Skip to main content
Erschienen in: Gastric Cancer 1/2015

Open Access 01.01.2015 | Original Article

New molecular staging with G-factor supplements TNM classification in gastric cancer: a multicenter collaborative research by the Japan Society for Gastroenterological Carcinogenesis G-Project committee

verfasst von: Tetsuji Sawada, Masakazu Yashiro, Kazuhiro Sentani, Naohide Oue, Wataru Yasui, Kohji Miyazaki, Keita Kai, Sachio Fushida, Takashi Fujimura, Masaichi Ohira, Yoshihiro Kakeji, Shoji Natsugoe, Ken Shirabe, Sachiyo Nomura, Yutaka Shimada, Naohiro Tomita, Kosei Hirakawa, Yoshihiko Maehara

Erschienen in: Gastric Cancer | Ausgabe 1/2015

Abstract

Background

The G-Project committee was erected by the Japan Society for Gastroenterological Carcinogenesis with an aim of establishing a new classification scheme based on molecular biological characteristics that would supplement the conventional TNM classification to better predict outcome.

Methods

In a literature search involving 822 articles on gastric cancer, eight molecules including p53, vascular endothelial growth factor (VEGF)-A, VEGF-C, matrix metalloproteinase-7 (MMP-7), human epidermal growth factor receptor 2, Regenerating islet-derived family, member 4, olfactomedin-4 and Claudin-18 were selected as candidates to be included in the new molecular classification scheme named G-factor. A total of 210 cases of gastric cancer who underwent curative R0 resection were registered from four independent facilities. Immunohistochemical staining for the aforementioned molecules was performed for the surgically resected specimens of the 210 cases to investigate the correlation between clinicopathological factors and expression of each molecule.

Results

No significant correlation was observed between the immunostaining expression of any of the eight factors and postoperative recurrence. However, the expressions of p53 and MMP-7 were significantly correlated with overall survival (OS). When 210 gastric cancer patients were divided into three groups based on the expression of p53 and MMP-7 (G0 group: negative for both p53 and MMP-7, n = 69, G1 group: positive for either p53 or MMP-7, n = 97, G2 group: positive for both of the molecules, n = 44), G2 group demonstrated significantly higher recurrence rate (59 %) compared to 38 % in G0 (p = 0.047). The multivariate regression analysis revealed that G2 group was independently associated with a shorter disease-free survival (DFS) (hazard ratio 1.904, 95 % CI 1.098–3.303; p = 0.022), although the association with OS was not significant. Stage II patients among the G2 group had significantly inferior prognosis both in terms of OS and DFS when compared with those among the G0/G1 group, with survival curves similar to those of Stage III cases.

Conclusions

G-factor based on the expression of p53 and MMP-7 was found to be a promising factor to predict outcome of Stage II/III gastric cancer, and possibly to help select the treatment for Stage II cancer, thus supplementing the conventional TNM system.

Introduction

To date, staging classification of gastric cancers has been performed by the International Union against Cancer (UICC)/TNM classification system [1] and the general rules of the Japanese classification of gastric carcinoma edited by Japanese Gastric Cancer Association [2]. These stage classifications based on clinicopathological factors are the global gold standard for clinical decision-making. In Japan, the technique and procedure for gastrectomy with lymph node dissection in gastric cancer patients have been established, along with adjuvant postoperative chemotherapy for those who have undergone R0 resection [3]. However, some patients suffer from unexpectedly early recurrence among those who underwent curative surgery even in Stage II cancer, implicating differences in biological characteristics. Tumor biology may not always be reflected in the clinical stage, which represents the extent of cancer spread at the time the disease was diagnosed. Therefore, identification of a new factor that reflects biological characteristics is warranted so as to supplement the clinicopathological factors for more precise prediction of the outcome. The G-Project committee was established by the Japan Society for Gastroenterological Carcinogenesis during their 2005 annual meeting, with an aim of proposing a new TNM-G classification that contains G-factor, a category that evaluates expression of relevant molecules that might influence the outcome. While evaluating the convenience of implementing the TNM-G classification system into daily practice, analysis of expression at the genetic level was considered too challenging. Thus, analysis of protein expression by immunohistological staining became the method of choice for this project. Here, we conducted a multicenter collaborative study to identify an appropriate molecule as a G-factor.

