Skip to main content
Erschienen in: BMC Cancer 1/2018

Open Access 01.12.2018 | Research article

Venous invasion as a risk factor for recurrence after gastrectomy followed by chemotherapy for stage III gastric cancer

verfasst von: Keiji Nishibeppu, Shuhei Komatsu, Daisuke Ichikawa, Taisuke Imamura, Toshiyuki Kosuga, Kazuma Okamoto, Hirotaka Konishi, Atsushi Shiozaki, Hitoshi Fujiwara, Eigo Otsuji

Erschienen in: BMC Cancer | Ausgabe 1/2018

Abstract

Background

Although adjuvant chemotherapy with S-1 after curative gastrectomy has been performed as a standard treatment for Stage II and III gastric cancer (GC) in Japan, patients with Stage III GC still have a high incidence of recurrence and a poor prognostic outcome. The aim of this study was to investigate risk factors for recurrence in patients with Stage III GC despite of curative gastrectomy followed by adjuvant chemotherapy, suggesting an indicator for more intensive management.

Methods

A total of 97 patients with pathological Stage III GC underwent adjuvant chemotherapy after curative gastrectomy between 2001 and 2014, were enrolled in this study. We retrospectively analyzed their hospital records from our hospital.

Results

The 5-year relapse-free survival (RFS) rates of patients with pStage III GC were 42.0%. Univariate and multivariate analyses for RFS revealed that venous invasion (v+) was an independent factor predicting a shorter RFS (v + vs. v-, 36.5% vs. 47.4%, P = 0.034, HR 1.82, 95% CI: 1.01–3.37). Venous invasion also predicted a shorter overall survival (OS) (v + vs. v-, 33.7% vs. 50.4%, P = 0.027). Regarding the patterns of recurrence, hematogenous recurrence was significantly occurred in patients with v + GC than those without (P = 0.022).

Conclusions

Stage III GC with venous invasion is a high-risk subgroup for hematogenous recurrence after curative surgery followed by adjuvant chemotherapy. More intensive and effective adjuvant chemo and/or molecular targeted therapy for Stage III GC patients with venous invasion should be considered to improve their outcomes.

Background

Gastric cancer (GC) is a major cause of death worldwide [1]. Treatments for GC have certainly improved with recent advances in surgical procedures and adjuvant chemotherapy [25]. However, treatment of the primary tumor and regional lymph nodes is recognized as the only chance for macroscopic tumor clearance and cure for GC; the effects of such surgical resection is restricted to locally controlling the primary tumor [2, 3], and cannot prevent recurrence due to micro-metastasis, which could be residual and/or occur at the time of surgery [6, 7]. Therefore, perioperative systemic chemotherapy has been recommended to achieve microscopic tumor clearance.
Cunningham et al. [8] demonstrated that perioperative chemotherapy with a regimen of epirubicin, cisplatin, and fluorouracil improves overall survival (OS) and relapse-free survival (RFS) among patients with resectable adenocarcinoma of the stomach as compared with surgery alone. Intergroup 0116 demonstrated a notable benefit in OS and RFS by performing adjuvant chemo-radiotherapy following curative GC resection in patients with ≥ T3 and/or node-positive GC [9, 10]. In Japan, adjuvant chemotherapy following curative gastrectomy for stage II or III GC has been recommended as a standard treatment, based on the result of the ACTS-GC (Adjuvant Chemotherapy Trial of TS-1 for Gastric Cancer) [11, 12]. The ACTS-GC demonstrated the overall survival benefit (HR 0.67, 95% CI: 0.54–0.83) of adjuvant chemotherapy with S-1 in patients with stage II or III GC. With the proven survival benefit of perioperative adjuvant chemotherapy, it is now globally used [13].
According to the results of the ACTS-GC trial, the five-year OS rate of patients with stage II GC was 84.2%, whereas the five-year OS rate in stage IIIA and IIIB patients was only 57.3% and 44.1%, respectively [12]. Therefore, the efficacy of S-1 may be limited to stage II GC, with a need to improve the prognostic markers in stage III GC patients after combined curative gastrectomy and adjuvant S-1 chemotherapy in Japan. Several studies have recently been conducted to investigate the safety and efficacy of a more intensive combination chemotherapy as adjuvant therapy [1416]. To validate these more intensive adjuvant chemo- and/or molecular-targeted therapies, stage III GC patients, with a high-risk factor for recurrence following adjuvant chemotherapy, need to be identified. In this study, to promote more intensive adjuvant treatment, we aimed to investigate surrogate pathologic factors for recurrence in stage III GC after curative gastrectomy followed by adjuvant chemotherapy.

