Skip to main content
Erschienen in: Gastric Cancer 6/2023

Open Access 28.09.2023 | Original Article

SOX combined with sintilimab versus SOX alone in the perioperative management of locally advanced gastric cancer: a propensity score–matched analysis

verfasst von: Xingmao Huang, Jingquan Fang, Ling Huang, Hang Chen, Han Chen, Tengjiao Chai, Zeyao Ye, Hanguang Chen, Qi Xu, Yian Du, Pengfei Yu

Erschienen in: Gastric Cancer | Ausgabe 6/2023

Abstract

Objectives

To evaluate the efficacy of SOX combined with a programmed cell death protein-1 (PD-1) inhibitor compared with SOX alone in the perioperative management of locally advanced gastric cancer and to explore biomarkers that may predict response to anti-PD-1 therapy.

Methods

Data of patients with clinical stage T3–4aN0–3M0 (IIb–III) gastric cancer were reviewed to create a primary database. Patients treated with perioperative SOX combined with sintilimab were included in Group A, while those treated with SOX alone were included in Group B. After one-to-one propensity score matching, pathological response and short-term survival outcomes were compared between the two groups. In addition, potential efficacy-related biomarkers were analyzed.

Results

Between January 2018 and December 2022, a total of 150 patients were included in the analysis, with 75 patients in each group. The rates of pathological complete response (21.3% vs. 4.0%; P = 0.001) and major pathological response (45.3% vs. 22.7%; P = 0.003) in Group A were statistically higher than those in Group B. There was no significant difference in 1-year overall survival (92.8% vs. 92.0%; P = 0.392) and disease-free survival (88.9% vs. 88.0%; P = 0.357) between the two groups. Subgroup analysis of Group A showed that the pathological complete response (40.6% vs. 8.6%; P = 0.002) and major pathological response (65.6% vs. 28.6%; P = 0.002) rates were significantly higher in programmed death ligand-1-positive patients with a combined positive score of ≥ 5. A pathological complete response was achieved in 42.9% patients (3/7) with mismatch repair deficiency. For the two patients confirmed as Epstein-Barr virus-positive, one patient achieved a pathological complete response and the other achieved a major pathological response.

Conclusions

The adoption of SOX combined with a PD-1 inhibitor may improve the pathological response rate of patients with locally advanced gastric cancer, especially those with programmed death ligand-1 combined positive score ≥ 5, Epstein–Barr virus-positivity and mismatch repair deficiency. However, further prospective studies are still warranted to confirm the long-term survival benefit.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10120-023-01431-z.
Xingmao Huang, Jingquan Fang and Ling Huang contributed equally to this work and should be considered as co-first authors.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BSA
Body surface area
CPS
Combined positive score
DFS
Disease-free survival
dMMR
MMR deficiency
EBV
Epstein–Barr virus
EBVaGC
EBV-associated gastric cancer
ICI
Immune checkpoint inhibitor
irAEs
Immune-related adverse events
LAGC
Locally advanced gastric cancer
MLH1
MutL homolog 1
MMR
Mismatch repair
MPR
Major pathological response
MSH2
MutS homolog 2
MSH6
MutS homolog 6
MSI
Microsatellite instability
OS
Overall survival
pCR
Pathological complete response
PD-1
Programmed cell death protein-1
PD-L1/2
Programmed death ligand-1/2
PMS2
PMS1 homolog 2
PSM
Propensity score matching
TRG
Tumor regression grading

Introduction

Gastric cancer ranks as the fifth most common malignancy and the fourth most common cause of cancer death around the world [1]. Most patients are diagnosed at an advanced stage. Radical gastrectomy is the main treatment strategy for locally advanced gastric cancer (LAGC) [2, 3]. However, postoperative tumor recurrence and distant metastasis are common and can lead to poor prognosis [4, 5].
Different management strategies have been proposed to improve the prognosis of patients with LAGC, and perioperative chemotherapy has increasingly become an important treatment modality [6]. Studies have shown that preoperative chemotherapy can eliminate occult micrometastases, achieve tumor downstaging, and improve the curative resection rate [79]. Additional studies have shown that perioperative chemotherapy can improve the prognosis of patients with LAGC compared with adjuvant chemotherapy alone [8, 10]. However, the relatively low pathological complete response (pCR) rate was unsatisfactory, which may be caused by the high heterogeneity of gastric cancer.
The introduction of immune checkpoint inhibitors (ICIs) has provided clinical benefits for patients with gastric cancer, especially those with late-stage disease [1113]. As ICIs, programmed cell death protein-1 (PD-1) inhibitors elicit a strong immune response by blocking the interaction of PD-1 with its ligands, programmed death ligand-1 [PD-L1] and PD-L2, which are highly expressed on cancer cells [14]. The CheckMate-649 trial reported that PD-1 inhibitors in combination with chemotherapy improved the overall survival (OS) and progression free survival (PFS) of patients with advanced gastric, gastroesophageal junction, or esophageal adenocarcinoma, especially for those with PD-L1 combined positive score (CPS) ≥ 5 [12]. Anti-PD-1 therapy also exerts a positive effect on patients with LAGC. The addition of PD-1 inhibitors to cytotoxic chemotherapy as neoadjuvant treatment for LAGC showed encouraging results in terms of pathological response [1518]. However, most studies were single-arm studies with small sample sizes, and there is a lack of effective biomarkers for predicting the efficacy of anti-PD-1 therapy.
We conducted a retrospective study to evaluate the efficacy of SOX combined with sintilimab compared with SOX alone in the perioperative management of patients with stage IIb–III LAGC using propensity score matching (PSM) and to explore biomarkers that may predict response to anti-PD-1 therapy.

