Play all audios:
ABSTRACT BACKGROUND: Epidermal growth factor receptor (EGFR) is overexpressed in a subset of human epidermal growth factor receptor 2 (HER2)-positive breast cancers, and coexpression of HER2
and EGFR has been reported to be associated with poor clinical outcome. Moreover, interaction between HER2 and EGFR has been suggested to be a possible basis for trastuzumab resistance.
METHODS: We analysed the clinical significance of EGFR overexpression and _EGFR_ gene copy number alterations in 242 HER2-positive primary breast cancers. In addition, we examined the
correlations between EGFR overexpression, trastuzumab response and clinical outcome in 447 primary, and 112 metastatic HER2-positive breast cancer patients treated by trastuzumab. RESULTS:
Of the 242 primary cases, the level of EGFR overexpression was 2+ in 12.7% and 3+ in 11.8%. High _EGFR_ gene copy number was detected in 10.3%. Epidermal growth factor receptor
overexpression was associated with hormone receptor negativity and high Ki-67 proliferation index. In survival analyses, EGFR overexpression, but not high _EGFR_ copy number, was associated
with poor disease-free survival in all patients, and in the subgroup not receiving adjuvant trastuzumab. In 447 HER2-positive primary breast cancer patients treated with adjuvant
trastuzumab, EGFR overexpression was also an independent poor prognostic factor. However, EGFR overexpression was not associated with trastuzumab response, progression-free survival or
overall survival in the metastatic setting. CONCLUSIONS: Epidermal growth factor receptor overexpression, but not high _EGFR_ copy number, is a poor prognostic factor in HER2-positive
primary breast cancer. Epidermal growth factor receptor overexpression is a predictive factor for trastuzumab response in HER2-positive primary breast cancer, but not in metastatic breast
cancer. SIMILAR CONTENT BEING VIEWED BY OTHERS DIFFERENTIAL RESPONSE OF HER2-POSITIVE BREAST CANCER TO ANTI-HER2 THERAPY BASED ON HER2 PROTEIN EXPRESSION LEVEL Article Open access 22
September 2023 PROGNOSTIC SIGNIFICANCE OF HER2-LOW STATUS IN HR-POSITIVE/HER2-NEGATIVE ADVANCED BREAST CANCER TREATED WITH CDK4/6 INHIBITORS Article Open access 17 April 2023 PROGNOSTIC
IMPACT OF HER2-LOW POSITIVITY IN PATIENTS WITH HR-POSITIVE, HER2-NEGATIVE, NODE-POSITIVE EARLY BREAST CANCER Article Open access 11 November 2023 MAIN Epidermal growth factor receptor (EGFR,
ERBB1) expression has been widely studied in breast cancer by immunohistochemistry (IHC), and the frequency of EGFR overexpression in breast cancer is quite variable, ranging from 7 to 43%
(Wrba et al, 1988; Suo et al, 2002; Bhargava et al, 2005; DiGiovanna et al, 2005; Nieto et al, 2007; Koletsa et al, 2010; Lv et al, 2011b; Hwangbo et al, 2013). Epidermal growth factor
receptor overexpression is associated with hormone receptor negativity, large tumour size, high histologic grade and poor clinical outcome (DiGiovanna et al, 2005; Nieto et al, 2007; Koletsa
et al, 2010; Lv et al, 2011b). In particular, coexpression of human epidermal growth factor receptor 2 (HER2) and EGFR is associated with worse survival in patients with HER2-positive
breast cancer (Suo et al, 2002; DiGiovanna et al, 2005; Nieto et al, 2007). Interaction between HER2 and other ERBB coreceptors, such as EGFR and HER3 (ERBB3), has been suggested as a
possible mechanism of resistance to trastuzumab, a humanised monoclonal antibody against the extracellular portion of the HER2 protein (Franklin et al, 2004; Diermeier et al, 2005). Upon
ligand binding, ERBB receptors form homo- or heterodimers, and these phosphorylate their cytoplasmic tyrosine kinase domains and activate intracellular signalling, leading to cell division,
motility, survival and angiogenesis (Yarden and Sliwkowski, 2001). Ritter et al (2007)generated trastuzumab-resistant BT-474 cells _in vivo_; these contained higher levels of phosphorylated
EGFR and EGFR/HER2 heterodimers. Application of the EGFR tyrosine kinase inhibitors, erlotinib and gefitinib, was associated with decreased phosphorylation of HER2, suggesting that
amplification of ligand-induced activation of ERBB receptors through heterodimerisation is a plausible mechanism for resistance to trastuzumab (Ritter et al, 2007). Actually, it was shown
that trastuzumab is not able to block ligand-induced EGFR/HER2 and HER2/HER3 heterodimers (Cho et al, 2003; Franklin et al, 2004). However, the significance of EGFR overexpression for
trastuzumab response is not clear in clinical settings. Gori et al (2009) evaluated the expression of EGFR in 45 HER2-positive metastatic breast cancer patients treated with trastuzumab, and
found that EGFR overexpression was not associated with response to trastuzumab, time to progression or overall survival. In contrast, in another study, EGFR expression was associated with
decreased overall survival of HER2-positive metastatic breast cancer patients treated with trastuzumab (Gallardo et al, 2012). _Epidermal growth factor receptor_ copy number alteration, one
of the mechanisms of EGFR overexpression, is also highly variable in breast cancer, with amplification frequencies up to 24% in triple-negative breast cancer (Bhargava et al, 2005; Gumuskaya
et al, 2010; Koletsa et al, 2010; Shao et al, 2011; Lv et al, 2011a; Martin et al, 2012). Recently, we reported that high _EGFR_ copy number because of _EGFR_ amplification or high polysomy
is an independent prognostic factor for poor disease-free survival in patients with triple-negative breast cancer (Park et al, 2014). However, the prognostic significance of _EGFR_ copy
number in HER2-positive breast cancer is not clear. In this study, we assessed the clinicopathologic significance of EGFR overexpression and _EGFR_ copy number alteration in a large series
of HER2-positive primary breast cancers from two institutions. In addition, we examined the correlation of EGFR expression with trastuzumab response and clinical outcome in HER2-positive
primary and metastatic breast cancer patients treated by trastuzumab. MATERIALS AND METHODS PATIENTS AND TISSUE SAMPLES We used three different sets of breast cancer samples in this study.
