<<

Oncogene (2009) 28, S14–S23 & 2009 Macmillan Publishers Limited All rights reserved 0950-9232/09 $32.00 www.nature.com/onc REVIEW Overview of molecular testing in non-small-cell lung : mutational analysis, gene copy number, protein expression and other biomarkers of EGFR for the prediction of response to inhibitors

T John, G Liu and M-S Tsao

University Health Network, Princess Margaret Hospital Site, Ontario Cancer Institute, University of Toronto, Toronto, Ontario, Canada

Most patients with non-small-cell (NSCLC) and seem to be more reliable predictors of response than present with advanced disease. Current treatment para- clinical features. This review discusses candidate mole- digms are shifting from cytotoxic alone to cular predictive markers of anti-EGFR and the single-agent and combination biological and targeted different methodologies used to assess them. . As patient responses to these therapies vary, predictive biomarkers will be an important facet of a patient’s diagnostic workup in , as there is accumulating evidence that they may enable the Molecular predictors of response to EGFR antagonists prognostication and prediction of therapeutic response. Potential biomarkers for the selection of patients with Protein expression by immunohistochemistry NSCLC most likely to benefit from epidermal growth There are several ways to detect EGFR protein and factor (EGFR) tyrosine kinase inhibitors (TKIs), measure its levels. These include radioactive-labeled ligand such as gefitinib and , include , gene binding, competitive immunoassay, western blotting and copy number increase and single-nucleotide polymorph- immunohistochemistry. The first three methods require isms of the EGFR gene, EGFR protein expression and protein extraction from fresh or snap-frozen samples. oncogenic on the KRAS gene. Many techniques Although these methods are quantitative, they require are available to assay for these biomarkers. In this review, special laboratory setups that are more suitable for basic we present the current weight of evidence for using these research. In addition, as protein is extracted from whole methods as biomarkers for anti-EGFR therapy in patients tissue, normalization of results for tumor cell content can with NSCLC. be problematic. Immunohistochemistry has several ad- Oncogene (2009) 28, S14–S23; doi:10.1038/onc.2009.197 vantages over the above assays: (1) protein expression level can be evaluated directly on tumor cells or on other Keywords: erlotinib; gefitinib; polymerase chain reaction; specific cells of interest; (2) the technique can be performed biomarker; survival on formalin-fixed, paraffin-embedded (FFPE) tumor tissues, which represent the largest archival bank of human tissue samples; and (3) immunohistochemistry is a technique that is routinely used in clinical pathology Introduction practice. Countering these advantages, however, is the fact that quantification of expression levels by visual inspection Targeting the epidermal receptor (EGFR) canberegardedassemiquantitativeatbestandachallenge using tyrosine kinase inhibitors (TKIs) has proven to be to standardize. an effective treatment strategy for advanced non-small- Most laboratories use the indirect immunolabeling cell lung cancer (NSCLC). However, not all patients method for immunohistochemistry. The factors that are benefit from TKI treatment, and several investigators most likely to influence immunoreactivity and staining have sought to identify a priori markers that will predict include types of tissue fixative and condition of fixation, a response to, and benefit from treatment. Certain as well as the reactivity and specificity of the primary clinical characteristics have been shown to be associated antibody (Atkins et al., 2004; Dei Tos and Ellis, 2005; with an increased likelihood of responsiveness to EGFR Penault-Llorca et al., 2006; Buckley and Kakar, 2007). TKIs (female gender, nonsmoking history, adenocarci- Several commercial antibodies for EGFR are available. noma histology and East Asian race) (Miller et al., The two most commonly used are the DakoCytomation 2004). In addition, several molecular predictors are (Carpentaria, CA, USA) clone 18C9, which is used in the associated with responsiveness to EGFR TKI treatment, pharmDx , and the Zymed Laboratories Inc. (South San Francisco, CA, USA) clone 31G7 (Bhargava et al., 2006; Penault-Llorca et al., 2006). To obtain consistent Correspondence: Dr M-S Tsao, Room 7-611, 610 University Avenue, Toronto, Ontario, Canada M5G 2M9. results, immunohistochemistry studies require carefully E-mail: [email protected] controlled and validated standard operating procedures. Molecular testing of EGFR in NSCLC T John et al S15 Immunohistochemistry results can be evaluated qua- chromosome corresponding to the gene, with a specific litatively for the absence or presence of staining of the fluorescent dye. As multiple fragments can be labeled studied cells, and semiquantitatively for the intensity of using fluorophores with different emission wavelengths, staining, percentage of stained cells or both. In addition, multiple FISH probes can be co-hybridized to detect by multiplying the staining intensity by the percentage different genes in a single assay. The fluorescent labels of cells stained, an H-score can be derived. A simple enable the visualization of the number of copies and H-score may be based on a single predominant staining location of the target genes in metaphase as well as intensity, whereas more complex H-scores can include interphase nuclei. The technique for the EGFR FISH the sum of individual H-scores for each intensity level assay is well described (Varella-Garcia, 2006) and observed. The sample can then be scored as positive or performed routinely using B4-mm-thick FFPE tissue negative on the basis of specific adopted cutoff points. sections. The most commonly used probe is the LSI The method for determining such a cutoff is one of the EGFR SpectrumOrange/CEP 7 SpectrumGreen probe controversial issues in immunohistochemistry scoring. set (Vysis Inc., Downers Grove, IL, USA). This probe set includes directly labeled DNA FISH probes for the EGFR immunohistochemistry as a prognostic and EGFR gene and the centromere of chromosome 7. There predictive biomarker are several technical considerations that could compro- Studies using different antibody sources have reported a mise the FISH assay, including lengths of proteinase K high prevalence of EGFR immunostaining in NSCLCs digestion and probe hybridization duration and condi- (Rusch et al., 1997; Fontanini et al., 1998; Hsieh et al., tion (Varella-Garcia, 2006). 2000; Hirsch et al., 2003). The rate is consistently higher in The currently used EGFR FISH scoring system was squamous-cell (55–100%) than in adenocarci- developed by the Colorado group (Cappuzzo et al., nomas (35–60%) or large-cell carcinomas (20–60%). 2005) and requires the use of an epifluorescence Several studies have shown an association between EGFR microscope equipped with a genetic workstation. The protein expression and response to EGFR TKIs; however, cutoffs used for this scoring system were determined in a there have been conflicting results with regard to the value retrospective study of gefitinib-treated patients with of EGFR protein expression in predicting survival benefit advanced NSCLC (Cappuzzo et al., 2005). Unlike the with EGFR TKIs (see Hirsh et al., 2009). FISH assay for the HER2 gene in breast cancer, the scoring system for the EGFR gene copy changes in NSCLC is more complex, perhaps reflecting the EGFR immunohistochemistry as a clinical test different biological impact of the two genes in the two Several major issues with technical aspects of immuno- diseases. The system stratifies tumors into six categories histochemistry require standardization, although the on the basis of the quantity of tumor cells with a specific most important variables include the type and source of EGFR gene copy number compared with the chromo- antibody and the scoring system used (Dei Tos and Ellis, some 7 copy number, which is scored using the CEP7 2005; Eberhard et al., 2008; Hirsch et al., 2008a). probe. Tumors are considered FISH positive if they Discordance between antibodies may be significant, show amplification (the presence of tight gene clusters ranging from 12 to 24% (Buckley and Kakar, 2007; and a ratio of X2 genes/chromosomes per cell or X15 Hirsch et al., 2008a). EGFR immunohistochemistry is gene copies per cell in X10% of analysed cells) or high therefore not yet optimal in determining a patient’s polysomy (X4 copies of the gene in X40% of cells). eligibility to receive EGFR TKI therapy (Ciardiello and Tumors considered FISH negative are those with no Tortora, 2008), despite results indicating that EGFR amplification as defined above or with 40% of cells immunohistochemistry-negative patients demonstrate o showing X4 copies of the EGFR gene per cell (Varella- reduced responses and clinical benefit. Garcia, 2006). EGFR gene copy or dosage The EGFR gene is located on the short arm of EGFR FISH as a prognostic and predictive biomarker chromosome 7 (7p21), which is commonly overrepre- The association between the high EGFR gene copy by sented or amplified in NSCLC. There are several FISH (FISH-positive), as defined by the Colorado methods for detecting and determining EGFR gene group (Cappuzzo et al., 2005), and the clinical end copy number and dosage, including fluorescence in situ points of EGFR TKI therapies has been investigated in hybridization (FISH), chromogenic in situ hybridization a number of studies. In the Italian Expanded Access (CISH) and real-time quantitative polymerase chain Study of gefitinib, the BR.21 and the ISEL studies, an reaction (qPCR). association was observed between EGFR FISH positiv- ity and either increased response rates or an improved FISH survival benefit (Hirsch et al., 2003, 2006; Tsao et al., The development of fluorescent-labeling techniques has 2005; see Hirsch et al., 2009). The high EGFR gene copy enabled multiple targets on the same or separate number detected by FISH may also be predictive of chromosomes to be analysed simultaneously, thereby benefit from combined with allowing control probes to be hybridized and used as a (Hirsch et al., 2008b). reference (Trask et al., 1985). This involves labeling the Compared with reports in Caucasian patients, only a DNA fragment of a gene, or the bacterial artificial limited number of FISH studies have been conducted in

