Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

International Journal of Surgery 51 (2018) 120–127

Contents lists available at ScienceDirect

International Journal of Surgery


journal homepage: www.elsevier.com/locate/ijsu

Review

Neoadjuvant chemotherapy in advanced gastric and esophago-gastric T


cancer. Meta-analysis of randomized trials
Federico Coccolinih,∗, Matteo Nardib, Giulia Montoria, Marco Ceresolia, Andrea Celottic,
Stefano Cascinud, Paola Fugazzolaa, Matteo Tomasonia, Olivier Glehene, Fausto Catenaf,
Yutaka Yonemurag, Luca Ansalonih
a
General Surgery Dept., Papa Giovanni XXIII Hospital, Bergamo, Italy
b
General Surgery Dept., La Sapienza University Hospital, Roma, Italy
c
Second Surgical Dept., Civili Hospital, Brescia, Italy
d
Medical Oncology Dept., University Hospital, Modena, Italy
e
General Surgery Dept., Centre Hospitalier Lyon Sud, Hospices Civils de Lyon and EMR 3738, Université Lyon 1, France
f
General Surgery Dept., Ospedale Maggiore, Parma, Italy
g
General Surgery Dept., Kusatsu General Hospital, Yabase 1660, Japan
h
General, Emergency and Trauma Surgery Dept., Bufalini Hospital, Cesena, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: Introduction: Even in after curative surgery and adequate linfoadenectomy the survival of advanced gastric
Gastric cancer cancer (AGC) remains poor. At present some data have been published on the effects of NACT and perioperative
Esophago-gastric cancer chemotherapy on AGC and Esophago-gastric cancer (EGC) but not definitive ones. The present meta-analysis
Perioperative aims to evaluate the effects of neoadjuvant chemotherapy (NACT) on the AGC and EGC.
Neoadjuvant chemotherapy
Material and methods: A systematic review with meta-analysis of randomized controlled trials (RCTs) of
Survival
Management
NACT + surgery vs. Surgery in patients with AGC and EGC was performed.
Treatment Results: 15 RCTs have been included (2001 patients: 977 into NACT + surgery arm and 1024 into control arm).
Recurrence NACT + Surgery reduces the overall mortality at 1, 3 and 5-year in cumulative analysis (RR = 0.78; 0.81; 0.88
Post-operative respectively), at 1, 2, 3 and 5-years in EGC (RR = 0.79; 0.83; 0.84; 0.91 respectively) and at 3 and 5-years in
Mortality AGC (RR = 0.74; 0.82 respectively). Morbidity and perioperative mortality rate are not influenced by NACT.
Morbidity Recurrence rate is reduced by NACT + surgery in EGC (RR = 0.80).
Conclusions: NACT reduces the mortality in gastric and esophago-gastric cancer. Morbidity and perioperative
mortality are not influenced by NACT. The overall recurrence rate is reduced by NACT in esophago-gastric
cancer.

1. Introduction potentially increase the curative resection rate, improve the tumor
downstaging possiblities and reduce the tumor-related symptoms if the
Gastric cancer (GC) represents the fourth form of cancer in term of choosen drugs have effect on the specific tumor biology [6]. Hoever has
diffusion and formally the second cause of cancer death [1,2]. Recently, been reported as NACT could potentially increase the surgical related
has been observed as the esophago-gastric cancer (EGC) incidence has complications and as a consequence the perioperative mortality, if it
increased in North America and western European countries; as a doesn't delay surgical resection. At present some data have been pub-
counterpart, the GC incidence has decreased [3,4]. Even after curative lished on the effects of NACT on AGC and EGC but not definitive ones.
surgery and adequate linfoadenectomy the survival of advanced gastric Moreover the new TNM 8th stadiation modified the division of the GC
cancer (AGC) remains poor. Various therapeutical approaches have and EGC formally including these last among the GC [7].
been studied and applied to improve survival. Neoadjuvant che- The present meta-analysis aims to evaluate the effects of NACT on
motherapy (NACT) has been defined as any preoperative chemotherapy the AGC and EGC.
scheme aiming to convert unresectable into resectable tumors and/or to
increase microscopic complete tumor resection rates [5]. NACT could


Corresponding author. General, Emergency and Trauma Surgery Dept., Bufalini Hospital, Viale Giovanni Ghirotti, 286, 47521 Cesena, Italy. Tel.: +39 05473522132.
E-mail address: federico.coccolini@gmail.com (F. Coccolini).

