer, Haddican, Luckett, Ruiz, Axelrad, Backes and Fontham2002; Riely et al., Reference Riely, Kris, Rosenbaum, Marks, Li, Chitale, Nafa, Riedel, Hsu, Pao, Miller and Ladanyi2008; Arrieta et al., Reference Arrieta, Cardona, Bramuglia, Gallo, Campos-Parra, Serrano, Castro, Avilés, Amorin, Kirchuk, Cuello, Borbolla, Riemersma, Becerra and Rosell2011). Mutations in KRAS and EGFR that appear to be mutually exclusive are currently being used as molecular biomarkers for determining both prognosis and therapeutic targets in NSCLC (Murray et al., Reference Murray, Timotheadou, Linardou, Vrettou, Kostopoulos, Skrickova, Papakostantinou, Christodoulou, Pectasides, Samantas, Papakostas, Skarlos, Kosmidis and Fountzilas2006; Irmer et al., Reference Irmer, Funk and Blaukat2007; Mok et al., Reference Mok, Wu, Thongprasert, Yang, Chu, Saij, Sunpaweravong, Han, Margono, Ichinose, Nishiwaki, Ohe, Yang, Chewaskulyong, Jiang, Duffield, Watkins, Armour and Fukuoka2009; Brevet et al., Reference Brevet, Johnson, Azzoli and Ladanyi2011; Heigener & Reck, Reference Heigener and Reck2011). Tyrosine kinase inhibitors (TKIs) are widely used as an adjuvant treatment to chemotherapy in advanced stage NSCLC cases that specifically display activating EGFR mutations (Keedy et al., Reference Keedy, Temin, Somerfield, Beasley, Johnson, McShane, Milton, Strawn, Wakelee and Giaccone2011). NSCLC harbouring activating KRAS mutations, are resistant to EGFR-TKIs treatment and patients have shorter survival and response rates (Pao et al., Reference Pao, Wang, Riely, Miller, Pan, Ladanyi, Zakowski, Heelan, Kris and Varmus2005; Borràs et al., Reference Borràs, Jurado, Hernan, Gamundi, Dias, Martí, Mañé, Arcusa, Agúndez, Blanca and Carballo2011; Gaughan & Costa, Reference Gaughan and Costa2011).

BRAF is mutated in a wide range of human cancers, including lung cancers (Xing, Reference Xing2005; Dhomen & Marais, Reference Dhomen and Marais2009; Gaughan & Costa, Reference Gaughan and Costa2011). Ninety per cent of BRAF mutations are represented by a single somatic mutation BRAF V600E (Cantwell-Dorris, Reference Cantwell-Dorris2011; Gaughan & Costa, Reference Gaughan and Costa2011) and approximately 50% of lung adenocarcinomas harbour the recurrent somatic oncogenic mutations in EGFR, KRAS and BRAF (Girard, Reference Girard2013; Oxnard et al., Reference Oxnard, Binder and Jänne2013).

Mutations in other genes are involved in modulating therapeutic response to inhibitors of the EGFR/PI3K/AKT pathway (Su et al., Reference Su, Dias-Santagata, Duke, Hutchinson, Lin, Borger, Chung, Massion, Vnencak-Jones, Iafrate and Pao2011). PTEN is a dual specificity phosphatase which directly antagonizes the phosphatidylinositol-3 kinase (PI3K) signalling pathway (Endersby & Baker, Reference Endersby and Baker2008; Tang et al., Reference Tang, Iijima, Kamimura, Chen, Long and Devreotes2011). PTEN is a tumour suppressor gene that is frequently mutated in human cancer, with most mutations leading to an inactivation of the gene (Cantley & Neel, Reference Cantley and Neel1999; Simpson & Parsons, Reference Simpson and Parsons2001; Pandolfi, Reference Pandolfi2008). Although inactivating PTEN mutations are present in approximately 10% in NSCLC, PTEN expression is diminished in a larger proportion of these tumours (almost 70%) possibly by epigenetic mechanisms (Tang et al., Reference Tang, He, Guo and Chang2006; Li et al., Reference Li, Zhao, Peng, Liang, Deng and Chen2012).

TTF-1 is a DNA-binding protein found to be expressed in lung cells, regulates the activity of proliferating cells and also plays a role in angiogenesis (Berghmans et al., Reference Berghmans, Mascauxa, Hallerb, Meerta, Van Houttec and Sculier2008; Wislez et al., Reference Wislez, Antoine, Baudrin, Poulot, Neuville, Pradere, Longchampt, Isaac-Sibille, Lebitasy and Cadranel2010). TTF-1 was reportedly over-expressed in approximately 80% of lung adenocarcinomas (Maeshima et al., Reference Maeshima, Omatsu, Tsuta, Asamura and Matsuno2008). An activating EGFR mutation is associated with TTF-1 over-expression in lung adenocarcinoma (Yatabe et al., Reference Yatabe, Kosaka, Takahashi and Mitsudomi2005; Tapia et al., Reference Tapia, Savic, Bihl, Rufle, Zlobec, Terracciano and Bubendorf2009) and the pattern of EGFR(+)/TTF-1(–) seems to be an exclusive signature for NSCLC, especially squamous-cell carcinoma (SCC) subtype (Berghmans et al., Reference Berghmans, Mascauxa, Hallerb, Meerta, Van Houttec and Sculier2008).

Ethnicity has been shown to affect risk for developing lung cancer. African Americans have higher incidence rates for lung cancer and family history of lung cancer, compared with pack/year smokers matched Caucasians (Cote et al., Reference Cote, Kardia, Wenzlaff, Ruckdeschel and Schwartz2005; Haiman et al., Reference Haiman, Stram, Wilkens, Pike, Kolonel, Henderson and Le Marchand2006). Moreover, for lung cancer patients, being of Japanese ethnicity and never-smoker are independent favourable prognostic factors for overall survival compared with Caucasians (Kawaguchi et al., Reference Kawaguchi, Matsumura, Fukai, Tamura, Saito, Zell, Maruyama, Ziogas, Kawahara and Ignatius2010). These ethnic differences are in all likelihood the result of the combined differences in the rate of germline and somatic sequence alterations in genes involved in NSCLC pathogenesis.

The spectrum of somatic EGFR, KRAS, BRAF, PTEN mutations and TTF-1 expression and its potential associations in genetically heterogeneous Brazilian lung cancer patients has not been previously reported, and that was the focus of this study.