Methods

Patients and surgical procedures

The study was approved by the Kyoto Prefectural University of Medicine and have therefore been performed in accordance with the ethical standards laid down in an appropriate version of the Declaration of Helsinki. Written informed consent was obtained from all patients for research purposes.
A total of 127 patients underwent curative gastrectomies, followed by adjuvant chemotherapy for pathological stage III GC at our institute between January 2001 and December 2014. All of the enrolled patients had undergone D2 or D2+ lymphadenectomies. In the D2 dissection, the perigastric lymph nodes and all second-tier lymph nodes were completely retrieved. Depending on the location of the tumor, the lymphadenectomy was done along the distal side of the splenic artery (No.11d) and at the splenic hilum (No.10), together with a splenectomy, or splenectomy with a distal pancreatectomy [17]. Enrolled patients also underwent macroscopic and pathologically curative resection (R0), and had negative results for peritoneal washing cytology.
Of these 127 patients, 30 patients were excluded from the study because of neoadjuvant chemotherapy (n = 28) and insufficient follow-up (n = 2). Consequently, a total of 97 patients were enrolled in this study. Resected specimens were examined by pathologists, and evaluated based on classifications of the 14th JCGC and 7th AJCC-UICC staging systems. All dissected lymph nodes were fixed in buffered formalin and embedded in paraffin and subjected to pathological examination. Pathologists in our institution examined embedded lymph nodes by sectioning slices in the plane of the largest node dimension to confirm the presence of metastasis. The clinicopathological findings of these patients were determined retrospectively on the basis of their hospital records.

Follow-up after curative gastrectomy followed by adjuvant chemotherapy

Post-operative follow-ups were performed every three months after surgery in the outpatient clinic. Blood chemistry was measured every three months. Endoscopic examinations were performed annually, and computed tomography (CT) examinations were performed every three-to-six months for five years after surgery.

Statistical analysis

The Chi-square test and Fisher’s exact probability test were used to analyze categorical variables to compare the clinicopathological characteristics between the two groups. For the analysis of survival, Kaplan-Meier survival curves were constructed for groups based on univariate predictors, and differences between the groups were tested with a generalized Wilcoxon test. The Cox proportional hazards model was used for further evaluations of multivariate survival analysis. A p-value < 0.05 was considered significant. Statistical analyses were conducted using JMP 10 (SAS Institute Inc., Cary, NC).

Results

Clinicopathological characteristics of stage III GC patients after curative gastrectomy followed by adjuvant chemotherapy

Table 1 shows the characteristics and treatment of patients with stage III GC in this study. The median age was 65 years old. Of these, 61 patients (63%) were male and 36 patients (37%) were female. Total gastrectomy was performed in 53 patients (55%) and distal gastrectomy in 44 patients (45%) as curative resection according to the location of the tumor to secure a sufficient resection margin. Of 97 stage III GC patients, 41 patients (42%) received S-1 alone, 12 patients (12%) received tegafur-uracil, 12 patients (12%) received methotrexate plus 5-fluorouracil, 14 patients (14%) received S-1 plus cisplatin, 5 patients (5%) received 5-fluorouracil alone, 7 patients (7%) received S-1 plus paclitaxel, 5 patients (5%) received 5-fluorouracil plus cisplatin and 1 patient (1%) received paclitaxel alone as adjuvant chemotherapy. None of the patients received adjuvant radiotherapy or chemo-radiotherapy.
Table 1
Clinical characteristics and treatment of stage III GC patients
Variables
Number
Percent
Total
97
 