https://doi.org/10.1016/j.ijsu.2018.01.008
Received 18 December 2017; Accepted 7 January 2018
Available online 20 February 2018
1743-9191/ © 2018 IJS Publishing Group Ltd. Published by Elsevier Ltd. All rights reserved.
F. Coccolini et al. International Journal of Surgery 51 (2018) 120–127

2. Material and method 3. Results

2.1. Literature search strategy Fifteen RCTs fulfilled the inclusion criteria and were included in the
meta-analysis (publication dates 1987–2014). There were a total of
Electronic searches were performed using Medline, Embase 2001 patients (977 randomized to receive NACT + radical resection
(1988–March 2017), PubMed (January 1980–March 2017), Cochrane and 1024 randomized to receive radical resection without NACT)
Central Register of Controlled Trials (CCTR), Cochrane Database of (Table 2).
Systematic Reviews (CDSR) and CINAHL from (1966–March 2017). The
search terms were: ‘neoadjuvant chemotherapy’, ‘stomach’, ‘gastric 3.1. Quality of trials
cancer’, ‘advanced’, ‘randomized trial’, ‘meta-analysis’ combined with
AND/OR. Research included also all the MeshTerms. No search re- There was good agreement between the reviewers (FeCo, MN) about
strictions were imposed. The reference lists of all retrieved articles were the eligibility and quality of the studies. Table 1 demonstrates the
reviewed for further identification of potentially relevant studies. quality of the 15 included RCTs [6,10–23].
Review articles were also obtained to determine other possible studies. In eight RCTs [6,11–13,17,19,20], the method of allocation con-
Duplicate published trials with accumulating numbers of patients or cealment was adequate; randomisation was performed on a central site
increased lengths of follow-up, were considered only in the last or at and transmitted to treatment providers by telephone, fax or sealed
least in the more complete version. opaque envelopes. In the remaining 7 RCTs [10,14–16,18,21–23] the
information regarding approaches to allocation concealment could not
be determined. The baseline features were similar between treatment
2.2. Selection criteria groups in all 15 RCTs thus reducing the clinical heterogeneity. All RCTs
specified the eligibility criteria for patients to be enrolled. All RCTs
Studies which have been judged eligible for this systematic review specified numbers lost to follow-up in each treatment group. All RCTs
and consequent meta-analysis are those in which patients with AGC or analysed the data on an intention-to-treat (ITT) basis, whereby parti-
EGC both without peritoneal carcinosis were randomly assigned to re- cipants were analysed in the groups to which they were initially ran-
ceive either NACT + surgery or surgery without NACT. All included domised. Blinding after allocation was impossible because of the nature
patients must have histologically-proven gastric or gastro-oesophageal- of the trials. All fifteen RCTs were considered to be at acceptable risk of
junction adenocarcinoma and underwent potentially curative resection. bias in the important domains (Table 1).
All forms of NACT in addition to surgery were included. No language
restrictions have been applied. Eligibility for study inclusion into the 3.2. 1-year mortality
meta-analysis and study quality assessment were performed in-
dependently by two authors (MN, FeCo). Study data were extracted Eight studies reported 1-year mortality in AGC and three in EGC
onto standard forms independently by two authors (MN, FeCo). [6,10,12,13,17–19] 291 and 436 patients received the surgical treat-
Discrepancies between the two investigators were resolved by discus- ment alone and 236 and 435 NACT + surgery in the two groups re-
sion and evaluation of the question with the other investigators. spectively (Fig. 1). There was no statistical heterogeneity between
The primary outcome measures for the meta-analysis were the im- studies. In the fixed-effects model, the 1-year mortality rate was sig-
pact of NACT on 1, 2, 3 and 5-year mortality and the effect of NACT on nificantly favourable to the NACT + surgery arm in the cumulative
perioperative mortality, morbidity and recurrence in AGC and EGC. analysis (RR = 0.78, 95%CI = 0.67–0.94) in the EGC (RR = 0.79,
95%CI = 0.64–0.97) and was not significantly favourable to the
2.3. Assessment of risk of bias NACT + surgery arm in AGC (RR = 0.81, 95%CI = 0.61–1.09).

