Original Article Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Apr 14, 2013; 19(14): 2171-2178
Published online Apr 14, 2013. doi: 10.3748/wjg.v19.i14.2171
Correlation of human epidermal growth factor receptor 2 expression with clinicopathological characteristics and prognosis in gastric cancer
Chao He, Xing-Zhi Ni, Dan-Ping Shen, Hua Liu, Zhi-Yong Shen, Department of General Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
Xue-Yi Bian, Department of General Surgery, Suzhou Jiulong Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215021, Jiangsu Province, China
Yan-Ying Shen, Qiang Liu, Department of Pathology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
Author contributions: He C, Bian XY and Ni XZ designed the research; He C, Bian XY, Shen DP, Shen YY, Liu H, Shen ZY and Liu Q performed the research; He C, Bian XY, Shen DP and Ni XZ analyzed the data; He C, Bian XY and Ni XZ wrote the paper.
Correspondence to: Xing-Zhi Ni, MD, Professor of Medicine, Department of General Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, 1630 Dongfang, Pudong district, Shanghai 200127, China. niyin@yahoo.com
Telephone: +86-21-68383731 Fax: +86-21-58394262
Received: August 26, 2012
Revised: January 9, 2013
Accepted: February 8, 2013
Published online: April 14, 2013

Abstract

AIM: To investigate human epidermal growth factor receptor 2 (HER2) gene amplification and protein expression in Chinese patients with resectable gastric cancer and the association with clinicopathological characteristics and survival.

METHODS: One hundred and ninety-seven gastric cancer patients who underwent curative surgery procedures were enrolled into this study. HER2 gene amplification and protein expression were examined using fluorescence in-situ hybridization (FISH) and immunohistochemistry (IHC) analysis on formalin-fixed paraffin-embedded gastric cancer samples from all patients. For scoring, Hofmann’s HER2 gastric cancer scoring system was adopted. All cases showing IHC3+ or FISH positivity were defined as HER2 positive. Patient clinicopathological data and survival information were collected. Finally, χ2 statistical analysis was performed to analyze the HER2 positivity rate amongst the subgroups with different clinicopathological characteristics including; gender, age, tumor location, Lauren classification, differentiation, TNM staging, depth of invasion, lymph node metastases and distant metastasis. The probability of survival for different subgroups with different clinicopathological characteristics was calculated using the Kaplan-Meier method and survival curves plotted using log rank inspection.

RESULTS: According to Hofmann’s HER2 gastric cancer scoring criteria, 31 cases (15.74%) were identified as HER2 gene amplified and 19 cases (9.64%) were scored as strongly positive for HER2 membrane staining (3+), 25 cases (12.69%) were moderately positive (2+) and 153 cases (77.66%) were HER2 negative (0/1+). The concordance rate between IHC and FISH analyses was 88.83% (175/197). Thirty-six cases were defined as positive for HER2 gene amplification and/or protein expression, with 24 of these cases being eligible for Herceptin treatment according to United States recommendations, and 29 of these cases eligible according to EU recommendations. Highly consistent results were detected between IHC3+, IHC0/1 and FISH (73.68% and 95.42%), but low consistency was observed between IHC2+ and FISH (40.00%). The positivity rates in intestinal type and well-differentiated gastric cancer were higher than those in diffuse/mixed type and poorly-differentiated gastric cancer respectively (28.57% vs 13.43%, P = 0.0103; 37.25% vs 11.64%, P < 0.0001), but were not correlated with gender, age, tumor location or TNM stage, depth of invasion, lymph node metastases and distant metastasis. In poorly-differentiated gastric cancer patients, those without lymph node metastasis showed a higher HER2 positivity rate than those with lymph node metastasis (26.47% vs 7.14%, P = 0.0021). This association was not present in those patients with well-differentiated gastric cancer (28.57% vs 43.33%, P = 0.2832). Within our patient cohort, 26 cases were lost to follow-up. The median survival time for the remaining 171 patients was 18 mo. The median survival times of the HER2 positive and negative groups were 17 and 18.5 mo respectively. Overall survival was not significantly different between HER2-positive and negative groups (χ2 = 0.9157, P = 0.3386), but in patients presenting well-differentiated tumors, the overall survival of the HER2-positive group was significantly worse than that of the HER2-negative group (P = 0.0123). In contrast, patients with poorly differentiated and diffuse/mixed subtype gastric cancers showed no significant differences in overall survival associated with HER2. Furthermore, the median survival time of the HER2 positive group did not show any statistically significant differences when compared to the subgroups of gender, age, tumor location, TNM classification, lymph node metastases and distant metastasis.