Gender
 Male
61
63%
 Female
36
37%
Age
  ≥65
51
53%
  <65
46
47%
Tumor location
 U
19
20%
 M
28
29%
 L
32
33%
 Whole
18
19%
Tumor major axis(mm)
  ≥90
33
34%
  <90
64
66%
T-stage
 T2 / T3
27
28%
 T4
70
72%
N-stage
 N1/ N2
47
48%
 N3
50
52%
Histopathological type
 Differentiated
31
32%
 Undifferentiated
66
68%
Venous invasion
 Present
49
51%
 Absent
48
49%
Lymphatic invasion
 Present
87
90%
 Absent
10
10%
Surgical procedure
 Total gastrectomy
53
55%
 Distal gastrectomy
44
45%

Clinical outcomes and prognostic factors for relapse-free survival of stage III GC patients after curative gastrectomy followed by adjuvant chemotherapy

The average observation period was 43.9 months. The cumulative five-year RFS and OS rates in the 97 patients with stage III GC were 42.0%, and 42.6%, respectively. Univariate analysis revealed venous invasion (P = 0.034) as a prognostic factor for RFS after curative gastrectomy followed by adjuvant chemotherapy. Multivariate analysis using the Cox proportional hazard model demonstrated that venous invasion was the only independent factor predicting a shorter RFS in stage III GC (P = 0.048, HR 1.82, 95% CI: 1.01–3.37) (Table 2). Fig. 1 shows the RFS curves according to GC patients with or without venous invasion. A significant difference was observed between patients with and without venous invasion (Figs. 1, 5-year RFS rates, venous invasion + vs. venous invasion -; 36.5% vs. 47.4%, P = 0.034). Furthermore, GC patients with venous invasion exhibited a shorter OS than those with without GC (Figs. 2, 5-year OS rates, venous invasion + vs. venous invasion -; 33.7% vs. 50.4%, P = 0.027). Additional file 1: Figure S1 shows the RFS and OS curves for GC patients after curative gastrectomy followed by S-1 treatment alone with or without venous invasion. No significant difference was observed between patients with or without venous invasion (Additionral file 1: Figure S1a, 5-year RFS rates, venous invasion + vs. venous invasion -, 41.8% vs. 41.5%, P = 0.089; Additional file 2: Figure S1b, 5-year OS rates, venous invasion + vs. venous invasion -, 36.4% vs. 58.0%, P = 0.163). However, the prognoses of GC patients after curative gastrectomy followed by S-1 treatment alone with venous invasion tended to be poorer than those of patients without venous invasion.
Table 2
Univariate and multivariate analysis for relapse-free survival (RFS) in stage III GC patients after curative gastrectomy followed by adjuvant chemotherapy
 
Univariatea
Multivariateb
 
N
5-year RFSc
P-value
HRd
95% CIe
P-value
Sex
Male
61
46.1%
0.577
1.20
0.63–2.31
0.581
 
Female
36
36.0%
    
Age
<65
46
42.2%
0.824
1.39
0.76–2.53
0.285
 
≥65
51
41.9%
    
Tumor major axis(mm)
≥90
33
30.8%
0.092
1.46
0.71–3.21
0.109
 
<90
64
48.8%
    
T-stage
T4
70
38.9%
0.326
1.46
0.79–3.63
0.311
 
T2/ T3
27
50.1%
    
N-stage
N3
50
33.4%
0.339
1.51
0.82–2.84
0.187
 
N1/ N2
47
50.9%
    
Venous invasion
Present
48
36.5%
0.034
1.82
1.01–3.37
0.048
 
Absent
49
47.4%
    
Lymphatic invasion
Present
87
39.8%
0.168
1.51
0.56–5.28
0.445
 
Absent
10
40.0%
    
aKaplan-Meier method: significance was determined by Wilcoxon test
bMultivariate survival analysis was performed using Cox’s proportional hazard model
cRFS Relative free survival
dHR Hazard ratio
eCI Confidence interval
Significant values are in bold

