Skip to main content
Erschienen in: World Journal of Surgical Oncology 1/2023

Open Access 01.12.2023 | Research

The value of lymph nodes ratios in the prognosis of resectable remnant gastric cancer through the retrospective propensity score matching analysis

verfasst von: Biao Yang, Tao Liu, Hangtian Cui, Zhengmao Lu, Guoen Fang, Xuchao Xue, Tianhang Luo

Erschienen in: World Journal of Surgical Oncology | Ausgabe 1/2023

Abstract

Purpose

Currently, the characteristics and prognosis of remnant gastric cancer (RGC) are not fully understood yet. The present study aimed to describe the details of clinicopathological features of resectable RGC and investigated the factors affecting survival after the curative operation.

Methods

From Jan. 2006 to Dec. 2015, a total of 118 resectable RGC patients (the RGC group) and 236 age-, sex- and TNM stages-matched resectable gastric cancer (GC) patients (the control group) were recruited retrospectively. Clinicopathological characteristics and overall survival were compared between the two groups.

Results

The overall survival rate was 46.61% for RGC patients compared to 55.08% for control groups (P < 0.01), and the mean overall survival time of RGC patients was 40.23 ± 32.27 months, compared to 55.06 ± 34.29 months in the control group (P = 0.023 after matching). The overall survival (OS) of RGC patients with stage IIb was much worse than IIa (P < 0.001) and similar to IIIa (P = 0.463) and IIIb (P = 0.014). Multivariate Cox proportional hazards model analysis revealed that TNM stage (HR: 3.899, P < 0.001) and lymph nodes ratio (LNR) (HR: 2.405, P = 0.028) were independent prognostic significance to OS.

Conclusions

The OS of RGC was much worse than GC with similar TNM stages, and LNR might consider a highly reliable indicator to evaluate the prognostic in RGC.
Hinweise
Biao Yang and Tao Liu contributed equally to this work.

Publisher’s Note

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

Introduction

Residual gastric cancer (RGC) is a specific type of gastric cancer, that originally refers to cancers after gastric resection for either benign or malignant diseases, which develops 5 years following the initial procedure [13]. After continuous research and development, cancerous changes that occur in the stomach more than 10 years after partial gastrectomy due to malignant cancers have also been included in the scope of RGC [4]. RGC differs from the concept of "carcinoma in the remnant stomach (CRS)" proposed by the Japanese Gastric Cancer Association [5, 6]. Regarding the exact definition of RGC, there is currently no consensus worldwide. According to the 2018 Surgical Expert Consensus on the Definition of Residual Gastric Cancer in China, the RGC in clinical work is defined as new cancer that occurs in the residual stomach more than 5 years after gastrectomy for benign diseases or more than 10 years after gastrectomy for gastric cancer (GC) [7]. With the increasing number of patients undergoing gastrectomy, the incidence of RGC is increasing year by year [8]. Therefore, it is necessary to further strengthen the understanding of the clinical characteristics of RGC and improve the level of diagnosis and treatment.
At present, most scholars believe that the main pathogenesis of RGC is related to the anatomical changes of the residual stomach after partial gastrectomy, and the reflux stimulation of bile, intestinal juice, pancreatic juice, and other alkaline fluids [9]. This leads to the development of the residual gastric mucosa into chronic atrophic gastritis, intestinal metaplasia, and dysplasia, which may eventually lead to RGC [10]. The interval between RGCs after surgery for benign gastric disease is longer than for malignant disease. The time for RGC to develop after resection of benign gastric disease is 20–36 years, and the time after resection of malignant disease is 2–9 years [1113]. The incidence of RGC is higher in European and American populations than in Asian populations. This may be due to the higher incidence of primary gastric diseases (including primary GC) in Asia than in other parts of the world [3, 14, 15]. With the continuous improvement of the early diagnosis rate of GC, the overall survival time of patients after surgery has been significantly prolonged, and the incidence of RGC after radical subtotal gastrectomy has increased year by year. Due to the atypical clinical manifestations of early RGC patients, most of them have progressed at the time of consultation. A study in Europe suggests that RGC needs to be addressed early and properly [16]. Therefore, the early diagnosis and treatment of RGC are particularly important.
Clinical manifestations in patients with RGC are generally non-specific and are easily missed or misdiagnosed by clinicians. Early RGCs generally have no obvious clinical signs and symptoms and often require gastroscopy to confirm the diagnosis. In clinical practice, RGC has a higher rate of invasion to adjacent organs, and lymph node metastasis was commonly observed [17], which may cause the prognosis of RGC much worse than primary GC [18]. But some articles argued that the prognosis of RGC was similar to primary GC [19]. Several previous studies have discussed the clinical characteristics of resectable RGC in a limited number of cases, but factors influencing the prognosis of patients with RGC still remained unclear or controversial [2022]. In this study with a relatively larger sample size, we described the clinicopathological characteristics of resectable RGC and investigated factors affecting prognosis after the curative operation, attempting to address the inherent controversies more reliably.