Patients and methods

Patient extraction

This study was conducted after obtaining approval from the society’s ethics committee at the annual meeting in 2007, and then requesting approval from the ethics committee of each of the four institutions, Department of Surgical Oncology, Osaka City University graduate School of Medicine (Osaka, Japan); Department of Gastroenterological Surgery, Kanazawa University (Kanazawa, Japan); Department of Surgery and Science, Graduate School of Medicine, Kyushu University (Fukuoka, Japan); and Department of Gastroenterological Surgery, Saga University, Faculty of Medicine (Saga, Japan), that supplied resected specimens. Each institution provided samples according to an implementation planning report. The study protocol conformed to the ethical guidelines of the Declaration of Helsinki, 1975. Recent reports suggest that there was a difference of at least 20 % in 5-year survival when patients were stratified according to the expression of useful prognostic factors [411]. To detect as much difference in survival in a retrospective analysis, approximately 100 cases who had recurrence and 100 cases who were recurrence-free after a sufficient follow up were deemed necessary.
The aforementioned four institutions ultimately registered 210 cases of gastric cancer who underwent curative R0 resection by gastrectomy with more than D1 lymph node dissection (D1+ to D2). Of the 210 cases, 104 were Stage II and 106 were Stage III according to the Japanese Classification of Gastric Carcinoma (the 13th edition). In addition, 104 cases were confirmed to have postoperative recurrence or death within 5 years, whereas 106 cases were confirmed to have been recurrence-free for 5–10 years. Upon gaining approval from the ethics committees of each of the aforementioned institutions, tissue samples were obtained, along with clinicopathologic data such as age, gender, occupation, operative procedure, degree of penetration into the wall (pT), lymph node metastasis (pN), final stage, ly and v factors, histological type, presence or absence of adjuvant therapy and regime, recurrence type, postoperative disease-free survival (DFS) and postoperative overall survival (OS). The data were subsequently analyzed by the Department of Oncology at the Institute of Geriatrics and Medical Science, Osaka City University, Graduate School of Medicine.

Selection of molecules for factor analysis

The G project committee was elected, and as a preliminary step, the members conducted a PubMed literature search of articles published between 1990 and 2005 using the key words “gastric cancer” and “independent prognostic factor.” A total of 822 articles on gastric cancer were extracted and reviewed (Table 1). Of the 50 molecules identified, the molecules or other factors frequently highlighted in both univariate and multivariate analyses as prognostic factors were epidermal growth factor (EGF) (35 papers), p53 (21), vascular endothelial growth factor (VEGF) (18), microsatellite instability (12), TGF-β (6), urokinase-type plasminogen activator (u-PA) (5) and E-cadherin (5). Based on these results and considering the known functions of the molecules, the committee selected p53 and VEGF (VEGF-A and VEGF-C) as candidate molecules to be nominated as G-factor. More recent articles were reviewed in the meantime, and the committee decided to add the following five molecules as candidates, based on their emerging roles in cancer biology reported after 2005: matrix metalloproteinase-7 (MMP-7, a member of the metalloprotease family) [12], human epidermal growth factor receptor 2 (HER2) [13], Regenerating islet-derived family, member 4 (Reg IV) [14, 15], olfactomedin 4 (GW112, an anti-apoptotic factor) [16], and Claudin-18 (the tight junction molecule) [17].
Table 1
Paper review of prognostic factors in gastric cancer
Category
Molecules
Number of papers by multivariate (M) or univariate (U) analysis
Oncogene
k-ras
 
c-myc
Tumor suppressor gene
p53
(M-8a, U-13b)
DCC
 
p27
 
RUNX3
 
MSI (MMR gene)
MSI/BAT
(M-3, U-3)
Cell proliferation
Cyclin D1
 
PCNA
(M-4)
MIB-1
 
Ki-67
 
Growth factor/cytokine and those receptor
EGF/erbB2
(M-15, U-20)
VEGF
(M-12, U-6)
c-met
(M-6)
TGF-b/Smad
(M-6)
b-FGF
(U-2)
PDGF
 
TGF-α
 
IL-6
 
IL-18
 
CCR7
 
TNFR
 
Apoptosis signal pathway
Bcl-2
(U-2)
Bax
 
Caspase 3
 
NF kappa B
 
Cell invasion and adhesion
TIMP
(M-4)
uPA
(M-3, U-2)
E-cadherin
(M-3, U-2)
CD4
(U-2)
MMPs
 
PAI-1, 2
 
AMF
 
KISS-1
 
Angiogenesis
Thymidine phosphorylase
(U-4)
Others
COX2
(M-2, U-2)
Microvessel count
(U-4)
Metastatic LN ration
 
aNo. of papers by multivariate analysis
bNo. of papers by univariate analysis. The list of prognostic factors reported in the 822 published articles between 1990 and 2005