There is a potential risk of overestimating the beneficial treatment 3.3. 2-years mortality
effects of RCT with a resultant risk of bias. The risk of bias was assessed
comprehensively according to guidelines of The Cochrane Three studies reported 2-years mortality in EGC [6,11,13] 436 pa-
Collaboration [8] and six items have been considered relevant tients received the surgical treatment alone and 435 NACT + surgery
(Table 1): 1) whether the method of allocation was truly random; 2) (Fig. 1). There was no statistical heterogeneity between studies. In the
whether there was proper allocation concealment; 3) whether the fixed-effects model, the 2-years mortality rate was significantly fa-
groups were similar at baseline; 4) whether the eligibility criteria were vourable to the NACT + surgery arm (RR = 0.83,
documented; 5) whether loss to follow-up in each treatment arm was 95%CI = 0.73–0.93).
specified; 6) whether intention-to-treat analysis was conducted.
Therefore the evaluation of the quality level of the study was conducted 3.4. 3-years mortality
as follows: Positive answer to all six questions was required for a trial to
be rated as high quality. With a positive answer to five or four questions Five studies reported 3-year mortality in AGC and three in EGC
the study was considered of fair quality. With a positive answer to three [6,10–13,17,18,22] 315 and 436 patients received the surgical treat-
or fewer questions the study was registered as low quality. ment alone and 254 and 435 NACT + surgery in the two groups re-
spectively (Fig. 1). There was statistical heterogeneity between studies.
In the fixed-effects model, the 3-year mortality rate was significantly
2.4. Statistical analysis favourable to the NACT + surgery arm in the cumulative analysis
(RR = 0.81, 95%CI = 0.74–0.89), in the EGC (RR = 0.84,
Data from the individual eligible studies were entered into a spread 95%CI = 0.76–0.92) and in AGC (RR = 0.74, 95%CI = 0.60–0.91).
sheet for further analysis. Review Manager (RevMan) (Version 5.3.
Copenhagen: The Nordic Cochrane Centre, The Cochrane 3.5. 5-year mortality
Collaboration, 2011) was used to perform the statistical analysis. Risk
Ratio (RR) was calculated for discrete variables. The fixed-effects and Eight studies reported 5-year mortality in AGC and three in EGC
random-effects models were used to calculate the outcomes [8,9]. [6,10–13,15,18,20–23] 472 and 436 patients received the surgical
Heterogeneity amongst the trials was determined by means of the Co- treatment alone and 422 and 435 NACT + surgery in the two groups
chrane Q value and quantified using the I2 inconsistency test. respectively (Fig. 1). There was statistical heterogeneity between

121
F. Coccolini et al. International Journal of Surgery 51 (2018) 120–127

Table 1
Included trials risk of bias and quality assessment.

Study (Ref.) Randomization Allocation Homogeneous Baseline Eligibility criteria Loss to follow-up and drop-out Intention-to-treat
Year concealment characteristic described analysis

Imano (10) YES Unclear YES YES YES YES


2010
Cunningham(6) YES YES NO YES YES YES
2006
Schumacher (11) YES YES YES YES YES YES
2010
Hartgrink (12) YES YES YES YES YES YES
2004
Ychou (13) YES YES YES YES YES YES
2011
Hashemzadeh (14) YES Unclear YES YES YES YES
2014
Zhang (15) YES Unclear YES YES YES YES
2004
Biffi (16) YES Unclear YES YES YES YES
2010
Yonemura (17) YES YES YES YES YES YES
1993
Nio (18) YES Unclear YES YES YES YES
2004
Sun (19) YES YES YES YES YES YES
2011
Kobayashi (20) YES YES YES YES YES YES
2000
Lygidakis (21) YES Unclear YES YES YES YES
1999
Shchepotin (22) YES Unclear YES YES YES YES
1999
Wang (23) YES Unclear YES YES YES YES
2000