Comparison of recurrence patterns according to stage III GC patients with or without venous invasion

Table 3 shows postoperative adjuvant treatments according to the status of venous invasion in stage III GC patients. There was no significant difference between patients with or without venous invasion in the regimens of adjuvant chemotherapy. Regarding the patterns of recurrence, the incidence of hematogenous recurrence was significantly higher in patients with venous invasion than in those without (venous invasion + vs. venous invasion -: 10% vs. 2%, P = 0.022; Table 4).
Table 3
The regimens of adjuvant chemotherapy
  
Venous invasion
 
 
N
Present
Absent
P-value
Regimen
     
 
S-1
41
23(24%)
18(19%)
0.346
S-1 plus cisplatin
14
10(10%)
4(4%)
0.086
5-FU plus methotrexate
12
4(4%)
8(8%)
0.200
UFT
12
5(5%)
7(7%)
0.511
S-1 plus paclitaxel
7
2(2%)
5(5%)
0.221
5-FU plus cisplatin
5
3(3%)
2(2%)
0.662
5-FU
5
1(1%)
4(4%)
0.148
Paclitaxel
1
1(1%)
0(0%)
0.241
Table 4
Comparison of recurrence patterns according to stage III GC patients with or without venous invasion
 
Venous invasion
Univariatea
 
N
Present
Absent
P-value
Total
97
49 (51%)
48 (49%)
0.615
Hematogenous recurrence
12
10 (10%)
2 (2%)
0.022
 
Liver
8
7 (7%)
1 (1%)
0.021
Lung
2
2 (2%)
0 (0%)
0.096
Bone
2
1 (1%)
1 (1%)
0.988
Lymphatic recurrence
12
7 (7%)
5 (5%)
0.562
Peritoneal recurrence
34
18 (19%)
16 (16%)
0.726
Local recurrence
4
1 (1%)
3 (3%)
0.287
aKaplan-Meier method: significance was determined by the Wilcoxon test
Significant values are in bold

Discussion

This study demonstrated that stage III GC patients with venous invasion have a markedly poor prognosis despite curative gastrectomy followed by adjuvant chemotherapy. Furthermore, this study clearly identified stage III GC patients with venous invasion as a high-risk subgroup for hematogenous recurrence. These results strongly indicate that GC with venous invasion could be specifically targeted in an effort to improve the prognosis of patients with stage III GC, suggesting an indication for more intensive adjuvant chemo- and/or molecular targeted therapy.
Based on the results of the ACTS-GC trial for stage II or III GC, conventional adjuvant chemotherapy with S-1 has the potential to reduce the incidence of peritoneal and lymphatic recurrences. However, this trial also indicated the limitations of the inhibitory effects of S-1 on hematogenous recurrence [11], particularly in stage III GC. To further improve the prognostic outcomes, the prevention of hematogenous recurrence after surgery should be a pivotal treatment target in advanced GC patients. Hematogenous metastasis results when cancer cells derived from the primary lesion enter blood vessels and are transported to distant organs where they can proliferate and form secondary tumors. This leads to hematogenous recurrence. Several previous reports demonstrated that vascular invasion in resected specimens was a risk factor for hematogenous recurrence and a poor prognostic factor in GC [18, 19]. These results strongly support our results that GC with venous invasion is a high-risk subgroup for hematogenous recurrence after curative gastrectomy followed by adjuvant chemotherapy. Thus, we suggest that venous invasion is the surrogate biomarker for an indication of more intensive chemo- and/or molecular-targeted therapy in order to prevent hematogenous recurrence.
Recently, several promising studies have been conducted to investigate the efficacy of combination chemotherapy as a more intensive adjuvant therapy for stage III GC [1416, 20, 21]. Of these, the results of the CLASSIC trial indicated that adjuvant treatment with capecitabine plus oxaliplatin may have the potential to reduce the incidence of hematogenous recurrence [22]. Currently, this regimen is already included in standard adjuvant chemotherapy for stage II and III GC in Japan. Furthermore, the efficacy of molecular targeted drugs against tumor angiogenesis, which is essential for hematogenous metastasis, has been demonstrated [2325]. Trastuzumab in combination with chemotherapy for patients with HER2-positive advanced gastric or gastro-esophageal junction cancer was proven to be more effective for measurable tumors such as hematogenous recurrence [23]. Ohtsu et al. [24] demonstrated that adding bevacizumab, a monoclonal antibody targeting VEGF-A, to fluoropyrimidine-cisplatin improved progression-free survival and overall response rate in the first-line treatment of advanced GC. The Rainbow trial indicated that adding ramucirumab, a monoclonal antibody VEGFR-2 antagonist, to paclitaxel improved OS in 665 GC patients (median 9.6 months vs 7.4 months stratified) (P = 0.017, HR 0.81, 95% CI: 0.68–0.96) [25]. These treatment strategies for advanced GC may become treatment candidates as more intensive adjuvant therapies for stage III GC with venous invasion.
This study is limited because the results were obtained from a retrospective evaluation with a small number of patients and inconsistent treatments at a single institute. A large-scale or multicenter prospective cohort study is warranted to validate the significance of the strategy.