Oncogene Molecular testing of EGFR in NSCLC TJohnet al S16 East Asian patients with NSCLC treated with EGFR scoring system proposed for FISH (Cappuzzo et al., TKIs (Ichihara et al., 2007; Sone et al., 2007). In the 2005) may not be extrapolated automatically to CISH. studies by Ichihara et al. (2007) and Sone et al. (2007), Nevertheless, with a scoring system that depends only 41% of patients were FISH positive, but FISH positivity on an EGFR gene signal, CISH is an alternative method was not associated with survival benefit, suggesting a for assessing EGFR gene copy status. discrepancy between East Asian and Caucasian ethni- In a recent study comparing the performance of FISH cities. It is hoped that the FISH analysis from the and CISH for the detection of increased EGFR copy recently presented I-PASS study will further clarify this number in 77 patients with NSCLC treated by surgery issue (Mok et al., 2008). alone (Figure 1), a significant correlation was found between the number of signals detected by both methods (Spearman’s r ¼ 0.81; Po0.0001) (Sholl et al., 2007). CISH Discrepancies between the two tests were seen in only CISH is an alternative assay to FISH for determining 7% of cases. At a signal of 4.5, CISH had very high gene copy number and amplification in tissue sections. sensitivity (89%) and specificity (89%) for the discrimi- The major difference between the two methods is that nation of low and high EGFR polysomy (Po0.0001). At FISH requires an epifluorescent system, whereas CISH a signal of 7.1, CISH was also effective at distinguishing can be evaluated by the pathologist using bright-field between high polysomy and amplification (P ¼ 0.0003). microscopy and correlated directly with histopathology. Another study comparing CISH and FISH in 58 Furthermore, as with immunohistochemistry, CISH- NSCLC samples showed similar results, with a high stained slides can be archived and stored permanently. concordance between EGFR copy number determined The DNA probes are labeled with digoxigenin or by the two tests (Gallegos Ruiz et al., 2007). Overall, biotin (Gallegos Ruiz et al., 2007; Sholl et al., 2007; these data support the feasibility of using CISH to Chang et al., 2008; Li et al., 2008) and then detected evaluate the function of EGFR gene copy as a predictive using a CISH Detection Kit (Zymed Laboratories, marker for EGFR TKI therapy, although further South San Francisco, CA, USA). Gene/chromosome evaluation and validation in samples is probes are visualized as dark brown dots, which are needed before this technique can be adopted clinically. counted in parallel sections using standard light micro- scopy and are converted into the number of nuclear signals per cell. CISH may be useful in samples that are qPCR difficult to interpret by fluorescence microscopy because Several different methods of performing qPCR mea- of sample heterogeneity, high autofluorescent back- surements can be used to detect small amounts of RNA ground signal or both (Daniele et al., 2007). The main or DNA (Bustin, 2002). Using probes that are labeled potential drawback of CISH compared with FISH is with both a fluorophore and fluorescence-quenching that most CISH studies have been performed on frozen molecule, amplicon generation can be detected and sections and only a single color can be developed per quantified in real time during a PCR. An alternative and section, meaning that one is not able to evaluate a less expensive method uses the double-stranded DNA control probe for aneusomy. Therefore, the complex intercalating agent, SYBR Green, as the signal emitter.

Figure 1 Fluorescent in situ hybridization (FISH) and chromogenic in situ hybridization (CISH) for EGFR gene copy in NSCLC. EGFR FISH probe (green) and 7q control probe (red) show EGFR gene high polysomy (a) and amplification (c). The corresponding slides stained with the EGFR gene probe by the CISH method are shown in (b and d). Adapted with permission from Sholl et al. (2007).

Oncogene Molecular testing of EGFR in NSCLC T John et al S17 Using multiple primers labeled with different fluoro- anced PCR amplification and the relative amount of phores, current qPCR technology can quantitate multi- contaminating wild-type allele of non-tumor cells can ple genes, including control genes, simultaneously in one influence the sensitivity of mutant detection by direct reaction. sequencing (Engelman et al., 2006). In addition, DNA The greatest pitfall for using qPCR to measure gene extracted from FFPE tissues could give rise to sequen- dosage is the inability to control for contamination from cing artifacts, thus requiring a confirmation by the normal host cells that result in the dilution of tumor cell sequencing of independent PCR products. gene copy. Performing laser-capture microdissection can Owing to concern regarding the sensitivity of the reduce this problem, but this is laborious and clinically direct-sequencing method, a variety of other methods impractical. The other issue with qPCR-based gene have been investigated to detect low abundance muta- dosage estimation is normalization. Normalization tions and increase the sensitivity of the mutation assay. using another region of the same chromosome as the These include high-resolution melting analysis (Nomoto studied gene, such as centromeric sequences, will detect et al., 2006; Willmore-Payne et al., 2006; Fukui et al., true amplification, but will not detect increased gene 2008), single-stranded conformational polymorphism copy number due to polysomy. Normalization using and denaturing high-performance liquid chromatogra- centromeric sequences of multiple chromosomes may phy analysis of PCR amplicons after digestion by CEL1 allow the assessment of the degree of aneusomy. endonuclease (Ja¨nne et al., 2006). These methods allow There have been two studies on EGFR gene dosage by the rapid mutation screening of large numbers of qPCR to predict treatment benefit in patients with samples with increased sensitivity but still require direct NSCLC receiving TKI therapy. Both studies failed to sequencing to identify or confirm mutations. The demonstrate its clinical applicability (Bell et al., 2005; amplification refractory mutation system (ARMS) Dziadziuszko et al., 2006). EGFR qPCR was not (Newton et al., 1989; Whitcombe et al., 1999), combined predictive of response to treatment, disease control, with the scorpion method (Kimura et al., 2006) and the progression-free survival or overall survival, nor did it peptide nucleic acid-locked clamping method (Nagai correlate with FISH, immunohistochemistry or mRNA et al., 2005), are alternative sensitive methods that may expression (Bell et al., 2005; Takano et al., 2005; be used to detect rapidly specific EGFR mutations using Dziadziuszko et al., 2006, 2007). real-time PCR-based technology.

