Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

Effect of neoadjuvant chemotherapy combined with arterial chemoembolization on short-term clinical outcome of locally advanced gastric cancer

verfasst von: Jianguo Yang, Juncai Li, Qican Deng, Zhenzhou Chen, Kuan He, Yajun Chen, Zhongxue Fu

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Background

The purpose of this study was to explore the short-term efficacy and safety of neoadjuvant chemotherapy combined with arterial chemoembolization for locally advanced gastric cancer (LAGC).

Methods

We retrospectively analyzed the clinical data of 203 patients with LAGC who received neoadjuvant therapy from June 2019 to December 2021. The patients were divided into a neoadjuvant chemotherapy combined with arterial chemoembolization group (combined group, n = 102) and a neoadjuvant chemotherapy group (conventional group, n = 101). The adverse events of chemotherapy, postoperative complications and pathological complete response (pCR) rate were compared between the two groups. Univariate and multivariate analyses were performed to evaluate the potential factors affecting pCR.

Results

A total of 78.8% of the patients were in clinical stage III before neoadjuvant therapy. A total of 52.2% of the patients underwent surgery after receiving two cycles of neoadjuvant therapy. There were 21.2% patients with ≥ grade 3 (CTCAE 4.0) adverse events of chemotherapy and 11.3% patients with Clavien-Dindo classification ≥ grade 3 postoperative complications. Compared with the conventional group, the combination group did not experience an increase in the adverse events of chemotherapy or postoperative complications. The pCR rate in the combined group was significantly higher than that in the conventional group (16.7% vs. 4.95%, P = 0.012). The multivariate analysis showed that arterial chemoembolization, pre-treatment neutrophil-to-lymphocyte ratio (NLR) and pre-treatment platelet-to-lymphocyte ratio (PLR) were independent factors affecting pCR.

Conclusion

Neoadjuvant chemotherapy combined with arterial chemoembolization contributed to improving the pCR rate of LAGC patients. Arterial chemoembolization, pre-treatment NLR and pre-treatment PLR were also predictors of pCR.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-023-10712-w.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Gastric cancer is a global disease with high morbidity and mortality [1]. With the improvement of quality of life and treatment methods, a steady downward trend in the incidence and mortality rates of gastric cancer has been observed [2]. Nonetheless, over 1 million new cases of gastric cancer and 760,000 deaths are still reported worldwide each year, making it the fifth most diagnosed cancer and the third most common cause of cancer-related death globally [3].
Although progress has been made in biological research on the occurrence and development of gastric cancer, radical surgery is still the most important treatment approach for resectable gastric cancer [4, 5]. Studies have reported that 50–70% of patients are diagnosed with locally advanced or advanced gastric cancer at the first visit [6, 7]. Moreover, the 5-year overall survival of resectable locally advanced gastric cancer (LAGC) is 20–30% after radical surgery [810]. In recent years, the treatment of LAGC has been transformed from a single operation mode to a comprehensive treatment mode based on surgery combined with neoadjuvant chemoradiotherapy, targeted therapy and immunotherapy [1115]. Compared with surgery alone, the conceivable advantages of preoperative neoadjuvant therapy involve downstaging, increasing the radical resection rate, and improving survival outcomes [1113, 16]. However, the optimal regimen of neoadjuvant chemotherapy is inconclusive, and there are East-West differences. The PRODIGY study in South Korea showed that DOS for 3 cycles before surgery for LAGC could downstage tumors and significantly improve PFS [12]. The FLOT4-AIO trial found that compared with the ECF/ECX chemotherapy regimen, the FLOT regimen could acquire higher pathological complete response (pCR) rate and R0 resection rate without increasing adverse effects, thereby improving the 5-year overall survival (OS) of patients (45% vs. 36%) and disease-free survival (DFS) (41% vs. 31%) [13]. Transcatheter arterial chemoembolization has been widely used in the treatment of hepatocellular carcinoma and has become the first-line treatment strategy for intermediate stage liver cancer [17]. Arterial chemotherapy or chemoembolization has rarely been reported in gastric cancer. Some studies have shown that systemic chemotherapy combined with arterial chemotherapy can significantly improve the outcomes of patients with advanced gastric cancer [1821].
However, whether neoadjuvant chemotherapy combined with arterial chemoembolization is superior to neoadjuvant chemotherapy in efficacy and adverse events in patients with LAGC has not yet been evaluated. Therefore, we conducted this retrospective study to assess the short-term efficacy, safety, and feasibility of neoadjuvant chemotherapy combined with arterial chemoembolization for resectable LAGC.

Materials and methods

Patients

We retrospectively reviewed the medical records of gastric cancer patients hospitalized in the Department of Gastrointestinal Surgery of the First Affiliated Hospital of Chongqing Medical University from June 2019 to December 2021. The inclusion criteria were as follows: (1) Pathologically confirmed gastric adenocarcinoma or mucinous adenocarcinoma, (2) Locally advanced gastric cancer (cT1 − 2 N+M0 or cT3 − 4b Any NM0) and resectable, [22, 23] (3) two or more cycles of neoadjuvant therapy (chemotherapy only or chemotherapy combined with arterial chemoembolization), and (4) Completed radical surgery [24]. The exclusion criteria were as follows: 1. synchronous other malignancies, 2, incomplete medical records, and 3. palliative surgery or emergency surgery. Finally, 203 patients with LAGC were enrolled, including 101 in the conventional group and 102 in the combined group (Fig. 1). This study was approved by the Ethics Review Committee of the First Affiliated Hospital of Chongqing Medical University, and written informed consent was obtained from the patients (Approval number: 20,192,801).