CONCLUSION: Patients with intestinal type gastric cancer (GC), well-differentiated GC and poorly-differentiated GC without lymph node metastasis, may all represent suitable candidates for targeted therapy using Herceptin.

Key Words: Gastric cancer, Human epidermal growth factor receptor 2, Gene amplification, Protein expression, Clinicopathological characteristics



INTRODUCTION

Human epidermal growth factor receptor 2 (HER2) is a 185-kDa transmembrane tyrosine kinase receptor[1] and its gene amplification and protein overexpression play an important role in the proliferation, apoptosis, adhesion, angiogenesis and aggressiveness of many solid tumors[2], including; breast[3], colon[4], bladder[4], ovarian[5], uterine cervix[6], esophageal[7] and gastric cancer.

Herceptin (trastuzumab) has been approved[8] in the European Union and the United States for use in combination with 5-fluorouracil (5-FU) or capecitabine plus cisplatin for the first-line treatment of patients with HER2-positive metastatic adenocarcinoma of the stomach or gastroesophageal junction according to the results of the 2010 trastuzumab for gastric cancer (ToGA) trial. However, precise patient inclusion criteria for Herceptin treatment is still not fully defined due to the lack of a standardized HER2 scoring system for gastric cancer[9,10]. For a clinician, defining the relationships between HER2 and clinicopathological characteristics can help to select suitable candidates.

Our study aimed to investigate the relationship between HER2 gene amplification and protein overexpression in resectable gastric cancer patients and determine any correlations with relevant clinicopathological characteristics. Furthermore, we explored the influence of HER2 on disease prognosis in gastric cancer patients. Our study was conducted with a view towards the future introduction of Herceptin targeted therapy for the treatment of gastric cancer patients.

MATERIALS AND METHODS
Patients and tissue specimens

From July 2009 to January 2012, 197 gastric cancer patients who underwent curative surgery at Renji hospital, Shanghai Jiaotong University were enrolled into our study. Formalin-fixed, paraffin-embedded samples of tumors and corresponding normal stomach tissues from 197 gastric cancer patients were evaluated for HER2 protein and gene amplification using immunohistochemistry (IHC) and fluorescence in-situ hybridization (FISH) analysis. None of the patients had undergone prior preoperative radiation, chemotherapy or targeted therapy.

The study included 65 women and 132 men, with ages ranging from 22 to 88 years. The median age was 62 years. The tumor sample characteristics of all 197 cases are shown in Table 1. Of all the tumors examined, 31 (15.74%) were located in the cardiac region, 42 (21.32%) in the body, and 122 (61.93%) in the pylorus. The majority (98.98%) of the samples were primary tumors with only 2 recurrent tumors identified. According to Lauren classification, 63 (31.98%) tumors were intestinal-type and 134 (68.02%) were diffuse-type or mixed-type carcinomas. Poorly differentiated tumors (grades I and II) comprised 25.89%, whilst 74.11% of tumors were moderately differentiated (grades III and IV). TNM classification revealed that 13 cases were stage I (6.60%), 46 were stage II (23.35%), 98 were stage III (49.75%) and 40 were stage IV (20.30%). Postoperative follow-up ended in April, 2012.

Table 1 Correlation of human epidermal growth factor receptor 2 expression with clinicopathological characteristics n (%).
Clinicopathological characteristicsnHER2
χ2P value
PositiveNegative
Sex1.27360.2591
Male13227 (20.45)105 (79.55)
Female659 (13.85)56 (86.15)
Age (yr)1.30560.2532
< 608813 (14.77)75 (85.23)
≥ 6010923 (21.10)86 (78.90)
Tumor site10.04090.9798
Cardiac316 (19.35)25 (80.65)
Body428 (19.05)34 (80.96)
Pylorus12222 (18.03)100 (81.97)
Lauren classification6.57590.0103
Intestinal6318 (28.57)45 (71.43)
Diffuse/mixed13418 (13.43)116 (86.57)
Tumor differentiation16.6003< 0.0001
Well-differentiated5119 (37.25)32 (62.75)
Poorly-differentiated14617 (11.64)129 (88.36)
TNM classification0.67540.879
I132 (15.38)11 (84.62)
II467 (15.22)39 (84.78)
III9820 (20.41)78 (79.59)
IV407 (17.50)33 (82.50)
FISH detection for HER2 gene amplification

