Background

The role of human epidermal growth factor receptor 2 (HER2) overexpression has been well established in breast cancer, with corresponding targeted therapies. In contrast, the clinicopathologic features and prognosis of HER2 overexpression in gastric cancer remain inconclusive.

Methods

In this study, 334 patients with gastric cancer who received surgical resection between May 2017 and June 2021 were enrolled at a single medical center in Taiwan. HER2 status was determined by immunohistochemistry (IHC) staining or fluorescence in situ hybridization (FISH). The clinicopathologic features and survival curves of the HER2-positive and HER2-negative gastric cancer patients were analyzed.

Results

The HER2-positive ratio was 7.2%. HER2-positive gastric cancer was associated with more differentiated tumors (P = 0.016), more Lauren intestinal type (P = 0.010), and a higher portion of Ming expanding type (P = 0.033) in the univariate analysis, but only Lauren intestinal type was an independent factor in the logistic regression model (P = 0.015). The overall survival and disease-free survival between the HER2-positive and HER2-negative groups were not significantly different. Patients with HER2-positive gastric cancer were more likely to have distant lymphatic recurrence than those with HER2-negative gastric cancer (P = 0.026).

Conclusions

HER2-positive gastric cancer is associated with intestinal histologic type and distant lymphatic recurrence, but HER2 is not an independent prognostic factor.

Gastric cancer is the fifth most common cancer and the fourth most common cause of cancer death worldwide.1  The treatment for gastric cancer is surgical resection with curative intention for patients without distant metastasis. For advanced or metastatic disease, survival is generally poor. However, improvements in chemotherapy with add-on targeted therapy or immunotherapy have led to survival benefits, which means that tumor biology is more important than ever.2 

Overexpression of human epidermal growth factor receptor 2 (HER2) is well known to promote oncogenesis in several cancers. The HER2 protein is a 185-kDa transmembrane tyrosine kinase (TK) receptor and a member of the epidermal growth factor receptor (EGFR) family. It is encoded by the Her2/neu oncogene located on chromosome 17q21 and is associated with tumor cell proliferation by activating downstream signals, such as PI3K/Akt/mTOR and MAPK.3,4  In breast cancer, the role of HER2 has been well established. Numerous targeted agents have been approved for the treatment of HER2-positive breast cancer. In contrast, the role of HER2 overexpression in gastric cancer is still inconclusive. The HER2-positive rate in gastric cancer has been reported to range widely from 3.8% to 50% by 2 meta-analyses. The 2 meta-analyses have demonstrated that HER2 overexpression is associated with male sex, intestinal type, and well/moderate cell differentiation.5,6  However, the prognostic value of HER2 in gastric cancer is still controversial.7–10 

This study aims to investigate the clinicopathologic features and prognosis of HER2-positive gastric cancer in the Taiwanese population.

Patient collection

A total of 334 gastric cancer patients with adenocarcinoma who underwent surgical resection between May 2017 and June 2021 at the Department of Surgery in Taipei Veterans General Hospital were enrolled in this study. Patients with pathology other than adenocarcinoma were excluded from this study.

Prior to surgery, all patients underwent upper gastrointestinal endoscopy, chest radiography, and computerized tomography scans of the abdomen. Radical total gastrectomy was performed for proximal-third lesions, and radical subtotal gastrectomy was performed for middle- or distal-third lesions. For early gastric cancer, at least D1+ lymph node dissection was performed. For advanced gastric cancer, D2 lymph node dissection was performed.11 

The gross features of the pathologic specimens were evaluated according to the tumor location, tumor size, and Borrmann classification. The microscopic features of histology, pathology, and cell differentiation were analyzed according to the cell grade of tumor differentiation, Lauren histology (intestinal or diffuse type), Ming histology (expanding or infiltrating type), and lymphovascular invasion patterns. The pathologic staging was defined according to the 8th American Joint Committee on Cancer/Union for International Cancer Control Tumor-Node-Metastasis (TNM) classification of malignant tumors.12 

After the surgery, oral fluoropyrimidine S-1 or adjuvant systemic therapy as per the physician’s choice was given for stage II or stage III gastric cancer patients after curative surgery. For patients with palliative resection, salvage concurrent chemoradiotherapy or systemic therapy was given by an oncologist.