Immunohistochemical staining and evaluation

The surgical specimens were formalin-fixed and paraffin-embedded, and sent to the Department of Molecular Pathology at Hiroshima University (Hiroshima, Japan). Immunohistochemical staining was performed at this facility using eight primary antibodies against the eight candidate factors. The Envision+ staining kit (Dako Corp., Glostrup, Denmark) was used for immunostaining. Paraffin-embedded specimens were sectioned at 4 μm, hydrophilized, and microwaved for 30 min in pH 6.0 citric acid buffer or autoclaved in ethylenediaminetetraacetic acid (EDTA) buffer to activate the antigen. Intrinsic peroxidase was deactivated by incubation with 3 % H2O2 for 10 min. After rinsing, blocking was performed using sheep serum and reacting with each primary antibody. Anti-p53 primary antibody was cloned name DO-7 (Dako); VEGF-A, polyclonal antibody (Santa Cruz); VEGF-C, polyclonal antibody (American Research Products); Reg IV, polyclonal antibody (see Ref. [14]); GW112, N212 (see Ref. [15]); Claudin-18, polyclonal antibody (Invitrogen); MMP-7, 141-7B2 (Daiichi Fine Chemicals). All primary antibodies were diluted 1:50 and incubated at room temperature for 1 h, washed with phosphate buffered saline (PBS), and incubated with a secondary antibody at room temperature for 1 h. The Envision+ Rabbit Peroxidase Detection System (Dako Corp.) contained the primary rabbit antibodies VEGF-A, VEGF-C, Reg IV, and Claudin-18, whereas the Envision+ Mouse Peroxidase Detection System (Dako Corp.) was used to detect p53, olfactomedin 4, and MMP-7. After washing with PBS, the samples were incubated in diaminobenzidine (DAB) solution for 10 min, stained, washed with PBS, and counter-stained with hematoxylin. Expression levels of p53, VEGF-A, VEGF-C, MMP-7, HER2, Reg IV, olfactomedin 4 and Claudin-18, were analyzed by immunostaining using one representative specimen from the center of the tumor. The stained area was scored by the percentage of immunopositive cells as an index of the expression of each molecule. Cases that were not at all stained were scored as 0, cases with < 10 % of stained tumor cells were 1+, cases with 10–50 % of stained tumor cells were 2+, while cases with > 50 % of stained tumor cells were 3+. Evaluation of immunostaining was conducted independently by two pathologists, and any discrepancies in assessment were discussed and reassessed by microscopy. Scores of 2+ and 3+ were determined as positive for immunostaining.

Data and statistical analysis

The correlation between a clinicopathological factor and immunostaining result was analyzed by the Chi-square test or Fisher’s exact test. The survival duration was calculated using the Kaplan–Meier method and analyzed by the log-rank test and Cox’ regression analysis to compare the cumulative survival durations in the patient groups. In all tests, a p value of <0.05 was considered to represent statistical significance. SPSS statistical software (SPSS Japan, Tokyo, Japan) was used for all analyses.

Results

Expression of various molecules as evaluated by immunostaining

Each representative positive expression in histological image for gastric cancer is depicted in Fig. 1. The positive staining rate of each molecule was 41.4 % for p53, 55.2 % for VEGF-A, 73.3 % for VEGF-C, 25.7 % for Reg IV, 66.2 % for olfactomedin 4, 63.4 % for Claudin-18, 46.7 % for MMP-7, and 13.0 % for HER2, respectively.

Correlation of postoperative recurrence and clinicopathological factors or the expression of the candidate molecular factors