Laboratory and imaging examination

One week before neoadjuvant therapy, all patients received routine blood tests, liver and kidney function tests, and serum tumor-related antigen tests (e.g., CEA, CA19-9). Previous studies reported that pre-treatment blood biomarkers can predict tumor regression response [25, 26]. Therefore, we analyzed the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) before neoadjuvant therapy. Gastroscopy, gastrointestinal ultrasonography, and contrast-enhanced CT of the chest and abdomen were performed to evaluate the clinical stage of the tumor and the effect of neoadjuvant therapy.

Treatment

Chemotherapy

Patients with LAGC underwent 2–4 cycles of chemotherapy before radical surgery. The preoperative chemotherapy regimen included the DOS regimen: docetaxel and oxaliplatin (50 mg/m2 and 100 mg/m2) were given intravenously on Day 1, and S-1 (40 mg/m2) was given orally twice a Days on day 1–14; The NPOS regimen: included intravenous nab-paclitaxel (260 mg/m2) and oxaliplatin (85 mg/m2) on Day 1, oral S-1 (40 mg/m2) twice a day from Days 1 to 14.

Arterial chemoembolization

A 5 F vascular sheath was inserted by retrograde puncture of the right femoral artery by the Seldinger technique, [27] and then was placed into the celiac axis. The contrast medium was injected to show the celiac trunk and its branches. A 2.9 F microcatheter and a 2.7 F microguide wire were used to superselect the main blood supply arteries of the tumor. Angiographic vessels were selected according to tumor location. For cardia and fundus carcinomas, catheters were inserted into the left gastric artery and the left inferior phrenic artery. Lesser curvature gastric catheters were inserted into the left and right gastric arteries. For tumors of the greater curvature of the stomach, right gastroepiploic artery and gastroduodenal arteriography were performed. Gastroduodenal and right gastric arteriograms were performed if the tumor was located in the gastric antrum. Arteries for chemotherapy and embolization were identified based on angiographic findings. The chemotherapy regimen was oxaliplatin (100 mg/m2) and docetaxel (50 mg/m2)/nab-paclitaxel (260 mg/m2). After arterial chemotherapy was completed, lipiodol (5 ml) was used to embolize the blood vessels and reimaging was performed to ensure complete embolization of the blood vessels supplying the tumor. After arterial chemoembolization was completed, the patients took oral S-1 from Day 1 to Day 14. Then, 1–3 cycles of intravenous chemotherapy were performed before surgery. (Fig. S1)

Surgical procedure

The imaging evaluation was re-evaluated within 3–4 weeks after the completion of 2 or more neoadjuvant treatment cycles. If the tumor regressed well, the operation was performed within 3–4 weeks after the completion of neoadjuvant therapy after the evaluation of 2 gastrointestinal tumor surgeons with 10 years of experience. The decision to use laparoscopic gastrectomy was based on tumor stage, history of abdominal surgery, and tolerability of laparoscopic surgery. Surgical schemes for gastric cancer after neoadjuvant therapy include: distal gastrectomy with No. 1/3/4sb/4d/5/6/7/8a/9/11p/12a lymph node dissection; and total gastrectomy with No. 1/2/3/4sa/4sb/4d/5/6/7/8a/9/11p/11d/12a lymph node dissection. Billroth I gastroduodenostomy, Billroth II gastrojejunostomy, and Roux-en-Y gastrojejunostomy were employed for gastrointestinal reconstruction after distal gastrectomy. The Roux-en-Y esophagojejunostomy was used to reconstruct the digestive tract after total gastrectomy [24, 28].

Neoadjuvant therapy adverse events and pathological response assessment

The Common Terminology Criteria for Adverse Events (CTCAE 4.0) was utilized to record the adverse effects of chemotherapy. The main adverse events of neoadjuvant therapy included: leukopenia, neutropenia, thrombocytopenia, liver and kidney dysfunction, febrile neutropenia, nausea and vomiting, etc. Postoperative morbidity was defined as complications occurring within 30 days of surgery or during hospitalization. Postoperative complications were assessed using the Clavien-Dindo classification system [29]. Pathological response to neoadjuvant therapy was evaluated in accordance with tumor regression grade (TRG) [30]. Patients with TRG 0–1 have a good response, while those with TRG 2–3 have a poor response. pCR was defined as the absence of any residual tumor cells in gastric and dissected lymph node specimens on postoperative histological evaluation after neoadjuvant therapy. HER-2 positivity was defined as IHC 3 + or FISH positivity [24].

Statistical analysis

Receiver operating characteristic (ROC) analysis and relative area under the curve (AUC) statistics were applied to select the ratio of the point of maximum sensitivity and specificity as the optimal cut-off values for pre-treatment NLR and pre-treatment PLR. Categorical variables were analyzed using the Chi-square or Fisher’s exact test. Continuous variables were expressed as the median (interquartile range), and differences between the two groups were analyzed by unpaired t test or the Mann-Whitney U rank sum test. Univariate and multivariate analyses were performed using binary logistic regression models to explore the factors affecting pCR and tumor regression. Variables with P < 0.10 in the univariate analysis were included in the multivariate analysis. Nomograms were drawn based on predictors of pCR and TRG in the multivariate analysis, and the performance of the nomograms was assessed using internal validation and AUC. Moreover, the discriminative power of the nomogram was evaluated by the C-index. P < 0.05 was considered statistically significant. Statistical analysis was performed by the SPSS statistical package version 22.0 (SPSS, Chicago, IL, USA) and R software (Version 4.0.1.).

