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

Open Access 01.12.2018 | Research article

Effect of neoadjuvant chemotherapy in patients with gastric cancer: a PRISMA-compliant systematic review and meta-analysis

verfasst von: Zhi-Feng Miao, Xing-Yu Liu, Zhen-Ning Wang, Ting-Ting Zhao, Ying-Ying Xu, Yong-Xi Song, Jin-Yu Huang, Hao Xu, Hui-Mian Xu

Erschienen in: BMC Cancer | Ausgabe 1/2018

Abstract

Background

Neoadjuvant chemotherapy (NAC) is extensively used in the treatment of patients with gastric cancer (GC), particularly in high risk, advanced gastric cancer. Previous trials testing the efficacy of NAC have reported inconsistent results.

Methods

This study compares the combined use of NAC and surgery with surgery alone for GC by using a meta-analytic approach. We performed an electronic search of PubMed, EmBase, and the Cochrane Library to identify randomized controlled trials (RCTs) on NAC published before Oct 2015. The primary outcome of the studies was data on survival rates for patients with GC. The summary results were pooled using the random-effects model. We included 12 prospective RCTs reporting data on 1538 GC patients.

Results

Patients who received NAC were associated with significant improvement of OS (P = 0.001) and PFS (P < 0.001). Furthermore, NAC therapy significantly increased the incidence of 1-year survival rate (SR) (P = 0.020), 3-year SR (P = 0.011), and 4-year SR (P = 0.001). Similarly, NAC therapy was associated with a lower incidence of 1-year (P < 0.001), 2-year (P < 0.001), 3-year (P < 0.001), 4-year (P = 0.001), and 5-year recurrence rate (P = 0.002). Conversely, patients who received NAC also experienced a significantly increased risk of lymphocytopenia (P = 0.003), and hemoglobinopathy (P = 0.021).

Conclusions

The findings of this study suggested that NAC is associated with significant improvement in the outcomes of survival and disease progression for GC patients while also increasing some toxicity.
Abkürzungen
GC
Gastric cancer
NAC
Neoadjuvant chemotherapy
RCTs
Randomized controlled trials
SR
Survival rate

Background

Although cancer-related incidence and mortality have been decreasing in the past few years, gastric cancer (GC) remains the fourth most common malignancy in world [1]. The incidence of early gastric cancer were highest in China, Japan, and Korea, which accounting for greater than 50% of the world totals [2]. The prognosis of GC patients is determined relative to their cancer stage. Such as, for patients with advanced stages of GC (III and IV preoperative TNM staging), the 5-year survival rate of approximately 25 % [3]. It is estimated that local recurrence or distant metastases will happen in about 60% of GC patients even if they undergo macroscopic resection [4].
Multimodality therapy including neoadjuvent chemotherapy (NAC) therapy and D1+/D2 gastrectomy is regarded as standard of care across Europe and Australasia and is increasing accepted in North America [5]. D2 Gastrectomy with adjuvant therapy is practised routinely in Asia, whereas patients with advanced gastric tumors needed to received NAC therapy [3]. A previous meta-analysis of 6 randomized controlled trial (RCT) has no significant effect on overall survival or complete resection [6]. However, one trial [7] was included in a previous meta-analysis despite its use of imbalanced postoperative chemotherapy, resulting in obvious bias [6]. Additionally, a misjudged trial [8] for RCT and two researches [9, 10] with unmatched postoperative treatment led to an ineligible criteria in Ge’s analysis [11]. Moreover, the meta-analysis of Li et al. [12] and Wu et al. [13] included non-RCTs and few qualified RCTs. Finally, the potential role of NAC as treatment in patients with GC on year-specific survival rate has not been investigated by previous meta-analyses.
In order to reach a higher level of meta-analysis, the pooled data for this study will consist entirely of RCTs. Using only these qualified RCTs, we carried out a meta-analysis and systemic review to demonstrate the survival outcomes related to NAC.

Methods

Data sources, search strategy, and selection criteria

This review was conducted and reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Statement issued in 2009 [14]. Ethics approval was not necessary for this study, as only de-identified pooled data from individual studies were analyzed. Following the Cochrane Handbook for systematic review and meta-analysis, electronic databases including the Cochrane online library, PubMed and Embase were utilized for the comprehensive search, and the following terms were used for the identification of relevant trials: (“gastric cancer” OR “gastric carcinoma” OR “gastric neoplasm” OR “stomach cancer” OR “stomach neoplasm” OR “stomach carcinoma” OR “gastroesophageal junction neoplasm” OR “cancer of stomach”) AND (“neoadjuvant chemotherapy” OR “preoperative chemotherapy”). We also conducted manual searches of reference lists from all relevant original research and review articles to identify additional eligible studies. The medical subject heading, methods, patient population, design, intervention, control, and outcome variables of these articles were used to identify relevant studies.
We introduced a two-stage process to select eligible studies based on the above eligibility criteria. Studies selected via systematic identification were evaluated for consistency through their title, abstract and full text, and those that failed to meet the inclusion criteria were rejected. For the articles with only the abstract available, we tried to contact the corresponding author in an effort to obtain the full text. Trials were included if they compared NAC versus Surgery Alone (SA) in patients with GC and at least one of following reported outcomes: resectability, OS, PFS, year-specific survival rate (SR) and recurrence, and Grade 3 or 4 adverse events. Furthermore, all included studies followed a proper RCT design. There was no restriction for language or publication status. Data expressed as medians were not included and case series, case reports, reviews and duplicates were excluded. Finally, studies that reported data comparing outcomes of patients with or without postoperative chemotherapy were excluded.