First, we retrospectively examined 242 cases of HER2-positive primary breast cancers, which comprised 105 cases operated on at Seoul National University Bundang Hospital (SNUBH) between 2003
and 2009, and 137 cases operated on at Asan Medical Center (AMC) between 2003 and 2004. Expression of HER2 was scored according to the 2007 ASCO/CAP guidelines: 0, no staining; 1+, weak and
incomplete membranous staining in ⩾10% of the tumour cells; 2+, weak to moderate, complete membranous staining in ⩾10% of the tumour cells and 3+, strong, complete membranous staining in
⩾30% of the tumour cells (Wolff et al, 2007). Human epidermal growth factor receptor2 positivity was defined as a score of 3+ in the IHC test, or amplification shown by fluorescence _in
situ_ hybridisation. Baseline characteristics of the patients are summarised in Supplementary Table S1. The second set comprised 447 cases of HER2-positive primary breast cancer treated with
chemotherapy and with adjuvant trastuzumab for 1 year at Asan Medical Center from 2006 to 2011, which were used for a previous study (Lee et al, 2014a). Of the 450 cases analysed in the
previous study (Lee et al, 2014a), three cases were occult breast cancers, and thus were excluded for this study. The third set was 112 cases of HER2-positive metastatic breast cancer
treated with trastuzumab at Seoul National University Bundang Hospital, Seoul National University Hospital and Asan Medical Center from 2001 to 2011 (Lee et al, 2014b). In 80 of the 112
metastatic cases, the tissue originated from the primary cancer site, and in the remaining cases it was obtained from metastatic sites. Trastuzumab was administered as the first-line
treatment for metastatic breast cancer in 99 patients (88.4%). Taxane was used most frequently used in trastuzumab-based combination chemotherapy (in 107 patients, 95.5%), and other agents
(i.e. gemcitabine, capecitabine, and vinorelbine) were combined with trastuzumab in three cases. All cases were independently reviewed by two breast pathologists (SYP and HJL), and the
following histopathologic variables were examined: histologic subtype, T stage, nodal status, Nottingham combined histologic grade and lymphvascular invasion. This study was approved by the
institutional review boards of Seoul National University Bundang Hospital, Asan Medical Center and Seoul National University Hospital; the requirement for informed consent was waived. TISSUE
MICROARRAY CONSTRUCTION For the first set, tissue microarrays (TMAs) were constructed from archival formalin-fixed, paraffin-embedded tissue blocks. Briefly, a representative tumour area
was carefully selected for each tumour from haematoxylin- and eosin-stained sections. The designated zone of each donor block was punched with a 2 mm diameter tissue cylinder, and the sample
was transferred to a recipient block. Each sample was arrayed in triplicate. For the second set, we used TMAs that were constructed in a previous study (Lee et al, 2014a). For metastatic
cases, TMAs were constructed from archival formalin-fixed, paraffin-embedded tissue blocks from resected cases. After selection of a representative tumour area, each donor block was punched
with a tissue cylinder 4 mm in diameter. For biopsy specimen, whole sections were evaluated. IMMUNOHISTOCHEMISTRY AND SCORING Formalin-fixed and paraffin-embedded tissue sections were cut,
dried, dewaxed in xylene and rehydrated through graded alcohol. Epidermal growth factor receptor expression was detected using EGFR pharmDx Kit (Dako, Capinteria, CA, USA) and scored as
follows: 0, no staining, or weak membranous staining in <10% of the tumour cells; 1+, weak membranous staining in ⩾10% of the tumour cells; 2+, moderate, membranous staining in ⩾10% of
the tumour cells; 3+, strong membranous staining in ⩾10% of the tumour cells. Both complete and incomplete membranous staining was accepted. If the triplicate TMA cores yielded different
scores, the highest score was used. In the first set, we classified tumours into two groups according to the staining pattern of EGFR: homogeneous expression (0/1+ or 2+/3+ in all TMA cores)
and heterogeneous expression (mixed pattern of 0/1+ and 2+/3+ in three TMA cores). Oestrogen and progesterone receptors were regarded as positive if there were at least 1% positive tumour
nuclei (Hammond et al, 2010). Cases with 10% or more positive staining were grouped as positive for p53. The Ki-67 proliferation index was defined as the percentage of tumour cells showing
nuclear positivity. FLUORESCENCE _IN SITU_ HYBRIDISATION ASSAYS FOR EGFR Fluorescence _in situ_ hybridisation (FISH) was performed on the TMA samples with commercially available
locus-specific and chromosome enumeration probes (CEPs) (LSI EGFR SpectrumOrange probe (7p12) and CEP 7 SpectrumGreen probe (7p11.1–q11.1); Abbott Molecular, Des Plaines, IL, USA). Briefly,
4-_μ_m deparaffinised TMA sections were incubated in the pretreatment solution (Abbott Molecular) at 80 °C for 30 min, and then in the protease solution (Abbott Molecular) for 25 min at 37
°C. Probes were diluted in tDen-Hyb-2 hybridisation buffer (InSitus Biotechnologies, Albuquerque, NM, USA). Codenaturation of the probes and DNA was achieved by incubating at 75 °C for 5 min
in a HYBrite hybridisation chamber (Abbott Molecular) followed by 16-h hybridisation at 37 °C. Posthybridisation washes were performed according to the supplier protocols. Slides were
mounted in 4′,6-diamidino-2-phenylindole/antifade and viewed with a fluorescence microscope. A total of 50 tumour cells were evaluated for each core, and the genetic variables evaluated