Somatic EGFR gene mutations EGFR mutations as biomarkers In May 2004, two independent groups of investigators Exon 19 deletion and exon 21 L858R substitution reported the discovery of somatic mutations in the TK account for 85–90% of the drug-sensitive EGFR domain (exons 18–23) of EGFR (Lynch et al., 2004; mutations seen in NSCLC (Riely et al., 2006; Sakurada Paez et al., 2004). Practically all mutations that have et al., 2006). The mutations are more common in been reported are on exons 18 through 21. These , in persons of East Asian ethnicity, mutations affect the (ATP)- women and in never smokers. These phenotypic features binding cleft of EGFR, which is also where TKIs bind. had already been marked as clinical predictors of In vitro studies have shown that EGFR mutants have response to TKIs before the discovery of the mutations constitutive TK activity and, therefore, a greater (Fukuoka et al., 2003; Kris et al., 2003). The prevalence sensitivity to anti-EGFR inhibition. Among all muta- of EGFR mutations varies by ethnicity with ranges from tions, four predominantly result in TKI drug sensitivity 20–40% in Asian populations (Huang et al., 2004; by in vitro and in vivo studies. These include point Kosaka et al., 2004; Han et al., 2005; Shigematsu et al., mutations in exons 18 (G719A/C) and 21 (L858R and 2005; Tokumo et al., 2005) to 5–20% among Caucasians L861Q) and in-frame deletions in exon 19, which (Cappuzzo et al., 2005; Marchetti et al., 2005). eliminate four amino acids (LREA) downstream of the The association between the presence of activating lysine residue at position 745; other mutations appear to EGFR mutations and high response rates to TKIs was be associated with variable or less sensitivity (Eberhard demonstrated in a number of phase II studies in which et al., 2005; Chen et al., 2006; Sharma et al., 2007; see all patients received EGFR TKI therapy. The overall Gazdar, 2009). response rates were as high as 80% in patients harboring The most commonly used method to detect mutations mutations compared with p10% in patients with wild- is direct sequencing of the PCR-amplified exon se- type EGFR (Huang et al., 2004; Lynch et al., 2004; Mu quences. Tumor sections are initially stained with et al., 2004; Paez et al., 2004; Pao et al., 2004; Bell et al., hematoxylin and eosin (H&E) or methylene blue to 2005; Cappuzzo et al., 2005; Chou et al., 2005; Cortes- determine tumor cell percentage and to account for all Funes et al., 2005; Eberhard et al., 2005; Han et al., cellular components present. For the direct sequencing 2005; Kim et al., 2005; Kondo et al., 2005; Mitsudomi method, samples with tumor cellularity o40% should et al., 2005; Takano et al., 2005; Taron et al., 2005; be enriched by microdissection. DNA is extracted from Tomizawa et al., 2005; Tsao et al., 2005; Zhang et al., the selected tumor tissue using standard methods or a 2005; Shih et al., 2006; Uramoto et al., 2006). In commercially available kit. After PCR amplification of addition, the presence of mutations has also been each exon of the EGFR TK domain, amplicons are associated with improved progression-free survival and sequenced. The copy number of mutant allele, imbal- overall survival with EGFR TKI treatment compared

Oncogene Molecular testing of EGFR in NSCLC TJohnet al S18 with patients with wild-type EGFR (Cappuzzo et al., American study (Liu et al., 2008) found associations 2005; Cortes-Funes et al., 2005; Han et al., 2005; with lung cancer survival or toxicity with gefitinib, Mitsudomi et al., 2005; Takano et al., 2005; Tsao whereas others found no significant association (Ichi- et al., 2005); however, several studies have since reported hara et al., 2007; Gregorc et al., 2008). Shorter CA that non-TKI-treated patients with NSCLC and EGFR repeat lengths were associated with poorer survival in mutations have a more favorable prognosis than do the absence of therapy with an EGFR TKI, which is a patients with wild-type EGFR (Eberhard et al., 2005; reversal of expectations in TKI-treated patients (Dubey Riely et al., 2006; Sasaki et al., 2006; Kosaka et al., 2007; et al., 2006). Yoshida et al., 2007; see Gazdar, 2009). The multidrug transporter ABCG2 was shown to be Mutations in the kinase domain of EGFR have also active in removing gefitinib from cells (Elkind et al., been associated with an acquired resistance to EGFR 2005). In published reports, ABCG2 polymorphisms TKIs. A secondary mutation involving the substitution have been identified as being associated with increased of methionine for threonine in codon 790 () is EGFR TKI concentrations, toxicity or both, in patients present in approximately 50% of cases of acquired treated with gefitinib and erlotinib (Cusatis et al., 2006; resistance (Kobayashi et al., 2005; Pao et al., 2005a; Li et al., 2007; Rudin et al., 2008). Kosaka et al., 2006; see Gazdar, 2009). Screening for the In summary, genetic polymorphisms may be impor- emergence of the T790M mutation during treatment tant contributors to variability in response to EGFR may allow for the earlier identification of acquired TKI therapy, but evidence is thus far contradictory. resistance. A recent study showed that T790M can be Several reasons exist to explain the inconsistent data: detected before EGFR TKI treatment with a highly (1) studies have been underpowered, with sample sizes sensitive allele-specific assay (Maheswaran et al., 2008). generally lower than 200 patients; (2) different definitions The investigators found that the pretreatment presence are used for key variables, such as ‘short’ and ‘long’ of T790M did not preclude responses with EGFR TKIs, intron 1 CA repeat lengths and ‘clinical toxicity’; (3) but was associated with significantly shorter progres- confounders such as race, polymorphism allele frequen- sion-free survival compared with patients without cies, mutation rates, copy number changes and other detectable T790M before TKI treatment (7.7 vs 16.5 clinical and molecular prognostic and predictive factors months; Po0.001). These results suggest that T790M have rarely been accounted for in these polymorphism may be a useful pretreatment biomarker for identifying analyses. Comprehensive analyses are needed to deter- patients who are not likely to experience long-term mine whether genetic polymorphisms are independently benefits with reversible EGFR TKIs (that is, erlotinib predictive with EGFR TKI therapy, are simply corre- and gefitinib). lated with other molecular or clinical prognostic factors, or are prognostic but not predictive.