Results

Patient characteristics

A total of 203 patients with LAGC were enrolled in this study, including 101 in the conventional group and 102 in the combined group. The baseline characteristics of the patients are summarized in Table 1. Among the 203 patients, the median age was 58 years, and most of them (75.37%) were male. There were no significant differences in hemoglobin level, neutrophil count, lymphocyte count, CEA level or CA19-9 level between the two groups before neoadjuvant therapy. The majority of the patients (74.88%) had T4 stage disease, 175 (86.21%) patients had lymph node metastasis, 43 (30.46%) had clinical II stage disease, and 160 (69.54%) had clinical III stage disease. A total of 53.48% of the patients received NPOS chemotherapy, and 46.52% received DOS chemotherapy. In the combined group, 11 patients underwent 2 sessions of arterial chemoembolization therapy. Moreover, approximately half of the patients underwent surgery after 2 cycles of neoadjuvant therapy.
Table 1
Patient characteristics
Characteristic
Conventional group (n = 101)
Combined group (n = 102)
P
Sex
  
0.254
 
Male
80 (79.2%)
73 (71.6%)
 
 
Female
21 (20.8%)
29 (28.4%)
 
Age
Median (IQR, years)
57 (50–65)
61 (51-67.25)
0.151
 
< 60 years
59 (58.4%)
49 (48%)
0.16
BMI
Median (IQR, kg/m2)
21.64 (20.24–24.22)
22.15 (20.48–24.22)
0.637
Smoking
59 (58.4%)
60 (58.8%)
1
Drinking
53 (52.5%)
59 (57.8%)
0.482
Comorbidity
    
 
Hypertension
14 (13.9%)
16 (15.7%)
0.844
 
Coronary heart disease
3 (3%)
4 (3.9%)
1
 
Diabetes
14 (13.9%)
8 (7.8%)
0.183
 
Other
15 (14.9%)
12 (11.8%)
0.542
Family history
4 (4%)
7 (6.9%)
0.537f
ASA
   
0.622
 
1
2 (2%)
1 (1%)
 
 
2
63 (62.4%)
56 (54.9%)
 
 
3
35 (34.6%)
44 (43.1%)
 
 
4
1 (1%)
1(1%)
 
Pretreatment Hb (IQR, g/L)
118 (86–133)
125 (92–139)
0.17
Pretreatment neutrophils (IQR, *108)
3.74 (3.09–4.76)
3.72 (2.92–4.92)
0.969
Pretreatment lymphocyte (IQR, *108)
1.37 (1.06–1.68)
1.41 (1.13–1.73)
0.574
Pretreatment platelet (IQR, *108)
243 (203–351)
235 (181–307)
0.235
Pretreatment albumin (IQR, g/L)
38 (34–41)
31 (33–40)
0.567
Pretreatment CEA(>5.2 ng/ml)
36 (35.6%)
26 (25.5%)
0.129
Pretreatment CA19-9 (> 27 U/ml)
34 (33.7%)
29 (28.4%)
0.451
Tumor location
  
0.766
 
Upper stomach
35 (34.6%)
36 (35.3%)
 
 
Middle stomach
22 (21.8%)
26 (25.5%)
 
 
Lower stomach
44 (43.6%)
40 (39.2%)
 
Tumor size (IQR, cm)
6 (5-7.7)
5.57 (4.6–6.8)
0.236
Borrmann typing
  
0.49
 
I
26 (25.7%)
34 (33.3%)
 
 
II
12 (11.9%)
12 (11.8%)
 
 
III
51 (34.6%)
49 (48%)
 
 
IV
12 (11.9%)
7 (6.9%)
 
Clinical T stage
  
0.859
 
T3
24 (23.8%)
27 (26.5%)
 
 
T4a
70 (69.3%)
67 (65.7%)
 
 
T4b
7 (6.9%)
8 (7.8%)
 
Clinical N stage
  
0.374
 
N0
11 (10.9%)
17 (16.7%)
 
 
N1
29 (28.7%)
27 (26.5%)
 
 
N2
42 (41.6%)
46 (45.1%)
 
 
N3
19 (18.8%)
12 (11.8%)
 
Clinical TNM stage
  
0.825
 
IIa
2 (2%)
3 (2.9%)
 
 
IIb
16 (15.8%)
22 (21.6%)
 
 
IIIa
31 (30.7%)
30 (29.4%)
 
 
IIIb
35 (34.7%)
33 (32.4%)
 
 
IIIc
17 (16.8%)
14 (13.7%)
 
Chemotherapy regimen
  
0.152
 
NPOS
56 (55.4%)
67 (65.7%)
 
 
DOS
45 (44.6%)
35 (34.3%)
 
Number of neoadjuvant therapy cycles
  
0.175
 
2
49 (48.5%)
57 (55.9%)
 
 
3
40 (39.6%)
40 (39.2%)
 
 
4
12 (11.9%)
5(4.9%)
 
Number of chemoembolization
   
 
1
 
91 (89.2%)
 