were: _EGFR_ gene copy number, chromosome 7 copy number and average _EGFR_ gene : chromosome 7 ratio. The University of Colorado Cancer Center criteria for the _EGFR_ gene were used as
follows: disomy (⩽2 copies in ⩾90% of cells), low trisomy (⩽2 copies in ⩾40% of cells, 3 copies in 10–40% of cells, ⩾4 copies in <10% of cells), high trisomy (⩽2 copies in ⩾40% of cells,
3 copies in ⩾40% of cells, ⩾4 copies in <10% of cells), low polysomy (⩾4 copies in 10–40% of cells) and high polysomy (⩾4 copies in ⩾40% of cells). For gene amplification, the criterion
was a ratio of _EGFR_ to chromosome 7 of ⩾2 or ⩾15 copies of _EGFR_ per cell in ⩾10% of the analysed cells (Cappuzzo et al, 2005). For further analysis, the patients were divided into two
groups according to _EGFR_ copy number as follows: low _EGFR_ copy number (disomy, low trisomy, high trisomy and low polysomy) and high _EGFR_ copy number (high polysomy and gene
amplification). ASSESSMENT OF TRASTUZUMAB RESPONSE FOR HER2-POSITIVE METASTATIC BREAST CANCER Responses to trastuzumab-based therapy were evaluated every 8 to 12 weeks using Response
Evaluation Criteria in Solid Tumours criteria version 1.1 (Eisenhauer et al, 2009). Clinical benefit was defined as complete response, partial response or stable disease over at least 6
months. Time to progression was defined as the time from the initiation of trastuzumab treatment to disease progression, and overall survival as the time from initiation of trastuzumab
treatment to patient’s death from any cause. STATISTICAL ANALYSIS Statistical significance was assessed using Statistical Package, SPSS version 18.0 for Windows (SPSS, Chicago, IL, USA). The
correlation between EGFR overexpression and _EGFR_ gene : chromosome 7 ratio was determined by Spearman’s correlation analysis. The associations of EGFR overexpression or high _EGFR_ copy
number with clinicopathological characteristics of the tumours were analysed using Student’s _t_-test, Fisher’s exact test or the _χ_2 test, depending on the test conditions. Survival curves
were estimated using the Kaplan–Meier method, and the significance of differences between survival curves was determined using the log-rank test. Covariates that were statistically
significant in the univariate analysis were included in a multivariate analysis using a Cox proportional hazards regression model, and the hazard ratio (HR) and its 95% confidence interval
(CI) were assessed for each factor. _P_-values of <0.05 were considered statistically significant. All _P_-values reported are two-sided. RESULTS CORRELATION BETWEEN EGFR OVEREXPRESSION
AND EGFR COPY NUMBER ALTERATION IN HER2-POSITIVE PRIMARY BREAST CANCER The EGFR IHC and FISH results obtained in the 242 HER2-positive primary breast cancers are shown in Supplementary Table
S2. Epidermal growth factor receptor protein expression was not evaluable in 5 of the 242 cases, because of the loss of TMA cores. Of the other 237 cases, 149 (62.9%) did not express EGFR
protein (IHC score 0); 30 (12.7%) were scored as 1+, another 30 (12.7%) as 2+ and 28 cases (11.8%) were scored as 3+. Of the 58 cases with EGFR expression of 2+ or more, 27 showed
heterogeneous EGFR expression in three TMA cores. _Epidermal growth factor receptor_ copy number data were available in 232 cases; they were unavailable in the remainder because of the
detachment of TMA cores or inadequate hybridisation. _Epidermal growth factor receptor_ gene amplification and high polysomy were detected in 11 (4.7%) and 13 cases (5.6%), respectively. The
distribution of EGFR IHC scores and _EGFR_ copy number did not differ between the two institutions (_χ_2 square test, _P_=0.723, _P_=0.664, respectively). We compared the results of EGFR
IHC and _EGFR_ FISH in each TMA core. Epidermal growth factor receptor protein overexpression of 3+ and 2+ or more was associated with high _EGFR_ copy number (all _P_<0.001; Table 1). In
a correlation analysis, a weak but significant positive correlation was found between EGFR IHC score and _EGFR_: chromosome 7 ratio (_ρ_=0.282, _P_<0.001). Of the 11 cases in which
_EGFR_ was amplified, five scored as 3+, three as 2+ and three as 0 (Supplementary Table S3 and Figure 1). CLINICOPATHOLOGIC CHARACTERISTICS OF CASES WITH EGFR OVEREXPRESSION OR HIGH EGFR
COPY NUMBER We examined the clinicopathologic significance of EGFR overexpression by two different criteria (3+; 2+ or more), because there is no standardised cutoff point for EGFR
overexpression in breast cancer. Clinicopathologic characteristics of tumours showing EGFR overexpression or high _EGFR_ copy number are summarised in Table 2. Epidermal growth factor
receptor overexpression was significantly correlated with hormone receptor negativity, and higher Ki-67 proliferation index, irrespective of the criteria for positivity (EGFR overexpression
of 3+, _P_<0.001, _P_=0.014, respectively; EGFR overexpression of 2+ or more, _P_<0.001, _P_=0.003, respectively). High _EGFR_ copy number tended to be associated with hormone receptor
negativity, but this did not reach statistical significance (_P_=0.078). PROGNOSTIC SIGNIFICANCE OF EGFR OVEREXPRESSION AND EGFR COPY NUMBER INCREASE At the time of the analysis, the median
follow-up for patients was 98.3 months (range, 1–152 months). There were 41 (16.9%) recurrences and 1 (0.4%) cancer-related death as the first event. In survival analysis, EGFR