Genetic polymorphisms and EGFR-targeted drugs The EGFR gene contains numerous genetic polymorphic KRAS gene mutation variants. The evaluation of germ line genetic variants RAS is considered a ‘master switch’ signal transduction uses DNA extracted from any non-tumor source, protein downstream of multiple types of cell surface including leukocytes from whole blood. Several genetic receptors, including tyrosine kinase and G-protein- variants are associated with alterations in the mRNA coupled receptors (Aviel-Ronen et al., 2006; Molina gene expression (Liu et al., 2005). Both the EGFR and Adjei, 2006). A point mutation on codons 12, 13 and –216G/T and –191C/A polymorphisms are located in the 61 of all RAS family genes will lead to the inability of the transcriptional start site region of the promoter, wherein RAS–guanosine triphosphatase (RAS–GTPase)-activat- multiple nuclear regulatory affinity sites are located (Liu ing protein to hydrolyse bound GTP to guanosine et al., 2005). Two studies in Caucasian patients reported diphosphate, thus resulting in a constitutive activation that the –216G/T variant, alone or in combination with of the RAS protein. In NSCLC, >90% of RAS –191C/A, was associated with improved outcome, mutations involve the KRAS gene, with most occurring greater toxicity to gefitinib or both (Cusatis et al., in codon 12. The most direct way to detect KRAS codon 2006; Liu et al., 2008), whereas a third found no 12 and 13 mutations is by PCR amplification of exon 2 association (Gregorc et al., 2008). Among Asians, these and by direct sequencing of the amplicon. However, all variants are rare (Ichihara et al., 2007). the previously discussed caveats for the detection of One of the enhancer elements for EGFR is located in EGFR mutations using this technique on FFPE samples intron 1 (Maekawa et al., 1989; Haley and Waterfield, apply, especially the need to enrich samples to X50% 1991). Shorter alleles of the dinucleotide CA repeat tumor cellularity. The need for repeat PCR to confirm sequence polymorphism in intron 1 of EGFR are mutations is less of a concern, as any mutations detected associated with a greater EGFR expression than are outside codons 12, 13 and 61 can be ignored. the longer repeats (Gebhardt et al., 1999; Etienne- Pao et al. (2005b) first suggested that patients with Grimaldi et al., 2005). In Asian patients, a shorter CA NSCLC whose tumors harbor mutations are resistant to repeat length was associated with a greater EGFR EGFR TKI. To date, among 75 NSCLC patients expression and copy number (Zhou et al., 2006). Asian harboring KRAS mutations who were treated with patients tend to have longer CA repeat lengths. Two TKIs (Pao et al., 2005b; Hirsch et al., 2006; Massarelli Asian studies (Han et al., 2007; Nie et al., 2007) and one et al., 2007; Miller et al., 2008; Zhu et al., 2008), only

Oncogene Molecular testing of EGFR in NSCLC T John et al S19 one patient has been reported to respond (Zhu et al., pretreated metastatic NSCLC and in 40–50% of East 2008). Interestingly, this patient’s tumor also had an Asian patients. The ability to predict significant clinical EGFR gene amplification. On the basis of TKI response benefits with appropriate biomarkers would be a major rates of up to 10% in patients with wild-type EGFR, one advancement in the achievement of personalized med- would expect that at least seven to eight of these 75 icine for patients with NSCLC, as it would allow the patients should have responded, suggesting that mu- tailoring of treatment with TKIs to patients who are tated KRAS results in TKI resistance. In the TRIBUTE most likely to benefit, whereas other patients would study, patients with KRAS mutant tumors, who were receive alternative therapies that may also be effective. treated with a combination of erlotinib and chemother- To date, several such biomarkers and various techno- apy, showed poorer survival than did patients treated logies to assay them have been described and studied, with chemotherapy alone or patients with wild-type but none is currently ready for clinical implementation. KRAS, regardless of treatment type (Eberhard et al., This is largely because of the fact that most biomarker 2005). In the BR.21 study, KRAS mutations were found study results have been derived from phase II trials in in 30 (15%) of 206 patients tested. The HR of erlotinib which all patients were treated, whereas very limited to placebo treatment in 30 patients with a KRAS data are available from the results of phase III trials that mutation was 1.67 (95% CI, 0.62–4.50; P ¼ 0.31), included both drug-treated and placebo-controlled whereas for 176 patients with wild-type KRAS it was patients. The latter is important when one is trying to 0.69 (95% CI, 0.49–0.97; P ¼ 0.03), with an insignificant identify significant predictive markers that truly demon- interaction P-value of 0.09. In a multivariate analysis, strate differential benefits of therapies between marker- KRAS mutation was neither prognostic for poorer negative and marker-positive patients (Shepherd and survival in untreated patients nor predictive of differ- Tsao, 2006). ential treatment effect (Zhu et al., 2008). Overall, these To maximize the therapeutic index and cost-effective- results suggest that patients with NSCLC who have ness of anti-EGFR therapy, both negative and positive KRAS mutations are unlikely to derive any benefit from predictive biomarkers should be considered. For nega- current TKI therapy, although the number of reported tive markers that may potentially exclude patients from cases remains limited. receiving therapy, the candidate markers are EGFR immunohistochemical negativity, KRAS mutation and MET amplification. Each of these has its respective MET amplification shortcomings. For immunohistochemistry, a standar- Using gefitinib-resistant NSCLC cell lines, Engelman dized assay and quantitative scoring procedures need to et al. (2007) recently found that amplification of the be established and validated prospectively. Despite MET proto-oncogene may be involved in resistance to accumulating evidence for KRAS tumors being resistant EGFR TKIs by enabling cells to activate the PI3K and to EGFR TKIs, the number of reported cases remains Akt-signaling pathway independently of EGFR through low ( 100 cases), especially considering that only 10% HER3 activation. MET is commonly overexpressed in o of tumors that are negative for both KRAS and EGFR NSCLC (Tsao et al., 1998; Ma et al., 2005) and may be a mutations still respond to TKIs. et al. et al. poor prognostic marker (Cheng , 2005; Rossi , Both EGFR mutation and a high gene copy number 2005). It has been speculated that MET amplification (FISH positivity) are promising positive selection accounts for approximately 10–20% of acquired resis- biomarkers (that is, markers to select patients who are tance to gefitinib and erlotinib (Engelman and Janne, ¨ likely to respond to TKI therapy). The current adoption 2008). In addition, preclinical data suggest that inhibi- of EGFR mutation as a selection marker would exclude tion of MET can restore sensitivity to gefitinib (Engel- EGFR TKI therapy in 80–90% of non-East Asian man et al., 2007). Although this requires further patients, whereas 50–60% would be excluded when elucidation in a clinical setting, the dual inhibition of using EGFR gene copy (FISH positivity) as a marker. EGFR and MET may be an approach to overcome However, the value of mutational analysis for predicting resistance to EGFR TKIs. response may not be uniform across various ethnicities. MET amplification can be detected either by qPCR For example, in Caucasian patients, whose overall or FISH (Engelman et al., 2007). Along with the response rate is 10%, it seems highly sensible to use general challenges that accompany these two techniques, mutational status as a marker to select the 10–20% of an added difficulty centers on the threshold level of patients who are most likely to benefit from anti-EGFR MET amplification, which is clinically relevant for therapy. Yet, many studies have consistently reported EGFR TKI treatment. Currently, this is not well 10% response rates to gefitinib and erlotinib in patients defined and will clearly be important if this molecular with wild-type EGFR, and the HR of erlotinib to factor is to be used as a biomarker for EGFR TKI placebo is 0.74 (Zhu et al., 2008), which is comparable to treatment. the overall survival benefit of erlotinib in all BR.21 patients (Shepherd et al., 2005). Therefore, it may be that one unifying predictive model does not apply to all Practical considerations patients with NSCLC, and that it is more practical to define different models for patients of various ethnicities. EGFR TKIs are associated with significant clinical Finally, the adoption of biomarker analysis into responses in 10–20% of Caucasian patients with heavily clinical practice would be significantly facilitated if such