Conclusion

We demonstrated that stage III GC with venous invasion presented a worse prognosis and higher rate of hematogenous recurrence despite adjuvant chemotherapy. Stage III GC with venous invasion could be specifically targeted in an effort to improve the prognosis with stage III GC, suggesting an indication for additional adjuvant chemotherapy.

Acknowledgements

None.

Funding

There is no funding to be declared.

Availability of data and materials

The datasets used during the current study are available from the corresponding author on reasonable request.
This study was designed in accordance with the Declaration of Helsinki and was approved by the Institutional Review Board of Kyoto Prefectural University of Medicine. All patients received sufficient explanation of the study, and written informed consent was obtained.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
2.
Zurück zum Zitat Gunderson LL. Gastric cancer—patterns of relapse after surgical resection. Semin Radiat Oncol. 2002;12:150–61.CrossRefPubMed Gunderson LL. Gastric cancer—patterns of relapse after surgical resection. Semin Radiat Oncol. 2002;12:150–61.CrossRefPubMed
4.
Zurück zum Zitat Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893–917.CrossRefPubMed Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893–917.CrossRefPubMed
5.
Zurück zum Zitat Paoletti X, Oba K, Burzykowski T, Michiels S, Ohashi Y, Pignon JP, et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA. 2010;303:1729–37.CrossRefPubMed Paoletti X, Oba K, Burzykowski T, Michiels S, Ohashi Y, Pignon JP, et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA. 2010;303:1729–37.CrossRefPubMed
6.
Zurück zum Zitat Iwatsuki M, Mimori K, Fukagawa T, Ishii H, Yokobori T, Sasako M, et al. The clinical significance of vimentin-expressing gastric cancer cells in bone marrow. Ann Surg Oncol. 2010;17:2526–33.CrossRefPubMed Iwatsuki M, Mimori K, Fukagawa T, Ishii H, Yokobori T, Sasako M, et al. The clinical significance of vimentin-expressing gastric cancer cells in bone marrow. Ann Surg Oncol. 2010;17:2526–33.CrossRefPubMed
7.
Zurück zum Zitat Kurashige J, Kamohara H, Watanabe M, Hiyoshi Y, Iwatsuki M, Tanaka Y, et al. MicroRNA-200b regulates cell proliferation, invasion, and migration by directly targeting ZEB2 in gastric carcinoma. Ann Surg Oncol. 2012;19:656–64.CrossRef Kurashige J, Kamohara H, Watanabe M, Hiyoshi Y, Iwatsuki M, Tanaka Y, et al. MicroRNA-200b regulates cell proliferation, invasion, and migration by directly targeting ZEB2 in gastric carcinoma. Ann Surg Oncol. 2012;19:656–64.CrossRef
8.
Zurück zum Zitat Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed
9.
Zurück zum Zitat Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–30.CrossRefPubMed Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725–30.CrossRefPubMed
10.
Zurück zum Zitat Smalley SR, Benedetti JK, Haller DG, Hundahl SA, Estes NC, Ajani JA, et al. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30:2327–33.CrossRefPubMedPubMedCentral Smalley SR, Benedetti JK, Haller DG, Hundahl SA, Estes NC, Ajani JA, et al. Updated analysis of SWOG-directed intergroup study 0116: a phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J Clin Oncol. 2012;30:2327–33.CrossRefPubMedPubMedCentral