overexpression defined as 3+ (_P_<0.001; Figure 2A) and 2+ or more (_P_=0.025; Figure 2B) was associated with worse disease-free survival of the patients. However, high _EGFR_ copy number
was not associated with disease-free survival (_P_=0.521; Figure 2C). The overall survival of the patients was not affected by EGFR expression or _EGFR_ copy number alteration (Figures
2D–F). Pattern of EGFR overexpression (heterogeneous or homogeneous) was not associated with survival of the patients, either. Besides EGFR overexpression (3+, 2+ or more), high T stage and
lymph node metastasis were associated with poor disease-free survival (Table 3). In multivariate analysis including EGFR overexpression (3+), high T stage and lymph node metastasis, both
lymph node metastasis (pN0 _vs_ pN1∼pN3; HR, 5.611; 95% CI, 2.262–13.357; _P_<0.001) and EGFR overexpression (0∼2+ _vs_ 3+; HR, 3.013; 95% CI, 1.481–6.131; _P_=0.002) were found to be
independent prognostic factors of poor disease-free survival. However, in multivariate analysis including EGFR overexpression (2+ or more), only lymph node metastasis (HR, 5.465; 95% CI,
2.235–13.363; _P_<0.001) was an independent prognostic indicator of poor disease-free survival. In the subgroup analyses, according to adjuvant trastuzumab therapy, EGFR overexpression of
3+ was also an independent prognostic factor of poor disease-free survival (0∼2+ _vs_ 3+; HR, 3.065; 95% CI, 1.480–6.348; _P_=0.003) in the subgroup not receiving adjuvant trastuzumab
(_n_=209), but EGFR overexpression of 2+ or more and high _EGFR_ copy number were not associated with disease-free survival. However, as only a small number of patients (_n_=33) were treated
with adjuvant trastuzumab, we could not determine the prognostic and predictive significance of EGFR overexpression in this subgroup. Therefore, we additionally analysed EGFR overexpression
in an independent set of HER2-positive primary breast cancer patients treated with adjuvant trastuzumab. CORRELATION BETWEEN EGFR OVEREXPRESSION AND CLINICAL OUTCOME IN HER2-POSITIVE
PRIMARY BREAST CANCER PATIENTS TREATED WITH ADJUVANT TRASTUZUMAB In the second set, which comprised 447 HER2-positive primary breast cancer patients treated with adjuvant trastuzumab for 1
year, EGFR expression data were not available in five cases, because of the loss of TMA cores. Of the other 442 cases, 42 (9.5%) were scored as 1+, 46 (10.4%) as 2+ and 30 cases (6.8%) as
3+. Epidermal growth factor receptor overexpression also showed correlations with high histologic grade, hormone receptor negativity and high Ki-67 labelling index (Supplementary Table S4).
At the time of the analysis, the median follow-up for patients was 49 months (range, 18–104 months). There were 34 (7.6%) recurrences. In survival analysis, the patients with
EGFR-overexpressing tumour had shorter disease-free survival time than those without it (3+, _P_=0.013; 2+ or more, _P_=0.002; Figures 3A and B). The overall survival of the patients was
also significantly affected by EGFR overexpression, showing poor overall survival (3+, _P_<0.001; 2+ or more, _P_<0.001; Figures 3C and D). In this second set, high T stage, lymph node
metastasis and lymphovascular invasion were also associated with poor disease-free or overall survival of the patients (Supplementary Table S5). In multivariate analysis, both EGFR
overexpression (3+; 2+ or more) and lymphovascular invasion were found to be independent prognostic factors for poor disease-free and overall survival of the patients (Table 4). CORRELATION
BETWEEN EGFR OVEREXPRESSION AND RESPONSE TO TRASTUZUMAB-BASED THERAPY IN HER2-POSITIVE METASTATIC BREAST CANCER IN THE THIRD SET We also examined EGFR overexpression in HER2-positive
metastatic breast cancers treated with trastuzumab-based chemotherapy to evaluate the predictive significance of EGFR overexpression, as well as its prognostic significance. Epidermal growth
factor receptor IHC failed in 5 of the 112 cases. Epidermal growth factor receptor expression was 2+ in 7.5% (8 out of 107) and 3+ in 17.8% (19 out of 107). Epidermal growth factor receptor
overexpression was also associated with hormone receptor negativity (3+, _P_=0.002; 2+ or more, _P_<0.001; Supplementary Table S6). The clinical benefit of trastuzumab treatment did not
differ between the cases in which there was EGFR overexpression and those in which there was not (3+, 84.2% _vs_ 84.3%, _P_=1.000; 2+ or more, 84.6% _vs_ 84.2%, _P_=1.000). Epidermal growth
factor receptor overexpression was not associated with progression-free survival irrespective of the criterion used (3+; HR, 1.177; 95% CI, 0.682–2.033; _P_=0.559 (Figure 4A); 2+ or more,
HR, 1.140; 95% CI, 0.694–1.870; _P_=0.605 (Figure 4B)). Although EGFR overexpression tended to be associated with decreased overall survival, the effect did not reach statistical
significance (3+; HR, 1.733; 95% CI, 0.925–3.244; _P_=0.086 (Figure 4C); 2+ or more; HR, 1.561; 95% CI, 0.870–2.800; _P_=0.135 (Figure 4D)). DISCUSSION Epidermal growth factor receptor
alterations occur at an advanced stage of malignancy characterised by metastatic competence, and EGFR is thought to promote cancer cell migration and invasion (Masuda et al, 2012). In the
present study, we have shown that EGFR overexpression is a poor prognostic factor in two independent sets of HER2-positive primary breast cancer patients. In the first set, patients with
EGFR-overexpressing tumours showed poor disease-free survival, and the poor prognostic impact of EGFR overexpression (3+) was also applied to the subgroup not receiving trastuzumab.