Methods

Study design

This was a single-center, retrospective study conducted using PSM. The study aimed to compare the short-term effects of perioperative chemotherapy plus a PD-1 inhibitor versus chemotherapy alone in patients with stage IIb–III gastric cancer.
The data of 897 patients with LAGC who received perioperative treatment at the Department of Gastric Surgery, Zhejiang Cancer Hospital, China, between January 2018 and December 2022 were collected and reviewed to create an analytical database. The main selection criteria were: aged 18 to 75 years, histological diagnosis of gastric cancer, clinical stage T3–4aN0–3M0 (IIb–III) disease, received perioperative treatment (SOX plus sintilimab/SOX alone) and radical gastrectomy, Eastern Cooperative Oncology Group performance status 0–1, with adequate organ function to tolerate chemoimmunotherapy or surgery. Patients were excluded if they had remnant gastric cancer, concomitant malignancies or refractory autoimmune diseases, a long-term history of steroid or immunosuppressant use, or missing clinicopathological data. A total of 658 patients were excluded due to the adoption of other treatment modalities (n = 419), loss to follow-up (n = 97), palliative-intent gastrectomy (n = 49), insufficient data (n = 81), and incomplete treatment (n=12). The remaining 239 patients who met the selection criteria were enrolled. Patients treated with perioperative SOX plus sintilimab were included in Group A (n = 79), while those treated with SOX alone were included in Group B (n = 160).
In order to reduce selection bias and ensure comparability between the two groups, PSM without replacement was performed in a 1:1 ratio with a 0.05 caliper width. The resulting propensity score-matched pairs were used in subsequent analyses. Covariates included age, sex, and clinical stage, which were considered to have an effect on the study endpoints. Balance was checked by computing absolute standardized mean differences (SMD) between the two groups for each covariate in the PSM sample. A SMD threshold of 0.2 was considered to detect substantial imbalance. After PSM, the propensity score-matched cohort comprised 75 patients in each group and the baseline variables between the groups were comparable (P > 0.05, SMD < 0.200, Table 1). The process of patient selection is presented in Fig. 1.
Table 1
Baseline characteristics before and after propensity score matching (PSM)
Variable
Before PSM
After PSM
Group A (n = 79)
Group B (n = 160)
P-value
SMD
Group A (n = 75)
Group B (n = 75)
P-value
SMD
Age (years)
0.088
0.235
  
0.866
0.029
 ≤ 60
33 (41.8)
49 (30.6)
  
29 (38.7)
28 (37.3)
  
 > 60
46 (58.2)
111 (69.4)
  
46 (61.3)
47 (62.7)
  
Gender
0.790
0.037
  
0.707
0.062
 Male
59 (74.7)
122 (76.3)
  
55 (73.3)
57 (76.0)
  
 Female
20 (25.3)
38 (23.8)
  
20 (26.7)
18 (24.0)
  
Clinical stage (cTNM)
0.570
0.078
  
0.754
0.050
 IIb
7 (8.9)
18 (11.3)
  
5 (6.7)
6 (8.0)
  
 III
72 (91.1)
142 (88.8)
  
70 (93.3)
69 (92.0)
  
Preoperative CEA
0.787
0.039
  
0.402
0.136
 ≤ 5
61 (77.2)
126 (78.8)
  
59 (78.7)
63 (84.0)
  
 > 5
18 (22.8)
34 (21.3)
  
16 (21.3)
12 (16.0)
  
Preoperative CA125
0.167
0.185
  
0.239
0.191
 ≤ 35
65 (82.3)
142 (88.8)
  
62 (82.7)
67 (89.3)
  
 > 35
14 (17.7)
18 (11.3)
  
13 (17.3)
8 (10.7)
  
Preoperative CA199
0.371
0.123
  
0.577
0.091
 ≤ 37
56 (70.9)
122 (76.3)
  
54 (72.0)
57 (76.0)
  
 > 37
23 (29.1)
38 (23.8)
  
21 (28.0)
18 (24.0)
  
CEA carcinoembryonic antigen, CA125 carbohydrate antigen 125, CA199 carbohydrate antigen199
This study was approved by the Ethics Committee of Zhejiang Cancer Hospital (approval number: IRB-2022-592) and was conducted in accordance with the Declaration of Helsinki. Informed consent was obtained from each patient enrolled in the study.

Treatment

Perioperative  chemotherapy and immunotherapy

SOX plus sintilimab was administered to patients in Group A, while those in Group B were treated with SOX alone. Patients received 2–4 cycles of preoperative treatment and 4–6 cycles of postoperative treatment, with each cycle lasting 3 weeks. The dose of SOX regimen was calculated according to body surface area (BSA): oxaliplatin (130 mg/m2) was administered intravenously on Day 1 and S-1 (BSA < 1.25 m2, 80 mg/day; 1.25 ≤ BSA < 1.50 m2, 100 mg/day; BSA ≥ 1.50m2, 120 mg/day) was administered orally on Days 1–14, followed by a 1-week rest. Sintilimab (200 mg every 3 weeks) was administered intravenously on Day 1 after the beginning of each cycle in Group A.

Surgery

All patients underwent D2 radical gastrectomy according to the Japanese Gastric Cancer Treatment Guidelines [19, 20]. Distal or total gastrectomy was performed according to tumor location. Different reconstruction methods, including Roux-en-Y esophagojejunostomy, Billroth II gastrojejunostomy, and Billroth I gastroduodenostomy, were performed according to the extent of gastrectomy. Resected specimens were assessed by two experienced pathologists to determine the overall response and pathological staging after neoadjuvant treatment. The residual tumor (R) classification was used to categorize tumor status after treatment. Surgical margins with no residual tumor, microscopic residual tumor, and macroscopic residual tumor were defined as R0, R1, and R2 resection, respectively [21]. Postoperative complications were graded according to the Clavien–Dindo classification [22].

Endpoints and assessment

The pCR rate was the primary endpoint, and the major pathological response (MPR), R0 resection, disease-free survival (DFS) and OS rates were analyzed as the secondary endpoints.
Adverse events were graded according to the latest version of the National Cancer Institute’s Common Terminology Criteria for Adverse Events (CTCAE, V.3.0) [23]. Tumor response was assessed in accordance with the Response Evaluation Criteria in Solid Tumors (RECIST) guidelines (version 1.1) [24]. The responses were categorized as follows: complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD). Tumor regression grading (TRG) was adopted to evaluate the tumor regression of resected specimens. TRG was defined as follows: TRG1a, no residual tumor cells; TRG1b, < 10% residual tumor cells; TRG2, 10–50% residual tumor cells; TRG3, > 50% residual tumor cells [25]. Resected specimens without viable tumor cells were considered as pCR (TRG1a). Less than 10% residual tumor cells after preoperative treatment were considered as MPR (TRG1a/b). Downstaging was assessed by comparing the differences between clinical and pathological staging.
PD-L1 expression, mismatch repair (MMR) status, and Epstein-Barr virus (EBV)-positivity were retrospectively assessed in formalin-fixed, paraffin-embedded biopsy samples from Group A patients. PD-L1 expression was determined using immunohistochemistry (PD-L1, Clone 22C3; Dako) and described using CPS. CPS was defined as the total number of PD-L1-positive cells, including tumor cells, lymphocytes, and macrophages, divided by the total number of living tumor cells multiplied by 100. Patients with a CPS ≥ 1 were considered PD-L1-positive. MMR proteins, including MLH1 (clone ES05; Dako), MSH2 (clone FE11; Dako), MSH6 (clone EP49; Dako), and PMS2 (clone EP51; Dako), were stained by immunohistochemistry to determine MMR status. Loss of any of the four MMR proteins was defined as MMR deficiency (dMMR). Additionally, in situ hybridization was performed to evaluate the EBV status (EBV-0050; EBER ISH Kit, MXB).