2. Materials and methods

(i) Subjects

The study cohort encompassed 88 patients diagnosed with NSCLC who were eligible for surgery, with no previous history of chemotherapy or radiotherapy. Patients were recruited from a referral centre of thoracic surgery (Hospital Julia Kubitscheck – FHEMIG, Belo Horizonte, Brazil) between 1 January 2006 and 31 December 2011. Controls were 28 healthy individuals older than 55 years with no previous personal or family history of cancer, randomly recruited from the outpatient clinic in the same medical centre during the same time period. A group of 96 healthy Brazilian individuals, representative of Southeastern Brazil, were used as controls for genomic ancestry.

(ii) Ethics statement

The Ethics Committee of Universidade Federal de Minas Gerais (Comitê de Ética em Pesquisa da UFMG, # 373-05) approved the study protocol and all participants signed a written informed consent.

(iii) EGFR/KRAS/BRAF/PTEN genotyping

Genomic DNA of participating NSCLC patients was isolated from fresh tumour tissue samples as well as from peripheral blood, according to a proteinase K-based standard protocol (Miller et al., Reference Miller, Dykes and Polesky1988). Peripheral blood was collected in vacuum tubes and genomic DNA was isolated using the high salt method of Lahiri and Nurnberger (Lahiri & Nurnberger, Reference Lahiri and Nurnberger1991) and was extracted from all study participants – NSCLC cases and controls. Genotyping for germline and somatic alterations was carried out for the following genes and mutations: exons 18 (G719S), 19 (746_750del, D761Y and L747S), 20 (insertions and T790M) and 21 (L858R and L861Q) of EGFR, exons 2 and 3 of KRAS (codons 12, 13 and 61), exon 15 of BRAF (BRAFV600E) and all nine exons of PTEN (to evaluate any inactivating mutation in the entire coding regions of the gene) were amplified by PCR with primers specific for each region (Table 1). For PCR reactions 2 μl of DNA at 30 ng/μl were mixed with 2·5 μl of 10X IIB buffer (40 mm NaCl; 10 mm Tris–HCl pH 8·4; 0·1% Triton X-100; 1·5 mm MgCl2), 2·5 μl of 0·2 mm dNTP, 0·5 μl of each primer at 10 pmol/μl and 0·25 μl of Taq polymerase (Invitrogen, Brazil) 0·625 U, on a final volume of 25 μl. Samples were placed on an Eppendorf Mastercycler® (Hamburg, Germany) at 94°C for 3 min and then 35 cycles of 94°C for 30 s, 55°C for 30 s and 72°C for 30 s and a final extension time at 72°C for 5 min. PCR products were purified using Illustra GFX PCR DNA and Gel Band Purification Kit (GE Healthcare, São Paulo, Brazil) following manufacturer's protocol and visualized on a silver-stained 6·5% polyacrylamide gel. To improve the sensitivity of the KRAS mutation detection we used the COLD-PCR method as previously described (Zuo et al., Reference Zuo, Chen, Chandra, Galbincea, Soape, Doan, Barkoh, Koeppen, Medeiros and Luthra2009).

Table 1. Characteristics of PCR amplification of EGFR and KRAS

a Tm: annealing temperature.

Sequences were obtained on ABI 3130 Genetic Analyzer (Applied Biosystems, Foster City, CA). Bidirectional sequence data were analysed using Sequencer 4.9 software (http://genecodes.com). Positive findings for EGFR exon 19 deletions were also confirmed by fragment analysis. The genomic fragment including all exon 19 was amplified in 10 μl final PCR volume of the following: 1X PCR buffer (10 mm Tris–HCl pH 8·3 or pH 9·2, 75 mm KCl, 3·5 mm MgCl2), 200 μ m dNTPs, 1·0 U of Platinum Taq DNA polymerase (Life Technologies, São Paulo, Brazil), 20 ng of genomic DNA, 1·5 μ m of M13-40 forward primer labelled with the FAM dye, 1·5 μ m of each unlabelled reverse primer and 0·1 μ m of each unlabelled forward primer.

(iv) Immunohistochemistry of TTF-1

Tissue sections from 27 samples previously diagnosed by pathological report as SCCs, three adenocarcinomas and four diagnosed as NSCLC poorly differentiated carcinoma were stained with TTF-1 antiserum. Briefly, 4 μm paraffin-embedded sections were dewaxed in xylene and hydrated with graded ethanol. Endogenous peroxidase activity was blocked with 3% H2O2 in water for 10 min. Heat-induced epitope retrieval was performed with 1 mm EDTA buffer pH 8·0 for 30 min in a steamer at 96°C. Primary polyclonal rabbit antiserum was used at 1 : 100 for 18 h at 4°C. This was followed by incubation with the labelled streptavidin-biotin kit NovoLinkTM Max Polymer (Novocastra, UK). Peroxidase activity was developed with DAB (Sigma, St Louis, MO) with timed monitoring using a positive control sample. The sections were then counterstained with haematoxylin, dehydrated and mounted. All slides were examined under light microscopy and staining for TTF-1 was evaluated according to the presence or not of the protein by two pathologists who were blinded to the clinical course of the patient.

(v) Genomic ancestry analysis

Germline DNA of all 88 lung cancer patients and 96 ethnically diverse Brazilian controls were genotyped with a set of 40 biallelic short insertion/deletion polymorphisms (In/Dels), as previously described (Bastos-Rodrigues et al., Reference Bastos-Rodrigues, Pimenta and Pena2006). Amplicons were size fractionated using an ABI 3130 DNA sequencer (Applied *Biosystems) and analysed using the GeneMapper® Software (version 3.7). To estimate the proportion of European, African and Amerindian biogeographical ancestry of each individual we used the Structure program, version 2.3 (http://pritch.bsd.uchicago.edu/structure.html).

(vi) Statistical analysis

The proportion of European, African and Amerindian bio-geographical ancestry of each individual was used for stratifying statistical analysis. For statistical comparisons between cases and controls, the two-tailed Mann–Whitney U test was used. Single-marker allelic and genotypic association tests were performed using the Unphased software package version 3.0.12 (www.mrcbsu.cam.ac.uk/personal/frank/software/unphased/). A value of P ⩽ 0·05 was considered statistically significant. Odds ratios and 95% confidence interval were calculated.

3. Results

(i) Sample characteristics

Demographics and relevant clinical and pathological data of all 88 NSCLC cases are shown in Table 2. The control group (for EGFR germline mutation genotyping) encompassed 28 healthy individuals, comprising 64% women and 36% men with a mean age of 72·8 ± 9·15 years (range 55–89 years).