FISH was conducted with the HER2 DNA Probe Kit (InvitrogenTM by Life Technologies) according to the manufacturer’s instructions. Four-μm-thick sections were baked overnight at 56 °C, deparaffinized in three 10 min changes of xylene and then rehydrated through two 5-min changes of 100% ethanol. The slides were then reduced for 18 min in SPOT-Light tissue pretreatment solution at > 98 °C, and briefly washed in 3 × PBS at room temperature. The slides were then incubated for 16 min in enzyme reagent solution at 37 °C and washed in 3 × PBS at room temperature, dehydrated through 70%, 85%, and 100% ethanol, and allowed to air dry. After open air drying, the HER2 DNA probe kit (PathVysion HER2 DNA Probe Kit, Abbott Laboratories) which was denatured at 79 °C for 6 min, was applied onto each slide, a cover slip was added and then sealed with rubber cement. After 16 to 18 h of hybridization at 37 °C, the slides were washed with 73 °C preheated post hybridization buffer for 5 min and dehydrated through 70%, 85% and finally 100% ethanol. After air drying, the slides were counter-stained with 14 μL diamidino-phenyl-indole, cover slips applied and then slides chilled for 30 min at 4 °C. Finally, the slides were observed through a fluorescence microscope (OLYMPUS BX61).

Immunohistochemical staining

HER2 IHC analysis was performed on 4 μm thick tissue sections. Briefly, after deparaffinization and rehydration steps, the tissue samples were incubated in antigen retrieval solution at 99 °C for 40 min. Endogenous peroxidase activity was quenched by 5 min incubation with hydrogen peroxide. Sections were then incubated with HER2 antibody (HerceptestTM, DAKO) for 30 min. Both the primary and secondary antibodies against human HER2 protein were applied for 30 min at room temperature and then the immunocomplexes were visualized with diaminobenzidine for 10 min and placed under a cover slip. Finally, the slides were viewed using light microscopy (LEICA DM2500).

Results scoring

An absolute HER2 gene copy number lower than 6 or a HER2/Chr17 ratio of less than 2 was considered HER2 negative, whilst cases showing average gene copy numbers of HER2 ≥ 6 or a gene/CEN17 fluorescence ratio ≥ 2 were considered positive for gene amplification. Additionally, tight gene clustering of HER2 signals was also defined as gene amplification. The above criteria are based on Hofmann’s criteria in gastric cancer[9].

In the present study, the IHC score criteria on human gastric cancer also followed Hofmann’s criteria[9]: no staining or < 10% tumor cell positive staining as 0/negative; faintly or barely perceptible staining on > 10% tumor cell membrane as 1+/negative; weak to moderate positive staining on > 10% tumor cells as 2+/(equivocal) positive; cohesive moderate to strong staining on the membrane will be scored as 3+/positive. All cases with IHC3+ or FISH positivity were defined as HER2 positive.

Statistical analysis

χ2 statistical analysis was performed to assess the HER2 positivity rate amongst the subgroups with different clinicopathological characteristics. The probability of survival for different subgroups was calculated using the Kaplan-Meier method and the survival curves plotted using log rank inspection. All statistics were performed using 2-sided analysis, with a significance level of P < 0.05, using the “SAS9.13” statistical software package.

RESULTS
HER2 gene amplification and protein expression

The FISH and IHC analysis results for all 197 gastric cancer tissues are shown in Table 2. According to Hofmann’s HER2 FISH scoring criteria, 31 cases (15.74%) were identified as HER2 gene amplified and the other 166 cases (84.26%) were HER2 gene amplification negative (Figure 1). Of the 197 samples examined by IHC (following Hofmann’s criteria), 19 cases (9.64%) were scored as strongly positive for HER2 membrane staining (3+), 25 cases (12.69%) were moderately positive (2+), and 153 cases (77.66%) were HER2 negative (0/1+) (Figure 2).

Figure 1
Figure 1 Fluorescent in-situ hybridization analysis of human epidermal growth factor receptor 2 gene amplification (× 600). A: Normal human epidermal growth factor receptor 2 (HER2) gene expression: Red signals (HER2 gene), green signals [chromosome enumeration probe 17 (CEP17)], blue signals (nuclei lining dye); B: Positive HER2 gene amplification: HER2:CEP17 > 2; C: Positive HER2 gene amplification: HER2:CEP17 > 2 with clear red cluster signals observed.
Table 2 Immunochemistry-fluorescence in situ hybridization concordance n (%).
FISHIHC
Total
3+2+1+0
Positive14107031 (15.74)
Negative51521125166 (84.26)
Total19 (9.64)25 (12.69)28 (14.21)125 (63.45)197
Figure 2
Figure 2 Immunohistochemical analysis of human epidermal growth factor receptor 2 protein expression (× 200). A: Immunohistochemical (IHC) 0: No staining on tumor cell membrane; B: IHC1+: Faintly perceptible staining on > 10% tumor cell membrane; C: IHC2+: Moderate staining on > 10% tumor cell membrane; IHC3+: Strong staining on > 10% tumor cell membrane.