studies. In the fixed-effects model, the 5-year mortality rate was fa- 4. Discussion
vourable to the NACT + surgery arm in the cumulative analysis
(RR = 0.88, 95%CI = 0.83–0.93), in the EGC (RR = 0.91, The real effect of NACT on AGC and EGC is still greatly unknown.
95%CI = 0.86–0.96) and in AGC (RR = 0.82, 95%CI = 0.71–0.95). Encouraging results have been published in term of survival. Definitive
results on survival and effect on short-term complications are needed.
3.6. Morbidity In fact the occurrence of short-term complications could potentially
delay the surgical intervention and definitively reduce the survival and
Six studies reported the morbidity in AGC and three in EGC disease free outcomes. The present paper aims to investigate the effect
[6,10,11,13–16,18,23] 380 and 436 patients received the surgical of NACT on survival and post-operative morbidity and short-term
treatment alone and 272 and 435 NACT + surgery in the two groups mortality in both EGC and AGC. A few papers have been published
respectively (Fig. 2). There was acceptable statistical heterogeneity about the effect of NACT but no conclusive data exist [5,24–26].
between studies. In the fixed-effects model, the morbidity rate was not Moreover the comparison between NACT and perioperative che-
influenced by the different treatment in AGC (RR = 0.99, motherapy in term of efficacy haven't yet given definitive results. An
95%CI = 0.65–1.51) in the EGC (RR = 1.12, 95%CI = 0.94–1.33) and active matter of debate is the different NACT regimens utilized by the
in the cumulative analysis (RR = 1.09, 95%CI = 0.93–1.28). different groups [6,27]. In fact several chemotherapy regimens have
been published with good results. Few data are available to evaluate the
best NACT regimen. A previously published meta-analysis showed that
3.7. Perioperative mortality
combination regimen and IV route of NACT had a high efficiency on
AGC [24]. In present study the different regimens have not been dis-
Six studies reported the post-operative mortality in AGC and three
tinguished and they have all been considered as part of NACT, re-
in EGC [6,11–17,23] 227 and 436 patients received the surgical treat-
producing the daily clinical practice of the different realities.
ment alone and 179 and 435 NACT + surgery in the two groups re-
The present analysis shows as the NACT doesn't give any adverse
spectively (Fig. 3). There was acceptable statistical heterogeneity be-
effect during the perioperative period. In fact it doesn't increase the
tween studies. In the fixed-effects model, the post-operative mortality
complications and post-operative mortality rate. The effect on EGC and
rate was not influenced by the different treatment in AGC (RR = 1.26,
AGC are positive and prolonged at all time-points in term of survival.
95%CI = 0.36–4.40) in the EGC (RR = 1.30, 95%CI = 0.70–2.42) and
The cumulative analysis demonstrated that all survival results are sig-
in the cumulative analysis (RR = 1.29, 95%CI = 0.74–2.25).
nificant confirming the effective role of NACT especially in considera-
tion of the new anathomo-pathological TNM division of esophageal and
3.8. Overall recurrence gastric cancer.
The post-operative morbidity is of fundamental importance because
Two studies reported recurrence rate in EGC [6,13] 364 patients of its occurrence could potentially delay the adjuvant chemotherapy or
received the surgical treatment alone 364 NACT + surgery (Fig. 4). some times preclude the possibility to prosecute the chemotherapy after
There was low statistical heterogeneity between studies. In the fixed- the surgical procedures. This brings detrimental effects on the short and
effects model, the recurrence rate was significantly favourable to the long-term prognosis. Moreover the reduction of tumor load for many
NACT + surgery arm in the EGC (RR = 0.80, 95%CI = 0.65–0.98).

122
Table 2
F. Coccolini et al.

Summary of the 15 included trials. (Preop: preoperative, Postop: postoperative, FU: Fluoruracil, CDDP: Cisplatin, Cht: Chemotherapy, nd: not defined).

Study (Ref.) Number of Study period Neoadjuvant Chemotherapy + Surgery arm Surgery arm Kind of tumor Lynphadenectomy
Year patients N = 977 N = 1024
N = 2001

1 Imano (10) 63 1992–2002 N = 47 N = 16 Gastric cancer D2


2010 PREOP: 72 h before surgery
Group F = 16 pz single cycle 5-FU
(330 mg/m2/24 h) for 72 h - 80 h before surgery
Group C = 15 pz single cycle CDDP
3cycles (6 mg/m2/each time) for 30 min at 68 h 44 h and 20 h before surgery
Group FC = 16 pz single cycle 5-FU + CDDP
2 Hartgrink (12) 56 1993–1996 N = 27 N = 29 Gastric cancer D1
2004 PREOP: FAMTX (Methotrexate 1500 mg/m2 + 5-FU 1500 mg/m2 + 30 mg Leucovorin every 6 h)
3 Hashemzadeh (14) 60 2011–2014 N = 30 N = 30 Gastric cancer D1 59%
2014 PREOP: Docetaxel 75 mg/m Cisplatin 75 mg/m2, 5-Fluoruracil 750 mg/m/day every 3 weeks for D2 8,1%
6 courses.
4 Zhang (15) 91 nd N = 37 N = 54 Gastric cancer
2004 PREOP: Intrarterial infusion FAP (5-FU 1.0 g + Adriamicin 30–50 mg + Cysplatin 40–60 mg) or
FMP (5-FU 1.0 g + Mytomicin 8–10 mg + Cysplatin 40–60 mg
POSTOP: FAP or FMP
5 Biffi (16) 69 1999–2005 N = 34 N = 35 Gastric cancer D2
2010 PREOP: 21days cycle TCF (docetaxel 75 mg/m2 1 h IVday 1, cisplatin 75 mg/m 4 h IV day 1 5-Fu
300 mg/m per day continuous IV infusion day 1–14)