Oncogene Molecular testing of EGFR in NSCLC TJohnet al S20 tests could be performed using blood samples, especially clinical practice are important issues that need to be considering the fact that for patients with advanced addressed. This can be achieved only by dedicated disease, the diagnosis is often made on fine-needle tumor banking and further molecular profiling of the aspirates, which are not likely to yield adequate material tumors from patients enrolled in large phase III for mutational/FISH testing. Kimura et al. (2006, 2007) randomized studies. reported the ability to detect EGFR mutations in serum samples of patients with advanced NSCLC with approximately 75% sensitivity. More recently, Mahes- Conflict of interest waran et al. (2008) reported 92% sensitivity in the detection of EGFR mutations in isolated circulating M-S Tsao has received consulting/paid advisory board tumor cells, including the detection of the T790M- fees from Roche International, AstraZeneca and Roche resistant mutants in patients undergoing TKI therapy. Canada. M-S Tsao has received lecture fees from These approaches are becoming more feasible with the AstraZeneca. The remaining authors have declared no increasing availability of automated equipment neces- conflict of interest. sary for the isolation of circulating tumor cells and may circumvent the issue of limited tissue availability. The ability to personalize small molecule-based Acknowledgements therapies on the basis of molecular markers remains This article was funded by Boehringer Ingelheim Pharmaceu- elusive in patients with NSCLC, although enormous ticals Inc. Dr John is an International Association for Study of progress has been made in a relatively short period of Lung Cancer (IASLC) Translational Fellow in Lung Cancer time. The most appropriate biomarkers for predicting Research. Dr Liu is the Alan B Brown Chair in Molecular response and the preferred gene-sequencing technology Genomics, and Dr Tsao is the M Qasim Choksi Chair in Lung for the determination of mutational status in routine Cancer Translational Research.

References

Atkins D, Reiffen KA, Tegtmeier CL, Winther H, Bonato MS, Cheng TL, Chang MY, Huang SY, Sheu CC, Kao EL, Cheng YJ et al. Storkel S. (2004). Immunohistochemical detection of EGFR in (2005). Overexpression of circulating c-met messenger RNA is paraffin-embedded tumor tissues: variation in staining intensity due significantly correlated with nodal stage and early recurrence in non- to choice of fixative and storage time of tissue sections. J Histochem small cell lung cancer. Chest 128: 1453–1460. Cytochem 52: 893–901. Chou TY, Chiu CH, Li LH, Hsiao CY, Tzen CY, Chang KT et al. Aviel-Ronen S, Blackhall FH, Shepherd FA, Tsao MS. (2006). K-ras (2005). Mutation in the tyrosine kinase domain of epidermal growth mutations in non-small-cell lung : a review. Clin Lung factor receptor is a predictive and prognostic factor for gefitinib Cancer 8: 30–38. treatment in patients with non-small cell lung cancer. Clin Cancer Bell DW, Lynch TJ, Haserlat SM, Harris PL, Okimoto RA, Res 11: 3750–3757. Brannigan BW et al. (2005). Epidermal Ciardiello F, Tortora G. (2008). EGFR antagonists in cancer mutations and gene amplification in non-small-cell lung cancer: treatment. N Engl J Med 358: 1160–1174. molecular analysis of the IDEAL/INTACT gefitinib trials. J Clin Cortes-Funes H, Gomez C, Rosell R, Valero P, Garcia-Giron C, Oncol 23: 8081–8092. Velasco A et al. (2005). receptor activating Bhargava R, Chen B, Klimstra DS, Saltz LB, Hedvat C, Tang LH mutations in Spanish gefitinib-treated non-small-cell lung cancer et al. (2006). Comparison of two antibodies for immunohistochem- patients. Ann Oncol 16: 1081–1086. ical evaluation of epidermal growth factor receptor expression in Cusatis G, Gregorc V, Li J, Spreafico A, Ingersoll RG, Verweij J et al. colorectal carcinomas, adenomas, and normal mucosa. Cancer 106: (2006). Pharmacogenetics of ABCG2 and adverse reactions to 1857–1862. gefitinib. J Natl Cancer Inst 98: 1739–1742. Buckley AF, Kakar S. (2007). Comparison of the Dako EGFR Daniele L, Macri L, Schena M, Dongiovanni D, Bonello L, Armando pharmDx kit and Zymed EGFR antibody for assessment of EGFR E et al. (2007). Predicting gefitinib responsiveness in lung cancer by status in colorectal . Appl Immunohistochem Mol fluorescence in situ hybridization/chromogenic in situ hybridization Morphol 15: 305–309. analysis of EGFR and HER2 in biopsy and cytology specimens. Bustin SA. (2002). Quantification of mRNA using real-time reverse Mol Cancer Ther 6: 1223–1229. transcription PCR (RT-PCR): trends and problems. JMol Dei Tos AP, Ellis I. (2005). Assessing epidermal growth factor receptor Endocrinol 29: 23–39. expression in tumours: what is the value of current test methods? Cappuzzo F, Hirsch FR, Rossi E, Bartolini S, Ceresoli GL, Bemis L Eur J Cancer 41: 1383–1392. et al. (2005). Epidermal growth factor receptor gene and protein and Dubey S, Stephenson P, Levy DE, Miller JA, Keller SM, Schiller JH gefitinib sensitivity in non-small-cell lung cancer. J Natl Cancer Inst et al. (2006). EGFR dinucleotide repeat polymorphism as a 97: 643–655. prognostic indicator in non-small cell lung cancer. J Thorac Oncol Chang JW, Liu HP, Hsieh MH, Fang YF, Hsieh MS, Hsieh JJ et al. 1: 406–412. (2008). Increased epidermal growth factor receptor (EGFR) gene Dziadziuszko R, Holm B, Skov BG, Osterlind K, Sellers MV, Franklin copy number is strongly associated with EGFR mutations WA et al. (2007). Epidermal growth factor receptor gene copy and adenocarcinoma in non-small cell lung : a chromogenic number and protein level are not associated with outcome of non- in situ hybridization study of 182 patients. Lung Cancer 61: small-cell lung cancer patients treated with chemotherapy. Ann 328–329. Oncol 18: 447–452. Chen YR, Fu YN, Lin CH, Yang ST, Hu SF, Chen YT et al. (2006). Dziadziuszko R, Witta SE, Cappuzzo F, Park S, Tanaka K, Distinctive activation patterns in constitutively active and gefitinib- Danenberg PV et al. (2006). Epidermal growth factor receptor sensitive EGFR mutants. Oncogene 25: 1205–1215. messenger RNA expression, gene dosage, and gefitinib