Follow-up

After treatment, follow-up was performed every 3 months for the first 2 years and then every 6 months from years 3 to 5. Follow-up was mainly conducted by telephone and outpatient review. Physical examination, imaging studies, and blood tests (including tumor markers) were performed regularly in the outpatient clinic. DFS was calculated from the date of gastrectomy until the date of recurrence. OS was calculated from the date of diagnosis until the date of death or last follow-up. The last follow-up was performed on February 28, 2023.

Statistical analysis

Categorical variables were presented as frequencies and percentages, and analyzed using either the Chi-square test or Fisher's exact test depending on the situation. Continuous variables were compared using the Mann–Whitney U test. Survival curves were plotted using the Kaplan–Meier method and compared using the log-rank test. A two-tailed P-value < 0.05 was considered statistically significant. All statistical analyses were conducted using SPSS (version 26.0; SPSS Inc., Chicago, IL, USA).

Results

Clinicopathological characteristics

In the entire cohort, 112 patients (74.7%) were men. The median age was 64 (range: 31–75) years. One hundred and thirty-nine patients (92.7%) had stage III disease. Most patients (96.0%) had gastric adenocarcinoma. Furthermore, 88.7% of tumors were poorly differentiated. Regarding tumor location, 83.3% of tumors were located in the stomach, while the remaining 16.7% were located at the gastroesophageal junction. Detailed information is provided in Table 1 and 2.
Table 2
Clinicopathological characteristics before and after propensity score matching (PSM)
Variable
Before PSM
After PSM
Group A (n = 79)
Group B (n = 160)
P-value
Group A (n = 75)
Group B (n = 75)
P-value
Radical resection
0.174*
  
1.000*
 R0 resection
79 (100.0)
155 (96.9)
 
75 (100)
74 (98.7)
 
 Non-R0 resection
0 (0.0)
5 (3.1)
 
0 (0.0)
1 (1.3)
 
Surgical procedures
0.910
  
1.000
 Distal gastrectomy
31 (39.2)
64 (40.0)
 
29 (38.7)
29 (38.7)
 
 Total gastrectomy
48 (60.8)
96 (60.0)
 
46 (61.3)
46 (61.3)
 
Reconstruction method
0.111*
  
0.153*
 Billroth I
0 (0.0)
10 (6.3)
 
0 (0.0)
4 (5.3)
 
 Billroth II
31 (39.2)
54 (33.8)
 
29 (38.7)
25 (33.3)
 
 Roux-en-Y esophagojejunostomy
48 (60.8)
96 (60.0)
 
46 (61.3)
46 (61.3)
 
Tumor location
0.703*
  
0.421
 Upper
21 (26.6)
51 (31.9)
 
20 (26.7)
26 (34.7)
 
 Middle
22 (27.8)
39 (24.4)
 
22 (29.3)
16 (21.3)
 
 Lower
36 (45.6)
68 (42.5)
 
33 (44.0)
33 (44.0)
 
 Total stomach
0 (0.0)
2 (1.3)
 
0 (0.0)
0 (0.0)
 
Pathologic stage (ypTNM)
< 0.001
  
0.006
 ypT0N0M0
16 (20.3)
4 (2.5)
 
16 (21.3)
3 (4.0)
 
 I
19 (24.1)
34 (21.3)
 
18 (24.0)
14 (18.7)
 
 II
17 (21.5)
42 (26.3)
 
17 (22.7)
23 (30.7)
 
 III
27 (34.2)
80 (50.0)
 
24 (32.0)
35 (46.7)
 
Differentiation degree
0.610
  
0.440
 Poorly
70 (88.6)
138 (86.3)
 
68 (90.7)
65 (86.7)
 
 Moderately-highly
9 (11.4)
22 (13.8)
 
7 (9.3)
10 (13.3)
 
Pathological type
0.290*
  
0.557*
 Adenocarcinoma
74 (93.7)
156 (97.5)
 
71 (94.7)
73 (97.3)
 
 Signet-ring cell carcinoma
2 (2.5)
2 (1.3)
 
2 (2.7)
0 (0.0)
 
 Other
3 (3.8)
2 (1.3)
 
2 (2.7)
2 (2.7)
 
Nerve infiltration
0.722
  
0.479
 No
51 (64.6)
107 (66.9)
 
50 (66.7)
54 (72.0)
 
 Yes
28 (35.4)
53 (33.1)
 
25 (33.3)
21 (28.0)
 
Vascular tumor embolus
0.795
  
0.484
 No
51 (64.6)
106 (66.3)
 
49 (65.3)
53 (70.7)
 
 Yes
28 (35.4)
54 (33.8)
 
26 (34.7)
22 (29.3)
 
Postoperative CEA
0.309
  
0.290
 ≤ 5
67 (84.8)
143 (89.4)
 
65 (86.7)
69 (92.0)
 
 > 5
12 (15.2)
17 (10.6)
 
10 (13.3)
6 (8.0)
 
Postoperative CA125
0.542
  
0.723
 ≤ 35
54 (68.4)
103 (64.4)
 
51 (68.0)
53 (70.7)
 
 > 35
25 (31.6)
57 (35.6)
 
24 (32.0)
22 (29.3)
 
Postoperative CA199
0.688
  
0.575
 ≤ 37
71 (89.9)
141 (88.1)
 
67 (89.3)
69 (92.0)
 
 > 37
8 (10.1)
19 (11.9)
 
8 (10.7)
6 (8.0)
 
CEA carcinoembryonic antigen, CA125 carbohydrate antigen 125, CA199 carbohydrate antigen199
*Fisher’s test
Including adenosquamous carcinoma and squamous cell carcinoma

Treatment outcomes

Perioperative treatment

In both groups, the median number of preoperative treatment cycles was three, and the median number of postoperative treatment cycles was four. After neoadjuvant treatment, all patients underwent D2 radical gastrectomy. R0 resection was performed in 75 patients (100%) in Group A and 74 patients (98.7%) in Group B, with no significant difference between the groups (P = 1.000, Table 2). Four patients in Group B underwent multivisceral resection due to tumor invasion or lymph node metastasis (splenectomy in three patients, pancreatectomy in two patients, and colectomy in one patient).
The median time between preoperative treatment and surgery was 29 (range 22–54) days in Group A and 30 (range: 21–49) days in Group B (P = 0.265). The median time between surgery and postoperative treatment was 27 (range 21–32) and 26 (range 21–29) days, respectively (P = 0.307).
In the entire cohort, surgical complications were observed in 16 patients (10.7%), including nine patients in Group A (pneumonia in five patients, intraperitoneal infection in two patients, and abdominal hemorrhage and anastomotic leakage in one patient each) and seven patients in Group B (pneumonia in three patients, anastomotic leakage in two patients, and intraperitoneal and wound infection in one patient each). There was no significant difference between the two groups (P = 0.597). The postoperative condition of these patients improved after symptomatic treatment. No surgery-related deaths occurred in either group.