Furthermore, in the second set, EGFR overexpression was associated with poor disease-free and overall survival of the patients treated with adjuvant trastuzumab. Therefore, these results
indicate that EGFR overexpression has a prognostic and predictive value in HER2-positive primary breast cancers. However, there are, at present, no validated scoring systems for EGFR protein
overexpression assessed by IHC. We therefore analysed the clinical significance of EGFR overexpression using two different cutoff values (3+, 2+ or more). We found that prognostic impact of
EGFR overexpression using two different criteria was similar, although not same. Previous studies that showed prognostic significance of EGFR overexpression in HER2-positive breast cancer
also used different cutoff values for EGFR expression (Suo et al, 2002; DiGiovanna et al, 2005; Nieto et al, 2007), and even any membranous staining was found to be significant (DiGiovanna
et al, 2005). However, they used different clones and different staining methods for EGFR IHC. In this study, we used EGFR pharmDx Kit, which is approved for the identification of colorectal
cancer patients eligible for treatment with cetuximab or panitumumab, but any membranous staining or expression of 1+ or more had no prognostic impact on the survival of the patients. Thus,
most reliable cutoff values for EGFR overexpression and its standard staining method should be determined to predict the prognosis of HER2-positive breast cancer patients, but, in the
study, EGFR overexpression of 3+ was found to be most predictive. We also revealed that the clinicopathologic characteristics of EGFR-overexpressing HER2-positive breast cancers were same
irrespective of the cutoff value used. Especially, the association of EGFR overexpression with hormone receptor negativity was clearly demonstrated in the three different sets of
HER2-positive breast cancers. Recently, the Cancer Genome Atlas (TCGA) Network analysed primary breast cancers with various platforms, and showed that there were at least two types of
clinically HER2-positive breast cancers (Cancer Genome Atlas N, 2012). One corresponded to the subtype which is enriched for _HER2_ mRNA and in which there is high expression of receptor
tyrosine kinases including _EGFR_, _HER2_ and _FGFR4_. The other is the luminal mRNA subtype with high expression of _GATA3_, _BCL2_ and _ESR1_, the so-called luminal cluster of genes. In
our study, the correlation between EGFR overexpression and hormone receptor negativity is consistent with the results of TCGA analysis, and suggests that HER2-positive breast cancers have
different features depending on their hormone receptor status. The relationship between EGFR overexpression and high _EGFR_ copy number has not been clearly defined. In this study, analysis
of TMA cores revealed a positive correlation between _EGFR_ gene status and EGFR protein overexpression. In particular, cases with high levels of _EGFR_ amplification (case nos. 196 and 224;
Supplementary Table S3) yielded 3+ EGFR IHC results in all three cores evaluated. However, our data also revealed that EGFR overexpression had low specificity and low positive predictive
value for high _EGFR_ copy number as not all tumours with high _EGFR_ copy number overexpressed EGFR and not all EGFR-overexpressing tumours had high _EGFR_ copy numbers. Furthermore, while
EGFR overexpression was associated with poor prognosis in HER2-positive primary breast cancer patients, high _EGFR_ copy number did not have prognostic significance. These results suggest
that besides increased _EGFR_ gene copy number, EGFR overexpression may be induced by other mechanisms, such as mutation, aberrant transcription or translational modification. Epidermal
growth factor receptor overexpression did not affect the response to trastuzumab and progression-free survival in patients with HER2-positive metastatic breast cancers, although EGFR
overexpression tended to be associated with poor overall survival. In this study, predictive value of EGFR overexpression for trastuzumab response in metastatic setting seems to be different
from that in an adjuvant setting. However, the third metastatic breast cancer set has some limitations; not all patients were treated by the same protocol, and most of the tissues used for
analysis were primary tumours even though there were possibilities of change in EGFR status during tumour progression. In HER2-positive primary breast cancer, EGFR overexpression was
associated with worse survival in the patients not treated with trastuzumab, as well as those treated with adjuvant trastuzuamb. Therefore, the association of EGFR overexpression with poor
clinical outcome in patients receiving adjuvant trastuzuamb cannot be solely explained by resistance to trastuzumab caused by EGFR overexpression. The utility of EGFR overexpression as a
predictive biomarker for traststuzumab response should be validated carefully in further large-scale studies. In the light of the known interplay between EGFR and HER2, EGFR inhibitor