Table 2. Clinical data of non-small cell lung cancer (NSCLC) patients

a Tumours positive for TTF-1 marker (Travis et al., Reference Travis, Brambilla and Riely2013).

ADC, adenocarcinoma; SCC, squamous-cell carcinoma.

(ii) EGFR/KRAS/BRAF/PTEN mutation status

EGFR gene genotyping showed the presence of the 746_750del (LREA domain) in 3/88 patients (3·4%), and was exclusively detected in 3/45 (6·6%) of patients with adenocarcinoma. No sequence alterations, especially G719S, were noted in exon 18 of the EGFR gene. Only one patient diagnosed with adenocarcinoma (1·5%) showed the L585R variation in exon 21 of EGFR. In addition to these clearly pathogenic mutations in the EGFR gene, two previously reported polymorphisms were also identified in DNA of cases and controls: a silent base substitution (CAG>CAA) at c.2538 position (corresponding to rs1050171) was detected in 61·3% of the cases (n = 54) and a substitution at IVS-60T>C position (rs10241451) was noted in 20·4% of the cases (n = 18). The rs10241451 did not show a significant association with the disease both by allele and genotype frequency (P = 0·29 and P = 0·32, respectively) when compared with controls, whereas a significant association between rs1050171 and lung cancer compared with cancer-free controls was noted only by allele and not by genotype frequency (P = 0·04 and P = 0·11, respectively) (Table 3).

Table 3. Allele and genotype frequencies of rs1050171 and rs10241451 in patients and controls

Codon 12 KRAS mutations were noted in five male patients (5·7%) diagnosed with adenocarcinoma: three harboured the Gly12Cys (c.34G>T) mutation and two the Gly12Asp (c.35G>A) mutation. No mutations in codon 13 of KRAS were found. Two female smokers patients (3%) diagnosed with SCC at ages 60 and 63 years old showed a previously reported missense mutation (rs17851045) in codon 61 (c.182A>T), which leads to histidine for glutamine change (H61Q). No sequence alterations in exons 1–9 of the PTEN gene were noted and the BRAF V600E mutation was not detected in any of the samples analysed.

(iii) TTF-1 immunohistochemistry

All three adenocarcinoma samples, which served as positive controls for TTF-1 expression, demonstrated positivity, as expected. Eleven of the 27 SCCs analysed displayed positive TTF-1 expression and in 16 tumours no expression was present (Table 2). Four tumours previously described as poorly differentiated carcinoma also showed positive TTF-1 expression. All positive tumours for TTF-1 expression (n = 15) were considered NSCLC-favour adenocarcinoma.

(iv) Genomic ancestry analysis

We genotyped germline DNA from all 88 lung cancer patient samples and 96 controls, for 40 polymorphic In/Del loci which form a powerful ancestry informative test battery (Bastos-Rodrigues et al., Reference Bastos-Rodrigues, Pimenta and Pena2006). For the case group, the proportions of European, African and Amerindian ancestry were 0·87 ± 0·02 (mean±se), 0·09 ± 0·02 and 0·04 ± 0·007, respectively, whereas for the control group, the results were 0·85 ± 0·02, 0·08 ± 0·01 and 0·07 ± 0·001, respectively. The proportion of European, African and Amerindian ancestry in each group indicated that an African component was more prevalent in lung cancer patients than controls (P = 0·03) (Fig. 1).

Fig. 1. Analysis of genomic ancestry of patients with lung cancer. EU, Europeans; AF, African; AM, Amerindians. Significant difference was found between Africans when compared with control group (P = 0·004).