11.
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.CrossRefPubMed 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.CrossRefPubMed
12.
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.CrossRefPubMed 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.CrossRefPubMed
13.
Zurück zum Zitat Imamura T, Komatsu S, Ichikawa D, Kubota T, Okamoto K, Konishi H, et al. Poor prognostic subgroup in T3N0 stage IIA gastric cancer, suggesting an indication for adjuvant chemotherapy. J Surg Oncol. 2015;111:221–5.CrossRefPubMed Imamura T, Komatsu S, Ichikawa D, Kubota T, Okamoto K, Konishi H, et al. Poor prognostic subgroup in T3N0 stage IIA gastric cancer, suggesting an indication for adjuvant chemotherapy. J Surg Oncol. 2015;111:221–5.CrossRefPubMed
14.
Zurück zum Zitat Bang Y-J, Kim Y-W, Yang H-K, Chung HC, Park Y-K, Lee KH, et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379:315–21.CrossRefPubMed Bang Y-J, Kim Y-W, Yang H-K, Chung HC, Park Y-K, Lee KH, et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379:315–21.CrossRefPubMed
15.
Zurück zum Zitat Shitara K, Chin K, Yoshikawa T, Katai H, Terashima M, Ito S, et al. Phase II study of adjuvant chemotherapy of S-1 plus oxaliplatin for patients with stage III gastric cancer after D2 gastrectomy. Gastric Cancer. 2017;20:175–81.CrossRefPubMed Shitara K, Chin K, Yoshikawa T, Katai H, Terashima M, Ito S, et al. Phase II study of adjuvant chemotherapy of S-1 plus oxaliplatin for patients with stage III gastric cancer after D2 gastrectomy. Gastric Cancer. 2017;20:175–81.CrossRefPubMed
16.
Zurück zum Zitat Fuse N, Bando H, Chin K, Ito S, Yoshikawa T, Tsuburaya A, et al. Adjuvant capecitabine plus oxaliplatin after D2 gastrectomy in Japanese patients with gastric cancer: a phase II study. Gastric Cancer. 2017;20:332–40.CrossRefPubMed Fuse N, Bando H, Chin K, Ito S, Yoshikawa T, Tsuburaya A, et al. Adjuvant capecitabine plus oxaliplatin after D2 gastrectomy in Japanese patients with gastric cancer: a phase II study. Gastric Cancer. 2017;20:332–40.CrossRefPubMed
17.
Zurück zum Zitat Komatsu S, Ichikawa D, Miyamae M, Kosuga T, Okamoto K, Arita T, et al. Positive lymph node ratio as an indicator of prognosis and local tumor clearance in N3 gastric cancer. J Gastrointest Surg. 2016;20:1565–71.CrossRefPubMed Komatsu S, Ichikawa D, Miyamae M, Kosuga T, Okamoto K, Arita T, et al. Positive lymph node ratio as an indicator of prognosis and local tumor clearance in N3 gastric cancer. J Gastrointest Surg. 2016;20:1565–71.CrossRefPubMed
18.
Zurück zum Zitat Maehara Y, Kabashima A, Koga T, Tokunaga E, Takeuchi H, Kakeji Y, et al. Vascular invasion and potential for tumor angiogenesis and metastasis in gastric carcinoma. Surgery. 2000;128:408–16.CrossRefPubMed Maehara Y, Kabashima A, Koga T, Tokunaga E, Takeuchi H, Kakeji Y, et al. Vascular invasion and potential for tumor angiogenesis and metastasis in gastric carcinoma. Surgery. 2000;128:408–16.CrossRefPubMed
19.