gefitinib has been tried in combination with trastuzuamb to treat patients with HER2-positive metastatic breast cancer (Arteaga et al, 2008; Somlo et al, 2012). However, this combination of
gefitinib and trastuzumab was not found to be more effective than traststuzumab alone. Moreover, EGFR expression was not associated with response to treatment. However, in those studies, the
number of enrolled patients was too small to permit meaningful statistical analysis. Hence, the utility of EGFR overexpression as a predictive biomarker for EGFR-targeted therapy and as a
prognostic factor for HER2-positive metastatic breast cancer should be validated in large studies. There are treatments that may improve the clinical outcome of patients with HER2-positive
breast cancers overexpressing EGFR. A dual EGFR/HER2 tyrosine kinase inhibitor, lapatinib, has been shown to be useful after previous treatments such as trastuzumab in cases with progressive
metastatic HER2-positive disease (Geyer et al, 2006). Fabi et al (2013) evaluated _EGFR_ gene copy number and responses to lapatinib, and identified increased _EGFR_ copy number as a
positive predictive factor for lapatinib response. In addition to lapatinib, other small-molecule inhibitors targeting ERBB family members, such as afatinib and neratinib, are being
investigated in ongoing trials in the context of trastuzumab-resistant metastatic HER2-positive breast cancer (Gradishar, 2013). In conclusion, our study showed that EGFR protein
overexpression is an independent poor prognostic factor in HER2-positive primary breast cancer, and it is also a predictive factor for trastuzumab response in HER2-positive primary breast
cancer, but not in metastatic breast cancer. CHANGE HISTORY * _ 06 JANUARY 2015 This paper was modified 12 months after initial publication to switch to Creative Commons licence terms, as
noted at publication _ REFERENCES * Arteaga CL, O’Neill A, Moulder SL, Pins M, Sparano JA, Sledge GW, Davidson NE (2008) A phase I–II study of combined blockade of the ErbB receptor network
with trastuzumab and gefitinib in patients with HER2 (ErbB2)-overexpressing metastatic breast cancer. _Clin Cancer Res_ 14: 6277–6283. Article CAS Google Scholar * Bhargava R, Gerald WL,
Li AR, Pan Q, Lal P, Ladanyi M, Chen B (2005) EGFR gene amplification in breast cancer: correlation with epidermal growth factor receptor mRNA and protein expression and HER-2 status and
absence of EGFR-activating mutations. _Mod Pathol_ 18: 1027–1033. Article CAS Google Scholar * Cancer Genome Atlas N (2012) Comprehensive molecular portraits of human breast tumours.
_Nature_ 490: 61–70. Article Google Scholar * Cappuzzo F, Hirsch FR, Rossi E, Bartolini S, Ceresoli GL, Bemis L, Haney J, Witta S, Danenberg K, Domenichini I, Ludovini V, Magrini E,
Gregorc V, Doglioni C, Sidoni A, Tonato M, Franklin WA, Crino L, Bunn PA Jr, Varella-Garcia M (2005) Epidermal growth factor receptor gene and protein and gefitinib sensitivity in
non-small-cell lung cancer. _J Natl Cancer Inst_ 97: 643–655. Article CAS Google Scholar * Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW Jr, Leahy DJ (2003) Structure of
the extracellular region of HER2 alone and in complex with the Herceptin Fab. _Nature_ 421: 756–760. Article CAS Google Scholar * Diermeier S, Horvath G, Knuechel-Clarke R, Hofstaedter F,
Szollosi J, Brockhoff G (2005) Epidermal growth factor receptor coexpression modulates susceptibility to Herceptin in HER2/neu overexpressing breast cancer cells via specific erbB-receptor
interaction and activation. _Exp Cell Res_ 304: 604–619. Article CAS Google Scholar * DiGiovanna MP, Stern DF, Edgerton SM, Whalen SG, Moore D 2nd, Thor AD (2005) Relationship of
epidermal growth factor receptor expression to ErbB-2 signaling activity and prognosis in breast cancer patients. _J Clin Oncol_ 23: 1152–1160. Article CAS Google Scholar * Eisenhauer EA,
Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, Rubinstein L, Shankar L, Dodd L, Kaplan R, Lacombe D, Verweij J (2009) New response
evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). _Eur J Cancer_ 45: 228–247. Article CAS Google Scholar * Fabi A, Merola R, Ferretti G, Di Benedetto A,
Antoniani B, Ercolani C, Nistico C, Papaldo P, Ciccarese M, Sperduti I, Vici P, Marino M, Gori S, Botti C, Malaguti P, Cognetti F, Mottolese M (2013) Epidermal growth factor receptor gene
copy number may predict lapatinib sensitivity in HER2-positive metastatic breast cancer. _Expert Opin Pharmacother_ 14: 699–706. Article CAS Google Scholar * Franklin MC, Carey KD, Vajdos
FF, Leahy DJ, de Vos AM, Sliwkowski MX (2004) Insights into ErbB signaling from the structure of the ErbB2–pertuzumab complex. _Cancer Cell_ 5: 317–328. Article CAS Google Scholar *
Gallardo A, Lerma E, Escuin D, Tibau A, Munoz J, Ojeda B, Barnadas A, Adrover E, Sanchez-Tejada L, Giner D, Ortiz-Martinez F, Peiro G (2012) Increased signalling of EGFR and IGF1R, and
deregulation of PTEN/PI3K/Akt pathway are related with trastuzumab resistance in HER2 breast carcinomas. _Br J Cancer_ 106: 1367–1373. Article CAS Google Scholar * Geyer CE, Forster J,
Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, Skarlos D, Campone M, Davidson N, Berger M, Oliva C, Rubin SD, Stein S, Cameron D (2006)
Lapatinib plus capecitabine for HER2-positive advanced breast cancer. _N Engl J Med_ 355: 2733–2743. Article CAS Google Scholar * Gori S, Sidoni A, Colozza M, Ferri I, Mameli MG,
Fenocchio D, Stocchi L, Foglietta J, Ludovini V, Minenza E, De Angelis V, Crino L (2009) EGFR, pMAPK, pAkt and PTEN status by immunohistochemistry: correlation with clinical outcome in
HER2-positive metastatic breast cancer patients treated with trastuzumab. _Ann Oncol_ 20: 648–654. Article CAS Google Scholar * Gradishar WJ (2013) Emerging approaches for treating
HER2-positive metastatic breast cancer beyond trastuzumab. _Ann Oncol_ 24: 2492–2500. Article CAS Google Scholar * Gumuskaya B, Alper M, Hucumenoglu S, Altundag K, Uner A, Guler G (2010)
EGFR expression and gene copy number in triple-negative breast carcinoma. _Cancer Genet Cytogenet_ 203: 222–229. Article CAS Google Scholar * Hammond ME, Hayes DF, Wolff AC, Mangu PB,
Temin S (2010) American society of clinical oncology/college of american pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in
breast cancer. _J Oncol Pract_ 6: 195–197. Article Google Scholar * Hwangbo W, Lee JH, Ahn S, Kim S, Park KH, Kim CH, Kim I (2013) EGFR gene amplification and protein expression in
invasive ductal carcinoma of the breast. _Korean J Pathol_ 47: 107–115. Article Google Scholar * Koletsa T, Kotoula V, Karayannopoulou G, Nenopoulou E, Karkavelas G, Papadimitriou CS,
Kostopoulos I (2010) EGFR expression and activation are common in HER2 positive and triple-negative breast tumours. _Histol Histopathol_ 25: 1171–1179. CAS PubMed Google Scholar * Lee HJ,
Park IA, Park SY, Seo AN, Lim B, Chai Y, Song IH, Kim NE, Kim JY, Yu JH, Ahn JH, Gong G (2014a) Two histopathologically different diseases: hormone receptor-positive and hormone
receptor-negative tumors in HER2-positive breast cancer. _Breast Cancer Res Treat_ 145: 615–623. Article CAS Google Scholar * Lee HJ, Seo AN, Kim EJ, Jang MH, Suh KJ, Ryu HS, Kim YJ, Kim
JH, Im S, Gong G, Jung KH, Park IA, Park SY (2014b) HER2 heterogeneity affects trastuzumab responses and survival in patients with HER2-positive metastatic breast cancer. _Am J Clin Pathol_
in press. * Lv N, Xie X, Ge Q, Lin S, Wang X, Kong Y, Shi H, Wei W (2011a) Epidermal growth factor receptor in breast carcinoma: association between gene copy number and mutations. _Diagn
Pathol_ 6: 118. Article CAS Google Scholar * Lv N, Xie X, Ge Q, Lin S, Wang X, Kong Y, Shi H, Xie X, Wei W (2011b) Epidermal growth factor receptor in breast carcinoma: association
between gene copy number and mutations. _Diagn Pathol_ 6: 118. Article CAS Google Scholar * Martin V, Botta F, Zanellato E, Molinari F, Crippa S, Mazzucchelli L, Frattini M (2012)
Molecular characterization of EGFR and EGFR-downstream pathways in triple negative breast carcinomas with basal like features. _Histol Histopathol_ 27: 785–792. Article CAS Google Scholar
* Masuda H, Zhang D, Bartholomeusz C, Doihara H, Hortobagyi GN, Ueno NT (2012) Role of epidermal growth factor receptor in breast cancer. _Breast Cancer Res Treat_ 136: 331–345. Article
CAS Google Scholar * Nieto Y, Nawaz F, Jones RB, Shpall EJ, Nawaz S (2007) Prognostic significance of overexpression and phosphorylation of epidermal growth factor receptor (EGFR) and the
presence of truncated EGFRvIII in locoregionally advanced breast cancer. _J Clin Oncol_ 25: 4405–4413. Article Google Scholar * Park HS, Jang MH, Kim EJ, Kim HJ, Lee HJ, Kim YJ, Kim JH,
Kang E, Kim SW, Kim IA, Park SY (2014) High EGFR gene copy number predicts poor outcome in triple-negative breast cancer. _Mod Pathol_ 27: 1212–1222. Article CAS Google Scholar * Ritter
CA, Perez-Torres M, Rinehart C, Guix M, Dugger T, Engelman JA, Arteaga CL (2007) Human breast cancer cells selected for resistance to trastuzumab _in vivo_ overexpress epidermal growth
factor receptor and ErbB ligands and remain dependent on the ErbB receptor network. _Clin Cancer Res_ 13: 4909–4919. Article CAS Google Scholar * Shao MM, Zhang F, Meng G, Wang XX, Xu H,
Yu XW, Chen LY, Tse GM (2011) Epidermal growth factor receptor gene amplification and protein overexpression in basal-like carcinoma of the breast. _Histopathology_ 59: 264–273. Article
Google Scholar * Somlo G, Martel CL, Lau SK, Frankel P, Ruel C, Gu L, Hurria A, Chung C, Luu T, Morgan R Jr., Leong L, Koczywas M, McNamara M, Russell CA, Kane SE (2012) A phase I/II
prospective, single arm trial of gefitinib, trastuzumab, and docetaxel in patients with stage IV HER-2 positive metastatic breast cancer. _Breast Cancer Res Treat_ 131: 899–906. Article CAS
Google Scholar * Suo Z, Risberg B, Kalsson MG, Willman K, Tierens A, Skovlund E, Nesland JM (2002) EGFR family expression in breast carcinomas. c-erbB-2 and c-erbB-4 receptors have
different effects on survival. _J Pathol_ 196: 17–25. Article CAS Google Scholar * Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM,