Data collection and quality assessment

Two reviewers independently extracted data from eligible studies using a standardized data extraction table. Any disagreement was settled by discussion or, in the absence of a consensus, by a third reviewer. The data collected included the first author’s name, country, publication year, number of participants, mean age, percentage male, disease status, NAC chemotherapy regimen, and design of trials included. Reported outcomes included resectability, OS, PFS, 1-year SR, 2-year SR, 3-year SR, 4-year SR, 5-year SR, 1-year recurrence rate, 2-year recurrence rate, 3-year recurrence rate, 4-year recurrence rate, 5-year recurrence rate, and Grade 3 or 4 adverse events. The quality of the eligible studies was evaluated using the Jadad scale [15]. Randomization, blinding, withdrawals, generation of random numbers, and concealment of allocation as the essential parts to a RCT, were scored ranged 0 to 5. A threshold of ≥4 points was regarded as a high-quality study. Any inconsistencies were solved by group discussion for a consensus.

Statistical analysis

We assigned the results of each RCT as dichotomous frequency data. Relative risks (RR) and 95% confidence intervals (CI) were calculated for each study from event numbers and total patients extracted from each trial before data pooling. The overall HR or RR and 95% CI of resectability, OS, PFS, 1-year SR, 2-year SR, 3-year SR, 4-year SR, 5-year SR, 1-year recurrence rate, 2-year recurrence rate, 3-year recurrence rate, 4-year recurrence rate, 5-year recurrence rate, and Grade 3 or 4 adverse events were also calculated. Both fixed-effect and random-effect models were used to evaluate the pooled HR or RR for patients who received NAC compared with patients with surgery alone. Although both models yielded similar findings, results from the random-effect model, which assumes that the true underlying effect varies among included trials, are presented here [16, 17]. Sensitivity analysis was conducted by removing each individual study from the meta-analysis [18]. Subgroup analyses were conducted for resectability, OS and PFS on the basis of country, mean age, percentage male, percentage of tumor stages (I and II), and disease status. The Egger [19] and Begg tests [20] were also used to statistically assess publication bias for each outcome. All reported P values are 2-sided, and P values < 0.05 were considered statistically significant for all included studies. Statistical analyses were performed using STATA software (version 12.0; Stata Corporation, College Station, TX, USA).