 
2
 
11(10.8%)
 
Abbreviations: IQR, Interquartile range; BMI, Body mass index; ASA, American Society of Anesthesiologists; Hb, Hemoglobin; CEA, Carcinoembryonic antigen; CA19-9, Carbohydrate antigen 19 − 9; NPOS, Nab-paclitaxel, oxaliplatin and S-1; DOS, Docetaxel, oxaliplatin and S-1

Neoadjuvant therapy adverse events

The adverse events during neoadjuvant therapy were evaluated using the CTCAE 4.0. (Table 2). Hematologic toxicity was assessed by routine blood tests and liver and kidney function tests on Days 7, 14, and 21 of each treatment cycle. A total of 43 patients experienced grade 3–4 adverse events, the majority of which were hematological toxicity. The number of patients with grade ≥ 3 neutropenia in the combined group was greater than that in the conventional group (11.8% vs. 4%), but there was no significant difference (P = 0.065). In addition, a total of 5 patients developed grade ≥ 3 febrile neutropenia. The main adverse events of grade 3–4 non-hematologic toxicity during neoadjuvant therapy were nausea and vomiting (n = 7), liver function impairment (n = 7), infection (n = 3), and diarrhea (n = 3).
Table 2
Neoadjuvant therapy adverse events
Grade (CTCAE v 4.0)
Conventional group (n = 101)
 
Combined group (n = 102)
Grade 1–2
Grade 3
Grade 4
≥Grade 3
Grade 1–2
Grade 3
Grade 4
≥Grade 3
All adverse events
60 (59.4%)
16 (15.8%)
3 (3%)
17 (16.8%) *
 
66 (64.7%)
25 (24.5%)
5 (4.9%)
26(25.5%) *
Leukopenia
26 (25.7%)
1 (1%)
1 (1%)
2 (2%)
25 (24.5%)
4 (3.9%)
4 (3.9%)
8 (7.8%)
Neutropenia
31 (30.7%)
3 (3%)
1 (1%)
4 (4%)
22 (21.6%)
8 (7.8%)
4 (3.9%)
12 (11.8%)
Thrombocytopenia
7 (6.9%)
1 (1%)
0
1 (1%)
7 (6.86%)
2 (2%)
0
2 (2%)
Febrile neutropenia
0
2 (2%)
0
2 (2%)
0
2 (2%)
1 (1%)
3 (2.9%)
Anemia
13 (12.9%)
9 (8.9%)
0
9 (8.9%)
19 (18.6%)
6 (7.8%)
1 (1%)
7 (6.86%)
Nausea and vomiting
16 (15.8%)
4 (4%)
0
4 (4%)
29 (28.4%)
3 (2.9%)
0
3 (2.9%)
Anorexia
11 (10.9%)
0
0
0
19 (18.6%)
1 (1%)
0
1 (1%)
ALT or AST increased
18 (17.8%)
2 (2%)
0
2 (2%)
13 (12.7%)
5 (4.9%)
0
5 (4.9%)
Serum creatinine increased
3 (3%)
0
0
0
1 (1%)
0
0
0
Infection
1 (1%)
0
0
0
0
3 (2.9%)
0
3 (2.9%)
Diarrhea
3 (3%)
3 (3%)
0
3 (3%)
1 (1%)
0
0
0
Other
2 (2%)
0
1 (1%)
1 (1%)
2 (2%)
2 (2%)
0
2 (2%)
*: A patient may have one or more grade 3–4 adverse events

Details of surgery and postoperative clinical outcomes

Laparoscopic gastric resection was performed in 192 patients, of whom 57.8% underwent total gastrectomy with lymph node dissection (D2). Thirteen patients underwent combined organ resection (spleen, pancreas, or liver) due to tumor invasion into adjacent organs. The median operative time in the combined group was shorter than that in the conventional group (P = 0.029). Compared with the conventional group, the median intraoperative blood loss was less in the combined group (P = 0.011). Moreover, the patients in the combined group had less liquid diet time and hospital stay after operation than those in the conventional group. The details of the surgery are shown in Table 3.
Table 3
The detail of surgery
  
Conventional group (n = 101)
Combined group (n = 102)
P
Surgical approach
   
0.291
 
Laparoscopy
93 (92.1%)
99 (97.1%)
 
 
Open
3 (3%)
1 (1%)
 
 
Conversion to open
5 (4.9%)
2 (2%)
 
Gastrectomy type
   
0.261
 
Total gastrectomy
51 (50.5%)
60 (58.8%)
 
 
Distal gastrectomy
50 (49.5%)
42 (41.2%)
 
Combined resection
 
8 (7.9%)
5 (4.9%)
0.407
Residual tumor status
  
1
 
R0
98 (97%)
99 (97.1%)
 
 
R1
3 (3%)
3 (2.9%)
 