Postoperative follow-up assessments were performed as per local practice guidelines: every 3 months for the first 3 years and every 6 months thereafter until the patient’s death. The follow-up procedures included physical examinations, blood tests including tumor markers (e.g., carcinoembryonic antigen and carbohydrate antigen 19-9), liver function tests, chest films, abdominal sonography, and computerized tomography scans. Tumor recurrence was diagnosed by biopsies or by imaging studies when biopsies were not obtained. Tumor recurrence in the remnant stomach, hepatoduodenal ligament, celiac axis, or peripancreatic region is considered locoregional recurrence. We defined remote lymphatic metastasis (in the para-aortic, Virchow, and inguinal nodes) and pulmonary lymphangitic spread as distant lymphatic recurrence. Tumor recurrence is classified as locoregional, peritoneal, hematogenous, and distant lymphatic. Overall survival (OS) was defined as the period from the date of surgery to the date of death or the latest follow-up. Disease-free survival (DFS) was defined as the period from the date of surgery during which the patient survived without tumor recurrence. Patients with palliative resection were excluded from the DFS analysis.

Immunohistochemical staining and fluorescence in situ hybridization

For HER2 immunohistochemistry (IHC) staining, the antihuman c-erbB-2 A0485 polyclonal antibody (dilution 1:500; Dako, Denmark) was used, and IHC staining was performed using a BenchMark Ultra Platform (Ventana Medical Systems, Tucson, AZ) with the Optiview DAB Detection Kit (Ventana Medical Systems). According to the ToGA scoring systems, HER2 immunoreactivity was scored as 0, 1+, 2+, or 3+, with the following definition: 0, no reactivity or membranous reactivity in less than 10% of tumor cells; 1+, faint or barely perceptible membranous reactivity in at least 10% of tumor cells; 2+, weak to moderate complete, basolateral or lateral membranous reactivity in at least 10% of tumor cells; and 3+, strong complete, basolateral or lateral membranous reactivity in at least 10% of tumor cells.13  For specimens with an IHC score of 2+, fluorescence in situ hybridization (FISH) analysis of HER2 status was performed with the PathVysion HER2 DNA Probe Kit (Abbott, Abbott Park, IL). Positive HER2 gene amplification was defined as a HER2/CEP17 ratio ≥2.14  HER2 positivity was defined as an IHC score of 3+ or an IHC score of 2+ with a positive FISH result.

Ethical approval

Written informed consent for tissue collection was obtained from all patients before surgery. All samples used in this study had been previously collected from the biobank of Taipei Veterans General Hospital and were anonymized. The study was approved by the Institutional Review Board of Taipei Veterans General Hospital (IRB-TPEVGH no. 2022-10-003CC).

Statistical analysis

Statistical analyses were performed using IBM SPSS Statistics 19.0 (Armonk, NY). All the results in the text and tables are presented as the means ± SD. The clinicopathologic differences were compared using Pearson χ2 test or Fisher exact test. Corrections of data were performed using multiple testing in a logistic regression model. The distributions of OS and DFS were evaluated using the Kaplan-Meier method. Univariate analysis of the covariates (prognostic factors) of OS was performed first. The covariates with a P value <0.05 were selected for multivariate analysis using a Cox regression model with the forward stepwise method. A P value of less than 0.05 was considered statistically significant.

Among the 334 included gastric cancer patients, the HER2 status of the tumor samples were determined by IHC staining as follows: score 0, 233 (69.8%); score 1+, 72 (21.6%); score 2+, 11 (3.3%); and score 3+, 18 (5.4%) (Fig. 1). Of the 11 tumor samples with an IHC score of 2+, 6 (54.5%) were HER2-positive by FISH (Fig. 2). In total, 24 tumor samples (7.2%) were categorized as HER2-positive.

The patients’ clinicopathologic features according to HER2 status were analyzed (Table 1). The univariate analysis showed that the HER2-positive gastric cancer group was associated with more differentiated tumors (66.7% versus 41.3%; P = 0.016), more Lauren intestinal type (75% versus 44.7%; P = 0.010), and more Ming expanding type (37.5% versus 18.4%; P = 0.033) compared with the HER2-negative gastric cancer group. After multiple testing in a logistic regression model, only Lauren classification was an independent factor associated with HER2-positive gastric cancer (odds ratio, 3.264; P = 0.015).

The OS and DFS were compared between the HER2-positive and HER2-negative groups, and there was no significant difference found in either OS (P = 0.190) or DFS (P = 0.236; Fig. 3). Univariate analysis of prognostic factors with OS as an end point showed that tumor size, gross appearance, lymphovascular invasion, and TNM stage were associated with prognosis. After multivariate adjustment, only TNM stage was an independent prognostic factor. HER2 status failed to be a prognostic factor (Table 2).