123
6 Yonemura (17) 55 1988–1991 N = 29 N = 26 Gastric cancer
1993 PREOP: Cisplatin 75 mg/m + Mytomicin 10 mg/m2 + Etoposide 150 mg/body + UFT
(Tegafur + Uracil) 400 mg/d every 3 weeks
7 Nio (18) 295 1991–1999 N = 102 N = 193 Gastric cancer D2
2004 PPREOP and POSTOP: UFT (tegafur 8 mg/kg/d + uracil 6 mg/kg/d) over 2 weeks
8 Sun (19) 55 2008–2010 N = 29 N = 26 Gastric cancer NR
2011 3 cycles PREOP and 3 Cycles POSTOP
Docetaxel 75 mg/m2 + 5 fluoruracil 500 mg/m2 + Leucovirin 200 mg/m2
9 Kobayashi (20) 171 1990–1993 N = 91 N = 80 Gastric cancer D2
2000 PREOP and POSTOP
5-FU 610 mg/m2 day > 10 d
10 Lygidakis (21) 58 1995–1999 N = 39 N = 19 Gastric cancer D3 76%
1999 Neo Abdominal stop flow hypoxic perfusion CHT: Mytomicin C 5 Fluoruracil Leucovorin D2 24%
Farmorubicin + systemic chemiotherapy Group A N = 20;
Without CHT Group B N = 19
11 Shchepotin (22) 97 NR N = 47 N = 50 Gastric cancer
1995
12 Wang (23) 60 1987–1988 N = 30 N = 30 Gastric cancer
2000 FPLC oral
13 Cunningham (6) 503 1994–2002 N = 250 N = 253 Gastric cancer + Oesophagogastric D2 mainly
2006 PERIOP: 3 cycles preoperatively + 3 cycles postoperatively junction
Each cycle ECF (Epirubicin 50 mg/m2 + Cisplatin 60 mg/m2 + Fluoruracil 200 mg/m2
14 Schumacher (11) 144 1994–2004 N = 72 N = 72 Gastric cancer + Oesophagogastric D2
2010 PREOP: Two −48-day cycles of Cisplatin 50 mg/m2 + L-Folinic Acid 500 mg/m2 junction
IV + Fluoruracil 2000 mg/m2
15 Ychou (13) 224 1995–2003 N = 113 N = 111 Gastric cancer + Oesophagogastric D2
2011 PREOP: 3 cycles of Cisplatin 100 mg/m2 + 5-FU 800 mg/m2 for five days every 28 days and 3 or junction
4 postoperative same cycles
International Journal of Surgery 51 (2018) 120–127
F. Coccolini et al. International Journal of Surgery 51 (2018) 120–127

Fig. 1. Mortality (A: 1-year, B: 2-years, C: 3-years,


D: 5-years).

aspects may reduce the risk of complications by reducing the surgical the NACT; for this reason it should be considered a re-staging of the
effort. As a counterpart, however, if the NACT is not effective on the disease during the NACT.
specific tumor biology the risk is to have a disease progression during Recently the new TNM 8th classification of neoplastic diseases

124
F. Coccolini et al. International Journal of Surgery 51 (2018) 120–127

Fig. 2. Morbidity.