Results

The results of our study selection process are shown in Fig. 1. We identified 435 articles in our initial electronic search, of which 400 were excluded as duplicates or irrelevant studies. A total of 35 potentially eligible studies were selected for further judging. After detailed evaluations, 12 RCTs were selected for the final meta-analysis of the efficacy and safety of NAC and SA [7, 9, 10, 2129]. A manual search of the reference lists of these studies did not yield any new eligible studies. The general characteristics of the included studies are presented in Table 1.
Table 1
Baseline characteristics of studies included in the final meta-analysis
First author
Publication year
Country
Age
Male (%)
Sample size
Chemotherapy regimen
Disease status
Tumor stage (I and II)
Jadad score
NAC
SA
Total
Cunningham [21]
2006
UK
62.0
78.7
250
253
503
Cisplatin; fluorouracil
Resectable Gastroesophageal Cancer
43.8%
5
Hartgrink [22]
2004
Netherland
NG
NG
27
29
56
Methotrexate; 5-fluorouracil; leucovorin
Resectable GC
53.6%
4
Hashemzadeh [23]
2014
Iran
59.2
75.7
22
52
74
Docetaxel; cisplatin; 5-fluorouracil
Locally advanced GC
28.0%
1
Lygidakis [24]
1999
Greece
61.0
47.4
19
19
38
Mitomycin C; 5-fluorouracil; leucovorin; farmorubicin
Resectable GC
36.8%
2
Qu [25]
2010
China
56.0
61.5
39
39
78
Docetaxel
Advanced GC
0.0%
3
Schuhmacher [10]
2010
Europe
57.0
69.4
72
72
144
Cisplatin; fluorouracil
Locally Advanced Cancer of the Stomach and Cardia
0.0%
3
Sun [26]
2011
China
NG
NG
29
26
55
Docetaxel; dexamethasone; cimetidine; phenergan
Borrmann Type IV GC
NG
1
Wang [27]
2000
China
54.5
83.3
30
30
60
5-fluorouracil
Gastric cardia cancer
18.3%
1
Ychou [9]
2011
France
63.0
84.0
113
111
224
Fluorouracil; cisplatin
Resectable Gastroesophageal Adenocarcinoma
31.1%
3
Yonemura [7]
1993
Japan
60.5
74.5
26
29
55
Cisplatin; mitomycin C; etoposide;l-(2-tetrahydrofuryl)-5-fluorouracil; uracil
High-Grade Advanced GC
16.4%
2
Zhang [28]
2012
China
NG
60.0
38
42
80
Calcium folinate; oxaliplatin; 5-fluorouracil
Advanced GC
0.0%
2
Kobayashi [29]
2000
Japan
NG
NG
91
80
171
5-fluorouracil
Resectable GC
NG
2
NAC neoadjuvant chemotherapy, SA surgery alone, GC gastric cancer NG not given
The 12 included trials involve a total of 1538 GC patients. The sample sizes ranged from 38 to 503, with mean ages ranging from 54 to 64 years. Five trials were conducted in Europe [9, 1021, 22, 24], and the remaining 7 were conducted in Asia [723, 2529]. Study quality was evaluated using the Jadad scale. Overall, 1 trial [21] had a score of 5, 1 trial [22] had a score of 4, 3 trials [9, 1025] had a score of 3, 4 trials [924, 28, 29] had a score of 2, and the remaining 3 trials [23, 26, 27] had a score of 1.
Data for the effect of NAC on the incidence of resectability were available from 8 trials. The summary RR showed no significant difference between NAC and SA for resectability (RR: 1.08; 95%CI: 0.97–1.19; P = 0.168; Fig. 2). Substantial heterogeneity was detected across included trials (P < 0.001). As a result, a sensitivity analysis was conducted for resectability and, after excluding Cunningham et al.’s trial which specifically included patients with gastroesophageal cancer, we noted that patients receiving NAC were associated with a non-significant increase in the incidence of resectability (RR: 1.12; 95%CI: 1.00–1.26; P = 0.058).
Data for OS and PFS were available from 3 trials. NAC was associated with a statistically significant improvement in OS (HR: 0.74; 95%CI: 0.63–0.88; P = 0.001; Fig. 3) and PFS (HR: 0.67; 95%CI: 0.57–0.79; P < 0.001) as compared with SA. There was no significant heterogeneity across the included trials. Sensitivity analyses were conducted with the sequential exclusion of each trial, with no effect on the conclusions for OS and PFS.
Data for the effects of NAC on the incidence of year-specific SR were organized by increased SR per year and listed in Fig. 4. The combined RR suggests that patients who received NAC experienced a significantly increased incidence of 1-year SR (RR: 1.11; 95%CI: 1.02–1.21; P = 0.020), 3-year SR (RR: 1.30; 95%CI: 1.06–1.59; P = 0.011), and 4-year SR (RR: 1.45; 95%CI: 1.15–1.81; P = 0.001). However, there was no significant effect on the incidence of 2-year SR (RR: 1.14; 95%CI: 0.96–1.37; P = 0.137), and 5-year SR (RR: 1.33; 95%CI: 0.92–1.92; P = 0.130). Moderate heterogeneity was detected in 2-year SR and 5-year SR, while negligible heterogeneity was observed in 1-year SR, 3-year SR, and 4-year SR.