Examination of the all 210 gastric cancer cases revealed a significant correlation between the postoperative recurrence and pT stage (p = 0.008), pN stage (p = 0.078), final stage (p < 0.001), ly factor (p = 0.002), and v factor (p < 0.001), but not with surgical procedure or the type of adjuvant chemotherapy received. As for the prognostic significance of candidate molecular factors, no significant correlation was observed between the immunostaining expression of eight aforementioned factors and postoperative recurrence (Table 2).
Table 2
Correlation between postoperative recurrence and clinicopathologic features and G-factors expression in 210 patients with gastric cancer
Clinicopathological factors and G-factors expression
Recurrence
p value
Negative
n = 106 (50 %)
Positive
n = 104 (50 %)
Gender
 Male
76 (50 %)
72 (50 %)
1.000
 Female
30 (50 %)
32 (50 %)
Location
 U
24 (41 %)
30 (59 %)
0.290
 M
47 (54 %)
42 (46 %)
 L
35 (51 %)
32 (49 %)
pT
 1
8 (69 %)
1 (31 %)
0.008
 2
51 (59 %)
35 (41 %)
 3
47 (43 %)
63 (57 %)
 4
0 (0 %)
5 (100 %)
pN
 Negative
26 (64 %)
10 (36 %)
0.078
 Positive
80 (47 %)
94 (53 %)
pN
 0
26 (64 %)
10 (36 %)
0.041
 1
55 (53 %)
48 (47 %)
 2
24 (36 %)
45 (64 %)
 3
1 (50 %)
1 (50 %)
Stage
 II
76 (73 %)
28 (27 %)
<0.001
 III
30 (28 %)
76 (71 %)
Histologic type
 Diffuse type
64 (51 %)
62 (49 %)
0.911
 Intestinal type
42 (50 %)
42 (50 %)
Lymphatic invasion
 Negative
32 (69 %)
10 (31 %)
0.002
 Positive
74 (45 %)
94 (55 %)
Venous invasion
 Negative
76 (64 %)
40 (36 %)
<0.001
 Positive
30 (32 %)
64 (68 %)
p53
 Negative
67 (55 %)
56 (45 %)
0.100
 Positive
39 (43 %)
48 (57 %)
VEGF-A
 Negative
55 (56 %)
40 (44 %)
0.105
 Positive
51 (45 %)
64 (55 %)
VEGF-C
 Negative
28 (48 %)
28 (52 %)
0.651
 Positive
78 (51 %)
76 (49 %)
Reg IV
 Negative
76 (49 %)
80 (51 %)
0.536
 Positive
30 (54 %)
24 (46 %)
Olfactomedin 4
 Negative
30 (42 %)
41 (58 %)
0.089
 Positive
76 (54 %)
63 (46 %)
Claudin-18
 Negative
37 (47 %)
39 (53 %)
0.485
 Positive
69 (52 %)
45 (48 %)
MMP-7
 Negative
64 (56 %)
48 (44 %)
0.058
 Positive
42 (43 %)
56 (57 %)
HER2
 Negative
87 (48 %)
96 (52 %)
0.073
 Positive
19 (66 %)
8 (34 %)

Prognostic analysis of OS and DFS in expression of the candidate molecular factors

In terms of OS for all gastric cancer cases (Stage II and III), cases with positive p53 expression had significantly worse prognosis (p = 0.02) when compared with p53-negative cases. Cases positive for MMP7 tended to have worse prognosis in comparison with the negative cases, although not significantly so (p = 0.09) (Fig. 2a). In terms of DFS, p53-positive and MMP7-positive cases had significantly worse prognosis (p = 0.006 and p = 0.04, respectively) (Fig. 2b). No significant difference in prognosis was observed between patients positive and negative for four other factors (VEGF-A, Reg IV, olfactomedin 4, and Claudin-18) (data not shown).
Figure 3 reveals prognostic evaluation (OS and DFS) in Stage II gastric cancer cases (n = 104). MMP7-positive cases suffered from significantly worse prognosis both in terms of OS (p = 0.027) and DFS (p = 0.014). In addition, VEGF-C positive cases had more favorable prognosis (p = 0.044 for OS and p = 0.048 for DFS) compared with VEGF-C negative cases. The similar analysis of OS and DFS revealed no significant difference in survival among Stage III patients (n = 106) for any of the eight factors.