Oncogene Molecular testing of EGFR in NSCLC T John et al S21 sensitivity in non-small cell lung cancer. Clin Cancer Res 12: 3078– responsiveness in non-small-cell lung cancer. Pharmacogenet 3084. Genomics 17: 313–319. Eberhard DA, Giaccone G, Johnson BE. (2008). Biomarkers of Han SW, Kim TY, Hwang PG, Jeong S, Kim J, Choi IS et al. (2005). response to epidermal growth factor receptor inhibitors in Non- Predictive and prognostic impact of epidermal growth factor Small-Cell Lung Cancer Working Group: standardization for use in receptor mutation in non-small-cell lung cancer patients treated the clinical trial setting. J Clin Oncol 26: 983–994. with gefitinib. J Clin Oncol 23: 2493–2501. Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Hirsch FR, Dziadziuszko R, Thatcher N, Mann H, Watkins C, Herbst RS et al. (2005). Mutations in the epidermal growth factor Parums DV et al. (2008a). Epidermal growth factor receptor receptor and in KRAS are predictive and prognostic indicators in immunohistochemistry: comparison of antibodies and cutoff points patients with non-small-cell lung cancer treated with chemotherapy to predict benefit from gefitinib in a phase 3 placebo-controlled alone and in combination with erlotinib. J Clin Oncol 23: study in advanced non-small-cell lung cancer. Cancer 112: 5900–5909. 1114–1121. Elkind NB, Szentpetery Z, Apati A, Ozvegy-Laczka C, Varady G, Hirsch FR, Herbst RS, Olsen C, Chansky K, Crowley J, Kelly K et al. Ujhelly O et al. (2005). Multidrug transporter ABCG2 prevents (2008b). Increased EGFR gene copy number detected by fluorescent tumor cell death induced by the epidermal growth factor receptor in situ hybridization predicts outcome in non-small-cell lung cancer inhibitor Iressa (ZD1839, Gefitinib). Cancer Res 65: 1770–1777. patients treated with cetuximab and chemotherapy. J Clin Oncol 26: Engelman JA, Ja¨nne PA. (2008). Mechanisms of acquired resistance to 3351–3357. epidermal growth factor inhibitors in non- Hirsch FR, Varella-Garcia M, Bunn Jr PA, Di Maria MV, Veve R, small cell lung cancer. Clin Cancer Res 14: 2895–2899. Bremmes RM et al. (2003). Epidermal growth factor receptor in Engelman JA, Mukohara T, Zejnullahu K, Lifshits E, Borras AM, non-small-cell lung carcinomas: correlation between gene copy Gale CM et al. (2006). Allelic dilution obscures detection of a number and protein expression and impact on prognosis. J Clin biologically significant resistance mutation in EGFR-amplified lung Oncol 21: 3798–3807. cancer. J Clin Invest 116: 2695–2706. Hirsch FR, Varella-Garcia M, Bunn Jr PA, Franklin WA, Dziad- Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, ziuszko R, Thatcher N et al. (2006). Molecular predictors of Park JO et al. (2007). MET amplification leads to gefitinib resistance outcome with gefitinib in a phase III placebo-controlled study in in lung cancer by activating ERBB3 signaling. Science 316: advanced non-small-cell lung cancer. J Clin Oncol 24: 5034–5042. 1039–1043. Hirsch FR, Varella-Garcia M, Cappuzzo F. (2009). Predictive value of Etienne-Grimaldi MC, Pereira S, Magne N, Formento JL, Francoual EGFR and HER2 overexpression in advanced non-small-cell lung M, Fontana X et al. (2005). Analysis of the dinucleotide repeat cancer. Oncogene 28(Suppl 1): S32–S37. polymorphism in the epidermal growth factor receptor (EGFR) Hsieh ET, Shepherd FA, Tsao MS. (2000). Co-expression of epidermal gene in patients. Ann Oncol 16: 934–941. growth factor receptor and transforming growth factor-alpha is Fontanini G, De Laurentiis M, Vignati S, Chine S, Lucchi M, Silvestri independent of ras mutations in lung adenocarcinoma. Lung Cancer V et al. (1998). Evaluation of epidermal growth factor-related 29: 151–157. growth factors and receptors and of neoangiogenesis in completely Huang SF, Liu HP, Li LH, Ku YC, Fu YN, Tsai HY et al. (2004). resected stage I-IIIA non-small-cell lung cancer: and High frequency of epidermal growth factor receptor mutations with microvessel count are independent prognostic indicators of survival. complex patterns in non-small cell lung cancers related to gefitinib Clin Cancer Res 4: 241–249. responsiveness in Taiwan. Clin Cancer Res 10: 8195–8203. Fukui T, Ohe Y, Tsuta K, Furuta K, Sakamoto H, Takano T et al. Ichihara S, Toyooka S, Fujiwara Y, Hotta K, Shigematsu H, Tokumo (2008). Prospective study of the accuracy of EGFR mutational M et al. (2007). The impact of epidermal growth factor receptor analysis by high-resolution melting analysis in small samples gene status on gefitinib-treated Japanese patients with non-small- obtained from patients with non-small cell lung cancer. Clin Cancer cell lung cancer. Int J Cancer 120: 1239–1247. Res 14: 4751–4757. Ja¨nne PA, Borras AM, Kuang Y, Rogers AM, Joshi VA, Liyanage H Fukuoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard et al. (2006). A rapid and sensitive enzymatic method for epidermal JY et al. (2003). Multi-institutional randomized phase II trial of growth factor receptor mutation screening. Clin Cancer Res 12: gefitinib for previously treated patients with advanced non-small- 751–758. cell lung cancer (the IDEAL 1 trial) (corrected). J Clin Oncol 21: Kim KS, Jeong JY, Kim YC, Na KJ, Kim YH, Ahn SJ et al. (2005). 2237–2246. Predictors of the response to gefitinib in refractory non-small cell Gallegos Ruiz MI, Floor K, Vos W, Grunberg K, Meijer GA, lung cancer. Clin Cancer Res 11: 2244–2251. Rodriguez JA et al. (2007). Epidermal growth factor receptor Kimura H, Kasahara K, Kawaishi M, Kunitoh H, Tamura T, (EGFR) gene copy number detection in non-small-cell lung cancer; Holloway B et al. (2006). Detection of epidermal growth factor a comparison of fluorescence in situ hybridization and chromogenic receptor mutations in serum as a predictor of the response to in situ hybridization. Histopathology 51: 631–637. gefitinib in patients with non-small-cell lung cancer. Clin Cancer Res Gazdar AF. (2009). Activating and resistance mutations of EGFR in 12: 3915–3921. non-small-cell lung cancer: role in clinical response to EGFR Kimura H, Suminoe M, Kasahara K, Sone T, Araya T, Tamori S et al. tyrosine kinase inhibitors. Oncogene 28(Suppl 1): S24–S31. (2007). Evaluation of epidermal growth factor receptor mutation Gebhardt F, Zanker KS, Brandt B. (1999). Modulation of epidermal status in serum DNA as a predictor of response to gefitinib growth factor receptor gene transcription by a polymorphic (IRESSA). Br J Cancer 97: 778–784. dinucleotide repeat in intron 1. J Biol Chem 274: 13176–13180. Kobayashi S, Boggon TJ, Dayaram T, Ja¨nne PA, Kocher O, Meyerson Gregorc V, Hidalgo M, Spreafico A, Cusatis G, Ludovini V, Ingersoll M et al. (2005). EGFR mutation and resistance of non-small cell RG et al. (2008). Germ line polymorphisms in EGFR and survival lung cancer to gefitinib. N Engl J Med 352: 786–792. in patients with lung cancer receiving gefitinib. Clin Pharmacol Ther Kondo M, Yokoyama T, Fukui T, Yoshioka H, Yokoi K, 83: 477–484. Nagasaka T et al. (2005). Mutations of epidermal growth factor Haley JD, Waterfield MD. (1991). Contributory effects of de novo receptor of non-small cell lung cancer were associated with transcription and premature transcript termination in the regulation sensitivity to gefitinib in recurrence after surgery. Lung Cancer 50: of human epidermal growth factor receptor proto-oncogene RNA 385–391. synthesis. J Biol Chem 266: 1746–1753. Kosaka T, Yatabe Y, Endoh H, Kuwano H, Takahashi T, Mitsudomi Han SW, Jeon YK, Lee KH, Keam B, Hwang PG, Oh DY et al. T. (2004). Mutations of the epidermal growth factor receptor gene (2007). Intron 1 CA dinucleotide repeat polymorphism and in lung cancer: biological and clinical implications. Cancer Res 64: mutations of epidermal growth factor receptor and gefitinib 8919–8923.