Operation time (IQR, min)
222 (175-267.5)
200 (160–245)
0.029
Blood loss (IQR, ml)
100 (50–200)
75 (50-175.25)
0.011
Blood transfusion
17 (16.8%)
18 (17.6%)
1
Intraoperative complications
3 (3%)
3 (2.9%)
1
Time of liquid diet (IQR, day)
7 (5–10)
6 (5–8)
0.006
Postoperative hospital stays (IQR, day)
11 (8–17)
10 (8–13)
0.012
Reoperation
4 (4%)
5 (4.9%)
1
Readmission
2 (2%)
1 (1%)
0.621
Mortality
1 (1%)
1 (1%)
1
Abbreviations: IQR, Interquartile range
Postoperative complications that occurred during hospitalization or within 30 days of surgery were recorded and classified using the Clavien-Dindo classification system (Table 4). A total of 23 patients experienced grade ≥ 3 postoperative complications. Major grade ≥ 3 postoperative complications included anastomotic leakage (n = 14), intra-abdominal infection (n = 11), and pulmonary infection (n = 6). In addition, 9 patients underwent reoperation due to postoperative complications, including anastomotic leakage (n = 5), hemorrhage (n = 2), and wound infection (n = 2). There was no significant difference in any complication events of grade ≥ 3 between the two groups (P = 0.659).
Table 4
Postoperative complications
Clavien–Dindo
classification
Conventional group (n = 101)
 
Combined group (n = 102)
Grade I-II
Grade III
Grade IV
Grade V
≥III grade
Grade I-II
Grade III
Grade IV
Grade V
≥III grade
Anastomotic leakage
8 (7.9%)
4 (4%)
1 (1%)
1 (1%)
6 (5.9%)
 
8 (7.8%)
5 (4.9%)
2 (2%)
1 (1%)
8 (7.9%)
Abdominal infection
26 (25.7%)
2 (2%)
2 (2%)
0
4 (4%)
 
18 (17.6%)
4 (3.9%)
3 (2.9%)
0
7 (6.9%)
Intestinal obstruction
15 (14.9%)
0
0
0
0
 
3 (2.9%)
2 (2%)
1 (1%)
0
3 (2.9%)
Hemorrhage
1 (1%)
0
2 (2%)
0
2 (2%)
 
1 (1%)
1 (1%)
2 (2%)
0
3 (2.9%)
Wound infection
3 (3%)
2 (2%)
0
0
2 (2%)
 
0
3 (2.9%)
0
0
3 (2.9%)
Pulmonary infection
23 (22.8%)
2 (2%)
0
0
2 (2%)
 
21 (20.6%)
3 (2.9%)
1 (1%)
0
4 (3.9%)
Cardiovascular events
0
1 (1%)
0
0
1 (2%)
 
4 (3.9%)
0
0
0
0
Thrombotic events
1 (1%)
0
0
0
0
 
3 (2.9%)
0
0
0
0
Urinary tract infection
3 (3%)
0
0
0
0
 
4 (3.9%)
0
0
0
0
Any complication events
44 (43.6%)
9 (8.9%)
3 (3%)
1 (1%)
10 (9.9%)
 
38 (37.3%)
11 (10.8%)
4 (3.9%)
1 (1%)
13 (12.7%)

Pathological characteristics

The predominant pathological type of the patients included in the study was adenocarcinoma (n = 186). After neoadjuvant therapy, 10.8% of the patients achieved ypT0, and 46.8% of the patients were negative for lymph node metastasis. In addition, TRG 0–1 was identified in 53 patients. Finally, twenty-two patients achieved pCR in the primary tumor and lymph nodes, and 17 patients were from the combined group (Table 5).
Table 5
Pathological Characteristics
  
Conventional group (n = 101)
Combined group (n = 102)
P
Pathological Type
  
0.314
 
Adenocarcinoma
94 (93.1%)
92 (90.2%)
 
 
Mucinous
7 (6.9%)
10 (9.8%)
 
Degree of differentiation
  
0.661
 
Poorly/Mucinous
49 (48.5%)
54 (52.9%)
 
 
Moderate
51 (50.5%)
46 (45.1%)
 
 
Well
1 (1%)
2 (2%)
 
T Stage
   
0.03
 
ypT0
5 (5%)
17 (16.7%)
 
 
ypT1
7 (6.9%)
9 (8.8%)
 
 
ypT2
23 (12.9%)
17 (16.7%)
 
 
ypT3
54 (53.5%)
42 (41.2%)
 
 
ypT4
12 (11.9%)
18 (17.6%)
 
N Stage
   
0.055
 
ypN0
38 (37.6%)
57 (55.9%)
 
 
ypN1
17 (16.8%)
15 (14.7%)
 
 
ypN2
26 (25.7%)
15 (14.7%)
 
 
ypN3
20 (19.8%)
15 (14.7%)
 
M Stage
   
0.748
 
ypM0
97 (96%)
96 (94.1%)
 
 
ypM1
4 (4%)
6 (5.9%)
 
TRG
   
0.028
 
Grade 0
5 (5%)
17 (16.7%)
 
 
Grade 1
18 (17.8%)
23 (22.5%)
 
 
Grade 2
56 (55.4%)
46 (45.1%)
 
 
Grade 3
22 (21.8%)
16 (15.7%)
 
T Downstage
79 (78.2%)
75 (73.5%)
0.512
 N Downstage
47 (46.5%)
54 (52.9%)
0.401
Pathological Complete Response
5 (5%)
17 (16.7%)
0.012
Angiolymphatic invasion
12 (11.9%)
18 (17.6%)
0.323
Tumor deposit
6 (5.9%)
11 (10.8%)
0.311
HER2 positive
13 (12.9%)
9 (8.8%)
0.376
Abbreviations: TRG, Tumor regression response