Data for the effects of NAC on the incidence of year-specific recurrence rate were grouped by increased recurrence rate per year and listed in Fig. 5. We noted that patients who received NAC had a significantly reduced risk of 1-year (RR: 0.69; 95%CI: 0.58–0.81; P < 0.001), 2-year (RR: 0.78; 95%CI: 0.71–0.86; P < 0.001), 3-year (RR: 0.87; 95%CI: 0.80–0.94; P < 0.001), 4-year (RR: 0.90; 95%CI: 0.85–0.96; P = 0.001), and 5-year recurrence rate (RR: 0.93; 95%CI: 0.88–0.97; P = 0.002). There was no significant heterogeneity detected across the included trials.
The combined results of WHO grade 3 or greater adverse events are presented in Table 2. Most specific adverse events were non-significant due to the low number of trials reporting this information. We noted that patients who received NAC were associated with an elevated risk of developing lymphocytopenia (RR: 2.02; 95%CI: 1.27–3.24; P = 0.003), and hemoglobinopathy (RR: 11.13; 95%CI: 1.45–85.58; P = 0.021) when compared with SA. No significant effect was detected across the included trials for other adverse events.
Table 2
Summarized of grade 3 or greater adverse events
Outcomes
NAC group
Control group
RR (95% CI)
P value
P value for heterogeneity
Granulocytopenia
67/222
52/240
1.23 (0.81–1.87)
0.338
0.209
Lymphocytopenia
46/204
23/230
2.02 (1.27–3.24)
0.003
Leukopenia
39/250
29/263
1.26 (0.59–2.66)
0.552
0.086
Hemoglobinopathy
11/239
1/252
11.13 (1.45–85.58)
0.021
Thrombocytopenia
2/287
6/286
0.35 (0.07–1.72)
0.195
0.677
Other hematologic abnormality
1/249
2/251
0.51 (0.05–5.55)
0.577
Nausea
25/264
29/263
0.86 (0.53–1.41)
0.559
0.889
Vomiting
13/237
14/239
0.94 (0.45–1.96)
0.868
Neurologic effects
10/279
7/285
1.46 (0.55–3.89)
0.453
0.329
Skin effects
8/242
2/251
4.05 (0.87–18.88)
0.075
Stomatitis
10/240
5/248
2.02 (0.70–5.84)
0.192
Diarrhea
6/244
5/248
1.21 (0.38–3.93)
0.746
Subgroup analyses were performed for resectability, OS, and PFS to evaluate the effect of NAC in specific subpopulations (Table 3). First, we noted NAC was associated with higher resectability if the patients included in individual trial were Asian. Second, patients who received NAC has no significant effect on OS if the mean age of patients less than 60, percentage male less than 70%, percentage of tumor stage (I and II) less than 30%, and patients with GC. Third, NAC was not associated with PFS if the mean age of patients less than 60, percentage male less than 70%, percentage of tumor stage (I and II) less than 30%, and patients with GC.
Table 3
Subgroup analysis
Outcomes
Group
RR (95% CI)
P value
P value for heterogeneity
Resectability
Country
Asian
1.23 (1.04–1.46)
0.015
0.243
Europe
1.00 (0.91–1.11)
0.950
0.008
Mean age (years)
60 or more
1.02 (0.81–1.29)
0.842
0.002
< 60
1.29 (0.84–1.97)
0.251
< 0.001
Percentage male (%)
70 or greater
1.13 (0.87–1.46)
0.355
< 0.001
< 70
1.14 (0.91–1.42)
0.257
0.020
Percentage of tumor stage (I and II) (%)
30 or greater
0.99 (0.83–1.18)
0.934
0.009
< 30
1.22 (0.94–1.59)
0.132
0.001
Disease status
Gastroesophageal cancer
1.02 (0.81–1.29)
0.842
0.002
Gastric cancer
1.12 (0.96–1.31)
0.136
0.018
OS
Country
Asian
Europe
0.74 (0.63–0.88)
0.001
0.792
Mean age (years)
60 or more
0.73 (0.61–0.88)
0.001
0.674
< 60
0.84 (0.52–1.35)
0.474
Percentage male (%)
70 or greater
0.73 (0.61–0.88)
0.001
0.674
< 70
0.84 (0.52–1.35)
0.474
Percentage of tumor stage (I and II) (%)
30 or greater
0.73 (0.61–0.88)
0.001
0.674
< 30
0.84 (0.52–1.35)
0.474
Disease status
Gastroesophageal cancer
0.73 (0.61–0.88)
0.001
0.674
Gastric cancer
0.84 (0.52–1.35)
0.474
PFS
Country
Asian
Europe
0.67 (0.57–0.79)
< 0.001
0.825
Mean age (years)
60 or more
0.66 (0.55–0.78)
< 0.001
0.936
< 60
0.76 (0.49–1.17)
0.212
Percentage male (%)
70 or greater
0.66 (0.55–0.78)
< 0.001
0.936
< 70
0.76 (0.49–1.17)
0.212
Percentage of tumor stage (I and II) (%)
30 or greater
0.66 (0.55–0.78)
< 0.001
0.936
< 30
0.76 (0.49–1.17)
0.212
Disease status
Gastroesophageal cancer
0.66 (0.55–0.78)
< 0.001
0.936
Gastric cancer
0.76 (0.49–1.17)
0.212
The Egger and Begg test results showed no evidence of publication bias for resectability, OS, PFS, 1-year SR, 2-year SR, 3-year SR, 4-year SR, 1-year recurrence, 2-year recurrence, 3-year recurrence, 4-year recurrence, or 5-year recurrence. Although the Begg test showed no evidence of publication bias for 5-year SR (P = 0.452), the Egger test showed potential evidence of publication bias for 5-year SR (P = 0.009) (Table 4). The conclusion was unchanged after adjustment for publication bias by using the trim and fill method [30].
Table 4
Publication bias
Outcomes
P value for Egger
P value for Begg
Resectability
0.363
0.711
OS
0.763
1.000
PFS
0.444
1.000
1-year survival rate
0.376
0.764
2-year survival rate
0.266
1.000
3-year survival rate
0.991
0.806
4-year survival rate
0.065
0.221
5-year survival rate
0.009
0.452
1-year recurrence
0.069
1.000
2-year recurrence
0.242
0.296
3-year recurrence
0.243
0.296
4-year recurrence
0.723
1.000
5-year recurrence
0.887
1.000