Oncogene Molecular testing of EGFR in NSCLC TJohnet al S22 Kosaka T, Yatabe Y, Endoh H, Yoshida K, Hida T, Tsuboi M et al. Mok T, Wu Y-L, Thongprasert S, Yang C-H, Chu D, Saijo N et al. (2006). Analysis of epidermal growth factor receptor gene mutation (2008). Phase III, randomised, open-label, first-line study of gefitinib in patients with non-small cell lung cancer and acquired resistance (G) vs carboplatin/paclitaxel (C/P) in clinically selected patients to gefitinib. Clin Cancer Res 12: 5764–5769. (pts) with advanced non-small cell lung cancer (NSCLC) (IPASS) Kosaka T, Yatabe Y, Onozato R, Mitsudomi T. (2007). Prognostic (abstract). Ann Oncol 19(Suppl 8): viii1. LBA2. implication of the EGFR gene mutations in a large cohort of Mu XL, Li LY, Zhang XT, Wang SL, Wang MZ. (2004). Evaluation Japanese patients with early stage lung adenocarcinoma (abstract). of safety and efficacy of gefitinib (‘Iressa’, ZD1839) as monotherapy J Clin Oncol 25: 7574. in a series of Chinese patients with advanced non-small-cell lung Kris MG, Natale RB, Herbst RS, Lynch Jr TJ, Prager D, Belani CP cancer: experience from a compassionate-use programme. BMC et al. (2003). Efficacy of gefitinib, an inhibitor of the epidermal Cancer 4: 51. growth factor receptor tyrosine kinase, in symptomatic patients Nagai Y, Miyazawa H, Huqun, Tanaka T, Udagawa K, Kato M et al. with non-small cell lung cancer: a randomized trial. JAMA 290: (2005). Genetic heterogeneity of the epidermal growth factor 2149–2158. receptor in non-small cell lung cancer cell lines revealed by a rapid Li AR, Chitale D, Riely GJ, Pao W, Miller VA, Zakowski MF et al. and sensitive detection system, the peptide nucleic acid-locked (2008). EGFR mutations in lung adenocarcinomas: clinical testing nucleic acid PCR clamp. Cancer Res 65: 7276–7282. experience and relationship to EGFR gene copy number and Newton CR, Graham A, Heptinstall LE, Powell SJ, Summers C, immunohistochemical expression. J Mol Diagn 10: 242–248. Kalsheker N et al. (1989). Analysis of any point mutation in DNA: Li J, Cusatis G, Brahmer J, Sparreboom A, Robey RW, Bates SE et al. the amplification refractory mutation system (ARMS). Nucleic (2007). Association of variant ABCG2 and the of Acids Res 17: 2503–2516. epidermal growth factor receptor tyrosine kinase inhibitors in Nie Q, Wang Z, Zhang GC, An SJ, Lin JY, Guo AL et al. (2007). The cancer patients. Cancer Biol Ther 6: 432–438. epidermal growth factor receptor intron1 (CA) n microsatellite Liu G, Gurubhagavatula S, Zhou W, Wang Z, Yeap BY, Asomaning polymorphism is a potential predictor of treatment outcome in K et al. (2008). Epidermal growth factor receptor polymorphisms patients with advanced lung cancer treated with Gefitinib. Eur J and clinical outcomes in non-small-cell lung cancer patients treated Pharmacol 570: 175–181. with gefitinib. J 8: 129–138. Nomoto K, Tsuta K, Takano T, Fukui T, Yokozawa K, Sakamoto H Liu W, Innocenti F, Wu MH, Desai AA, Dolan ME, Cook Jr EH et al. et al. (2006). Detection of EGFR mutations in archived cytologic (2005). A functional common polymorphism in a Sp1 recognition specimens of non-small cell lung cancer using high-resolution site of the epidermal growth factor receptor gene promoter. Cancer melting analysis. Am J Clin Pathol 126: 608–615. Res 65: 46–53. Paez JG, Janne PA, Lee JC, Tracy S, Greulich H, Gabriel S et al. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, (2004). EGFR mutations in lung cancer: correlation with clinical Brannigan BW et al. (2004). Activating mutations in the epidermal response to gefitinib therapy. Science 304: 1497–1500. growth factor receptor underlying responsiveness of non-small-cell Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I et al. lung cancer to gefitinib. N Engl J Med 350: 2129–2139. (2004). EGF receptor gene mutations are common in lung cancers Ma PC, Jagadeeswaran R, Jagadeesh S, Tretiakova MS, Nallasura V, from ‘never smokers’ and are associated with sensitivity of Fox EA et al. (2005). Functional expression and mutations of tumors to gefitinib and erlotinib. Proc Natl Acad Sci USA 101: c-Met and its therapeutic inhibition with SU11274 and small 13306–13311. interfering RNA in non-small cell lung cancer. Cancer Res 65: Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF 1479–1488. et al. (2005a). Acquired resistance of lung adenocarcinomas to Maekawa T, Imamoto F, Merlino GT, Pastan I, Ishii S. (1989). gefitinib and erlotinib is associated with a second mutation in the Cooperative function of two separate enhancers of the human EGFR kinase domain. PLoS Med 2: e73. epidermal growth factor receptor proto-oncogene. J Biol Chem 264: Pao W, Wang TY, Riely GJ, Miller VA, Pan Q, Ladanyi M et al. 5488–5494. (2005b). KRAS mutations and primary resistance of lung adeno- Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, Collura carcinomas to gefitinib or erlotinib. PLoS Med 2: e17. CV et al. (2008). Detection of mutations in EGFR in circulating Penault-Llorca F, Cayre A, Arnould L, Bibeau F, Bralet MP, lung-cancer cells. N Engl J Med 359: 366–377. Rochaix P et al. (2006). Is there an immunohistochemical technique Marchetti A, Martella C, Felicioni L, Barassi F, Salvatore S, Chella A definitively valid in epidermal growth factor receptor assessment? et al. (2005). EGFR mutations in non-small-cell lung cancer: Oncol Rep 16: 1173–1179. analysis of a large series of cases and development of a rapid and Riely GJ, Politi KA, Miller VA, Pao W. (2006). Update on epidermal sensitive method for diagnostic screening with potential implications growth factor receptor mutations in non-small cell lung cancer. Clin on pharmacologic treatment. J Clin Oncol 23: 857–865. Cancer Res 12: 7232–7241. Massarelli E, Varella-Garcia M, Tang X, Xavier AC, Ozburn NC, Rossi G, Cavazza A, Marchioni A, Longo L, Migaldi M, Sartori G Liu DD et al. (2007). KRAS mutation is an important predictor of et al. (2005). Role of chemotherapy and the receptor tyro- resistance to therapy with epidermal growth factor receptor tyrosine sine kinases KIT, PDGFRalpha, PDGFRbeta, and Met in large- kinase inhibitors in non-small-cell lung cancer. Clin Cancer Res 13: cell neuroendocrine carcinoma of the lung. J Clin Oncol 23: 2890–2896. 8774–8785. Miller VA, Kris MG, Shah N, Patel J, Azzoli C, Gomez J et al. (2004). Rudin CM, Liu W, Desai A, Karrison T, Jiang X, Janisch L et al. Bronchioloalveolar pathologic subtype and smoking history predict (2008). Pharmacogenomic and pharmacokinetic determinants of sensitivity to gefitinib in advanced non-small-cell lung cancer. J Clin erlotinib toxicity. J Clin Oncol 26: 1119–1127. Oncol 22: 1103–1109. Rusch V, Klimstra D, Venkatraman E, Pisters PW, Langenfeld J, Miller VA, Riely GJ, Zakowski MF, Li AR, Patel JD, Heelan RT et al. Dmitrovsky E. (1997). Overexpression of the epidermal growth (2008). Molecular characteristics of bronchioloalveolar carcinoma factor receptor and its ligand transforming growth factor alpha is and adenocarcinoma, bronchioloalveolar carcinoma subtype, pre- frequent in resectable non-small cell lung cancer but does not dict response to erlotinib. J Clin Oncol 26: 1472–1478. predict tumor progression. Clin Cancer Res 3: 515–522. Mitsudomi T, Kosaka T, Endoh H, Horio Y, Hida T, Mori S et al. Sakurada A, Shepherd FA, Tsao MS. (2006). Epidermal growth factor (2005). Mutations of the epidermal growth factor receptor gene receptor tyrosine kinase inhibitors in lung cancer: impact of predict prolonged survival after gefitinib treatment in patients with primary or secondary mutations. Clin Lung Cancer 7(Suppl 4): non-small-cell lung cancer with postoperative recurrence. J Clin S138–S144. Oncol 23: 2513–2520. Sasaki H, Shimizu S, Endo K, Takada M, Kawahara M, Tanaka H Molina JR, Adjei AA. (2006). The Ras/Raf/MAPK pathway. J Thorac et al. (2006). EGFR and erbB2 mutation status in Japanese lung Oncol 1: 7–9. cancer patients. Int J Cancer 118: 180–184.