Predictors of pCR to Neoadjuvant Therapy

In the ROC analysis, the AUCs of the pre-treatment NLR and pre-treatment PLR for pCR were 0.649 (P = 0.028) and 0.631 (P = 0.055), respectively, and the optimal cutoffs were 2.86 and 182, respectively (Fig. S2). Therefore, NLR < 2.86 was defined as low NLR, and PLR < 182 was defined as low PLR. In the univariate analysis, it was found that pCR was associated with pre-treatment NLR (OR = 0.355, 95% CI 0.133–0.948, P = 0.039) and pre-treatment PLR (OR = 0.137, 95% CI 0.039–0.478, P = 0.002). In addition, preoperative arterial chemoembolization therapy was also an important factor for pCR (OR = 3.84, 95% CI 1.359–10.853, P = 0.011). The characteristics of the variables with P < 0.1 in the univariate analysis were analyzed by multivariate analysis. The analysis results indicated that pre-treatment NLR (OR = 0.193, 95% CI 0.016–0.611, P = 0.005), pre-treatment PLR (OR = 0.077, 95% CI 0.018–0.333, P = 0.001) and arterial chemoembolization (OR = 3.766, 95% CI 1.177–12.054, P = 0.025) were critical predictive factors of pCR (Table 6).
Table 6
Univariate and multivariate logistic regression analysis for tumor response to neoadjuvant therapy
Patient characteristics
Univariate Analysis
Multivariate Analysis
OR
95% CI
P
OR
95% CI
P
Gender
      
 
Female
ref
     
 
Male
3.609
0.813–16.022
0.091
2.199
0.438–11.049
0.339
Age
      
 
<60 years
ref
     
 
≥ 60 years
1.744
0.71–4.284
0.225
   
BMI
      
 
<25 kg/m2
ref
     
 
≥ 25 kg/m2
1.966
0.71–5.445
0.194
   
Tumor location
  
0.988
   
 
Upper stomach
ref
     
 
Middle stomach
0.916
0.281–2.988
0.844
   
 
Lower stomach
0.945
0.344–2.593
0.913
   
Tumor size
      
 
<5.8 cm
ref
     
 
≥5.8 cm
0.824
0.339–2.004
0.67
   
Pre-treatment NLR
      
 
<2.86
ref
     
 
≥ 2.86
0.355
0.133–0.948
0.039
0.193
0.016–0.611
0.005
Pre-treatment PLR
      
 
<182
ref
     
 
≥ 182
0.137
0.039–0.478
0.002
0.077
0.018–0.333
0.001
CEA
      
 
<5.2ng/ml
ref
     
 
≥ 5.2ng/ml
1.628
0.657–4.035
0.292
   
CA19-9
      
 
<27U/ml
ref
     
 
≥ 27U/ml
0.624
0.219–1.773
0.376
   
Degree of differentiation
      
 
poor/mucinous
ref
     
 
moderate/well
0.994
0.409–2.417
0.989
   
HER-2
       
 
negative
ref
     
 
positive
2.756
0.352–21.561
0.334
   
Clinical T stage
  
0.907
   
 
T3
ref
     
 
T4a
0.854
0.309–2.357
0.76
   
 
T4b
1.154
0.208–6.413
0.87
   
Clinical N stage
  
0.062
  
0.064
 
N0
ref
  
ref
  
 
N1
0.208
0.048–0.905
0.036
0.445
0.069–2.875
0.395
 
N2
0.317
0.097–1.039
0.058
1.044
0.063–17.217
0.976
 
N3
0.88
0.248–3.128
0.843
5.391
0.303–95.933
0.251
Clinical TNM stage
      
 
II
ref
     
 
III
0.42
0.163–1.078
0.071
0.306
0.025–3.74
0.354
Chemotherapy regimen
      
 
DOS
ref
     
 
NPOS
1.156
0.461–2.896
0.757
   
Arterial chemoembolization
      
 
No
ref
     
 
Yes
3.84
1.359–10.853
0.011
3.766
1.177–12.054
0.025
Number of chemotherapy cycles
  
0.531
   
 
2
ref
     
 
3
0.541
0.186–1.579
0.261
   
 
4
0.806
0.215–3.024
0.749
   
Abbreviations: BMI, Body mass index; NLR, neutrophil-to-lymphocyte ratio; PLR, platelet-to-lymphocyte ratio; CEA, Carcinoembryonic antigen; CA19-9, Carbohydrate antigen 19 − 9; NPOS, Nab-paclitaxel, oxaliplatin and S-1; DOS, Docetaxel, oxaliplatin and S-1
Based on the results of the multivariate analysis, pre-treatment NLR, pre-treatment PLR and arterial chemoembolization were applied to develop a visual nomogram to predict the response of LAGC to neoadjuvant therapy (Fig. 2A). The patients with higher scores were more likely to achieve pCR after neoadjuvant therapy. The internal validation calibration curves showed good consistency between the predicted and actual probabilities of pCR (Fig. 2B). The C-index was performed to evaluate the discriminant ability of the model. The results revealed that the C-index of the nomogram was 0.81 (95% CI, 0.702–0.917). In addition, the ROC curve was also consistent with the C index (Fig. 2C).