Feasibility of the candidate factors for G-factor

According to the aforementioned results, we selected p53 and MMP-7 as G-factor that might serve our purpose. Consequently, we analyzed the association between the combination of p53 and MMP-7 expression and prognosis. All gastric cancer patients in the current series were stratified into three groups as follows, based on the expression of these molecules; G0 group (negative for both molecules, n = 69), G1 group (positive for either of the molecules, n = 97), and G2 group (positive for both of the molecules, n = 44). In all of the 210 gastric cancer cases, G2 cases demonstrated significantly higher recurrence rate (59 %) compared to 38 % in G0 cases (p = 0.047). In stage-by-stage analyses, the recurrence rate of G2 cases (50 %) tended to be higher in comparison with that of G0 and G1 cases for Stage II (25 and 20 %, respectively, p = 0.069), but not for Stage III (p = 0.414).
In the univariate analysis, G2 cases were associated with a shorter OS (hazard ratio 1.83, 95 % CI 1.15–2.90; p = 0.01) and DFS (hazard ratio 2.15, 95 % CI 1.26–3.68; p = 0.005) in comparison with G0 and G1 cases. The multivariate Cox’ regression analysis revealed that the G2 group was not an independent prognostic factor for OS (hazard ratio 1.59, 95 % CI 0.99–2.55; p = 0.052), but was independently associated with a shorter DFS (hazard ratio 1.90, 95 % CI 1.10–3.30; p = 0.022) (Table 3).
Table 3
Prognostic factors of overall survival and disease-free survival (univariate and multivariate analyses)
Variable
OS univariate analysis
OS multivariate analysis
DFS univariate analysis
DFS multivariate analysis
HR
95 % CI
p value
HR
95 % CI
p value
HR
95 % CI
p value
HR
95 % CI
p value
p53/MMP-7 expression
 G2 vs. G0/1
1.83
1.15–2.90
0.010
1.59
0.99–2.55
0.052
2.15
1.26–3.68
0.005
1.90
1.10–3.30
0.022
Depth of invasion
 T1, T2 vs. T3, T4
1.99
1.39–2.87
<0.001
2.40
1.65–3.51
<0.001
1.79
1.20–2.70
0.005
2.17
1.43–3.30
<0.001
Lymph node metastasis
 Positive vs. negative
2.61
1.22–4.82
0.011
3.68
1.75–7.70
0.001
2.86
1.32–6.21
0.008
4.33
1.85–10.15
0.001
Lymphatic invasion
 Positive vs. negative
2.61
1.36–5.03
0.004
1.89
0.95–3.76
0.068
4.41
1.79–10.91
0.001
2.92
1.15–7.41
0.024
Venous invasion
 Positive vs. negative
2.56
1.70–3.85
<0.001
1.90
1.23–2.93
0.004
2.77
1.77–4.34
<0.001
1.94
1.21–3.11
0.06
Cox’s regression analysis; HR hazard ratio; 95 % CI 95 % confidence interval
Survival analysis revealed that long term outcome of G0 cases was significantly better than that of G1 cases (p = 0.024 for OS and 0.019 for DFS) and G2 cases (p = 0.001 for OS and DFS) in all cases analyzed. In addition, G2 was particularly strong as marker of poor prognosis among Stage II cancer (p = 0.004 for comparison of OS with G0 and 0.029 for comparison with G1) (Fig. 4).

Discussion

The TNM staging system is widely used throughout the world to predict the prognosis of malignant tumors, in which staging is based on three clinicopathological factors: T factor, the degree of wall penetration of the primary tumor; N factor, the status of lymph node metastasis; and M factor, the presence of distant metastasis [1]. In the case of gastric cancer, 5-year survival rate of Stage I cancer is >90 %, whereas survivors rarely exist among patients with Stage IV cancer. However, the outcome of Stage II or III cases are varied, despite being treated by a similar strategy of radical surgery and adjuvant therapy, reflecting the fact that gastric cancer is a mixed population of cancers with various biologic characteristics that cannot be classified based only on conventional prognostic factors [1820]. The present study was therefore conducted through the collaboration of several institutions and the Japan Society for Gastroenterological Carcinogenesis, to investigate the feasibility of new molecular staging of a G-factor, which would supplement the conventional TNM staging system.
An extensive literature search was conducted during selection of candidate G-factors, and 50 prognostic factors were identified for gastric cancer, mostly from single institutions. However, no significant correlation with prognosis was observed in most of the molecules thus selected for the current validation set consisting of 210 gastric cancer cases with known prognosis, with the exception of p53. MMP7 was the only other molecule that was of interest, since it was associated with prognosis in the Stage II subset. Moreover, even with p53, the difference in the 5-year survival rate was <20 %. This difference did not meet the criterion generally suggested in resent published reports [411, 2124] that a prognostic difference of ≥20 % is required when predicting the prognosis, based on the biochemical characteristics of cancer, so as to supplement the conventional TNM stage classification. In other words, current study indicated that no single molecule could serve the role of G-factor as originally proposed.
Consequently, we decided to combine p53 and MMP-7 as a new marker of prognosis. Patients were stratified into three groups based on p53 and MMP-7 expression; G0 group (both negative group), G1 group (either positive group), and G2 group (both positive group). Difference in survival between G2 and G0 was shown to be quite significant, and G2 was an independent prognostic factor in terms of DFS and of borderline significance in terms of OS. In addition, a significant difference in OS and DFS was observed between G0/1 and G2 groups among the Stage II cancer subset. Furthermore, the OS and DFS of Stage II G2 cases were similar to those of Stage III cases, and were clearly distinct from other Stage II cancers. New clinical trials for Stage III gastric cancer are either currently ongoing or under preparation based on the fact that hazard ratio for the treatment group incrementally increased as the clinical stage advanced from II to IIIB in a randomized trial comparing adjuvant S-1 monotherapy versus surgery alone [25]. A novel classification with G-factor can now identify a high-risk group among Stage II cancers that may be eligible for these trials and may benefit from more intensive adjuvant treatments.
A number of new molecules or those hitherto unrelated to cancer have been identified in recent researches using various types of arrays. Further efforts to enhance the G2 factor or to find other relevant gene signatures are warranted to improve clinical staging schemes, and to ultimately establish an era of personalized therapy.