Oncogene Molecular testing of EGFR in NSCLC T John et al S23 Sharma SV, Bell DW, Settleman J, Haber DA. (2007). Epidermal Trask B, van den Engh G, Landegent J, in de Wal NJ, van der Ploeg growth factor receptor mutations in lung cancer. Nat Rev Cancer 7: M. (1985). Detection of DNA sequences in nuclei in suspension by 169–181. in situ hybridization and dual beam flow cytometry. Science 230: Shepherd FA, Rodrigues Pereira J, Ciuleanu T, Tan EH, Hirsh V, 1401–1403. Thongprasert S et al. (2005). Erlotinib in previously treated non- Tsao MS, Liu N, Chen JR, Pappas J, Ho J, To C et al. (1998). small-cell lung cancer. N Engl J Med 353: 123–132. Differential expression of Met/ receptor in Shepherd FA, Tsao MS. (2006). Unraveling the mystery of prognostic subtypes of non-small cell lung cancers. Lung Cancer 20: 1–16. and predictive factors in epidermal growth factor receptor therapy. Tsao MS, Sakurada A, Cutz JC, Zhu CQ, Kamel-Reid S, Squire J J Clin Oncol 24: 1219–1220. et al. (2005). Erlotinib in lung cancer: molecular and clinical Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II predictors of outcome. N Engl J Med 353: 133–144. et al. (2005). Clinical and biological features associated with Uramoto H, Sugio K, Oyama T, Ono K, Sugaya M, Yoshimatsu T epidermal growth factor receptor gene mutations in lung cancers. et al. (2006). Epidermal growth factor receptor mutations are J Natl Cancer Inst 97: 339–346. associated with gefitinib sensitivity in non-small cell lung cancer in Shih JY, Gow CH, Yu CJ, Yang CH, Chang YL, Tsai MF et al. Japanese. Lung Cancer 51: 71–77. (2006). Epidermal growth factor receptor mutations in needle Varella-Garcia M. (2006). Stratification of non-small cell lung cancer biopsy/aspiration samples predict response to gefitinib therapy and patients for therapy with epidermal growth factor receptor survival of patients with advanced non-small cell lung cancer. Int J inhibitors: the EGFR fluorescence in situ hybridization assay. Diagn Cancer 118: 963–969. Pathol 1: 19. Sholl LM, John Iafrate A, Chou YP, Wu MT, Goan YG, Su L et al. Whitcombe D, Theaker J, Guy SP, Brown T, Little S. (1999). (2007). Validation of chromogenic in situ hybridization for detection Detection of PCR products using self-probing amplicons and of EGFR copy number amplification in non-small cell lung fluorescence. Nat Biotechnol 17: 804–807. carcinoma. Mod Pathol 20: 1028–1035. Willmore-Payne C, Holden JA, Layfield LJ. (2006). Detection of Sone T, Kasahara K, Kimura H, Nishio K, Mizuguchi M, Nakatsumi epidermal growth factor receptor and human epidermal growth Y et al. (2007). Comparative analysis of epidermal growth factor factor receptor 2 activating mutations in lung adenocarcinoma by receptor mutations and gene amplification as predictors of gefitinib high-resolution melting amplicon analysis: correlation with gene efficacy in Japanese patients with non-small cell lung cancer. Cancer copy number, protein expression, and hormone receptor expression. 109: 1836–1844. Hum Pathol 37: 755–763. Takano T, Ohe Y, Sakamoto H, Tsuta K, Matsuno Y, Tateishi U et al. Yoshida K, Yatabe Y, Park JY, Shimizu J, Horio Y, Matsuo K et al. (2005). Epidermal growth factor receptor gene mutations and (2007). Prospective validation for prediction of gefitinib sensitivity increased copy numbers predict gefitinib sensitivity in patients with by epidermal growth factor receptor gene mutation in patients with recurrent non-small-cell lung cancer. J Clin Oncol 23: 6829–6837. J Thorac Oncol Taron M, Ichinose Y, Rosell R, Mok T, Massuti B, Zamora L et al. non-small cell lung cancer. 2: 22–28. (2005). Activating mutations in the tyrosine kinase domain of the Zhang XT, Li LY, Mu XL, Cui QC, Chang XY, Song W et al. (2005). epidermal growth factor receptor are associated with improved The EGFR mutation and its correlation with response of gefitinib in survival in gefitinib-treated chemorefractory lung adenocarcinomas. previously treated Chinese patients with advanced non-small-cell Clin Cancer Res 11: 5878–5885. lung cancer. Ann Oncol 16: 1334–1342. Tokumo M, Toyooka S, Kiura K, Shigematsu H, Tomii K, Aoe M Zhou Q, Cheung YB, Jada SR, Lim WT, Kuo WL, Gray JW et al. et al. (2005). The relationship between epidermal growth factor (2006). EGFR intron 1 polymorphism in Asian populations and its receptor mutations and clinicopathologic features in non-small cell correlation with EGFR gene expression and amplification in breast lung cancers. Clin Cancer Res 11: 1167–1173. tumor tissues. Cancer Biol Ther 5: 1445–1449. Tomizawa Y, Iijima H, Sunaga N, Sato K, Takise A, Otani Y et al. Zhu CQ, da Cunha Santos G, Ding K, Sakurada A, Cutz JC, Liu N (2005). Clinicopathologic significance of the mutations of the et al. (2008). Role of KRAS and EGFR as biomarkers of response epidermal growth factor receptor gene in patients with non-small to erlotinib in National Cancer Institute of Canada Clinical Trials cell lung cancer. Clin Cancer Res 11: 6816–6822. Group Study BR.21. J Clin Oncol 26: 4268–4275.

Oncogene