Among the 299 patients with curative resection, 57 patients (19%) had tumor recurrence. The initial recurrence pattern was compared according to the HER2 status (Table 3). The patients with HER2-positive gastric cancer were more likely to have distant lymphatic recurrence than those with HER2-negative gastric cancer (15.8% versus 2.9%, P = 0.026).

A subgroup analysis of 12 patients with stage III/IV HER2-positive gastric cancer was performed. Among those 12 patients, 5 patients (41.7%) received anti-HER2 targeted therapy. For stage III/IV HER2-positive gastric cancer, the OS was significantly better for the patients with anti-HER2 therapy than for those without anti-HER2 therapy (P = 0.019).

The HER2-positive ratio was 7.2% in our study using standardized testing methods according to the ToGA trial. The range of the HER2-positive ratio has been reported widely from 3.1% to 50%.5,6  The reported wide range could be a result of ethnic variation and differences in methodology. For studies using the HER2-positive definition of IHC scores of 2+ and 3+ without FISH, the positive ratio could be overestimated. When studying the same population using the same criteria, Hsu et al9  demonstrated a similar positive ratio of 6.1%.

The association between HER2-positive gastric cancer and intestinal type as well as differentiated histology has been widely reported in most published literature, including two meta-analyses.5–9,15–17  Our study demonstrated consistent findings that HER2-positive gastric cancer was associated with more differentiated tumors, more Lauren intestinal type, and a higher portion of Ming expanding type in the univariate analysis. After further logistic regression, only Lauren classification was an independent factor associated with HER2-positive gastric cancer. Lauren classification categorized gastric cancer into the intestinal type and diffuse type, and the two different types have distinct tumor behaviors.18  The intestinal type is associated with atrophic gastritis, intestinal metaplasia, preserved glandular appearance, and better differentiation. In contrast, the diffuse type lacks glandular architecture and is poorly differentiated. Because Lauren classification already plays a role in terms of differentiation, the significance of differentiation is likely to be adjusted in logistic regression, as is shown in our statistical results.

We found that HER2-positive gastric cancer patients were more likely to have distant lymphatic recurrence than those with HER2-negative gastric cancer. In addition, among the 5 patients with HER2-positive gastric cancer who received palliative resection, 3 patients had para-aortic lymph node metastasis. Both results imply that HER2 positivity is associated with distant lymphatic spreading. A similar finding of a higher HER2-positive rate in gastric cancer patients with extensive lymph node metastasis was reported by Matsumoto et al.19 

Our results showed that for stage III/IV HER2-positive gastric cancer, the OS was significantly better for patients with anti-HER2 therapy than for those without anti-HER2 therapy. Among the 5 patients treated with anti-HER2 therapy, 2 patients were Lauren intestinal type (2 of 5; 40%), and 3 patients were Lauren diffuse type (3 of 5; 60%). Among the 7 patients without anti-HER2 therapy, 5 patients were Lauren intestinal type (5 of 7; 71.4%), and 2 patients were Lauren diffuse type (2 of 7; 28.6%). Lauren diffuse type is associated with poorer survival compared to intestinal type.20  Despite the differences in Lauren classification, patients receiving anti-HER2 therapy showed significantly improved survival. For the 3 patients with distant lymph node metastasis who received anti-HER2 therapy and palliative resection, the outcome was even more encouraging. All 3 patients were disease-free with survival up to 58 months. Arigami et al21  demonstrated similar findings that HER2 overexpression could be used to determine the prognosis of patients with para-aortic lymph node metastasis from gastric cancer.21  From their study, a higher HER2-positive ratio (43.9%) was noted, and HER2-positive patients who received trastuzumab-based chemotherapy had a significantly better prognosis than HER2-negative patients in gastric cancer with para-aortic lymph node metastasis. It seems that patients with HER2-positive gastric cancer and distant lymph node metastasis can benefit the most from anti-HER2 therapy. However, this hypothesis needs further larger-scale prospective studies for validation.