The concordance rate between IHC and FISH analyses was 88.83% (175/197). Thirty-six cases were defined as HER2 positive and 24 cases were suitable for Herceptin treatment according to the recommendations of the United States[11]. However, when applying European Union[11] recomnmendations for Herceptin usage, 29 cases were identified as eligible for Herceptin treatment. This difference underscores the requirement for standardized and more precise eligibility criteria for correct identification of patients who are eligible for HER2 targeted therapy.

Of the 31 FISH-positive cases, 14 cases (45.16%) were IHC3+ with a 100% concordance between IHC3+ and FISH, and 10 (32.26%) cases were IHC2+. None of the IHC 0 tumors demonstrated FISH amplification, and only 7 tumors in the IHC1+ group were found to be FISH positive with a ratio of 22.58%. High consistency results was detected between IHC3+, IHC0/1, and FISH scores (73.68% and 95.42%), but low consistency was observed between IHC2+ and FISH (40.00%).

Correlation of HER2 with clinicopathological characteristics

Significantly different HER2 positivity rates were observed when comparing intestinal-type gastric cancers with diffuse/mixed-type cancers (28.57% vs 13.43%, P = 0.0103), and well-differentiated cases with poorly-differentiated cases (37.25% vs 11.64%, P < 0.0001). No relationship was observed between the HER2 positivity rate and sex, age, tumor site and TNM GC classification (P > 0.05; Table 1). Furthermore, within the subgroups, no relationship was observed between HER2 positivity and depth of invasion, lymph node metastasis or distant metastasis (Table 3).

Table 3 Correlation of human epidermal growth factor recept-or 2 expression with tumor node metastasis staging n (%).
Clinicopathological characteristicsnHER2
χ2P value
PositiveNegative
T0.57820.4470
T1-T2266 (23.08)20 (76.92)
T3-T417129 (16.96)142 (84.04)
N4.62740.2012
N0558 (14.55)47 (85.45)
N18320 (24.10)63 (75.90)
N2335 (15.15)28 (84.85)
N3262 (7.69)24 (92.31)
M0.00001.0000
M018533 (17.84)152 (82.16)
M1122 (16.67)10 (83.33)

Within the poorly-differentiated gastric cancer patient group, those without lymph node metastasis showed a higher HER2 positivity rate than those with lymph node metastasis (26.47% vs 7.14%, P = 0.0021). This association was not observed in the well-differentiated gastric cancer patient group (28.57% vs 43.33%, P = 0.2832).

Survival analysis

Of our 197 gastric cancer patients, 26 cases were lost in follow-up. The median survival time for the remaining 171 patients was 18 mo (range: 0-33 mo). During the follow-up time, 60 deaths occurred (35.09%), 57 of which were disease-related. One patient died of perioperative pulmonary infection, and two cases died of heart disease and multiple organ failure, respectively.

The median survival time of the HER2 positive (29 cases) and negative groups (142 cases) was 17 mo and 18.5 mo, respectively. Nevertheless, the HER2 positive gastric cancer patients did not show statistically significant reductions in mean survival times, nor lower 1-year or 2-year survival rates. Furthermore, no statistically significant differences were observed in overall survival times between the HER2 positive and negative groups (χ2 = 0.9157, P = 0.3386; Figure 3A).

Figure 3
Figure 3 Kaplan-Meier survival analysis. A: Overall survival curves of 171 gastric cancer patients according to human epidermal growth factor receptor 2 (HER2) detection (P = 0.3386); B: Survival curve of patients with well differentiated gastric cancer according to HER2 expression (P = 0.0123); C: Survival curve of patients with poorly differentiated gastric cancer according to HER2 expression (P = 0.0988); D: Survival curve of patients with the diffuse/mixed type gastric cancer according to HER2 expression (P = 0.6623).

Within the well differentiated gastric cancer patient group, patients with HER2 tumor positivity had poorer outcomes than those with HER2 negative tumors. The well differentiated HER2 positive patient group exhibited shorter mean survival time (18.5 mo vs 27.5 mo) and lower 1-year and 2-year survival rates compared to the HER2 negative group (84.42% vs 96.00%; 50.65% vs 86.89%; P = 0.0123; Figure 3B). The median survival time of the HER2 positive group did not show any statistical associations when compared to the subgroups of sex, age, tumor site, TNM classification, depth of invasion, lymph node metastases and distant metastasis in gastric cancer (Table 4). Within the poorly differentiated and diffuse/mixed type gastric cancer patient groups, no statistically significant differences were observed between the HER2 positive and HER2 negative groups (Figure 3C and D).