Langer A, McShane LM, Paik S, Pegram MD, Perez EA, Press MF, Rhodes A, Sturgeon C, Taube SE, Tubbs R, Vance GH, van de Vijver M, Wheeler TM, Hayes DF (2007) American Society of Clinical
Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. _J Clin Oncol_ 25: 118–145. Article CAS Google
Scholar * Wrba F, Reiner A, Ritzinger E, Holzner JH, Reiner G (1988) Expression of epidermal growth factor receptors (EGFR) on breast carcinomas in relation to growth fractions, estrogen
receptor status and morphological criteria. An immunohistochemical study. _Pathol Res Pract_ 183: 25–29. Article CAS Google Scholar * Yarden Y, Sliwkowski MX (2001) Untangling the ErbB
signalling network. _Nat Rev Mol Cell Biol_ 2: 127–137. Article CAS Google Scholar Download references ACKNOWLEDGEMENTS This research was supported by the Basic Science Research Program
through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (Grant No. NRF-2012R1A1B4000557) and by Seoul National University Bundang
Hospital Research Fund (Grant No. 11-2012-007). AUTHOR INFORMATION Author notes * H J Lee and A N Seo: These authors contributed equally to this work. AUTHORS AND AFFILIATIONS * Department
of Pathology, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam-si, Gyeonggi, 463-707, Korea H J Lee, A N Seo, E J Kim, M H Jang & S Y Park * Department of
Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-746, Korea H J Lee & G Gong * Department of Medical Oncology, Seoul
National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam-si, Gyeonggi, 463-707, Korea Y J Kim & J H Kim * Department of Medical Oncology, Seoul National University
College of Medicine, 28 Yeongon-dong, Jongno-gu, Seoul, 110-799, Korea Y J Kim, J H Kim & S-A Im * Department of Surgery, Seoul National University Bundang Hospital, 300 Gumi-dong,
Bundang-gu, Seongnam-si, Gyeonggi, 463-707, Korea S-W Kim * Department of Surgery, Seoul National University College of Medicine, 28 Yeongon-dong, Jongno-gu, Seoul, 110-799, Korea S-W Kim *
Department of Pathology, Seoul National University College of Medicine, 28 Yeongon-dong, Jongno-gu, Seoul, 110-799, Korea H S Ryu, I A Park & S Y Park * Department of Medical Oncology,
Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-746, Korea K H Jung * Department of Surgery, Asan Medical Center, University of
Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 138-746, Korea H J Kim Authors * H J Lee View author publications You can also search for this author inPubMed Google
Scholar * A N Seo View author publications You can also search for this author inPubMed Google Scholar * E J Kim View author publications You can also search for this author inPubMed Google
Scholar * M H Jang View author publications You can also search for this author inPubMed Google Scholar * Y J Kim View author publications You can also search for this author inPubMed Google
Scholar * J H Kim View author publications You can also search for this author inPubMed Google Scholar * S-W Kim View author publications You can also search for this author inPubMed Google
Scholar * H S Ryu View author publications You can also search for this author inPubMed Google Scholar * I A Park View author publications You can also search for this author inPubMed
Google Scholar * S-A Im View author publications You can also search for this author inPubMed Google Scholar * G Gong View author publications You can also search for this author inPubMed
Google Scholar * K H Jung View author publications You can also search for this author inPubMed Google Scholar * H J Kim View author publications You can also search for this author inPubMed
Google Scholar * S Y Park View author publications You can also search for this author inPubMed Google Scholar CORRESPONDING AUTHOR Correspondence to S Y Park. ADDITIONAL INFORMATION This
work is published under the standard license to publish agreement. After 12 months the work will become freely available and the license terms will switch to a Creative Commons
Attribution-NonCommercial-Share Alike 3.0 Unported License. Supplementary Information accompanies this paper on British Journal of Cancer website SUPPLEMENTARY INFORMATION SUPPLEMENTARY
TABLES (DOC 184 KB) RIGHTS AND PERMISSIONS From twelve months after its original publication, this work is licensed under the Creative Commons Attribution-NonCommercial-Share Alike 3.0
Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-sa/3.0/ Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Lee, H., Seo, A., Kim,
E. _et al._ Prognostic and predictive values of EGFR overexpression and _EGFR_ copy number alteration in HER2-positive breast cancer. _Br J Cancer_ 112, 103–111 (2015).
https://doi.org/10.1038/bjc.2014.556 Download citation * Revised: 23 August 2014 * Accepted: 02 October 2014 * Published: 28 October 2014 * Issue Date: 06 January 2015 * DOI:
https://doi.org/10.1038/bjc.2014.556 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not
currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative KEYWORDS * breast carcinoma * gene amplification * EGFR * HER2 *
trastuzumab * immunohistochemistry