Discussion

This meta-analysis of studies analyzing the efficacy and safety of NAC included updated data from previously published studies and additional new RCTs not reviewed in previously published works. This additional information allows for a more robust analysis of the effect of NAC on survival outcomes for GC. The results of this updated meta-analysis indicate that NAC could elicit improvements in OS, PFS, 1-, 3-, and 4-year SR, and 1-, 2-, 3-, 4-, and 5-year recurrence in treatment of patients with GC as compared with those received SA. Conversely, patients receiving NAC also experienced a significantly increased risk of developing lymphocytopenia, and hemoglobinopathy. No other significant differences were detected across included trials.
The methodological assessment of individual trial was the essential parts including randomization, blinding, withdrawals, generation of random numbers, and concealment of allocation. This meta-analysis provides clear information about randomization and withdrawals, whereas other forms were available in few trials and might contribute to heterogeneity in overall analysis. Therefore, we critically this recommendations for the treatment of patients with GC due to the unsatisfactory quality of included trials.
There were certain limitations present in previous meta-analysis articles exploring the efficacy and safety of NAC on survival outcomes for gastric carcinoma.. Liao et al. [6] suggested that NAC was associated with an insignificant increase in the incidence of overall survival, R0 resection, postoperative complications, and perioperative mortality. Furthermore, Xiong et al. [31] conducted an updated meta-analysis of RCTs and found that NAC can significantly improve SR, 3-year PFS, tumor down-staging rate and R0 resection rate, whereas it had no significant effect on relapse rates, operative complications, perioperative mortality and grade 3/4 adverse events. However, these studies did not report year-specifically SR and recurrence. Additionally, although several trials suggest that NAC can be used as a standard therapy for patients with GC, the superiority of NAC over SA remains unclear due to the greater adverse events detected in the NAC group. Therefore, it was necessary to conduct an updated meta-analysis to explore further information regarding the efficacy and safety of the NAC in treatment of patients with GC.
There was no significant overall difference for the incidence of resectability between NAC and SA groups. However, three trials included in our study reported inconsistent results. The MAGIC Trial [21] suggested that patients with resectable gastroesophageal cancer who received NAC were associated with a lower incidence of resectability, whereas two other trials [23, 25] indicated that NAC therapy significantly increased the incidence of resectability. A possible explanation could be that patients who received NAC therapy might have had their surgery postponed, allowing for the disease to progress, causing these patients to lose a chance to undergo curative surgery.
The findings of our study suggest that patients who received NAC therapy experienced significant improvement in OS, PFS, and year-specifically SR and recurrence, although there was no significant difference between NAC and SA for 2-year SR and 5-year SR. The cause of this could simply be the smaller number of trials reporting these outcomes. Further, the reason for no significant difference for 5-year SR might affected by the Kobayashi et al.’s study, which included patients received the low dose of 5′-deoxy-5-fluorouridine. Furthermore, the use of NAC was considered in order to lower the stage of the tumor and improve resectability and survival. Therefore, NAC might play a beneficial role in the treatment of patients with GC.
As expected, NAC therapy was associated with an increased risk of some toxicity. The improvement of survival outcomes should balance these risks if used on grade 3 or greater adverse events, which optimize the impact on the patients’ quality of life. However, data on specific adverse events were rarely available and these results may be variable due to the low number of trials included. Therefore, we only aim to provide a synthetic and comprehensive review for adverse events in aggregate.
In our study, patients received NAC was associated with a higher incidence of resectability when the study included Asians. These findings were inconsistent with the study included Europeans. This could be because the percentage of tumor stage (I and II) was higher in Europe, which associated with higher resectability rate. Further, the tumor stages was higher in Asia than Europe, and the treatment effect on resectability was obvious. Two of included trials provided higher weight (21.1% and 20.5%; Fig. 2) were conducted in Europe and reported no significant effect on resectability, which could affect the treatment effect of NAC on resectability to no statistically significant [21]. In addition, disease status, tumor stages were also play an important role on treatment effect. Although no significant difference were detected, the reason could be that the analysis included smaller patient cohorts, and the result may be unstable. Furthermore, the results of subgroup analyses for OS and PFS were restricted due to only three trials provided the data of OS and PFS.
Two strengths of our study should be highlighted. First, the large sample size allowed us to quantitatively assess the efficacy and safety of NAC in the treatment of GC patients, thus our findings are potentially more robust than those of any individual study. Second, we specifically reported year-specific SR and recurrence, and summarized grade 3 or greater adverse events, which allows for an accurate assessment of the benefits and harms for GC patients.
The limitations of our study are as follows: (1) in a meta-analysis of published studies, publication bias is an inevitable problem; (2) the analysis used pooled data (individual data were not available), which restricted us from performing a more detailed relevant analysis and obtaining more comprehensive results; (3) data on adverse events or quality of life were rarely available in included trials, so the conclusion may be variable; and (4) In the planning stages, we intend conducted subgroup analyses based on gender (men, women), and tumor stages (I, or II, and III or IV), whereas the results of stratified analysis in individual trial were not available.