Table 4 Relationship of different clinicopathological characteristics and prognosis.
Clinicopathological characteristicsHER2 positive
HER2 negative
χ2P value
Median survival time (mo)1-year survival rate2-year survival rateMedian survival time (mo)1-year survival rate2-year survival rate
Sex
Male2074.34%50.18%2083.96%69.00%2.25910.1328
Female10100.00%50.00%16.574.50%51.79%0.01820.8927
Age (yr)
≤ 6023100.00%57.14%2080.54%61.81%0.01040.9186
> 601567.55%49.13%1880.62%64.35%1.63560.2009
Tumor site
Cardiac1966.67%50.00%1569.51%49.15%0.04940.8242
Body16.562.50%62.50%1467.80%44.07%0.15610.6927
Pylorus1785.56%46.67%2087.00%73.25%2.32950.1269
Lauren classification
Intestinal1784.85%50.91%2789.17%76.53%2.36040.1244
Diffues/mixed1467.53%49.24%16.576.99%57.24%0.19070.6623
Tumor differentiation
Well-differentiated18.584.42%50.65%27.596.00%86.89%6.27010.0123
Poorly-differentiated1467.88%49.491776.56%56.71%0.09880.7532
TNM classification
I and II stages18.568.57%57.14%21.593.60%79.20%2.98130.0842
III and IV stages1782.59%45.88%16.573.32%54.12%0.02630.8711
T
T1-T1766.67%66.67%28100.00%92.31%3.45870.0629
T3-T41591.30%46.99%1777.47%58.26%0.29530.5869
N
N01468.57%51.43%2190.46%74.98%2.06670.1505
N1-N318.579.19%49.49%1775.73%57.27%0.05310.8177
M
M01778.67%54.69%2084.41%66.01%0.78420.3757
M111.550.00%0.00%50.00%0.00%0.59000.4424
DISCUSSION

HER2 gene amplification and protein overexpression in gastric cancer were first reported in 1986[12,13]and have since been confirmed by numerous studies, highlighting ranges in both HER2 gene amplification rates from 16%-27.1% by FISH analysis and HER2 protein overexpression from 8.2%-53.4% by IHC analysis. The variability within these results is likely due to several factors including sample size, study design and differences in geographic location[14]. However, the most important variability factor is likely a consequence of having no standardized HER2 test and scoring criteria[15]. In the present study, both FISH and IHC scoring criteria followed that of Hofmann[9] which is considered to be the most appropriate HER2 scoring system in human gastric cancer. Furthermore, to ensure the reliability of our results, we followed the guidelines on HER2 detection in breast cancer, recommended by the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP)[16] and used the test kit certified by the United States Food and Drug Administration.

Herceptin (trastuzumab) is a recombinant human monoclonal antibody designed to target and block the function of HER2 by directly binding to the extracellular domain of the receptor[1,17]. It has been used for the treatment of HER2 overexpressing breast cancer for more than 10 years and was approved by the European Medicines Agency[18] in 2010 for use in combination with capecitabine or 5-FU and cisplatin for metastatic gastric or GE junction cancers, based on data from the ‘ToGA’’ clinical trial. The exact anti-tumor mechanism of Herceptin is not fully understood, however some mechanisms have been postulated[17,19-23] including interruption of HER2 mediated cell signaling pathways and cell cycle progression; induction of antibody-dependent cell-mediated cytotoxicity and apoptosis; induction of anti-angiogenesis effects and increasing receptor turnover by endocytosis. As clinical surgeons, we should be readily and accurately able to identify which patients are suitable for Herceptin treatment. An accurate and reliable HER2 scoring system, together with clinical information, may help us to better determine whether a gastric cancer patient is a potential candidate for targeted therapy using Herceptin.