4. Discussion

In the present study that focused on Brazilian lung cancer patients, the EGFR 746_750del, a mutation that is associated with a better response of NSCLC patients to TKIs, such as Gefitinib and Erlotinib (Han et al., Reference Han, Kim, Jeon, Hwang, Im, Lee, Kim, Kim, Heo, Kim, Chung and Bang2006; Riely et al., Reference Riely, Politi, Miller and Pao2006; Irmer et al., Reference Irmer, Funk and Blaukat2007) was detected somatically in only three of 88 tumour samples (3·4%). This is a significantly lower rate than the rates reported for ethnically diverse populations worldwide, with rates ranging from 15 to 40% of NSCLC analysed, primarily adenocarcinomas (Irmer et al., Reference Irmer, Funk and Blaukat2007; Matsuo et al., Reference Matsuo, Ito, Yatabe, Hiraki, Hirose, Wakai, Kosaka, Suzuki, Tajima and Mitsudomi2007; Leidner et al., Reference Leidner, Fu, Clifford, Hamdan, Jin, Eisenberg, Boggon, Skokan, Franklin, Cappuzzo, Hirsch, Varella-Garcia and Halmos2009; Tapia et al., Reference Tapia, Savic, Bihl, Rufle, Zlobec, Terracciano and Bubendorf2009; Vlastos et al., Reference Vlastos, Zinszner, Hussenet, du Manoir, Vordonis, Nikolakopoulou, Hardavella, Lacomme, Vignaud and Martinet2010). A previous study of Latin American patients from biopsies taken from Argentinean, Colombian, Peruvian and Mexican lung cancer patients (Arrieta et al., Reference Arrieta, Cardona, Bramuglia, Gallo, Campos-Parra, Serrano, Castro, Avilés, Amorin, Kirchuk, Cuello, Borbolla, Riemersma, Becerra and Rosell2011) reported the same deletion in 48·4% patients (185/382 cases) and this was in agreement with data from Asian (60%) and European populations (62·2%) (Rosell et al., Reference Rosell, Moran, Queralt, Porta, Cardenal, Camps, Majem, Lopez-Vivanco, Isla, Provencio, Insa, Massuti, Gonzalez-Larriba, Paz-Ares, Bover, Garcia-Campelo, Moreno, Catot, Rolfo, Reguart, Palmero, Sánchez, Bastus, Mayo, Bertran-Alamillo, Molina, Sanchez and Taron2009). A previous study also detected a low incidence of mutations in a Caucasian population (12%) and suggested that the specific lung cancer subtype analysed as well as the technique used could explain the wide range of somatic mutations reported (Wislez et al., Reference Wislez, Antoine, Baudrin, Poulot, Neuville, Pradere, Longchampt, Isaac-Sibille, Lebitasy and Cadranel2010). The L858R EGFR somatic mutation was found in only one patient and other mutations such as L861Q and G719S were not noted in the present study. These results are in line with data reported by Vlastos et al. (Reference Vlastos, Zinszner, Hussenet, du Manoir, Vordonis, Nikolakopoulou, Hardavella, Lacomme, Vignaud and Martinet2010) that analysed Caucasian population, but contrasts other studies reporting high rates of these EGFR mutations in NSCLC in patients of Asian (40%) and European (37·8%) descent (Han et al., Reference Han, Kim, Jeon, Hwang, Im, Lee, Kim, Kim, Heo, Kim, Chung and Bang2006; Murray et al., Reference Murray, Timotheadou, Linardou, Vrettou, Kostopoulos, Skrickova, Papakostantinou, Christodoulou, Pectasides, Samantas, Papakostas, Skarlos, Kosmidis and Fountzilas2006; Riely et al., Reference Riely, Politi, Miller and Pao2006; Irmer et al., Reference Irmer, Funk and Blaukat2007; Matsuo et al., Reference Matsuo, Ito, Yatabe, Hiraki, Hirose, Wakai, Kosaka, Suzuki, Tajima and Mitsudomi2007; Rosell et al., Reference Rosell, Moran, Queralt, Porta, Cardenal, Camps, Majem, Lopez-Vivanco, Isla, Provencio, Insa, Massuti, Gonzalez-Larriba, Paz-Ares, Bover, Garcia-Campelo, Moreno, Catot, Rolfo, Reguart, Palmero, Sánchez, Bastus, Mayo, Bertran-Alamillo, Molina, Sanchez and Taron2009). Specifically, the L858R mutation that has been reportedly detected in 12·5 to 45% of NSCLC-associated EGFR mutations (Pan et al., Reference Pan, Pao and Ladanyi2005; Tapia et al., Reference Tapia, Savic, Bihl, Rufle, Zlobec, Terracciano and Bubendorf2009) was found in only one case (1·1%) in the present study. This patient has features more common to NSCLC patients with the L585R mutation in exon 21, i.e. female, non-smoker with adenocarcinoma histology (Kondo et al., Reference Kondo, Yokoyama, Fukui, Yoshioka, Yokoi, Nagasaka, Imaizumi, Kume, Hasegawa, Shimokata and Sekido2005; Shigematsu et al., Reference Shigematsu, Lin, Takahashi, Nomura, Suzuki, Wistuba, Fong, Lee, Toyooka, Shimizu, Fujisawa, Feng, Roth, Herz, Minna and Gazdar2005; Riely et al., Reference Riely, Politi, Miller and Pao2006). One plausible explanation for the disparities between the rates of the EGFR mutations in the present study and other studies is the type of tumours and patients analysed. The cases analysed in the present study are mostly men, smokers, with a high African ancestry. Other studies showed that the prevalence of EGFR mutations among African Americans have yielded conflicting results (Cote et al., Reference Cote, Haddad and Edwards2011; Reinersman et al., Reference Reinersman, Johnson, Riely, Chitale, Nicastri, Soff, Schwartz, Sima, Ayalew, Lau, Zakowski, Rusch, Ladanyi and Kris2011). Indeed, in a previous study focusing on Brazilian NSCLC cases, Amoedo et al. (Reference Amoedo, Castelo-Branco, Paschoal, Pezzuto, Esperança, Rumjanek and Rumjanek2009) studied only exon 19 of EGFR and reported one 746_750del in 64 paraffin-embedded tissue samples analysed (∼1·5%). These data combined with the data presented herein support the notion of a low prevalence of somatic EGFR mutations in Brazilian NSCLC cases.

Somatic activating KRAS mutations in NSCLC can usually be detected in codon 12 (Gly12Asp), less frequently in codon 13 (Gly13Asp) and rarely at codon 61 (Gln61His) (Riely et al., Reference Riely, Marks and Pao2009). The frequency and type of KRAS activating mutations in NSCLC are in part determined by the specific tumour histology, patients’ ethnicity and smoking history (Riely et al., Reference Riely, Kris, Rosenbaum, Marks, Li, Chitale, Nafa, Riedel, Hsu, Pao, Miller and Ladanyi2008; Amoedo et al., Reference Amoedo, Castelo-Branco, Paschoal, Pezzuto, Esperança, Rumjanek and Rumjanek2009); these mutations are more commonly encountered in Caucasians, smokers with adenocarcinoma histology (Suda et al., Reference Suda, Tomizawa and Mitsudomi2010). In the present study, the mutation in codon 12 was noted in male patients diagnosed with adenocarcinoma. Unlike the reported mutational spectrum in the KRAS gene in other world populations (Dearden et al., Reference Dearden, Stevens, Wu and Blowers2013), the sequence alteration described in codon 61 was detected in two female smokers, diagnosed with SCC. Although environmental factors could be involved, the aetiology of this alteration remains to be established.

The rate of somatic KRAS mutations in the present study was low (7·9%), similar to the low rates (11/173; 6·3%) reported by Lee et al. (Reference Lee, Kim, Jin, Yoo, Park, Choi, Jeon, Cho, Lee, Cha, Park, Kim, Jung and Park2010) who analysed 173 Korean cases. These rates are well below the rates reported for Japanese (12·8%) (Lee et al., Reference Lee, Kim, Jin, Yoo, Park, Choi, Jeon, Cho, Lee, Cha, Park, Kim, Jung and Park2010), Latin American (16·6%) (Arrieta et al., Reference Arrieta, Cardona, Bramuglia, Gallo, Campos-Parra, Serrano, Castro, Avilés, Amorin, Kirchuk, Cuello, Borbolla, Riemersma, Becerra and Rosell2011) and Caucasian populations (41·9%) (Borràs et al., Reference Borràs, Jurado, Hernan, Gamundi, Dias, Martí, Mañé, Arcusa, Agúndez, Blanca and Carballo2011). These apparent differences in somatic mutation rates in the KRAS gene may also be attributed to different ethnic background of the studied populations and may indicate that the malignant transformation process in Brazilian NSCLC cases may have different pathways than those in some other, ethnically diverse, populations.