Tumor regression and survival

Tumor responses were assessed in all patients. CR, PR, and SD were achieved in 5 (6.7%), 48 (64.0%), and 22 (29.3%) patients in group A, and 2 (2.7%), 46 (61.3%), and 27 (36.0%) patients in group B, respectively, without a significant difference (P = 0.434, Supplementary Table S1). After surgery, the pathological regression of resected specimens was evaluated according to the Becker criteria. In the entire cohort, 19 patients (12.7%) achieved pCR (TRG1a), including 16 patients (21.3%) in Group A and three patients (4.0%) in Group B, with a significant difference between the groups (P = 0.001). The MPR rate was significantly higher in Group A than in Group B (45.3% vs. 22.7%, respectively; P = 0.003). Significant downstaging of TNM stage and the T and N categories were observed in Group A. Detailed information is provided in Tables 3 and Supplementary Tables S2, S3.
Table 3
Pathological evaluation of response to neoadjuvant treatment in GC patients
Variable
Group A (No., %)
Group B (No.,%)
P-value
TRG
  
0.006
 1a
16 (21.3)
3 (4.0)
 
 1b
18 (24.0)
14 (18.7)
 
 2
32 (42.7)
43 (57.3)
 
 3
9 (12.0)
15 (20.0)
 
pCR
16 (21.3)
3 (4.0)
0.001
MPR
34 (45.3)
17 (22.7)
0.003
All discharged patients were followed-up, with a median duration of 26 (range: 6–52) months. Median OS and DFS data are not yet mature. The 1-year OS rate was 92.8% for Group A and 92.0% for Group B, with no statistical difference between the groups (P = 0.392). The 1-year DFS rate for Group A and Group B was 88.9% and 88.0%, respectively (P = 0.357; Fig. 2).

Safety profile

As shown in Table 4, grade 3 or 4 adverse events were identified in 17 (22.7%) patients in Group A and 13 (17.3%) patients in Group B during the neoadjuvant treatment period, with no significant difference (P = 0.414). Meanwhile, during the adjuvant treatment period, grade 3 or 4 adverse events were observed in 11 (14.7%) patients in Group A and 5 (6.7%) patients in Group B (P = 0.113). In the entire cohort, the most common hematological toxicities were leucopenia, neutropenia, and anemia, while the most common non-hematological toxicities were transaminase elevation, vomiting, and pneumonia. In Group A, two patients suffered from grade 3 hypothyroidism and were treated with thyroid hormone and steroid hormone. The condition of all patients with adverse events was well-controlled after conversion treatment. No treatment-related deaths occurred in either group.
Table 4
Perioperative treatment-related adverse events
 
Neoadjuvant treatment
Adjuvant treatment
Group A (n.%)
Group B (n.%)
Group A (n.%)
Group B (n.%)
Any grade
Grade 3–4
Any grade
Grade 3–4
Any grade
Grade 3–4
Any grade
Grade 3–4
Total
70 (93.3)
17 (22.6)
67 (89.3)
13 (17.3)
68 (90.7)
11 (14.7)
64 (85.3)
5 (6.7)
Hematological
 Leucopenia
39 (52.0)
4 (5.3)
37 (49.3)
3 (4.0)
32 (42.7)
2 (2.7)
28 (37.3)
2 (2.7)
 Neutropenia
33 (44.0)
4 (5.3)
34 (45.3)
3 (4.0)
36 (48.0)
3 (4.0)
23 (30.7)
1 (1.3)
 Anemia
32 (42.7)
2 (2.7)
34 (45.3)
2 (2.7)
25 (33.3)
1 (1.3)
30 (40.0)
2 (2.7)
 Thrombocytopenia
21 (28.0)
3 (4.0)
25 (33.3)
4 (5.3)
13 (17.3)
2 (2.7)
9 (12.0)
2 (2.7)
Non-hematological
 Elevated ALT/AST
18 (24.0)
4 (5.3)
19 (25.3)
3 (4.0)
21 (28.0)
3 (4.0)
13 (17.3)
1 (1.3)
 Nausea/vomiting
15 (20.0)
2 (2.7)
13 (17.3)
1 (1.3)
8 (10.7)
1 (1.3)
10 (13.3)
1 (1.3)
 Pneumonia
5 (6.7)
0 (0.0)
4 (5.3)
0 (0.0)
1 (1.3)
0 (0.0)
0 (0.0)
0 (0.0)
 Diarrhea
5 (6.7)
0 (0.0)
3 (4.0)
0 (0.0)
1 (1.3)
0 (0.0)
2 (2.7)
0 (0.0)
 Elevated creatinine
4 (5.3)
0 (0.0)
2 (2.7)
0 (0.0)
4 (5.3)
0 (0.0)
6 (8.0)
0 (0.0)
 Dermatitis
2 (2.7)
0 (0.0)
0 (0.0)
0 (0.0)
1 (1.3)
0 (0.0)
0 (0.0)
0 (0.0)
 Hypothyroidism
4 (5.3)
2 (2.7)
0 (0.0)
0 (0.0)
1 (1.3)
0 (0.0)
0 (0.0)
0 (0.0)
 Hyperthyroidism
1 (1.3)
0 (0.0)
0 (0.0)
0 (0.0)
0 (0.0)
0 (0.0)
0 (0.0)
0 (0.0)
ALT alanine aminotransferase, AST aspartate aminotransferase