The relationship between HER2 gene amplification and protein expression in gastric cancer patients is controversial[24,25]. Nevertheless, more recent studies have reported a high concordance between gene amplification and protein overexpression using FISH and IHC approaches[11,26,27]. Indeed, the ToGA trial[28] (which recruited the largest population of gastric cancer patients to date-3807) reported a HER2 FISH and IHC concordance rate of 87.5%, and further reported that HER2 IHC3+ cases were almost all entirely HER2 gene amplified (97.5% of cases). However, 22.5% of HER2 FISH positive cases in the ToGA trial were HER2 IHC negative, a finding which differs from the situation observed in breast cancer, where almost all HER2 IHC 0/1+ samples are HER2 FISH negative[14]. In our study, the overall HER2 positive rate (FISH and IHC combined) was 18.27% while 15.74% of cases showed HER2 gene amplification by FISH and 9.64% of patients showed HER2 protein overexpression by IHC analysis. The concordance between the two detection methods was 88.83%. Of the 31 FISH-positive cases, 14 cases (45.16%) were IHC3+, with a 100% concordance between IHC3+ and FISH, and 10 (32.26%) cases were IHC2+. None of the IHC0 tumors showed FISH amplification, and only 7 tumors in the IHC1+ group were found to be FISH positive with a ratio of 22.58%. A high degree of data consistency was observed between IHC3+ and IHC0/1 with FISH (73.68% and 95.42%); however, low scoring consistency was observed between IHC2+ and FISH (40.00%). Thus, our data highlights the need and importance of further clarifying the relationship between HER2 gene amplification and protein overexpression in gastric cancer.

In our study, no relationship was observed between HER2 positivity and sex, age and TNM classification (P > 0.05). However, intestinal-type and well-differentiated gastric cancer cases showed a higher HER2 positive rate than diffuse/mixed-type and poorly-differentiated cancer cases. This finding is in keeping with similar data from the ToGA trial and other published studies[29,30]. Of interest, the ToGA trial reported a higher HER2 positivity rate in GE junction cancers compared to other gastric cancers (33.2% vs 20.9%, P < 0.001)[17]. Our study, as well as that of another group[31], showed no statistically significant difference between HER2 positivity and the gastric tumor site. Within the poorly-differentiated gastric cancer patient group, those patients without lymph node metastasis showed a higher HER2 positivity rate when compared to those with lymph node metastasis (26.47% vs 7.14%, P = 0.0021). No difference in HER2 positivity was observed, however, when comparing lymph node metastasis status in the well-differentiated gastric cancer patient group (28.57% vs 43.33%, P = 0.2832). The underlying molecular mechanisms behind the varying HER2 positivity rates in the different histological GC subtypes are clearly complex and require further investigation.

The role of HER2 as a prognostic factor in gastric cancer has been controversial due to significant differences in historical study results. More recent studies, however, indicate that HER2 is a poor prognostic factor in gastric cancer patients[32-35], especially those with liver metastases[36]. Whilst our study did not show any correlation between HER2 status and overall survival, patients with well-differentiated HER2 positive tumors showed poorer survival times compared to patients with HER2 negative tumors. We speculate that HER2 status has a mild impact on gastric cancer patient survival and may not constitute an independent prognostic factor in gastric cancer patients. Clearly, further research is required to explain the impact of HER2 on development and prognosis of gastric cancer.

In conclusion, an accurate and standardized scoring system for HER2 expression in gastric cancer patients is of clear importance and utility in the optimal selection of patients for Herceptin therapy. Our studies highlight intestinal-type, well-differentiated and poorly-differentiated gastric cancer patients without lymph node metastasis as the three main candidate patient groups for targeted therapy using Herceptin. Finally, we advocate further detailed research on the mechanism(s) through which HER2 expression drives progression of gastric cancer and consideration of additional studies to explore the role of HER2 as an independent prognostic factor.

ACKNOWLEDGMENTS

We thank Dr. Paul R Gavine for valuable suggestions and critical reading of the paper.

COMMENTS
Background

Gastric cancer (GC) is one of the most prevalent cancers worldwide, with poor prognosis. Herceptin (trastuzumab) can improve overall survival without compromising safety in patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic gastric cancer. However, a standardized HER2 scoring system is still required. Studies on the correlation of HER2 and clinicopathological characteristics could help clinicians to optimally select suitable candidates for targeted therapy using Herceptin.

Research frontiers

HER2 inhibition is playing a significant role as a new treatment option for gastric cancer. Numerous countries have approved the use of Herceptin for the treatment of gastric cancer and increasingly, HER2 has become a ‘‘hot’’ research topic. An accurate and reliable HER2 scoring system is necessary to select suitable candidates for Herceptin targeted therapy.

Innovations and breakthroughs

To date, there have been limited studies to determine any correlations of HER2 expression with clinicopathological characteristics and prognosis in Chinese patients with resectable gastric cancer. Intestinal type gastric cancer patients, well-differentiated gastric cancer patients and poorly-differentiated gastric cancer patients without lymph node metastasis showed a higher HER2 positivity rate and thus could represent ideal candidates for targeted-therapy using Herceptin.