Although several studies have shown the relationship of BRAF and PTEN with the tumourigenesis of lung cancer, several studies confirm the low frequency of mutations of these genes in NSCLC (Forgacs et al., Reference Forgacs, Biesterveld, Sekido, Fong, Muneer, Wistuba, Milchgrub, Brezinschek, Virmani, Gazdar and Minna1998; Yokomizo et al., Reference Yokomizo, Tindall, Drabkin, Gemmill, Franklin, Yang, Sugio, Smith and Liu1998; Sasaki et al., Reference Sasaki, Kawano, Endo, Suzuki, Haneda, Yukiue, Kobayashi, Yano and Fujii2006; Pratilas et al., Reference Pratilas, Hanrahan, Halilovic, Persaud, Soh, Chitale, Shigematsu, Yamamoto, Sawai, Janakiraman, Taylor, Pao, Toyooka, Ladanyi, Gazdar, Rosen and Solit2008; Jin et al., Reference Jin, Kim, Jeon, Choi, Kim, Lee, Cha, Yoon, Kim, Jung and Park2010; Marchetti et al., Reference Marchetti, Felicioni, Malatesta, Grazia Sciarrotta, Guetti, Chella, Viola, Pullara, Mucilli and Buttitta2011). In the Brazilian population analysed here, we have not found any mutations in BRAF and EGFR genes thus confirming the rarity of this alteration in NSCLC. To our knowledge, our work represents the first data of a PTEN and BRAF V600E mutation search in Brazilian patients diagnosed with NSCLC.

Approximately 80% of adenocarcinomas express TTF-1, and this marker has been used to differentiate the histological subtypes of NSCLC (adenocarcinoma and SCC) (Maeshima et al., Reference Maeshima, Omatsu, Tsuta, Asamura and Matsuno2008). Our results showed that 40·7% of SCCs analysed were TTF-1 (+) demonstrating that a subset of SCCs should be considered NSCLC-favour adenocarcinoma (Travis et al., Reference Travis, Brambilla and Riely2013). According to Ordóñez (Reference Ordóñez2012), the introduction of target therapies can result in dramatically different outcomes based on histological subtype. Thus, the use of markers such as TTF-1 can help to discriminate between adenocarcinoma and SCC subtypes, what is indispensable to define a personalized treatment.

The Brazilian population has a major Caucasian contribution (Pena et al., Reference Pena, Bastos-Rodrigues, Pimenta and Bydlowski2009) but our study found a greater African component in patients with NSCLC than in the control group. African-American patients with NSCLC are significantly less likely to harbour activating mutations in EGFR genes and their signalling pathway when compared with Caucasians (Riely et al., Reference Riely, Politi, Miller and Pao2006). This notion is further supported by Reinersman et al. (Reference Reinersman, Johnson, Riely, Chitale, Nicastri, Soff, Schwartz, Sima, Ayalew, Lau, Zakowski, Rusch, Ladanyi and Kris2011), who reported that African Americans are less likely to have KRAS mutations in NSCLC when compared with Caucasians, a finding corroborated in our study.

As demonstrated, our data show a low frequency of mutations in EGFR, KRAS, BRAF and PTEN. This may be related to the high African component found in our patients, which has been discussed in previous studies (Maxwell et al., Reference Maxwell, Risinger, Hayes, Alvarez, Dodge, Barrett and Berchuck2000; Kumar et al., Reference Kumar, Brim, Giardiello, Smoot, Nouraie, Lee and Ashktorab2009; Leidner et al., Reference Leidner, Fu, Clifford, Hamdan, Jin, Eisenberg, Boggon, Skokan, Franklin, Cappuzzo, Hirsch, Varella-Garcia and Halmos2009; Pena et al., Reference Pena, Bastos-Rodrigues, Pimenta and Bydlowski2009; Reinersman et al., Reference Reinersman, Johnson, Riely, Chitale, Nicastri, Soff, Schwartz, Sima, Ayalew, Lau, Zakowski, Rusch, Ladanyi and Kris2011) but had not yet been analysed in the Brazilian population. Thus, our data add to a growing body of evidence that highlight the genetic heterogeneity of the EGFR pathway in NSCLC among different populations, which highlights the need to incorporate these differences in designing clinical therapies and agents for the inhibition of this pathway.

The limitations of the present study should be pointed out: this is a relatively small study from a single medical centre that serves a population that might not reflect the ethnic diversity of the entire Brazilian population. In addition, it must be said that patients are referred to treatment when surgical procedures are no longer acceptable and further investigations inappropriate. Hence it is important to stress that these results should be expanded and validated in larger studies focusing on Brazilian patients.

In conclusion, Brazilian lung cancer patients display a low frequency of somatic mutations in EGFR, KRAS, BRAF and PTEN. This may be related to the high African ancestry component found in our patients, but an expansion and validation of these preliminary data is needed.

Acknowledgements

We are grateful to all patients.

Financial Support

This work was partially supported by FAPEMIG; CNPq and INCT em Medicina Molecular; Brasil.

Statement of Interest

None.