Conclusion

Through a multi-institutional effort led by the Japan Society for Gastroenterological Carcinogenesis, a combination of p53/MMP-7 expression was found to be a promising factor to predict outcome of Stage II/III gastric cancer, and possibly to help select the treatment for Stage II cancer, thus supplementing the conventional TNM system.

Acknowledgments

We thank Emeritus professor Eiichi Tahara (Hiroshima Cancer Seminar Foundation, Hiroshima, Japan) and Emeritus professor Michio Kaminishi (Showa General Hospital, Tokyo, Japan) for helpful advice. This study is partially founded by the Japan Society for Gastroenterological Carcinogenesis.

Conflict of interest

None of the authors have conflicts of interest to disclose.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Sobin LH. TNM, sixth edition: new developments in general concepts and rules. Semin Surg Oncol. 2003;21:19–22.PubMedCrossRef Sobin LH. TNM, sixth edition: new developments in general concepts and rules. Semin Surg Oncol. 2003;21:19–22.PubMedCrossRef
2.
Zurück zum Zitat Japanese Gastric Cancer A. Japanese classification of gastric carcinoma—2nd English edition. Gastric Cancer. 1998;1:10–24.CrossRef Japanese Gastric Cancer A. Japanese classification of gastric carcinoma—2nd English edition. Gastric Cancer. 1998;1:10–24.CrossRef
3.
Zurück zum Zitat Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, et al. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med. 2007;357:1810–20.PubMedCrossRef Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, et al. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med. 2007;357:1810–20.PubMedCrossRef
4.
Zurück zum Zitat Tanigawa N, Amaya H, Matsumura M, Shimomatsuya T. Correlation between expression of vascular endothelial growth factor and tumor vascularity, and patient outcome in human gastric carcinoma. J Clin Oncol. 1997;15:826–32.PubMed Tanigawa N, Amaya H, Matsumura M, Shimomatsuya T. Correlation between expression of vascular endothelial growth factor and tumor vascularity, and patient outcome in human gastric carcinoma. J Clin Oncol. 1997;15:826–32.PubMed
5.
Zurück zum Zitat Maeda K, Kang SM, Onoda N, Ogawa M, Sawada T, Nakata B, et al. Expression of p53 and vascular endothelial growth factor associated with tumor angiogenesis and prognosis in gastric cancer. Oncology. 1998;55:594–9.PubMedCrossRef Maeda K, Kang SM, Onoda N, Ogawa M, Sawada T, Nakata B, et al. Expression of p53 and vascular endothelial growth factor associated with tumor angiogenesis and prognosis in gastric cancer. Oncology. 1998;55:594–9.PubMedCrossRef
6.
Zurück zum Zitat Maehara Y, Tomoda M, Hasuda S, Kabashima A, Tokunaga E, Kakeji Y, et al. Prognostic value of p53 protein expression for patients with gastric cancer—a multivariate analysis. Br J Cancer. 1999;79:1255–61.PubMedCentralPubMedCrossRef Maehara Y, Tomoda M, Hasuda S, Kabashima A, Tokunaga E, Kakeji Y, et al. Prognostic value of p53 protein expression for patients with gastric cancer—a multivariate analysis. Br J Cancer. 1999;79:1255–61.PubMedCentralPubMedCrossRef
7.
Zurück zum Zitat Kakeji Y, Koga T, Sumiyoshi Y, Shibahara K, Oda S, Maehara Y, et al. Clinical significance of vascular endothelial growth factor expression in gastric cancer. J Exp Clin Cancer Res. 2002;21:125–9.PubMed Kakeji Y, Koga T, Sumiyoshi Y, Shibahara K, Oda S, Maehara Y, et al. Clinical significance of vascular endothelial growth factor expression in gastric cancer. J Exp Clin Cancer Res. 2002;21:125–9.PubMed
8.