Applications

The study results suggest that an accurate HER2 scoring system plays an important role with clinical significance. Patients with intestinal-type gastric cancer, well-differentiated gastric cancer and poorly-differentiated gastric cancer without lymph node metastasis are ideal candidates for targeted therapy using Herceptin.

Peer review

The paper makes sense to search for the gastric cancer patients in Jiangsu province. The study design is valid and the data is sufficient.

Footnotes

P- Reviewer Wang GQ S- Editor Zhai HH L- Editor O’Neill M E- Editor Zhang DN

References
1.  Tai W, Mahato R, Cheng K. The role of HER2 in cancer therapy and targeted drug delivery. J Control Release. 2010;146:264-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 363]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
2.  Kaur A, Dasanu CA. Targeting the HER2 pathway for the therapy of lower esophageal and gastric adenocarcinoma. Expert Opin Pharmacother. 2011;12:2493-2503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
3.  Lambein K, Praet M, Forsyth R, Van den Broecke R, Braems G, Matthys B, Cocquyt V, Denys H, Pauwels P, Libbrecht L. Relationship between pathological features, HER2 protein expression and HER2 and CEP17 copy number in breast cancer: biological and methodological considerations. J Clin Pathol. 2011;64:200-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
4.  Li Q, Wang D, Li J, Chen P. Clinicopathological and prognostic significance of HER-2/neu and VEGF expression in colon carcinomas. BMC Cancer. 2011;11:277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 47]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
5.  Yan B, Choo SN, Mulyadi P, Srivastava S, Ong CW, Yong KJ, Putti T, Salto-Tellez M, Lim GS. Dual-colour HER2/chromosome 17 chromogenic in situ hybridisation enables accurate assessment of HER2 genomic status in ovarian tumours. J Clin Pathol. 2011;64:1097-1101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
6.  Yan B, Yau EX, Choo SN, Ong CW, Yong KJ, Pang B, Salto-Tellez M. Dual-colour HER2/chromosome 17 chromogenic in situ hybridisation assay enables accurate assessment of HER2 genomic status in gastric cancer and has potential utility in HER2 testing of biopsy samples. J Clin Pathol. 2011;64:880-883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
7.  Reichelt U, Duesedau P, Tsourlakis MCh, Quaas A, Link BC, Schurr PG, Kaifi JT, Gros SJ, Yekebas EF, Marx A. Frequent homogeneous HER-2 amplification in primary and metastatic adenocarcinoma of the esophagus. Mod Pathol. 2007;20:120-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 143]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
8.  Lordick F. Trastuzumab: a new treatment option for HER2-positive metastatic gastric and gastroesophageal junction cancer. Future Oncol. 2011;7:187-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
9.  Hofmann M, Stoss O, Shi D, Büttner R, van de Vijver M, Kim W, Ochiai A, Rüschoff J, Henkel T. Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology. 2008;52:797-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 777]  [Cited by in F6Publishing: 868]  [Article Influence: 54.3]  [Reference Citation Analysis (0)]
10.  Hicks DG, Whitney-Miller C. HER2 testing in gastric and gastroesophageal junction cancers: a new therapeutic target and diagnostic challenge. Appl Immunohistochem Mol Morphol. 2011;19:506-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
11.  Albarello L, Pecciarini L, Doglioni C. HER2 testing in gastric cancer. Adv Anat Pathol. 2011;18:53-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 119]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
12.  Fukushige S, Matsubara K, Yoshida M, Sasaki M, Suzuki T, Semba K, Toyoshima K, Yamamoto T. Localization of a novel v-erbB-related gene, c-erbB-2, on human chromosome 17 and its amplification in a gastric cancer cell line. Mol Cell Biol. 1986;6:955-958.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Sakai K, Mori S, Kawamoto T, Taniguchi S, Kobori O, Morioka Y, Kuroki T, Kano K. Expression of epidermal growth factor receptors on normal human gastric epithelia and gastric carcinomas. J Natl Cancer Inst. 1986;77:1047-1052.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Lorenzen S, Lordick F. How will human epidermal growth factor receptor 2-neu data impact clinical management of gastric cancer? Curr Opin Oncol. 2011;23:396-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
15.  Jørgensen JT. Targeted HER2 treatment in advanced gastric cancer. Oncology. 2010;78:26-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 111]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
16.  Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med. 2007;131:18-43.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Meza-Junco J, Au HJ, Sawyer MB. Trastuzumab for gastric cancer. Expert Opin Biol Ther. 2009;9:1543-1551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 33]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