Potential biomarkers

PD-L1 expression, MMR status, and EBV-positivity were analyzed in Group A patients. Detailed information is provided in Table 5 and Supplementary Tables S4–S6. PD-L1 data were available for 67 patients. The number of patients with a CPS < 1, 1 ≤ CPS < 5, 5 ≤ CPS < 10, and CPS ≥ 10 was 20 (29.9%), 15 (22.4%), 14 (20.9%), and 18 (26.9%), respectively. Forty-seven patients (70.1%) were PD-L1-positive (CPS ≥ 1). The pCR rate of PD-L1-positive and -negative patients was 27.7% and 15.0% (P = 0.215), and the MPR rate was 48.9% and 40.0% (P = 0.502), respectively. Further analysis showed that the pCR (40.6% vs. 8.6%; P = 0.002) and MPR (65.6% vs. 28.6%; P = 0.002) rates were higher in patients with a CPS ≥ 5 compared to those with a CPS < 5, with significant differences between the groups. MMR data were available for 71 patients, of whom seven (9.9%) had dMMR. Three patients (42.9%) with dMMR achieved pCR. EBV data were available for 65 patients. Only two patients (3.1%) were EBV-positive (one achieved pCR; the other achieved MPR). Both EBV-positive patients had a CPS of 5 and pMMR.
Table 5
Associations between the expression of PD-L1 and pathological response
Variable
CPS < 1 (n = 20)
CPS ≥ 1 (n = 47)
P-value
CPS < 5 (n = 35)
CPS ≥ 5 (n = 32)
P-value
TRG
0.738
  
0.008
 1a
3 (15.0)
13 (27.7)
 
3 (8.6)
13 (40.6)
 
 1b
5 (25.0)
10 (21.3)
 
7 (20.0)
8 (25.0)
 
 2
9 (45.0)
18 (38.3)
 
19 (54.3)
8 (25.0)
 
 3
3 (15.0)
6 (12.8)
 
6 (17.1)
3 (9.4)
 
pCR
3 (15.0)
13 (27.7)
0.215
3 (8.6)
13 (40.6)
0.002
MPR
8 (40.0)
23 (48.9)
0.502
10 (28.6)
21 (65.6)
0.002

Discussion

In this study, we compared the efficacy of SOX plus a PD-1 inhibitor and SOX alone for the perioperative management of patients with stage IIb–III LAGC, focusing on pathological response and potential biomarkers. The pCR and downstaging rates of patients treated with perioperative SOX plus sintilimab were significantly higher than those of patients treated with SOX alone, which met the endpoints of this study as expected. Further analysis revealed that PD-L1 CPS ≥ 5, dMMR, or EBV-positivity were associated with improved efficacy.
The clinical benefit of neoadjuvant chemotherapy in patients with gastric cancer has been widely studied. Randomized trials reported that the addition of chemotherapy before gastrectomy improves the radical resection rate and prognosis compared to surgery alone [9, 10, 26, 27]. However, the pathological response rate of patients receiving neoadjuvant chemotherapy was relatively low, and the long-term survival was still unsatisfactory. Therefore, more effective strategies are needed to improve the efficacy and prognosis of LAGC.
Previous studies have demonstrated that immunotherapy may enhance the antitumor activity of chemotherapeutic drugs by exerting a synergistic effect [28, 29]. Furthermore, several studies have shown that the combination of chemotherapy and ICIs can improve the efficacy and prognosis of patients with advanced gastric cancer [11, 12, 30]. Encouraged by these results, studies focusing on the efficacy and safety of preoperative chemotherapy in combination with a PD-1 inhibitor were performed in patients with LAGC. Jiang et al. [16] evaluated the benefit of preoperative oxaliplatin/capecitabine plus sintilimab for patients with resectable gastric and gastroesophageal junction cancer in a single-arm phase II trial. Improved pCR (19.4%, 7/36) and MPR (47.2%, 17/36) rates were achieved compared to traditional neoadjuvant chemotherapy. In our study, the pCR and MPR rates of patients treated with perioperative SOX plus sintilimab were significantly higher than those of patients treated with SOX alone, showing superiority in the pathological response to this combination strategy directly based on the PD-1 inhibitor. However, for patients treated with neoadjuvant nivolumab monotherapy followed by gastrectomy, Hasegawa et al. [31] reported that the pCR (3.2%, 1/31) and MPR (16.1%, 5/31) rates were lower than those of patients treated with combination therapy, indicating that the effect of immunotherapy based on a single agent may be limited and the synergistic effect of immunotherapy and chemotherapy may yield better results. However, it remains unclear whether the superiority in pathological regression could translate into a survival benefit for patients with LAGC treated with chemotherapy and PD-1 inhibitors. Jiang et al. [16] showed that the 1-year OS and DFS rates in patients treated with preoperative chemotherapy plus sintilimab were 94.1% and 90.3%, respectively. In our study, the survival curves gradually separated during the follow-up period, however, there was no significant difference in the 1-year OS and DFS rates between the two groups. Long-term follow-up is needed to determine the survival benefit of perioperative chemotherapy plus immunotherapy in these patients. Moreover, the ongoing phase III KEYNOTE-585 trial of the perioperative XP/FP/FLOT regimen combined with pembrolizumab, with the primary endpoint of OS, will provide additional data in the future [32].