Zurück zum Zitat Fondevila C, Metges JP, Fuster J, Grau JJ, Palacin A, Castells A, et al. p53 and VEGF expression are independent predictors of tumour recurrence and survival following curative resection of gastric cancer. Br J Cancer. 2004;90:206–15.PubMedCentralPubMedCrossRef Fondevila C, Metges JP, Fuster J, Grau JJ, Palacin A, Castells A, et al. p53 and VEGF expression are independent predictors of tumour recurrence and survival following curative resection of gastric cancer. Br J Cancer. 2004;90:206–15.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Resnick MB, Routhier J, Konkin T, Sabo E, Pricolo VE. Epidermal growth factor receptor, c-MET, beta-catenin, and p53 expression as prognostic indicators in stage II colon cancer: a tissue microarray study. Clin Cancer Res. 2004;10:3069–75.PubMedCrossRef Resnick MB, Routhier J, Konkin T, Sabo E, Pricolo VE. Epidermal growth factor receptor, c-MET, beta-catenin, and p53 expression as prognostic indicators in stage II colon cancer: a tissue microarray study. Clin Cancer Res. 2004;10:3069–75.PubMedCrossRef
10.
Zurück zum Zitat White JD, Hewett PW, Kosuge D, McCulloch T, Enholm BC, Carmichael J, et al. Vascular endothelial growth factor-D expression is an independent prognostic marker for survival in colorectal carcinoma. Cancer Res. 2002;62:1669–75.PubMed White JD, Hewett PW, Kosuge D, McCulloch T, Enholm BC, Carmichael J, et al. Vascular endothelial growth factor-D expression is an independent prognostic marker for survival in colorectal carcinoma. Cancer Res. 2002;62:1669–75.PubMed
11.
Zurück zum Zitat Des Guetz G, Uzzan B, Nicolas P, Cucherat M, Morere JF, Benamouzig R, et al. Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br J Cancer. 2006;94:1823–32.PubMedCentralPubMedCrossRef Des Guetz G, Uzzan B, Nicolas P, Cucherat M, Morere JF, Benamouzig R, et al. Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br J Cancer. 2006;94:1823–32.PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Ajisaka H, Yonemura Y, Miwa K. Correlation of lymph node metastases and expression of matrix metalloproteinase-7 in patients with gastric cancer. Hepatogastroenterology. 2004;51:900–5.PubMed Ajisaka H, Yonemura Y, Miwa K. Correlation of lymph node metastases and expression of matrix metalloproteinase-7 in patients with gastric cancer. Hepatogastroenterology. 2004;51:900–5.PubMed
13.
Zurück zum Zitat Ross JS, McKenna BJ. The HER-2/neu oncogene in tumors of the gastrointestinal tract. Cancer Investig. 2001;19:554–68.CrossRef Ross JS, McKenna BJ. The HER-2/neu oncogene in tumors of the gastrointestinal tract. Cancer Investig. 2001;19:554–68.CrossRef
14.
Zurück zum Zitat Oue N, Kuniyasu H, Noguchi T, Sentani K, Ito M, Tanaka S, et al. Serum concentration of Reg IV in patients with colorectal cancer: overexpression and high serum levels of Reg IV are associated with liver metastasis. Oncology. 2007;72:371–80.PubMedCrossRef Oue N, Kuniyasu H, Noguchi T, Sentani K, Ito M, Tanaka S, et al. Serum concentration of Reg IV in patients with colorectal cancer: overexpression and high serum levels of Reg IV are associated with liver metastasis. Oncology. 2007;72:371–80.PubMedCrossRef
15.
Zurück zum Zitat Oue N, Mitani Y, Aung PP, Sakakura C, Takeshima Y, Kaneko M, et al. Expression and localization of Reg IV in human neoplastic and non-neoplastic tissues: Reg IV expression is associated with intestinal and neuroendocrine differentiation in gastric adenocarcinoma. J Pathol. 2005;207:185–98.PubMedCrossRef Oue N, Mitani Y, Aung PP, Sakakura C, Takeshima Y, Kaneko M, et al. Expression and localization of Reg IV in human neoplastic and non-neoplastic tissues: Reg IV expression is associated with intestinal and neuroendocrine differentiation in gastric adenocarcinoma. J Pathol. 2005;207:185–98.PubMedCrossRef
16.
Zurück zum Zitat Oue N, Sentani K, Noguchi T, Ohara S, Sakamoto N, Hayashi T, et al. Serum olfactomedin 4 (GW112, hGC-1) in combination with Reg IV is a highly sensitive biomarker for gastric cancer patients. Int J Cancer. 2009;125:2383–92.PubMedCrossRef Oue N, Sentani K, Noguchi T, Ohara S, Sakamoto N, Hayashi T, et al. Serum olfactomedin 4 (GW112, hGC-1) in combination with Reg IV is a highly sensitive biomarker for gastric cancer patients. Int J Cancer. 2009;125:2383–92.PubMedCrossRef