Conclusions

The findings of this study indicate that NAC might play an important role on the outcomes of survival rate and disease progression for patients with GC. However, it may also associate with an increased risk in for adverse effects. Future trials should focus on specific disease status and record pre- and post-operative adverse events.

Acknowledgements

Not applicable

Funding

This work was supported by the National Natural Science Foundation of China (No. 81272718, 81302125 and 81372550). The sponsors played no role in the study design, data collection, or analysis, nor decision to submit the article for publication.

Availability of data and materials

The datasets used and/or analysed during the current study available from the corresponding author on reasonable request.
Not applicable
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.
Zurück zum Zitat Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.CrossRefPubMed Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.CrossRefPubMed
2.
Zurück zum Zitat Sugano K. Screening of gastric cancer in Asia. Best Pract Res Clin Gastroenterol. 2015;29:895–905.CrossRefPubMed Sugano K. Screening of gastric cancer in Asia. Best Pract Res Clin Gastroenterol. 2015;29:895–905.CrossRefPubMed
3.
Zurück zum Zitat Xu AM, Huang L, Liu W, Gao S, Han WX, Wei ZJ. Neoadjuvant chemotherapy followed by surgery versus surgery alone for gastric carcinoma: systematic review and meta-analysis of randomized controlled trials. PLoS One. 2014;9:e86941.CrossRefPubMedPubMedCentral Xu AM, Huang L, Liu W, Gao S, Han WX, Wei ZJ. Neoadjuvant chemotherapy followed by surgery versus surgery alone for gastric carcinoma: systematic review and meta-analysis of randomized controlled trials. PLoS One. 2014;9:e86941.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Sant M, Allemani C, Santaquilani M, Knijn A, Marchesi F, Capocaccia R, EUROCARE Working Group. EUROCARE-4. Survival of cancer patients diagnosed in 1995–1999. Results and commentary. Eur J Cancer. 2009;45:931–91.CrossRefPubMed Sant M, Allemani C, Santaquilani M, Knijn A, Marchesi F, Capocaccia R, EUROCARE Working Group. EUROCARE-4. Survival of cancer patients diagnosed in 1995–1999. Results and commentary. Eur J Cancer. 2009;45:931–91.CrossRefPubMed
5.
Zurück zum Zitat Mezhir JJ, Tang LH, Coit DG. Neoadjuvant therapy of locally advanced gastric cancer. J Surg Oncol. 2010;101:305–14.CrossRefPubMed Mezhir JJ, Tang LH, Coit DG. Neoadjuvant therapy of locally advanced gastric cancer. J Surg Oncol. 2010;101:305–14.CrossRefPubMed
6.
Zurück zum Zitat Liao Y, Yang ZL, Peng JS, Xiang J, Wang JP. Neoadjuvant chemotherapy for gastric cancer: a meta-analysis of randomized, controlled trials. J Gastroenterol Hepatol. 2013;28:777–82.CrossRefPubMed Liao Y, Yang ZL, Peng JS, Xiang J, Wang JP. Neoadjuvant chemotherapy for gastric cancer: a meta-analysis of randomized, controlled trials. J Gastroenterol Hepatol. 2013;28:777–82.CrossRefPubMed
7.
Zurück zum Zitat Yonemura Y, Sawa T, Kinoshita K, Matsuki N, Fushida S, Tanaka S, et al. Neoadjuvant chemotherapy for high-grade advanced gastric cancer. World J Surg. 1993;17:256–61. discussion 261–62CrossRefPubMed Yonemura Y, Sawa T, Kinoshita K, Matsuki N, Fushida S, Tanaka S, et al. Neoadjuvant chemotherapy for high-grade advanced gastric cancer. World J Surg. 1993;17:256–61. discussion 261–62CrossRefPubMed
8.
Zurück zum Zitat Zhang CW, Zou SC, Shi D, Zhao DJ. Clinical significance of preoperative regional intra-arterial infusion chemotherapy for advanced gastric cancer. World J Gastroenterol. 2004;10:3070–2.CrossRefPubMedPubMedCentral Zhang CW, Zou SC, Shi D, Zhao DJ. Clinical significance of preoperative regional intra-arterial infusion chemotherapy for advanced gastric cancer. World J Gastroenterol. 2004;10:3070–2.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Ychou M, Boige V, Pignon JP, Conroy T, Bouché O, Lebreton G, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–21.CrossRefPubMed Ychou M, Boige V, Pignon JP, Conroy T, Bouché O, Lebreton G, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–21.CrossRefPubMed