redefined the classification of the gastric and gastro-esophageal junc- published to demonstrate the efficacy of intra-peritoneal chemotherapy
tion tumors and formally included the EGC among the gastric cancers (IPC) [28]. The association of IPC and NACT is potentially very effective
[7]. This meta-analysis confirms the same behavior of the EGC and the as showed by the studies of the Peritoneal Surface Oncology Group
AGC according to the clinical answer to NACT in the long-term results. International (PSOGI) [29] that promotes the bidirectional approach to
The AGC and EGC considering the adenocarcinoma histology could be the AGC with peritoneal carcinosis combining novel comprehensive
considered very similar according to the biological behavior. The main treatment consisting of cytoreductive surgery and perioperative che-
difference is the anatomical diffusion due to the localization, to the motherapy for the treatment of peritoneal metastases from gastric
different anatomy of the two regions and the consequent lymphatic cancer with curative intent. The NACT could potentially have a role
drainage. This issue remains a matter of great interest but from the also in the prevention and treatment of peritoneal carcinosis (PC) but
chemotherapy point of view the unification of these previously differ- more effective could be the associated use with the neoadjuvant IPC to
entiated anatomical and hysto-pathological entities could facilitate the prevent the PC occurrence. In fact where serosal invasion exists the PC
therapeutic algorithms without detrimental effects on the long-term is practically unavoidable. A recent study showed promising results
survival. with the association of NACT and prophylactic hyperthermic in-
In order to further improve the outcomes of such hard disease to traperitoneal chemotherapy [30]. Lastly it could be considered the
treat with curative intent, several studies and meta-analysis have been positive effect of the IPC in all those patients who cannot complete the

Fig. 3. Perioperative mortality.

125
F. Coccolini et al. International Journal of Surgery 51 (2018) 120–127

Fig. 4. Recurrence rate in Esophago-Gastric Cancer.