17.
Zurück zum Zitat Sanada Y, Oue N, Mitani Y, Yoshida K, Nakayama H, Yasui W. Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J Pathol. 2006;208:633–42.PubMedCrossRef Sanada Y, Oue N, Mitani Y, Yoshida K, Nakayama H, Yasui W. Down-regulation of the claudin-18 gene, identified through serial analysis of gene expression data analysis, in gastric cancer with an intestinal phenotype. J Pathol. 2006;208:633–42.PubMedCrossRef
18.
19.
Zurück zum Zitat Zhang YJ, Fang JY. Molecular staging of gastric cancer. J Gastroenterol Hepatol. 2008;23:856–60.PubMedCrossRef Zhang YJ, Fang JY. Molecular staging of gastric cancer. J Gastroenterol Hepatol. 2008;23:856–60.PubMedCrossRef
20.
Zurück zum Zitat Eschrich S, Yang I, Bloom G, Kwong KY, Boulware D, Cantor A, et al. Molecular staging for survival prediction of colorectal cancer patients. J Clin Oncol. 2005;23:3526–35.PubMedCrossRef Eschrich S, Yang I, Bloom G, Kwong KY, Boulware D, Cantor A, et al. Molecular staging for survival prediction of colorectal cancer patients. J Clin Oncol. 2005;23:3526–35.PubMedCrossRef
21.
Zurück zum Zitat Siemer S, Lehmann J, Loch A, Becker F, Stein U, Schneider G, et al. Current TNM classification of renal cell carcinoma evaluated: revising stage T3a. J Urol. 2005;173:33–7.PubMedCrossRef Siemer S, Lehmann J, Loch A, Becker F, Stein U, Schneider G, et al. Current TNM classification of renal cell carcinoma evaluated: revising stage T3a. J Urol. 2005;173:33–7.PubMedCrossRef
22.
Zurück zum Zitat Goldstraw P. New staging system: how does it affect our practice? J Clin Oncol. 2013;31:984–91.PubMedCrossRef Goldstraw P. New staging system: how does it affect our practice? J Clin Oncol. 2013;31:984–91.PubMedCrossRef
23.
Zurück zum Zitat Yoon HM, Ryu KW, Nam BH, Cho SJ, Park SR, Lee JY, et al. Is the new seventh AJCC/UICC staging system appropriate for patients with gastric cancer? J Am Coll Surg. 2012;214:88–96.PubMedCrossRef Yoon HM, Ryu KW, Nam BH, Cho SJ, Park SR, Lee JY, et al. Is the new seventh AJCC/UICC staging system appropriate for patients with gastric cancer? J Am Coll Surg. 2012;214:88–96.PubMedCrossRef
24.
Zurück zum Zitat Kologlu M, Kama NA, Reis E, Doganay M, Atli M, Dolapci M. A prognostic score for gastric cancer. Am J Surg. 2000;179:521–6.PubMedCrossRef Kologlu M, Kama NA, Reis E, Doganay M, Atli M, Dolapci M. A prognostic score for gastric cancer. Am J Surg. 2000;179:521–6.PubMedCrossRef
25.
Zurück zum Zitat Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, et al. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29:4387–93.PubMedCrossRef Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, et al. Five-year outcomes of a randomized phase III trial comparing adjuvant chemotherapy with S-1 versus surgery alone in stage II or III gastric cancer. J Clin Oncol. 2011;29:4387–93.PubMedCrossRef
Metadaten
Titel
New molecular staging with G-factor supplements TNM classification in gastric cancer: a multicenter collaborative research by the Japan Society for Gastroenterological Carcinogenesis G-Project committee
verfasst von
Tetsuji Sawada
Masakazu Yashiro
Kazuhiro Sentani
Naohide Oue
Wataru Yasui
Kohji Miyazaki
Keita Kai
Sachio Fushida
Takashi Fujimura
Masaichi Ohira
Yoshihiro Kakeji
Shoji Natsugoe
Ken Shirabe
Sachiyo Nomura
Yutaka Shimada
Naohiro Tomita
Kosei Hirakawa
Yoshihiko Maehara
Publikationsdatum
01.01.2015
Verlag
Springer Japan
Erschienen in
Gastric Cancer / Ausgabe 1/2015
Print ISSN: 1436-3291
Elektronische ISSN: 1436-3305
DOI
https://doi.org/10.1007/s10120-014-0338-2

Weitere Artikel der Ausgabe 1/2015

Gastric Cancer 1/2015 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.