References

Amoedo, N. D., Castelo-Branco, M. T., Paschoal, M. E., Pezzuto, P., Esperança, A. B., Rumjanek, V. M. & Rumjanek, F. D. (2009). Expression of ABC transporters, p53, Bax, Bcl-2 in an archival sample of non-small cell lung cancer bearing a deletion in the EGFR gene. International Journal of Molecular Medicine 23, 609614.Google Scholar
Arrieta, O., Cardona, A. F., Bramuglia, G. F., Gallo, A., Campos-Parra, A. D., Serrano, S., Castro, M., Avilés, A., Amorin, E., Kirchuk, R., Cuello, M., Borbolla, J., Riemersma, O., Becerra, H. & Rosell, R. (2011). Genotyping non-small cell lung cancer (NSCLC) in Latin America. Journal of Thoracic Oncology 6, 19551959.Google Scholar
Bastos-Rodrigues, L., Pimenta, J. R. & Pena, S. D. (2006). The genetic structure of human populations studied through short insertion-deletion polymorphisms. Annals of Human Genetics 70, 658665.Google Scholar
Berghmans, T., Mascauxa, C., Hallerb, A., Meerta, A. P., Van Houttec, P. & Sculier, J. P. (2008). EGFR, TTF-1 and Mdm2 expression in stage III non-small cell lung cancer: a positive association. Lung Cancer 62, 3544.Google Scholar
Borràs, E., Jurado, I., Hernan, I., Gamundi, M. J., Dias, M., Martí, I., Mañé, B., Arcusa, A., Agúndez, J. A., Blanca, M. & Carballo, M. (2011). Clinical pharmacogenomic testing of KRAS, BRAF and EGFR mutations by high resolution melting analysis and ultra-deep pyrosequencing. BMC Cancer 11, 406.Google Scholar
Brevet, M., Johnson, M. L., Azzoli, C. G. & Ladanyi, M. (2011). Detection of EGFR mutations in plasma DNA from lung cancer patients by mass spectrometry genotyping is predictive of tumor EGFR status and response to EGFR inhibitors. Lung Cancer 73, 96102.Google Scholar
Cantley, L. C. & Neel, B. G. (1999). New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proceedings of the National Academy of Sciences USA 96, 42404245.Google Scholar
Cantwell-Dorris, E. R. (2011). BRAFV600E: Implications for carcinogenesis and molecular therapy. Molecular Cancer Therapeutics 10, 385394.Google Scholar
Cote, M. L., Kardia, S. L., Wenzlaff, A. S., Ruckdeschel, J. C. & Schwartz, A. G. (2005). Risk of lung cancer among white and black relatives of individuals with early-onset lung cancer. JAMA 293, 30363042.Google Scholar
Cote, M. L., Haddad, R. & Edwards, D. J. (2011). Frequency and type of epidermal growth factor receptor mutations in African Americans with non-small cell lung cancer. Journal of Thoracic Oncology 6, 627630.Google Scholar
Dearden, S., Stevens, J., Wu, Y. L. & Blowers, D. (2013). Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap). Annals of Oncology 24, 23712376.Google Scholar
Dhomen, N. & Marais, R. (2009). BRAF signaling and targeted therapies in melanoma. Hematology/Oncology Clinics of North America 23, 529545.Google Scholar
Endersby, R. & Baker, S. J. (2008). PTEN signaling in brain: neuropathology and tumorigenesis. Oncogene 27, 54165430.Google Scholar
Forgacs, E., Biesterveld, E. J., Sekido, Y., Fong, K., Muneer, S., Wistuba, I. I., Milchgrub, S., Brezinschek, R., Virmani, A., Gazdar, A. F. & Minna, J. D. (1998). Mutation analysis of the PTEN/MMAC1 gene in lung cancer. Oncogene 17, 15571565.Google Scholar
Gaughan, E. M. & Costa, D. B. (2011). Genotype-driven therapies for non-small cell lung cancer: focus on EGFR, KRAS and ALK gene abnormalities. Therapeutic Advances in Medical Oncology 3, 113125.Google Scholar
Girard, N. (2013). Other signalization targets. Target Oncology 8, 6977.Google Scholar
Haiman, C. A., Stram, D. O., Wilkens, L. R., Pike, M. C., Kolonel, L. N., Henderson, B. E. & Le Marchand, L. (2006). Ethnic and racial differences in the smoking-related risk of lung cancer. New England Journal of Medicine 354, 333342.CrossRefGoogle ScholarPubMed
Han, S. W., Kim, T. Y., Jeon, Y. K., Hwang, P. G., Im, S. A., Lee, K. H., Kim, J. H., Kim, D. W., Heo, D. S., Kim, N. K., Chung, D. H. & Bang, Y. J. (2006). Optimization of patient selection for gefitinib in non-small cell lung cancer by combined analysis of epidermal growth factor receptor mutation, K-ras mutation, and Akt phosphorylation. Clinical Cancer Research 12, 25382544.Google Scholar
Heigener, D. F. & Reck, M. (2011). Mutations in the epidermal growth factor receptor gene in non-small cell lung cancer: impact on treatment beyond gefitinib and erlotinib. Advances in Therapy 28, 126133.Google Scholar
Hunt, J. D., Strimas, A., Eyer, M., Haddican, M., Luckett, B. G., Ruiz, B., Axelrad, T. W., Backes, W. L. & Fontham, E. T. (2002). Differences in KRAS mutation spectrum in lung cancer cases between African Americans and Caucasians after occupational or environmental exposure to known carcinogens. Cancer Epidemiology, Biomarkers and Prevention 11, 14051412.Google ScholarPubMed
Irmer, D., Funk, J. O. & Blaukat, A. (2007). EGFR kinase domain mutations – functional impact and relevance for lung cancer therapy. Oncogene 26, 56935701.Google Scholar
Jin, G., Kim, M. J., Jeon, H. S., Choi, J. E., Kim, D. S., Lee, E. B., Cha, S. I., Yoon, G. S., Kim, C. H., Jung, T. H. & Park, J. Y. (2010). PTEN mutations and relationship to EGFR, ERBB2, KRAS, and TP53 mutations in non-small cell lung cancers. Lung Cancer 69, 279283.Google Scholar
Kadara, H., Kabbout, M. & Wistuba, I. (2011). Pulmonary adenocarcinoma: a renewed entity in 2011. Respirology 17, 5065.Google Scholar
Kawaguchi, T., Matsumura, A., Fukai, S., Tamura, A., Saito, R., Zell, J. A., Maruyama, Y., Ziogas, A., Kawahara, M. & Ignatius, S. H. (2010). Japanese ethnicity compared with Caucasian ethnicity and never-smoking status are independent favorable prognostic factors for overall survival in non-small cell lung cancer: a collaborative epidemiologic study of the National Hospital Organization Study Group for Lung Cancer (NHSGLC) in Japan and a Southern California Regional Cancer Registry databases. Journal of Thoracic Oncology 5, 10011010.Google Scholar