Regarding the safety of preoperative treatment, a meta-analysis reported that the incidence of grade 3–4 adverse events in patients treated with neoadjuvant chemoimmunotherapy and chemotherapy was 20.6% and 25.7%, respectively, with no significant difference (P = 0.380) [33]. Similarly, our results indicated that the addition of a PD-1 inhibitor to chemotherapy did not significantly increase the incidence of adverse events compared to chemotherapy alone. Therefore, the toxicity of PD-1 inhibitors in combination with chemotherapy may be tolerable. However, the potential immune-related adverse events (irAEs) should not be ignored. Previous studies indicate that organ-specific irAEs occur most frequently in the endocrine organs and skin, followed by the gastrointestinal and pulmonary tracts, most of which are grade 1–2 [16, 17]. In our study, a total of five patients (6.7%) in Group A experienced irAEs, including two cases of grade 3 hypothyroidism that required hormone replacement therapy. With the increased use of ICIs in the treatment of LAGC, it is important to be aware of potential adverse events and to become better at recognizing and managing these specific toxicities.
Biomarkers are increasingly driving systemic therapy. PD-L1 expression, MMR status, and EBV-positivity have been identified as potential biomarkers for immunotherapy benefit, however, evidence in the perioperative setting is still unclear [6]. Therefore, further exploratory analyses were performed in patients treated with PD-1 inhibitors in this study.
PD-L1 is a widely accepted biomarker for immunotherapy in gastric cancer. The CPS, which indicates PD-L1 expression levels on tumor cells and tumor-related immune cells, may be useful in predicting the efficacy of anti-PD-1 therapy [34]. However, the optimal cutoff value has not been established. Tang et al. [15] and Guo et al. [17] reported a higher pathological response rate in patients with a CPS ≥ 1 who were treated with preoperative chemoimmunotherapy. However, superior pCR and MPR rates were only observed in patients with a CPS ≥ 5 in the present study. Therefore, further research is needed to determine the appropriate cutoff value of PD-L1 CPS, which could improve patient selection for perioperative treatment.
Patients with microsatellite instability (MSI)/dMMR are characterized by increased PD-L1-positive T-cell infiltration and are more likely to respond to PD-1 inhibitors [35]. In this study, the pathological regression rate in patients with dMMR was higher than that in those with proficient MMR, indicating that the combination of chemotherapy and immunotherapy may be beneficial in these patients. However, whether patients with dMMR gastric cancer treated with perioperative chemoimmunotherapy have a superior prognosis compared to patients treated with chemotherapy alone is not clear, and there is still a lack of consensus on the treatment of these patients. An exploratory analysis of the MAGIC trial [36] revealed that preoperative chemotherapy may exert a negative effect on the prognosis of patients with MSI/dMMR LAGC. Meanwhile, the GERCOR NEONIPIGA phase II trial [37] reported a promising pCR rate (58.6%, 17/29) in patients with resectable MSI/dMMR gastric/gastroesophageal junction adenocarcinoma (T2–4NxM0) who were treated with neoadjuvant nivolumab plus ipilimumab. However, the most effective perioperative strategies for the management of patients with MSI/dMMR gastric cancer remain controversial, and further clinical studies are warranted.
As a distinct molecular subtype of gastric cancer, EBV-positivity has also been reported as a biomarker for anti-PD-1 therapy. Previous studies have indicated that EBV-associated gastric cancer (EBVaGC) exhibits higher expression of PD-L1 and lymphocytic infiltration [38, 39]. Patients with EBVaGC may be more susceptible to PD-1 blockade and may have a better prognosis [4042]. Tang et al. [15] reported an exceptional pCR rate of 33.3% (2/6) in patients with EBVaGC who were treated with neoadjuvant PD-1 blockade plus chemotherapy. In this study, both patients with EBV-positive gastric cancer had encouraging outcomes: one achieved MPR and the other achieved pCR. Therefore, patients with EBVaGC may be an additional population with high potential for clinical benefit to chemoimmunotherapy.
Although the baseline characteristics were balanced and comparable between the two groups after PSM, there are several limitations in the present study. First, this was a single-center study, and selection bias was unavoidable given the retrospective design of the study. Second, the safety results of perioperative chemotherapy plus PD-1 inhibitor may be compromised due to the exclusion of some patients with incomplete treatment. Third, long-term survival and recurrence could not be fully analyzed due to the relatively short follow-up period. Finally, we did not explore the efficacy of perioperative chemotherapy plus PD-1 inhibitors in patients with different genomic backgrounds (tumor mutation burden, high microsatellite instability, etc.). Despite these limitations, our results are valuable for future research on the perioperative management of patients with LAGC.
In conclusion, perioperative SOX plus sintilimab may improve tumor downstaging and pathological regression rate of LAGC. Patients with PD-L1 CPS ≥ 5, EBV-positivity, or dMMR responded better to chemoimmunotherapy. However, long-term survival requires further follow-up, and more prospective clinical studies with larger sample sizes are needed to validate the findings of our study.