10.
Zurück zum Zitat Schuhmacher C, Gretschel S, Lordick F, Reichardt P, Hohenberger W, Eisenberger CF, et al. Neoadjuvant chemotherapy compared with surgery alone for locally advanced cancer of the stomach and cardia: European Organisation for Research and Treatment of Cancer randomized trial 40954. J Clin Oncol. 2010;28:5210–8.CrossRefPubMedPubMedCentral Schuhmacher C, Gretschel S, Lordick F, Reichardt P, Hohenberger W, Eisenberger CF, et al. Neoadjuvant chemotherapy compared with surgery alone for locally advanced cancer of the stomach and cardia: European Organisation for Research and Treatment of Cancer randomized trial 40954. J Clin Oncol. 2010;28:5210–8.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Ge L, Wang HJ, Yin D, Lei C, Zhu JF, Cai XH, et al. Effectiveness of 5-flurouracil-based neoadjuvant chemotherapy in locally-advanced gastric/ gastroesophageal cancer: a meta-analysis. World J Gastroenterol. 2012;18:7384–93.CrossRefPubMedPubMedCentral Ge L, Wang HJ, Yin D, Lei C, Zhu JF, Cai XH, et al. Effectiveness of 5-flurouracil-based neoadjuvant chemotherapy in locally-advanced gastric/ gastroesophageal cancer: a meta-analysis. World J Gastroenterol. 2012;18:7384–93.CrossRefPubMedPubMedCentral
12.
13.
Zurück zum Zitat Wu AW, Xu GW, Wang HY, Ji JF, Tang JL. Neoadjuvant chemotherapy versus none for resectable gastric cancer. Cochrane Database Syst Rev. 2007;18:CD005047. Wu AW, Xu GW, Wang HY, Ji JF, Tang JL. Neoadjuvant chemotherapy versus none for resectable gastric cancer. Cochrane Database Syst Rev. 2007;18:CD005047.
14.
Zurück zum Zitat Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6:e1000097.CrossRefPubMedPubMedCentral Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6:e1000097.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials. 1996;17(1):1–12.CrossRefPubMed Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary? Control Clin Trials. 1996;17(1):1–12.CrossRefPubMed
16.
Zurück zum Zitat DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.CrossRefPubMed DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.CrossRefPubMed
17.
Zurück zum Zitat Ades AE, Lu G, Higgins JP. The interpretation of random-effects metaanalysis in decision models. Med Decis Mak. 2005;25:646–54.CrossRef Ades AE, Lu G, Higgins JP. The interpretation of random-effects metaanalysis in decision models. Med Decis Mak. 2005;25:646–54.CrossRef
18.
Zurück zum Zitat Tobias A. Assessing the influence of a single study in meta-analysis. Stata Tech Bull. 1999;47:15–7. Tobias A. Assessing the influence of a single study in meta-analysis. Stata Tech Bull. 1999;47:15–7.
20.
Zurück zum Zitat Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics. 1994;50:1088–101.CrossRefPubMed Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics. 1994;50:1088–101.CrossRefPubMed
21.
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(1):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(1):11–20.CrossRefPubMed
22.
Zurück zum Zitat Hartgrink HH, van de Velde CJ, Putter H, Songun I, Tesselaar ME, Kranenbarg EK, et al. Neo-adjuvant chemotherapy for operable gastric cancer: long term results of the Dutch randomized FAMTX trial. Eur J Surg Oncol. 2004;30:643–9.CrossRefPubMed Hartgrink HH, van de Velde CJ, Putter H, Songun I, Tesselaar ME, Kranenbarg EK, et al. Neo-adjuvant chemotherapy for operable gastric cancer: long term results of the Dutch randomized FAMTX trial. Eur J Surg Oncol. 2004;30:643–9.CrossRefPubMed
23.
Zurück zum Zitat Hashemzadeh S, Pourzand A, Somi MH, Zarrintan S, Javad-Rashid R, Esfahani A. The effects of neoadjuvant chemotherapy on resectability of locallyadvanced gastric adenocarcinoma: a clinical trial. Int J Surg. 2014;12:1061–9.CrossRefPubMed Hashemzadeh S, Pourzand A, Somi MH, Zarrintan S, Javad-Rashid R, Esfahani A. The effects of neoadjuvant chemotherapy on resectability of locallyadvanced gastric adenocarcinoma: a clinical trial. Int J Surg. 2014;12:1061–9.CrossRefPubMed