Discussion

In this study, we examined the effect of neoadjuvant chemotherapy combined with arterial chemoembolization on the short-term clinical outcomes of LAGC. Our data showed that compared with the conventional group, the combined group did not experienced increased adverse events of chemotherapy or postoperative complications. In contrast, the operation time and postoperative hospital stay in the combined group were shorter than those in the conventional group. Furthermore, this study demonstrated that pre-treatment NLR, pre-treatment PLR and arterial chemoembolization were significant predictors of pCR after neoadjuvant therapy.
Arterial chemoembolization is widely used in the treatment of advanced hepatocellular carcinoma [3133]. Arterial chemoembolization has rarely been applied in LAGC. Nakajima et al. revealed that arterial chemotherapy with cisplatin and etoposide on Days 6 and 20 after intravenous chemotherapy with fluorouracil and calcium folinate in patients with unresectable gastric cancer significantly improved tumor response and resection rates [34]. Zhang et al. compared the effect of different administration methods on patients with unresectable gastric cancer. The patients were divided into two groups: one group received chemotherapy with the XELOX regimen, and the other group received chemotherapy with the FLEEOX regimen (after 5 days of continuous intravenous infusion of fluorouracil and calcium folinate, oxaliplatin, epirubicin, and etoposide were injected intra-arterially on Days 6 and 20, respectively.). The study found that the FLEEOX regimen greatly improved the R0 resection rate, median OS and DFS, but had no significant impact on chemotherapy toxicity or postoperative complications [35]. A retrospective study involving 128 patients with unresectable advanced gastric cancer showed that compared with systemic chemotherapy, systemic chemotherapy combined with regional arterial chemoembolization did not increase the number of chemotherapy adverse events. In addition, combined therapy also effectively improved the OS, DFS and clinical response rate [18].
Similarly, in our study, the combined group did not exhibit increased chemotherapy adverse events or postoperative complications in the patients with LAGC. The patients in the combined group had a higher pCR rate than those in the conventional group.
Several studies have shown that patients with LAGC who achieve pCR after neoadjuvant therapy have better oncological outcomes [3638]. However, the influencing factors of pCR in LAGC remain unclear. Becker et al. suggested that pCR was related to tumor location, degree of differentiation, chemotherapy regimen and number of chemotherapy cycles [39]. In this study, the pCR rate in the combined group was significantly higher than that in the conventional group. Arterial chemoembolization enhanced the anticancer effect by increasing the concentration of chemotherapeutic drug in the tumor area and prolonging the drug reaction time [40]. Embolization of tumor trophoblastic vessels reduced the tumor blood supply, which resulted in necrosis of tumor cells. Moreover, arterial chemotherapy could inhibit tumor cell proliferation by inducing tumor cell apoptosis, thereby improving the efficacy of neoadjuvant therapy [41].
Inflammation, considered an important factor affecting the occurrence and progression of tumors, contributes to tumor growth, invasion, metastasis, angiogenesis, and chemoresistance [42, 43]. Several studies have found that pre-treatment NLR is an independent effect factor for pCR in breast cancer patients [4446]. Lore et al. also found that patients with locally advanced rectal cancer with pre-treatment NLR > 4.06 had poor tumor response and DFS to chemoradiotherapy [47]. In addition, Shi et al. also showed that pre-treatment NLR was an independent predictor of pCR after neoadjuvant chemoradiotherapy for locally advanced rectal cancer [26]. As with colorectal and breast cancers, studies have indicated that pre-treatment NLR, PLR and LMR (lymphocyte to monocyte ratio) were the predictors for tumor regression response and oncological outcomes in LAGC patients after neoadjuvant therapy [4851]. Unexpectedly, in the multivariate analysis, it was also revealed that pre-treatment NLR and pre-treatment PLR were independent predictors of pCR after neoadjuvant therapy in this study. It has been shown that neutrophils are capable of secreting chemokines, cytokines and matrix-degrading proteases. Cytokines stimulate tumor microangiogenesis, and matrix-degrading proteases increase tumor adhesion and promoted distant metastasis [52, 53]. Platelets are able to promote epithelial mesenchymal transformation and metastatic tumor progression through cytokines (e.g., VEGR, EGF, platelet-derived growth factor, hepatocyte growth factor, TGF-β.) [5456]. Lymphocytes may inhibit tumor cell proliferation and migration by inducing cytotoxic cell death. In addition, lymphocytes play a crucial role in the immune surveillance, recognition and destruction of cancer cells [5759].
It is worth mentioning that this study has some limitations. First, as a retrospective study with a small sample size, information bias and selection bias were difficult to avoid. Therefore, the results should be interpreted with caution. Second, postoperative complications after neoadjuvant therapy for gastric cancer were related to age. Patients > 60 years old had higher postoperative morbidity [60, 61]. Approximately half of the study population in our study was younger than 60 years old, and they may have a low comorbidity rate and a low rate of postoperative complications. However, whether chemoembolization would increase postoperative complications in patients aged >60 years old should be verified by subgroup analysis. Third, the inflammatory markers were non-specific and were influenced by a variety of factors, such as drugs, comorbidities, and infections [62]. Moreover, the optimal cut-off values for pre-treatment PLR and pre-treatment NLR were not known. In this study, the cutoff values of pre-treatment PLR and pre-treatment NLR were obtained according to effective statistical methods, but population-based research is still needed for verification. Finally, we failed to obtain enough follow-up data to evaluate the effect of arterial chemoembolization, inflammatory markers and pCR on DFS and OS. Therefore, large-scale multicenter study is needed for further confirmation.
In conclusion, this study demonstrated that neoadjuvant chemotherapy combined with arterial chemoembolization did not increase the adverse events of chemotherapy or postoperative complications in patients with LAGC. Arterial chemoembolization, pre-treatment NLR and pre-treatment PLR were independent predictors of pCR after neoadjuvant therapy. Therefore, arterial chemoembolization may be a safe and effective regimen of neoadjuvant therapy for LAGC.