Acknowledgements

We thank all the study participants and research staff who participated in this work.

Declarations

Conflict of interest

The authors declare no competing interests.

Ethical approval and informed consent

This study was approved by the Ethics Committee of Zhejiang Cancer Hospital (approval number: IRB-2022-592) and was conducted according to the Declaration of Helsinki. Informed consent was obtained from each patient enrolled in the study.
Open Access This 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/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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

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

e.Med Innere Medizin

Kombi-Abonnement

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

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.CrossRefPubMed Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.CrossRefPubMed
2.
Zurück zum Zitat Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020;396:635–48.CrossRefPubMed Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020;396:635–48.CrossRefPubMed
3.
Zurück zum Zitat Ajani JA, D’Amico TA, Bentrem DJ, Chao J, Cooke D, Corvera C, et al. Gastric Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022;20:167–92.CrossRefPubMed Ajani JA, D’Amico TA, Bentrem DJ, Chao J, Cooke D, Corvera C, et al. Gastric Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022;20:167–92.CrossRefPubMed
4.
Zurück zum Zitat Thomassen I, van Gestel YR, van Ramshorst B, Luyer MD, Bosscha K, Nienhuijs SW, et al. Peritoneal carcinomatosis of gastric origin: a population-based study on incidence, survival and risk factors. Int J Cancer. 2014;134:622–8.CrossRefPubMed Thomassen I, van Gestel YR, van Ramshorst B, Luyer MD, Bosscha K, Nienhuijs SW, et al. Peritoneal carcinomatosis of gastric origin: a population-based study on incidence, survival and risk factors. Int J Cancer. 2014;134:622–8.CrossRefPubMed
5.
Zurück zum Zitat Chen QY, Zhong Q, Zhou JF, Qiu XT, Dang XY, Cai LS, et al. Conditional survival and recurrence of remnant gastric cancer after surgical resection: a multi-institutional study. Cancer Sci. 2020;111:502–12.CrossRefPubMed Chen QY, Zhong Q, Zhou JF, Qiu XT, Dang XY, Cai LS, et al. Conditional survival and recurrence of remnant gastric cancer after surgical resection: a multi-institutional study. Cancer Sci. 2020;111:502–12.CrossRefPubMed
7.
Zurück zum Zitat Al-Batran SE, Homann N, Pauligk C, Goetze TO, Meiler J, Kasper S, 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;393:1948–57.CrossRefPubMed Al-Batran SE, Homann N, Pauligk C, Goetze TO, Meiler J, Kasper S, 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;393:1948–57.CrossRefPubMed
8.
Zurück zum Zitat Zhang X, Liang H, Li Z, Xue Y, Wang Y, Zhou 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;22:1081–92.CrossRefPubMed Zhang X, Liang H, Li Z, Xue Y, Wang Y, Zhou 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;22:1081–92.CrossRefPubMed
9.
Zurück zum Zitat Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed Cunningham D, Allum WH, Stenning SP, Thompson JN, Van de Velde CJ, Nicolson M, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20.CrossRefPubMed
10.
Zurück zum Zitat Kang YK, Yook JH, Park YK, Lee JS, Kim YW, Kim JY, et al. PRODIGY: a phase III study of neoadjuvant docetaxel, oxaliplatin, and S-1 plus surgery and adjuvant S-1 versus surgery and adjuvant S-1 for resectable advanced gastric cancer. J Clin Oncol. 2021;39:2903–13.CrossRefPubMedPubMedCentral Kang YK, Yook JH, Park YK, Lee JS, Kim YW, Kim JY, et al. PRODIGY: a phase III study of neoadjuvant docetaxel, oxaliplatin, and S-1 plus surgery and adjuvant S-1 versus surgery and adjuvant S-1 for resectable advanced gastric cancer. J Clin Oncol. 2021;39:2903–13.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Kang YK, Chen LT, Ryu MH, Oh DY, Oh SC, Chung HC, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022;23:234–47.CrossRefPubMed Kang YK, Chen LT, Ryu MH, Oh DY, Oh SC, Chung HC, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022;23:234–47.CrossRefPubMed
12.
Zurück zum Zitat Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021;398:27–40.CrossRefPubMedPubMedCentral Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021;398:27–40.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Boku N, Ryu MH, Kato K, Chung HC, Minashi K, Lee KW, et al. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4). Ann Oncol. 2019;30:250–8.CrossRefPubMed Boku N, Ryu MH, Kato K, Chung HC, Minashi K, Lee KW, et al. Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4). Ann Oncol. 2019;30:250–8.CrossRefPubMed
14.
Zurück zum Zitat Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–49.CrossRefPubMed Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–49.CrossRefPubMed
15.
Zurück zum Zitat Tang X, Li M, Wu X, Guo T, Zhang L, Tang L, et al. Neoadjuvant PD-1 blockade plus chemotherapy induces a high pathological complete response rate and anti-tumor immune subsets in clinical stage III gastric cancer. Oncoimmunology. 2022;11:2135819.CrossRefPubMedPubMedCentral Tang X, Li M, Wu X, Guo T, Zhang L, Tang L, et al. Neoadjuvant PD-1 blockade plus chemotherapy induces a high pathological complete response rate and anti-tumor immune subsets in clinical stage III gastric cancer. Oncoimmunology. 2022;11:2135819.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Jiang H, Yu X, Li N, Kong M, Ma Z, Zhou D, et al. Efficacy and safety of neoadjuvant sintilimab, oxaliplatin and capecitabine in patients with locally advanced, resectable gastric or gastroesophageal junction adenocarcinoma: early results of a phase 2 study. J Immunother Cancer. 2022;10: e003635.CrossRefPubMedPubMedCentral Jiang H, Yu X, Li N, Kong M, Ma Z, Zhou D, et al. Efficacy and safety of neoadjuvant sintilimab, oxaliplatin and capecitabine in patients with locally advanced, resectable gastric or gastroesophageal junction adenocarcinoma: early results of a phase 2 study. J Immunother Cancer. 2022;10: e003635.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Guo H, Ding P, Sun C, Yang P, Tian Y, Liu Y, et al. Efficacy and safety of sintilimab plus XELOX as a neoadjuvant regimen in patients with locally advanced gastric cancer: a single-arm, open-label, phase II trial. Front Oncol. 2022;12: 927781.CrossRefPubMedPubMedCentral Guo H, Ding P, Sun C, Yang P, Tian Y, Liu Y, et al. Efficacy and safety of sintilimab plus XELOX as a neoadjuvant regimen in patients with locally advanced gastric cancer: a single-arm, open-label, phase II trial. Front Oncol. 2022;12: 927781.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Xu C, Xie X, Kang N, Jiang H. Neoadjuvant PD-1 inhibitor and apatinib combined with S-1 plus oxaliplatin for locally advanced gastric cancer patients: a multicentered, prospective, cohort study. J Cancer Res Clin Oncol. 2022;149:4091–9.CrossRefPubMed Xu C, Xie X, Kang N, Jiang H. Neoadjuvant PD-1 inhibitor and apatinib combined with S-1 plus oxaliplatin for locally advanced gastric cancer patients: a multicentered, prospective, cohort study. J Cancer Res Clin Oncol. 2022;149:4091–9.CrossRefPubMed
19.
Zurück zum Zitat Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric Cancer. 2017;20:1–19.CrossRef Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric Cancer. 2017;20:1–19.CrossRef
20.
Zurück zum Zitat Japanese gastric cancer treatment guidelines 2018 (5th edition). Gastric Cancer. 2021;24:1–21. Japanese gastric cancer treatment guidelines 2018 (5th edition). Gastric Cancer. 2021;24:1–21.
21.
Zurück zum Zitat Hermanek P, Wittekind C. Residual tumor (R) classification and prognosis. Semin Surg Oncol. 1994;10:12–20.CrossRefPubMed Hermanek P, Wittekind C. Residual tumor (R) classification and prognosis. Semin Surg Oncol. 1994;10:12–20.CrossRefPubMed
22.
Zurück zum Zitat Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240:205–13.CrossRefPubMedPubMedCentral Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240:205–13.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.CrossRefPubMed Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.CrossRefPubMed
25.
Zurück zum Zitat Becker K, Mueller JD, Schulmacher C, Ott K, Fink U, Busch R, et al. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer. 2003;98:1521–30.CrossRefPubMed Becker K, Mueller JD, Schulmacher C, Ott K, Fink U, Busch R, et al. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer. 2003;98:1521–30.CrossRefPubMed