postoperative chemotherapy. In fact almost the 50% of operated pa- Author contribution
tients are not able to be undergone or to complete the adjuvant che-
motherapy. Two randomized studies by Chunningham and Ychou re- Federico Coccolini, Matteo Nardi, Giulia Montori, Marco Ceresoli,
ported a percentage of 65.6% and 41.6% and of 50% and 44% of Andrea Celotti, Stefano Cascinu, Paola Fugazzola, Matteo Tomasoni,
patients who respectively started and completed the postoperative Olivier Glehen, Fausto Catena, Yutaka Yonemura, Luca Ansaloni con-
chemo-therapy in the first and in the second study [6,13]. In this per- tributed to the design and implementation of the research, to the ana-
spective, to warrant the best chemotherapeutical coverage to all these lysis of the results and to the writing of the manuscript.
patients, the use of IPC could be a hypothesis to be taken into con-
sideration. In fact the eventual adjunct of adjuvant chemotherapy after Guarantor
the IPC would be the optimum in advanced tumors even in absence of
PC [28–30]. Federico Coccolini.
The main criticism of the present study is the incomplete patients
selection and staging (advanced and early GC) with lack of considera- Acknowledgment
tion for the intraperitoneal cytological status. In fact the presence of a
positive cytology in intraperitoneal lavage brings different overall and None.
disease free survival outcomes [31,32]. If the different studies would
had been homogeneously considered the staging and the cytological References
status, results could have been potentially different, with a hypothetical
improvement in efficacy of NACT in patients with negative cytology [1] J.R. Kelley, J.M. Duggan, Gastric cancer epidemiology and risk factors, J. Clin.
and with different subgroup analysis for the AGC patients. Moreover it Epidemiol. 56 (1) (2003) 1–9.
[2] Parkin, DM.; Pisani, P.; Ferlay, J., Global cancer statistics. CA Cancer J Clin. n.d.
isn't possible to perform subgroup analysis according to the histology of 49(1):33–64. 1.
the included patients. These factors could contribute to explain the [3] S.S. Devesa, W.J. Blot, J.F. Fraumeni, Changing patterns in the incidence of eso-
different short-term outcomes in AGC and EGC. It could be hypothe- phageal and gastric carcinoma in the United States, Cancer 83 (10) (1998)
2049–2053.
sized that the patients affected by AGC that deceased in the short-term, [4] M. Pera, Epidemiology of esophageal cancer, especially adenocarcinoma of the
could have been those with the more advanced stages, the positive esophagus and esophagogastric junction, Recent Results Canc. Res. 155 (2000)
cytology or those with more aggressive histology. As a consequence the 1–14.
[5] L. Jiang, K.H. Yang, Q.L. Guan, Y. Chen, P. Zhao, J.H. Tian, Survival benefit of
NACT could potentially have good survival results in AGC even in the neoadjuvant chemotherapy for resectable cancer of the gastric and gastro-
short-term with a complete patients selection and staging, meaning esophageal junction, J. Clin. Gastroenterol. 49 (5) (2015) 387–394, http://dx.doi.
with the use of intra-peritoneal cytology, explorative laparoscopy and org/10.1097/MCG.0000000000000212.
[6] D. Cunningham, W.H. Allum, S.P. Stenning, et al., Perioperative chemotherapy
eco-endoscopy.
versus surgery alone for resectable gastroesophageal cancer, N. Engl. J. Med. 355
(1) (2006) 11–20, http://dx.doi.org/10.1056/NEJMoa055531.
[7] J.A. Ajani, H. In, T. Sano, et al., Stomach, Amin MB E. AJCC Cancer Staging Manual,
5. Conclusion eighth ed., 2017.
[8] R. DerSimonian, N. Laird, Meta-analysis in clinical trials, Contr. Clin. Trials 7 (3)
(1986) 177–188.
NACT reduces the mortality in gastric and esophago-gastric cancer. [9] Demets, DL., Methods for combining randomized clinical trials: strengths and
Morbidity and perioperative mortality are not influenced by NACT. The limitations. Stat. Med. n.d. 6(3):341–350.
overall recurrence rate is reduced by NACT in esophago-gastric cancer. [10] M. Imano, T. Itoh, T. Satou, et al., Prospective randomized trial of short-term
neoadjuvant chemotherapy for advanced gastric cancer, Eur. J. Surg. Oncol. 36 (10)
(2010) 963–968, http://dx.doi.org/10.1016/j.ejso.2010.06.012.
[11] C. Schuhmacher, S. Gretschel, F. Lordick, et al., Neoadjuvant chemotherapy com-
Conflicts of interest pared 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. 28 (35) (2010) 5210–5218, http://dx.doi.org/10.1200/JCO.
All authors declare to have no conflict of interest. 2009.26.6114.
[12] H.H. Hartgrink, C.J.H. van de Velde, H. Putter, et al., Neo-adjuvant chemotherapy
for operable gastric cancer: long term results of the Dutch randomised FAMTX trial,
Sources of funding Eur. J. Surg. Oncol. 30 (6) (2004) 643–649, http://dx.doi.org/10.1016/j.ejso.2004.
04.013.
[13] M. Ychou, V. Boige, J.-P. Pignon, et al., Perioperative chemotherapy compared with
All authors declare to have no sources of funding for this research. surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and
FFCD multicenter phase III trial, J. Clin. Oncol. 29 (13) (2011) 1715–1721, http://
dx.doi.org/10.1200/JCO.2010.33.0597.
[14] S. Hashemzadeh, A. Pourzand, M.H. Somi, S. Zarrintan, R. Javad-Rashid,
Ethical approval
A. Esfahani, The effects of neoadjuvant chemotherapy on resectability of locally-
advanced gastric adenocarcinoma: a clinical trial, Int. J. Surg. 12 (10) (2014)
Not required. 1061–1069, http://dx.doi.org/10.1016/j.ijsu.2014.08.349.
[15] C.W. Zhang, S.C. Zou, D. Shi, D.J. Zhao, Clinical significance of preoperative re-
gional intra-arterial infusion chemotherapy for advanced gastric cancer, World J.
Gastroenterol. 10 (20) (2004) 3070–3072.
Research registration unique identifying number (UIN) [16] R. Biffi, N. Fazio, F. Luca, et al., Surgical outcome after docetaxel-based neoadju-
vant chemotherapy in locally-advanced gastric cancer, World J. Gastroenterol. 16
Reviewregistry412. (7) (2010) 868–874.