Acknowledgements

Not applicable.

Declarations

This retrospective cohort study was approved by the Medical Ethics Committee of the First Affiliated Hospital of Chongqing Medical University. (Approval number: 20192801) and carried out in accordance with the Declaration of Helsinki. The Medical Ethics Committee of the First Affiliated Hospital of Chongqing Medical University waived the requirement for informed consent from all research participants due to the retrospective and anonymous nature of this study.
Not applicable.

Competing interests

The authors declare no conficts of interest to in association with the present study.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
7.
Zurück zum Zitat Wang Y, Li Z, Shan F, et al. Current status of diagnosis and treatment of early gastric cancer in China–Data from China gastrointestinal Cancer surgery union. Chin J Gastrointest Surg. 2018;21(2):168–74. Wang Y, Li Z, Shan F, et al. Current status of diagnosis and treatment of early gastric cancer in China–Data from China gastrointestinal Cancer surgery union. Chin J Gastrointest Surg. 2018;21(2):168–74.
13.
Zurück zum Zitat Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019;393(10184):1948–57. https://doi.org/10.1016/S0140-6736(18)32557-1.CrossRefPubMed Al-Batran SE, Homann N, Pauligk C, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019;393(10184):1948–57. https://​doi.​org/​10.​1016/​S0140-6736(18)32557-1.CrossRefPubMed
15.
Zurück zum Zitat Hofheinz RD, Hegewisch-Becker S, Kunzmann V, et al. Trastuzumab in combination with 5-fluorouracil, leucovorin, oxaliplatin and docetaxel as perioperative treatment for patients with human epidermal growth factor receptor 2-positive locally advanced esophagogastric adenocarcinoma: a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie Gastric Cancer Study Group. Int J Cancer. 2021;149(6):1322–31. https://doi.org/10.1002/ijc.33696.CrossRefPubMed Hofheinz RD, Hegewisch-Becker S, Kunzmann V, et al. Trastuzumab in combination with 5-fluorouracil, leucovorin, oxaliplatin and docetaxel as perioperative treatment for patients with human epidermal growth factor receptor 2-positive locally advanced esophagogastric adenocarcinoma: a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie Gastric Cancer Study Group. Int J Cancer. 2021;149(6):1322–31. https://​doi.​org/​10.​1002/​ijc.​33696.CrossRefPubMed
16.
Zurück zum Zitat Zhang X, Liang H, Li Z, et al. Perioperative or postoperative adjuvant oxaliplatin with S-1 versus adjuvant oxaliplatin with capecitabine in patients with locally advanced gastric or gastro-oesophageal junction adenocarcinoma undergoing D2 gastrectomy (RESOLVE): an open-label, superiority and non-inferiority, phase 3 randomised controlled trial. Lancet Oncol. 2021;22(8):1081–92. https://doi.org/10.1016/S1470-2045(21)00297-7.CrossRefPubMed Zhang X, Liang H, Li Z, et al. Perioperative or postoperative adjuvant oxaliplatin with S-1 versus adjuvant oxaliplatin with capecitabine in patients with locally advanced gastric or gastro-oesophageal junction adenocarcinoma undergoing D2 gastrectomy (RESOLVE): an open-label, superiority and non-inferiority, phase 3 randomised controlled trial. Lancet Oncol. 2021;22(8):1081–92. https://​doi.​org/​10.​1016/​S1470-2045(21)00297-7.CrossRefPubMed
30.
Zurück zum Zitat Jaffer AA, Thomas AD, David JB, editors. Gastric Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. Springer International Publishing. American Joint Commission on Cancer; 2020. Jaffer AA, Thomas AD, David JB, editors. Gastric Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. Springer International Publishing. American Joint Commission on Cancer; 2020.
40.
Zurück zum Zitat Zhu ZD, Pu YD. The study of pharmacokinetics of 5-Fu after left gastric artery intra-arterial infusion in treatment of gastric carcinoma. Chin J Bases Clin General Surg. 2001;8:26–8. Zhu ZD, Pu YD. The study of pharmacokinetics of 5-Fu after left gastric artery intra-arterial infusion in treatment of gastric carcinoma. Chin J Bases Clin General Surg. 2001;8:26–8.
54.
Zurück zum Zitat Wada Y, Yoshida K, Tsutani Y, et al. Neutrophil elastase induces cell proliferation and migration by the release of TGF-alpha, PDGF and VEGF in esophageal cell lines. Oncol Rep. 2007;17(1):161–7.PubMed Wada Y, Yoshida K, Tsutani Y, et al. Neutrophil elastase induces cell proliferation and migration by the release of TGF-alpha, PDGF and VEGF in esophageal cell lines. Oncol Rep. 2007;17(1):161–7.PubMed
Metadaten
Titel
Effect of neoadjuvant chemotherapy combined with arterial chemoembolization on short-term clinical outcome of locally advanced gastric cancer
verfasst von
Jianguo Yang
Juncai Li
Qican Deng
Zhenzhou Chen
Kuan He
Yajun Chen
Zhongxue Fu
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10712-w

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Update Onkologie

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