26.
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
27.
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
28.
Zurück zum Zitat Principe DR, Kamath SD, Korc M, Munshi HG. The immune modifying effects of chemotherapy and advances in chemo-immunotherapy. Pharmacol Ther. 2022;236: 108111.CrossRefPubMedPubMedCentral Principe DR, Kamath SD, Korc M, Munshi HG. The immune modifying effects of chemotherapy and advances in chemo-immunotherapy. Pharmacol Ther. 2022;236: 108111.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Kim R, An M, Lee H, Mehta A, Heo YJ, Kim KM, et al. Early tumor-immune microenvironmental remodeling and response to first-line fluoropyrimidine and platinum chemotherapy in advanced gastric cancer. Cancer Discov. 2022;12:984–1001.CrossRefPubMed Kim R, An M, Lee H, Mehta A, Heo YJ, Kim KM, et al. Early tumor-immune microenvironmental remodeling and response to first-line fluoropyrimidine and platinum chemotherapy in advanced gastric cancer. Cancer Discov. 2022;12:984–1001.CrossRefPubMed
30.
Zurück zum Zitat Janjigian YY, Kawazoe A, Yañez P, Li N, Lonardi S, Kolesnik O, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021;600:727–30.CrossRefPubMedPubMedCentral Janjigian YY, Kawazoe A, Yañez P, Li N, Lonardi S, Kolesnik O, et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature. 2021;600:727–30.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Hasegawa H, Shitara K, Takiguchi S, Takiguchi N, Ito S, Kochi M, et al. A multicenter, open-label, single-arm phase I trial of neoadjuvant nivolumab monotherapy for resectable gastric cancer. Gastric Cancer. 2022;25:619–28.CrossRefPubMedPubMedCentral Hasegawa H, Shitara K, Takiguchi S, Takiguchi N, Ito S, Kochi M, et al. A multicenter, open-label, single-arm phase I trial of neoadjuvant nivolumab monotherapy for resectable gastric cancer. Gastric Cancer. 2022;25:619–28.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Bang YJ, Van Cutsem E, Fuchs CS, Ohtsu A, Tabernero J, Ilson DH, et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol. 2019;15:943–52.CrossRefPubMed Bang YJ, Van Cutsem E, Fuchs CS, Ohtsu A, Tabernero J, Ilson DH, et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol. 2019;15:943–52.CrossRefPubMed
33.
Zurück zum Zitat Xu H, Li T, Shao G, Wang W, He Z, Xu J, et al. Evaluation of neoadjuvant immunotherapy plus chemotherapy in Chinese surgically resectable gastric cancer: a pilot study by meta-analysis. Front Immunol. 2023;14:1193614.CrossRefPubMedPubMedCentral Xu H, Li T, Shao G, Wang W, He Z, Xu J, et al. Evaluation of neoadjuvant immunotherapy plus chemotherapy in Chinese surgically resectable gastric cancer: a pilot study by meta-analysis. Front Immunol. 2023;14:1193614.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Shitara K, Özgüroğlu M, Bang YJ, Di Bartolomeo M, Mandalà M, Ryu MH, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;392:123–33.CrossRefPubMed Shitara K, Özgüroğlu M, Bang YJ, Di Bartolomeo M, Mandalà M, Ryu MH, et al. Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet. 2018;392:123–33.CrossRefPubMed
35.
Zurück zum Zitat Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.CrossRefPubMedPubMedCentral Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Smyth EC, Wotherspoon A, Peckitt C, Gonzalez D, Hulkki-Wilson S, Eltahir Z, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the medical research council adjuvant gastric infusional chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197–203.CrossRefPubMed Smyth EC, Wotherspoon A, Peckitt C, Gonzalez D, Hulkki-Wilson S, Eltahir Z, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the medical research council adjuvant gastric infusional chemotherapy (MAGIC) trial. JAMA Oncol. 2017;3:1197–203.CrossRefPubMed
37.
Zurück zum Zitat André T, Tougeron D, Piessen G, Fouchardière CDL, Louvet C, Adenis A, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability-high gastric or esophagogastric junction adenocarcinoma: the GERCOR NEONIPIGA phase II study. J Clin Oncol. 2023;41:255–65.CrossRefPubMed André T, Tougeron D, Piessen G, Fouchardière CDL, Louvet C, Adenis A, et al. Neoadjuvant nivolumab plus ipilimumab and adjuvant nivolumab in localized deficient mismatch repair/microsatellite instability-high gastric or esophagogastric junction adenocarcinoma: the GERCOR NEONIPIGA phase II study. J Clin Oncol. 2023;41:255–65.CrossRefPubMed
38.
Zurück zum Zitat Lima Á, Sousa H, Medeiros R, Nobre A, Machado M. PD-L1 expression in EBV associated gastric cancer: a systematic review and meta-analysis. Discov Oncol. 2022;13:19.CrossRefPubMedPubMedCentral Lima Á, Sousa H, Medeiros R, Nobre A, Machado M. PD-L1 expression in EBV associated gastric cancer: a systematic review and meta-analysis. Discov Oncol. 2022;13:19.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Panda A, Mehnert JM, Hirshfield KM, Riedlinger G, Damare S, Saunders T, et al. Immune activation and benefit from avelumab in EBV-positive gastric cancer. J Natl Cancer Inst. 2018;110:316–20.CrossRefPubMed Panda A, Mehnert JM, Hirshfield KM, Riedlinger G, Damare S, Saunders T, et al. Immune activation and benefit from avelumab in EBV-positive gastric cancer. J Natl Cancer Inst. 2018;110:316–20.CrossRefPubMed
40.
Zurück zum Zitat Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef
41.
Zurück zum Zitat Greally M, Chou JF, Chatila WK, Margolis M, Capanu M, Hechtman JF, et al. Clinical and molecular predictors of response to immune checkpoint inhibitors in patients with advanced esophagogastric cancer. Clin Cancer Res. 2019;25:6160–9.CrossRefPubMedPubMedCentral Greally M, Chou JF, Chatila WK, Margolis M, Capanu M, Hechtman JF, et al. Clinical and molecular predictors of response to immune checkpoint inhibitors in patients with advanced esophagogastric cancer. Clin Cancer Res. 2019;25:6160–9.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Sohn BH, Hwang JE, Jang HJ, Lee HS, Oh SC, Shim JJ, et al. Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin Cancer Res. 2017;23:4441–9.CrossRefPubMedPubMedCentral Sohn BH, Hwang JE, Jang HJ, Lee HS, Oh SC, Shim JJ, et al. Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin Cancer Res. 2017;23:4441–9.CrossRefPubMedPubMedCentral
Metadaten
Titel
SOX combined with sintilimab versus SOX alone in the perioperative management of locally advanced gastric cancer: a propensity score–matched analysis
verfasst von
Xingmao Huang
Jingquan Fang
Ling Huang
Hang Chen
Han Chen
Tengjiao Chai
Zeyao Ye
Hanguang Chen
Qi Xu
Yian Du
Pengfei Yu
Publikationsdatum
28.09.2023
Verlag
Springer Nature Singapore
Erschienen in
Gastric Cancer / Ausgabe 6/2023
Print ISSN: 1436-3291
Elektronische ISSN: 1436-3305
DOI
https://doi.org/10.1007/s10120-023-01431-z

Weitere Artikel der Ausgabe 6/2023

Gastric Cancer 6/2023 Zur Ausgabe

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

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.

Deutlich weniger Infektionen: Wundprotektoren schützen!

08.05.2024 Postoperative Wundinfektion Nachrichten

Der Einsatz von Wundprotektoren bei offenen Eingriffen am unteren Gastrointestinaltrakt schützt vor Infektionen im Op.-Gebiet – und dient darüber hinaus der besseren Sicht. Das bestätigt mit großer Robustheit eine randomisierte Studie im Fachblatt JAMA Surgery.

Chirurginnen und Chirurgen sind stark suizidgefährdet

07.05.2024 Suizid Nachrichten

Der belastende Arbeitsalltag wirkt sich negativ auf die psychische Gesundheit der Angehörigen ärztlicher Berufsgruppen aus. Chirurginnen und Chirurgen bilden da keine Ausnahme, im Gegenteil.

Update Chirurgie

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

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

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

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

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

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

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

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

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

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

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