126
F. Coccolini et al. International Journal of Surgery 51 (2018) 120–127

[17] Yonemura, Y.; Sawa, T.; Kinoshita, K.; et al., Neoadjuvant chemotherapy for high- resectable gastric cancer, in: E. Wu (Ed.), The Cochrane Database of Systematic
grade advanced gastric cancer. World J Surg n.d. 17(2):256–261-2. Reviews, John Wiley & Sons, Ltd, Chichester, UK, 2007CD005047.
[18] Nio, Y.; Koike, M.; Omori, H.; et al., A randomized consent design trial of neoad- [27] S. Al-Batran, N. Homann, H. Schmalenberg, H. Kopp, G. Haag, K. Luley,
juvant chemotherapy with tegafur plus uracil (UFT) for gastric cancer–a single in- W. Schmiegel, G. Folprecht, S. Probst, N. Prasnikar, P. Thuss-Patience,
stitute study. Anticancer Res. n.d. 24(3b):1879–1887. W. Fischbach, J. Trojan, M. Koeni, RH, Perioperative Chemotherapy with
[19] X.-C. Sun, J. Lin, A.-H. Ju, Treatment of Borrmann type IV gastric cancer with a Docetaxel, Oxaliplatin, and Fluorouracil/leucovorin (FLOT) versus Epirubicin,
neoadjuvant chemotherapy combination of docetaxel, cisplatin and 5-fluorouracil/ Cisplatin, and Fluorouracil or Capecitabine (ECF/ECX) for Resectable Gastric or
leucovorin, J. Int. Med. Res. 39 (6) (2011) 2096–2102, http://dx.doi.org/10.1177/ Gastroesophageal Junction (GEJ) Adenocarcinoma (FLOT4-aio): a Mul, Available
147323001103900605. from: http://abstracts.asco.org/199/AbstView_199_191595.html , Accessed date:
[20] T. Kobayashi, T. Kimura, Long-term outcome of preoperative chemotherapy with 5’- 11 July 2017.
deoxy-5-fluorouridine (5’-DFUR) for gastric cancer, Gan To Kagaku Ryoho 27 (10) [28] F. Coccolini, E. Cotte, O. Glehen, et al., Intraperitoneal chemotherapy in advanced
(2000) 1521–1526. gastric cancer. Meta-analysis of randomized trials, Eur. J. Surg. Oncol. 40 (1) (2014)
[21] Lygidakis, NJ.; Sgourakis, G., Aphinives P., Upper abdominal stop-flow perfusion as 12–26, http://dx.doi.org/10.1016/j.ejso.2013.10.019.
a neo and adjuvant hypoxic regional chemotherapy for resectable gastric carci- [29] Y. Yonemura, E. Canbay, Y. Li, et al., A comprehensive treatment for peritoneal
noma. A prospective randomized clinical trial. Hepato-Gastroenterology n.d. metastases from gastric cancer with curative intent, Eur. J. Surg. Oncol. 42 (8)
46(27):2035–2038. (2016) 1123–1131, http://dx.doi.org/10.1016/j.ejso.2016.03.016.
[22] I. Shchepotin, S. Evans, V. Chorny, et al., Preoperative superselective intraarterial [30] F. Coccolini, A. Celotti, M. Ceresoli, et al., Hyperthermic intraperitoneal che-
chemotherapy in the combined treatment of gastric-carcinoma, Oncol. Rep. 2 (3) motherapy (HIPEC) and neoadjuvant chemotherapy as prophylaxis of peritoneal
(1995) 473–479. carcinosis from advanced gastric cancer—effects on overall and disease free sur-
[23] Wang, XL.; Wu, GX.; Zhang, MD.; Guo, M.; Zhang, H.; Sun, XF., A favorable impact vival, J. Gastrointest. Oncol. 7 (4) (2016) 523–529, http://dx.doi.org/10.21037/
of preoperative FPLC chemotherapy on patients with gastric cardia cancer. Oncol. jgo.2016.06.05.
Rep. n.d. 7(2):241–244. [31] Y. Yonemura, T. Kawamura, E. Bandou, G. Tsukiyama, Y. Endou, M. Miura, The
[24] W. Li, J. Qin, Y.-H. Sun, T.-S. Liu, Neoadjuvant chemotherapy for advanced gastric natural history of free cancer cells in the peritoneal cavity, Recent Results Canc.
cancer: a meta-analysis, World J. Gastroenterol. 16 (44) (2010) 5621–5628. Res. 169 (2007) 11–23.
[25] A.-M. Xu, L. Huang, W. Liu, S. Gao, W.-X. Han, Z.-J. Wei, Neoadjuvant che- [32] M. Pecqueux, J. Fritzmann, M. Adamu, et al., Free intraperitoneal tumor cells and
motherapy followed by surgery versus surgery alone for gastric carcinoma: sys- outcome in gastric cancer patients: a systematic review and meta-analysis,
tematic review and meta-analysis of randomized controlled trials, PLoS One 9 (1) Oncotarget 6 (34) (2015) 35564–35578, http://dx.doi.org/10.18632/oncotarget.
(2014) e86941, , http://dx.doi.org/10.1371/journal.pone.0086941. 5595.
[26] A. Wu, G. Xu, H. Wang, J. Ji, J. Tang, Neoadjuvant chemotherapy versus none for

127

You might also like