Keedy, V. L., Temin, S., Somerfield, M. R., Beasley, M. B., Johnson, D. H., McShane, L. M., Milton, D. T., Strawn, J. R., Wakelee, H. A. & Giaccone, G. (2011). American Society of Clinical Oncology provisional clinical opinion: epidermal growth factor receptor (EGFR) Mutation testing for patients with advanced non-small-cell lung cancer considering first-line EGFR tyrosine kinase inhibitor therapy. Journal of Clinical Oncology 29, 21212127.Google Scholar
Kondo, M., Yokoyama, T., Fukui, T., Yoshioka, H., Yokoi, K., Nagasaka, T., Imaizumi, K., Kume, H., Hasegawa, Y., Shimokata, K. & Sekido, Y. (2005). Mutations of epidermal growth factor receptor of non-small cell lung cancer were associated with sensitivity to gefitinib in recurrence after surgery. Lung Cancer 50, 385391.Google Scholar
Kumar, K., Brim, H., Giardiello, F., Smoot, D. T., Nouraie, M., Lee, E. L. & Ashktorab, H. (2009). Distinct BRAF (V600E) and KRAS mutations in high microsatellite instability sporadic colorectal cancer in African Americans. Clinical Cancer Research 15, 11551161.Google Scholar
Lahiri, D. K. & Nurnberger, J. I. (1991). A rapid non-enzymatic method for the preparation of HMW DNA from blood for RFLP studies. Nucleic Acids Research 11, 5444.Google Scholar
Lee, S. Y., Kim, M. J., Jin, G., Yoo, S. S., Park, J. Y., Choi, J. E., Jeon, H. S., Cho, S., Lee, E. B., Cha, S. I., Park, T. I., Kim, C. H., Jung, T. H. & Park, J. Y. (2010). Somatic mutations in EGFR signaling pathway genes. Journal of Thoracic Oncology 5, 17341740.Google Scholar
Leidner, R. S., Fu, P., Clifford, B., Hamdan, A., Jin, C., Eisenberg, R., Boggon, T. J., Skokan, M., Franklin, W. A., Cappuzzo, F., Hirsch, F. R., Varella-Garcia, M. & Halmos, B. (2009). Genetic abnormalities of the EGFR pathway in African American patients with non–small-cell lung cancer. Journal of Clinical Oncology 27, 56205626.Google Scholar
Li, G., Zhao, J., Peng, X., Liang, J., Deng, X. & Chen, Y. (2012). The mechanism involved in the loss of PTEN expression in NSCLC tumor cells. Biochemical and Biophysical Research Communications 418, 547552.Google Scholar
Li, T., Kung, H. J., Mack, P. C. & Gandara, D. R. (2013). Genotyping and genomic profiling of non-small-cell lung cancer: implications for current and future therapies. Journal of Clinical Oncology 31, 10391049.Google Scholar
Maeshima, A. M., Omatsu, M., Tsuta, K., Asamura, H. & Matsuno, Y. (2008). Immunohistochemical expression of TTF-1 in various cytological subtypes of primary lung adenocarcinoma, with special reference to intratumoral heterogeneity. Pathology International 58, 3137.Google Scholar
Marchetti, A., Felicioni, L., Malatesta, S., Grazia Sciarrotta, M., Guetti, L., Chella, A., Viola, P., Pullara, C., Mucilli, F. & Buttitta, F. (2011). Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. Journal of Clinical Oncology 29, 357435479.Google Scholar
Matsuo, K., Ito, H., Yatabe, Y., Hiraki, A., Hirose, K., Wakai, K., Kosaka, T., Suzuki, T., Tajima, K. & Mitsudomi, T. (2007). Risk factors differ for non-small-cell lung cancers with and without EGFR mutation: assessment of smoking and sex by a case-control study in Japanese. Cancer Science 98, 96101.Google Scholar
Maxwell, G. L., Risinger, J. I., Hayes, K. A., Alvarez, A. A., Dodge, R. K., Barrett, J. C. & Berchuck, A. (2000). Racial disparity in the frequency of PTEN mutations, but not microsatellite instability, in advanced endometrial cancers. Clinical Cancer Research 6, 29993005.Google Scholar
Miller, S. A., Dykes, D. D. & Polesky, H. F. (1988). A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Research 16, 1215.Google Scholar
Mok, T. S., Wu, Y. L., Thongprasert, S., Yang, C. H., Chu, D. T., Saij, N., Sunpaweravong, P., Han, B., Margono, B., Ichinose, Y., Nishiwaki, Y., Ohe, Y., Yang, J., Chewaskulyong, B., Jiang, H., Duffield, E. L., Watkins, C. L., Armour, A. A. & Fukuoka, M. (2009). Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. New England Journal of Medicine 361, 947957.Google Scholar
Murray, S., Timotheadou, E., Linardou, H., Vrettou, A. V., Kostopoulos, I., Skrickova, J., Papakostantinou, C., Christodoulou, C., Pectasides, D., Samantas, E., Papakostas, P., Skarlos, D. V., Kosmidis, P. & Fountzilas, G. (2006). Mutations of the epidermal growth factor receptor tyrosine kinase domain and associations with clinicopathological features in non-small cell lung cancer patients. Lung Cancer 52, 225233.Google Scholar
Ordóñez, N. G. (2012). Thyroid transcription factor-1 is not expressed in squamous cell carcinomas of the lung: an immunohistochemical study with review of the literature. Applied Immunohistochemistry and Molecular Morphology 20, 525530.Google Scholar
Oxnard, G. R., Binder, A., & Jänne, P. A. (2013). New targetable oncogenes in non-small-cell lung cancer. Journal of Clinical Oncology 31, 10971104.Google Scholar
Paez, J. G., Jänne, P. A., Lee, J. C., Tracy, S., Greulich, H., Gabriel, S., Herman, P., Kaye, F. J., Lindeman, N., Boggon, T. J., Naoki, K., Sasaki, H., Fujii, Y., Eck, M. J., Sellers, W. R., Johnson, B. E. & Meyerson, M. (2004). EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 304, 14971500.Google Scholar
Pan, Q., Pao, W., Ladanyi, M. (2005). Rapid polymerase chain reaction-based detection of epidermal growth factor receptor gene mutations in lung adenocarcinomas. The Journal of Molecular Diagnostics 7, 396403.CrossRefGoogle ScholarPubMed
Pandolfi, P. P. (2008). P-TEN exciting years: from the cytosol to the nucleus and back to keep cancer at bay. Oncogene 27, 5386.Google Scholar
Pao, W., Wang, T. Y., Riely, G. J., Miller, V. A., Pan, Q., Ladanyi, M., Zakowski, M. F., Heelan, R. T., Kris, M. G. & Varmus, H. E. (2005). KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Medicine 2, e17.Google Scholar
Pena, S. D., Bastos-Rodrigues, L., Pimenta, J. R. & Bydlowski, S. P. (2009). DNA tests probe the genomic ancestry of Brazilians. Brazilian Journal of Medical and Biological Research 42, 870876.Google Scholar