18.  Fujita T. Trastuzumab for gastric cancer treatment. Lancet. 2010;376:1735; author reply 1735-1736.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Fujimoto-Ouchi K, Sekiguchi F, Yasuno H, Moriya Y, Mori K, Tanaka Y. Antitumor activity of trastuzumab in combination with chemotherapy in human gastric cancer xenograft models. Cancer Chemother Pharmacol. 2007;59:795-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 125]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
20.  Kim SY, Kim HP, Kim YJ, Oh do Y, Im SA, Lee D, Jong HS, Kim TY, Bang YJ. Trastuzumab inhibits the growth of human gastric cancer cell lines with HER2 amplification synergistically with cisplatin. Int J Oncol. 2008;32:89-95.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1728]  [Cited by in F6Publishing: 1847]  [Article Influence: 108.6]  [Reference Citation Analysis (0)]
22.  Lange T, Nentwich MF, Lüth M, Yekebas E, Schumacher U. Trastuzumab has anti-metastatic and anti-angiogenic activity in a spontaneous metastasis xenograft model of esophageal adenocarcinoma. Cancer Lett. 2011;308:54-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
23.  Croxtall JD, McKeage K. Trastuzumab: in HER2-positive metastatic gastric cancer. Drugs. 2010;70:2259-2267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
24.  Lemoine NR, Jain S, Silvestre F, Lopes C, Hughes CM, McLelland E, Gullick WJ, Filipe MI. Amplification and overexpression of the EGF receptor and c-erbB-2 proto-oncogenes in human stomach cancer. Br J Cancer. 1991;64:79-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 121]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
25.  Kameda T, Yasui W, Yoshida K, Tsujino T, Nakayama H, Ito M, Ito H, Tahara E. Expression of ERBB2 in human gastric carcinomas: relationship between p185ERBB2 expression and the gene amplification. Cancer Res. 1990;50:8002-8009.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Yano T, Doi T, Ohtsu A, Boku N, Hashizume K, Nakanishi M, Ochiai A. Comparison of HER2 gene amplification assessed by fluorescence in situ hybridization and HER2 protein expression assessed by immunohistochemistry in gastric cancer. Oncol Rep. 2006;15:65-71.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Tafe LJ, Janjigian YY, Zaidinski M, Hedvat CV, Hameed MR, Tang LH, Hicks JB, Shah MA, Barbashina V. Human epidermal growth factor receptor 2 testing in gastroesophageal cancer: correlation between immunohistochemistry and fluorescence in situ hybridization. Arch Pathol Lab Med. 2011;135:1460-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
28.  Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, Lordick F, Ohtsu A, Omuro Y, Satoh T. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4615]  [Cited by in F6Publishing: 4831]  [Article Influence: 345.1]  [Reference Citation Analysis (0)]
29.  Tsapralis D, Panayiotides I, Peros G, Liakakos T, Karamitopoulou E. Human epidermal growth factor receptor-2 gene amplification in gastric cancer using tissue microarray technology. World J Gastroenterol. 2012;18:150-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
30.  Liu W, Zhong S, Chen J, Yu Y. HER-2/neu overexpression is an independent prognostic factor for intestinal-type and early-stage gastric cancer patients. J Clin Gastroenterol. 2012;46:e31-e37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
31.  Marx AH, Tharun L, Muth J, Dancau AM, Simon R, Yekebas E, Kaifi JT, Mirlacher M, Brümmendorf TH, Bokemeyer C. HER-2 amplification is highly homogenous in gastric cancer. Hum Pathol. 2009;40:769-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 178]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
32.  Ananiev J, Gulubova M, Manolova I, Tchernev G. Prognostic significance of HER2/neu expression in gastric cancer. Wien Klin Wochenschr. 2011;123:450-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
33.  Jørgensen JT, Hersom M. HER2 as a Prognostic Marker in Gastric Cancer - A Systematic Analysis of Data from the Literature. J Cancer. 2012;3:137-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 179]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
34.  Bouché O, Penault-Llorca F. [HER2 and gastric cancer: a novel therapeutic target for trastuzumab]. Bull Cancer. 2010;97:1429-1440.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Yan SY, Hu Y, Fan JG, Tao GQ, Lu YM, Cai X, Yu BH, Du YQ. Clinicopathologic significance of HER-2/neu protein expression and gene amplification in gastric carcinoma. World J Gastroenterol. 2011;17:1501-1506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 55]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
36.  Dang HZ, Yu Y, Jiao SC. Prognosis of HER2 over-expressing gastric cancer patients with liver metastasis. World J Gastroenterol. 2012;18:2402-2407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (1)]