HER2 overexpression was thought to be associated with a poorer prognosis in node-positive and node-negative breast cancer.22  With the development of anti-HER2 therapy, the role of HER2 has changed. For patients treated with anti-HER2 agents appropriately, HER2-positive breast cancers are associated with favorable outcomes compared with HER2-negative breast cancers in the 8th American Joint Committee on Cancer staging system.23  A similar result is anticipated but not yet established in the era of gastric cancer. The ToGA trial, a phase 3 randomized study by Bang et al,14  demonstrated a survival benefit of trastuzumab for HER2-positive metastatic gastric or gastroesophageal junction cancer.14  In contrast, in the NRG Oncology/RTOG 1010 randomized phase 3 trial, the addition of trastuzumab to neoadjuvant chemoradiotherapy was not effective.24 

The JACOB trial, a phase 3 randomized controlled study conducted by Tabernero et al25  showed that adding pertuzumab to trastuzumab and chemotherapy did not improve OS for HER2-positive metastatic gastric or gastroesophageal junction cancer. Despite the negative result of the JACOB trial, the efficacy of dual blockade in the perioperative setting is still being investigated. The PETRARCA phase 2 trial, even though it was closed prematurely, demonstrated that the addition of trastuzumab and pertuzumab to perioperative FLOT significantly improved pathologic complete remission (35% versus 12%; P = 0.019) and preliminary survival benefit in patients with HER2-positive resectable esophagogastric adenocarcinoma.26  The INNOVATION trial has been conducted to evaluate the effect of chemotherapy alone versus chemotherapy plus trastuzumab versus chemotherapy plus trastuzumab plus pertuzumab in the neoadjuvant setting.27  The pending results are essential to determine the role of neoadjuvant trastuzumab and pertuzumab.

The efficacy of trastuzumab-based antibody–drug conjugates is also controversial. The GATSBY trial concluded that trastuzumab emtansine (T-DM1) was not superior to taxane in patients with previously treated HER2-positive advanced gastric cancer.28  In the DESTINY-Gastric01 trial, the result of trastuzumab deruxtecan (T-DXd) was totally different. Trastuzumab deruxtecan significantly improved the response and OS compared with standard therapies among patients with treated HER2-positive gastric cancer.29  Although the results of anti-HER2 agents for HER2-positive gastric cancer are inconsistent, numerous trials with various settings are still ongoing to determine the potential benefit for HER2-positive gastric cancer.

According to our study, TNM stage remains the most crucial prognostic factor. None of the remaining studied factors can be predictive of prognosis. This result again emphasizes the importance of early detection and early management for gastric cancer.

There are limitations in our study. First, it is a single-center retrospective study with only Taiwanese individuals included. Therefore, potential selection bias exists. Second, the number of HER2-positive cases is limited because of the low positive ratio of 7.2%. Therefore, it is difficult to perform further analysis in terms of prognostic value. Nevertheless, we found that HER2-positive gastric cancer is highly correlated with distant lymphatic spreading and its potential role as a favorable prognostic factor if treated with anti-HER2 therapy.

HER2-positive gastric cancer was associated with intestinal histologic type. Patients with HER2-positive gastric cancer were more likely to have distant lymphatic recurrence, but HER2 was not an independent prognostic factor.

This study was supported with funding from Melissa Lee Cancer Foundation (MLCF-V111_A11107) and Taipei Veterans General Hospital (V111C-222, V111C-184). The funding did not play a role in the study design, data collection, analysis, interpretation, or manuscript writing.