Pratilas, C. A., Hanrahan, A. J., Halilovic, E., Persaud, Y., Soh, J., Chitale, D., Shigematsu, H., Yamamoto, H., Sawai, A., Janakiraman, M., Taylor, B. S., Pao, W., Toyooka, S., Ladanyi, M., Gazdar, A., Rosen, N. & Solit, D. B. (2008). Genetic predictors of MEK dependence in non-small cell lung cancer. Cancer Research 68, 93759383.Google Scholar
Reinersman, J. M., Johnson, M. L., Riely, G. J., Chitale, D. A., Nicastri, A. D., Soff, G. A., Schwartz, A. G., Sima, C. S., Ayalew, G., Lau, C., Zakowski, M. F., Rusch, V. W., Ladanyi, M. & Kris, M. G. (2011). Frequency of EGFR and KRAS mutations in lung adenocarcinomas in African Americans. Journal of Thoracic Oncology 6, 2831.Google Scholar
Reungwetwattana, T., Weroha, S. J. & Molina, J. R. (2012). Oncogenic pathways, molecularly targeted therapies, and highlighted clinical trials in non-small-cell lung cancer (NSCLC). Clinical Lung Cancer 13, 252266.Google Scholar
Riely, G. J., Politi, K. A., Miller, V. A. & Pao, W. (2006). Update on epidermal growth factor receptor mutations in non-small cell lung cancer. Clinical Cancer Research 12, 72327241.Google Scholar
Riely, G. J., Kris, M. G., Rosenbaum, D., Marks, J., Li, A., Chitale, D. A., Nafa, K., Riedel, E. R., Hsu, M., Pao, W., Miller, V. A. & Ladanyi, M. (2008). Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clinical Cancer Research 14, 57315734.Google Scholar
Riely, G. J., Marks, J. & Pao, W. (2009). KRAS mutations in non-small cell lung cancer. Proceedings of the American Thoracic Society 6, 201205.Google Scholar
Rosell, R., Moran, C., Queralt, C., Porta, R., Cardenal, F., Camps, C., Majem, M., Lopez-Vivanco, G., Isla, D., Provencio, M., Insa, A., Massuti, B., Gonzalez-Larriba, J. L., Paz-Ares, L., Bover, I., Garcia-Campelo, R., Moreno, M. A., Catot, S., Rolfo, C., Reguart, N., Palmero, R., Sánchez, J. M., Bastus, R., Mayo, C., Bertran-Alamillo, J., Molina, M. A., Sanchez, J. J. & Taron, M. (2009). Screening for epidermal growth factor receptor mutations in lung cancer. New England Journal of Medicine 361, 958967.Google Scholar
Sasaki, H., Kawano, O., Endo, K., Suzuki, E., Haneda, H., Yukiue, H., Kobayashi, Y., Yano, M. & Fujii, Y. (2006). Uncommon V599E BRAF mutations in Japanese patients with lung cancer. Journal of Surgical Research 133, 203206.Google Scholar
Shigematsu, H., Lin, L., Takahashi, T., Nomura, M., Suzuki, M., Wistuba, I. I., Fong, K. M., Lee, H., Toyooka, S., Shimizu, N., Fujisawa, T., Feng, Z., Roth, J. A., Herz, J., Minna, J. D. & Gazdar, A. F. (2005). Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. Journal of the National Cancer Institute 97, 339346.Google Scholar
Simpson, L. & Parsons, R. (2001). PTEN: life as a tumor suppressor. Experimental Cell Research 264, 2941.Google Scholar
Su, Z., Dias-Santagata, D., Duke, M., Hutchinson, K., Lin, Y. L., Borger, D. R., Chung, C. H., Massion, P. P., Vnencak-Jones, C. L., Iafrate, A. J. & Pao, W. (2011). A platform for rapid detection of multiple oncogenic mutations with relevance to targeted therapy in non–small-cell lung cancer. Journal of Molecular Diagnostics 13, 7484.Google Scholar
Suda, K., Tomizawa, K. & Mitsudomi, T. (2010). Biological and clinical significance of KRAS mutations in lung cancer: an oncogenic driver that contrasts with EGFR mutation. Cancer Metastasis Review 29, 4960.Google Scholar
Tang, J. M., He, Q. Y., Guo, R. X. & Chang, X. J. (2006). Phosphorylated Akt overexpression and loss of PTEN expression in non-small cell lung cancer confers poor prognosis. Lung Cancer 51, 181191.Google Scholar
Tang, M., Iijima, M., Kamimura, Y., Chen, L., Long, Y. & Devreotes, P. (2011). Disruption of PKB signaling restores polarity to cells lacking tumor suppressor PTEN. Molecular Biology of the Cell 22, 437447.Google Scholar
Tapia, C., Savic, S., Bihl, M., Rufle, A., Zlobec, I., Terracciano, L. & Bubendorf, L. (2009). EGFR mutation analysis in non-small-cell lung cancer: experience from routine diagnostics. Der Pathologe 30, 384392. [In German]Google Scholar
Travis, W. D., Brambilla, E. & Riely, G. J. (2013). New pathologic classification of lung cancer: relevance for clinical practice and clinical trials. Journal of Clinical Oncology 31, 9921001.Google Scholar
Vlastos, F., Zinszner, J., Hussenet, T., du Manoir, S., Vordonis, L., Nikolakopoulou, S., Hardavella, G., Lacomme, S., Vignaud, J. M. & Martinet, N. (2010). Mapping EGFR1 mutations in patients with lung adenocarcinoma. Diagnostic Molecular Pathology 19, 209217.Google Scholar
Wislez, M., Antoine, M., Baudrin, L., Poulot, V., Neuville, A., Pradere, M., Longchampt, E., Isaac-Sibille, S., Lebitasy, M. P. & Cadranel, J. (2010). Non-mucinous and mucinous subtypes of adenocarcinoma with bronchioloalveolar carcinoma features differ by biomarker expression and in the response to gefitinib. Lung Cancer 68, 185191.Google Scholar
Xing, M. (2005). BRAF mutation in thyroid cancer. Endocrine Related Cancer 12, 245262.Google Scholar
Yatabe, Y., Kosaka, T., Takahashi, T. & Mitsudomi, T. (2005). EGFR mutation is specific for terminal respiratory unit type adenocarcinoma. American Journal of Surgical Pathology 29, 633639.Google Scholar
Yokomizo, A., Tindall, D. J., Drabkin, H., Gemmill, R., Franklin, W., Yang, P., Sugio, K., Smith, D. I. & Liu, W. (1998). PTEN/MMAC1 mutations identified in small cell, but not in non-small cell lung cancers. Oncogene 17, 475479.Google Scholar
Zuo, Z., Chen, S. S., Chandra, P. K., Galbincea, J. M., Soape, M., Doan, S., Barkoh, B. A., Koeppen, H., Medeiros, L. J. & Luthra, R. (2009). Application of COLD-PCR for improved detection of KRAS mutations in clinical samples. Modern Pathology 22, 10231031.Google Scholar
Figure 0

Table 1. Characteristics of PCR amplification of EGFR and KRAS

Figure 1

Table 2. Clinical data of non-small cell lung cancer (NSCLC) patients

Figure 2

Table 3. Allele and genotype frequencies of rs1050171 and rs10241451 in patients and controls

Figure 3

Fig. 1. Analysis of genomic ancestry of patients with lung cancer. EU, Europeans; AF, African; AM, Amerindians. Significant difference was found between Africans when compared with control group (P = 0·004).