Zurück zum Zitat Nakanishi Y, Ohara M, Domen H, Shichinohe T, Hirano S, Ishizaka M. Differences in risk factors between patterns of recurrence in patients after curative resection for advanced gastric carcinoma. World J Surg Oncol. 2013;11:98.CrossRefPubMedPubMedCentral Nakanishi Y, Ohara M, Domen H, Shichinohe T, Hirano S, Ishizaka M. Differences in risk factors between patterns of recurrence in patients after curative resection for advanced gastric carcinoma. World J Surg Oncol. 2013;11:98.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Takahari D, Hamaguchi T, Yoshimura K, Katai H, Ito S, Fuse N, et al. Survival analysis of adjuvant chemotherapy with S-1 plus cisplatin for stage III gastric cancer. Gastric Cancer. 2014;17:383–6.CrossRefPubMed Takahari D, Hamaguchi T, Yoshimura K, Katai H, Ito S, Fuse N, et al. Survival analysis of adjuvant chemotherapy with S-1 plus cisplatin for stage III gastric cancer. Gastric Cancer. 2014;17:383–6.CrossRefPubMed
21.
Zurück zum Zitat Fujitani K, Tamura S, Kimura Y, Tsuji T, Matsuyama J, Iijima S, et al. Three-year outcomes of a phase II study of adjuvant chemotherapy with S-1 plus docetaxel for stage III gastric cancer after curative D2 gastrectomy. Gastric Cancer. 2014;17:348–53.CrossRefPubMed Fujitani K, Tamura S, Kimura Y, Tsuji T, Matsuyama J, Iijima S, et al. Three-year outcomes of a phase II study of adjuvant chemotherapy with S-1 plus docetaxel for stage III gastric cancer after curative D2 gastrectomy. Gastric Cancer. 2014;17:348–53.CrossRefPubMed
22.
Zurück zum Zitat Noh SH, Park SR, Yang H-K, Chung HC, Chung I-J, Kim S-W, et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:1389–96.CrossRefPubMed Noh SH, Park SR, Yang H-K, Chung HC, Chung I-J, Kim S-W, et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:1389–96.CrossRefPubMed
23.
Zurück zum Zitat Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, 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;376:687–97.CrossRefPubMed Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, 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;376:687–97.CrossRefPubMed
24.
Zurück zum Zitat Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol. 2011;29:3968–76.CrossRefPubMed Ohtsu A, Shah MA, Van Cutsem E, Rha SY, Sawaki A, Park SR, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol. 2011;29:3968–76.CrossRefPubMed
25.
Zurück zum Zitat Wilke H, Muro K, Van Cutsem E, Oh S-C, Bodoky G, Shimada Y, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224–35.CrossRefPubMed Wilke H, Muro K, Van Cutsem E, Oh S-C, Bodoky G, Shimada Y, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224–35.CrossRefPubMed
Metadaten
Titel
Venous invasion as a risk factor for recurrence after gastrectomy followed by chemotherapy for stage III gastric cancer
verfasst von
Keiji Nishibeppu
Shuhei Komatsu
Daisuke Ichikawa
Taisuke Imamura
Toshiyuki Kosuga
Kazuma Okamoto
Hirotaka Konishi
Atsushi Shiozaki
Hitoshi Fujiwara
Eigo Otsuji
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2018
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4052-z

Weitere Artikel der Ausgabe 1/2018

BMC Cancer 1/2018 Zur Ausgabe

Update Onkologie

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