24.
Zurück zum Zitat Lygidakis NJ, Sgourakis G, Aphinives P. Upper abdominal stop-flow perfusion as a neo and adjuvant hypoxic regional chemotherapy for resectable gastric carcinoma: a prospective randomized clinical trial. Hepato-Gastroenterology. 1999;46:2035–8.PubMed Lygidakis NJ, Sgourakis G, Aphinives P. Upper abdominal stop-flow perfusion as a neo and adjuvant hypoxic regional chemotherapy for resectable gastric carcinoma: a prospective randomized clinical trial. Hepato-Gastroenterology. 1999;46:2035–8.PubMed
25.
Zurück zum Zitat Qu JJ, Shi YR, Liu FR, Ma SQ, Ma FY. A clinical study of paclitaxel combined with FOLFOX4 regimen as neoadjuvant chemotherapy for advanced gastric cancer. Zhonghua Wei Chang Wai Ke Za Zhi. 2010;13:664–7. ChinesePubMed Qu JJ, Shi YR, Liu FR, Ma SQ, Ma FY. A clinical study of paclitaxel combined with FOLFOX4 regimen as neoadjuvant chemotherapy for advanced gastric cancer. Zhonghua Wei Chang Wai Ke Za Zhi. 2010;13:664–7. ChinesePubMed
26.
Zurück zum Zitat Sun XC, Lin J, Ju AH. Treatment of Borrmann type IV gastric cancer with a neoadjuvant chemotherapy combination of docetaxel, cisplatin and 5-Fluorouracil/Leucovorin. J Int Med Res. 2011;39:2096–102.CrossRefPubMed Sun XC, Lin J, Ju AH. Treatment of Borrmann type IV gastric cancer with a neoadjuvant chemotherapy combination of docetaxel, cisplatin and 5-Fluorouracil/Leucovorin. J Int Med Res. 2011;39:2096–102.CrossRefPubMed
27.
Zurück zum Zitat Wang XL, Wu GX, Zhang MD, Guo M, Zhang H, Sun XF. A favorable impact of preoperative FPLC chemotherapy on patients with gastric cardia cancer. Oncol Rep. 2000;7:241–4.PubMed Wang XL, Wu GX, Zhang MD, Guo M, Zhang H, Sun XF. A favorable impact of preoperative FPLC chemotherapy on patients with gastric cardia cancer. Oncol Rep. 2000;7:241–4.PubMed
28.
Zurück zum Zitat Zhang J, Chen RX, Zhang J, Cai J, Meng H, Wu GC, et al. Efficacy and safety of neoadjuvant chemotherapy with modified FOLFOX7 regimen on the treatment of advanced gastric cancer. Chin Med J. 2012;125:2144–50.PubMed Zhang J, Chen RX, Zhang J, Cai J, Meng H, Wu GC, et al. Efficacy and safety of neoadjuvant chemotherapy with modified FOLFOX7 regimen on the treatment of advanced gastric cancer. Chin Med J. 2012;125:2144–50.PubMed
29.
Zurück zum Zitat Kobayashi T, Kimura T. Long-term outcome of preoperative chemotherapy with 5′-deoxy-5-fluorouridine (5’-DFUR) for gastric cancer. Jpn J Cancer Chemother. 2000;27:1521–6. Kobayashi T, Kimura T. Long-term outcome of preoperative chemotherapy with 5′-deoxy-5-fluorouridine (5’-DFUR) for gastric cancer. Jpn J Cancer Chemother. 2000;27:1521–6.
30.
Zurück zum Zitat Duvall S, Tweedie R. A nonparametric “trim and fill” method for assessing publication bias in meta-analysis. J Am Stat Assoc. 2000;95:89–98. Duvall S, Tweedie R. A nonparametric “trim and fill” method for assessing publication bias in meta-analysis. J Am Stat Assoc. 2000;95:89–98.
31.
Zurück zum Zitat Xiong B, Ma L, Cheng Y, Zhang C. Clinical effectiveness of neoadjuvant chemotherapy in advanced gastric cancer: an updated meta-analysis of randomized controlled trials. Eur J Surg Oncol. 2014;40:1321–30.CrossRefPubMed Xiong B, Ma L, Cheng Y, Zhang C. Clinical effectiveness of neoadjuvant chemotherapy in advanced gastric cancer: an updated meta-analysis of randomized controlled trials. Eur J Surg Oncol. 2014;40:1321–30.CrossRefPubMed
Metadaten
Titel
Effect of neoadjuvant chemotherapy in patients with gastric cancer: a PRISMA-compliant systematic review and meta-analysis
verfasst von
Zhi-Feng Miao
Xing-Yu Liu
Zhen-Ning Wang
Ting-Ting Zhao
Ying-Ying Xu
Yong-Xi Song
Jin-Yu Huang
Hao Xu
Hui-Mian Xu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2018
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4027-0

Weitere Artikel der Ausgabe 1/2018

BMC Cancer 1/2018 Zur Ausgabe

Update Onkologie

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