1.
Sung
H
et al.
Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2021
;
71
(
3
):
209
249
2.
Joshi
SS,
Badgwell
BD.
Current treatment and recent progress in gastric cancer
.
CA Cancer J Clin
2021
;
71
(
3
):
264
279
3.
Gravalos
C,
Jimeno
A.
HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target
.
Ann Oncol
2008
;
19
(
9
):
1523
1529
4.
Meric-Bernstam
F
et al.
Advances in HER2-targeted therapy: novel agents and opportunities beyond breast and gastric cancer
.
Clin Cancer Res
2019
;
25
(
7
):
2033
2041
5.
Wang
HB,
Liao
XF,
Zhang
J.
Clinicopathological factors associated with HER2-positive gastric cancer: a meta-analysis
.
Medicine (Baltimore)
2017
;
96
(
44
):
e8437
6.
Lei
YY
et al.
The clinicopathological parameters and prognostic significance of HER2 expression in gastric cancer patients: a meta-analysis of literature
.
World J Surg Oncol
2017
;
15
(
1
):
68
7.
Park
DI
et al.
HER-2/neu amplification is an independent prognostic factor in gastric cancer
.
Dig Dis Sci
2006
;
51
(
8
):
1371
1379
8.
Kurokawa
Y
et al.
Multicenter large-scale study of prognostic impact of HER2 expression in patients with resectable gastric cancer
.
Gastric Cancer
2015
;
18
(
4
):
691
697
9.
Hsu
JT
et al.
Impact of HER-2 overexpression/amplification on the prognosis of gastric cancer patients undergoing resection: a single-center study of 1,036 patients
.
Oncologist
2011
;
16
(
12
):
1706
1713
10.
Jorgensen
JT,
Hersom
M.
HER2 as a prognostic marker in gastric cancer–a systematic analysis of data from the literature
.
J Cancer
2012
;
3
:
137
–1
44
11.
Japanese Gastric Cancer Association
.
Japanese gastric cancer treatment guidelines 2014 (ver. 4)
.
Gastric Cancer
2017
;
20
(
1
):
1
19
12.
American Joint Committee on Cancer
.
AJCC Cancer Staging Manual
.
8th ed
.
New York, NY, USA
:
Springer
;
2017
13.
Hofmann
M
et al.
Assessment of a HER2 scoring system for gastric cancer: results from a validation study
.
Histopathology
2008
;
52
(
7
):
797
805
14.
Bang
YJ
et al.
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
(
9742
):
687
697
15.
Barros-Silva
JD
et al.
Association of ERBB2 gene status with histopathological parameters and disease-specific survival in gastric carcinoma patients
.
Br J Cancer
2009
;
100
(
3
):
487
493
16.
Son
HS
et al.
Correlation between HER2 overexpression and clinicopathological characteristics in gastric cancer patients who have undergone curative resection
.
J Gastric Cancer
2014
;
14
(
3
):
180
186
17.
Li
F
et al.
Relationship between HER2 expression and tumor interstitial angiogenesis in primary gastric cancer and its effect on prognosis
.
Pathol Res Pract
2021
;
217
:
153280
18.
Lauren
P.
The two histologic main types of gastric carcinoma: diffuse and so-called intestinal type carcinoma, an attempt at a histo-clinical classification
.
Acta Parhol Microb Scan
1965
;
64
:
31
49
19.
Matsumoto
T
et al.
HER2 expression in locally advanced gastric cancer with extensive lymph node (bulky N2 or paraaortic) metastasis (JCOG1005-A trial)
.
Gastric Cancer
2015
;
18
(
3
):
467
475
20.
Chen
YC
et al.
Clinicopathological variation of Lauren classification in gastric cancer
.
Pathol Oncol Res
2016
;
22
(
1
):
197
202
21.
Arigami
T
et al.
Prognostic significance of HER2 expression for gastric cancer with clinically para-aortic lymph node metastasis
.
Anticancer Res
2021
;
41
(
6
):
3099
3107
22.
Krishnamurti
U,
Silverman
JF.
HER2 in breast cancer: a review and update
.
Adv Anat Pathol
2014
;
21
(
2
):
100
107
23.
Giuliano
AE
et al.
Breast cancer–major changes in the American Joint Committee on Cancer eighth edition cancer staging manual
.
CA Cancer J Clin
2017
;
67
(
4
):
290
303
24.
Safran
HP
et al.
Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): a multicentre, randomised, phase 3 trial
.
Lancet Oncol
2022
;
23
(
2
):
259
269
25.
Tabernero
J
et al.
Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): final analysis of a double-blind, randomised, placebo-controlled phase 3 study
.
Lancet Oncol
2018
;
19
(
10
):
1372
1384
26.
Hofheinz
RD
et al.
Perioperative trastuzumab and pertuzumab in combination with FLOT versus FLOT alone for HER2-positive resectable esophagogastric adenocarcinoma: Final results of the PETRARCA multicenter randomized phase II trial of the AIO
.
J Clin Oncol
2020
;
38
(
15
suppl):
4502
4502
27.
Wagner
AD
et al.
EORTC-1203-GITCG - the “INNOVATION”-trial: effect of chemotherapy alone versus chemotherapy plus trastuzumab, versus chemotherapy plus trastuzumab plus pertuzumab, in the perioperative treatment of HER2 positive, gastric and gastroesophageal junction adenocarcinoma on pathologic response rate: a randomized phase II-intergroup trial of the EORTC-Gastrointestinal Tract Cancer Group, Korean Cancer Study Group and Dutch Upper GI-Cancer group
.
BMC Cancer
2019
;
19
(
1
):
494
28.
Thuss-Patience
PC
et al.
Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study
.
Lancet Oncol
2017
;
18
(
5
):
640
653
29.
Shitara
K
et al.
Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer
.
N Engl J Med
2020
;
382
(
25
):
2419
2430