Methods

Study design and participants

From January 2010 to December 2015, the clinicopathological data of 195 patients with RGC who underwent curative gastric resection (including total resection of the gastric stump with lymphadenectomy) at Changhai Hospital were retrospectively collected. In this study, RGC was defined as a carcinoma arising in the gastric remnant after gastrectomy, regardless of the histology of the previous lesion (benign or malignant), its risk of recurrence, the extent of initial resection, or methods of reconstruction [23]. The TNM classification (7th edition) was reported as a practical staging system for RGC, so the RGC stage in this study was conducted according to the TNM classification (7th edition) by AJCC [24, 25]. All the patients enrolled received standard therapy, which meant curative operation and postoperative adjuvant chemotherapy if necessary according to the NCCN Clinical Practice Guidelines in Oncology Gastric Cancer. This study was approved by the Medical Ethics Committee of Changhai Hospital of the Second Military Medical University. The informed consent requirement was exempted.
Inclusion criteria: 1) age 18 to 85 years, life expectancy > 3 months; 2) Histopathological examination confirmed as stomach adenocarcinoma; 3) Benign gastric lesions above 5 years after the initial partial gastrectomy, new cancers on the remnant gastric; 4) gastric malignant lesions undergo partial gastric resection and more than 5 years after R0 resection, new cancers of the remnant gastric; 5) complete clinical-pathological data; 6) adequate organ functions (leukocyte count > 3,500/μl, platelet count > 100,000/μl, hemoglobin > 10.0 g/dl, serum creatinine < 1.25 times the upper limit of normal (ULN), transaminases and alkaline phosphatases < 2.5 times ULN or < 5 times ULN in patients with liver metastasis, bilirubin < 1.5 times ULN, and prothrombin time < 12.0 s). Exclusion criteria: 1) concomitant other primary cancers; 2) incomplete or unavailable medical record data, incomplete preoperative examination, or refusal of surgical treatment; 3) patients whose initial lesions are malignant and have not been removed by R0; 4) patients with incomplete medical records.
Between January 2010 and December 2015, a total of 195 patients with RGC and 2001 patients with GC were admitted. Under case selection criteria, a total of 77 patients with RGC and 624 patients with GC were excluded. Reasons for ineligibility included patients incompatible with the operation (8/71 cases), positive margin (3/41 cases), inadequate organ function (4/155 cases), incomplete clinical data (6/162 cases), palliative operation or M1 stage (51/195 cases), and wedge gastrectomy (5/0 cases). Finally, this study included 118 resectable RGC patients and 1377 patients with GC who performed the curative operation.

Follow-up and data collection

Follow-ups were conducted once every 28-day for half a year, once every 3-month for 2 years, once every 6-month for 3 years, and yearly thereafter. Follow-up terms consisted of physical examination, complete blood test, chest radiography, and ultrasound of the abdomen as clinically indicated. Computed tomography (CT) scanning or magnetic resonance imaging (MRI) would be performed if necessary. The follow-up data of patients were collected either through telephone or outpatient service.
Medical records of patients enrolled were collected retrospectively by two assistants from Medbanks Network Technology CO. Collected data included patients’ background, time intervals between primary and secondary operation, pathologic diagnosis of RGC, surgical data, and tumor characteristics. The data errors were corrected by the Department of General Surgery Changhai Hospital.

Statistical analysis

To minimize the influence of other confounders on the outcome, we used a propensity score analysis to match RGC patients with GC patients. RGC patients were matched in a 1:4 ratio with GC patients using the nearest neighbor matching and based on gender (male or female), age (≤ 60 or > 60 years), tumor Grade (well-differentiated, moderately differentiated, poorly differentiated), tumor size (≤ 3 cm or > 3 cm), number of lymph nodes (LN) harvested, TNM stage (7th edition, AJCC) and pathological lymph nodes positive ratio (pLNR, number of positive LN/number of resected LN).
Statistical analysis was performed using SPSS software (18th version, SPSS, Chicago, IL, USA). Enumeration data were presented in percentage, measurement data was presented in average number and standard deviation, and abnormal distribution data was displayed by median and quartile. The associations between categorical variables were analyzed by the Pearson chi-square test. Continuous variables were assessed using t test or Cochran-Mantel–Haenszel test as appropriate. Survival curves and univariate analysis were performed in accordance with the Kaplan–Meier method, and the log-rank test was used to evaluate statistical significance. Cox regression analysis was used in multivariate analysis to assess independent prognostic factors. A probability (P) value < 0.05 was considered to indicate statistical significance. All tests were two-sided.

Results

Enrollment and patient characteristics

The flowchart of the program for participants in this study is shown in Fig. 1. The baseline characteristics of patients with GC in the pre-match and post-match samples are presented in Table 1. The two groups did not differ significantly in terms of age, sex, tumor grade, and TNM stage. However, compared with the control group, the RGC group had significantly larger tumors, higher pLNR, and fewer LNs harvested (all p < 0.05).
Table 1
Clinicopathological features of RGC and GC before and after matching on the propensity score
Clinicopathological Features
Before matching
P-value
After matching
P-value
RGC (n = 118)
GC (n = 1377)
RGC (n = 113)
GC (n = 406)
Gender
  
0.0088#
  
0.864#
female
22 (18.64%)
414 (30.07%)
 
22 (19.47%)
82 (20.20%)
 
male
96 (81.36%)
963 (69.93%)
 
91 (80.53%)
324 (79.80%)
 
Age (year)
  
0.0258#
  
0.760#
 ≤ 60
49 (41.53%)
719 (52.21%)
 
48 (42.48%)
179 (44.09%)
 
 > 60
69 (58.47%)
658 (47.79%)
 
65 (57.52%)
227 (55.91%)
 
Tumor Grade
  
0.0031#
  
0.823#
poorly
70 (59.32%)
639 (47.47%)
 
66 (58.41%)
227 (55.91%)
 
moderately
38 (32.20%)
642 (47.70%)
 
37 (32.74%)
135 (33.25%)
 
well
10 (8.47%)
65 (4.83%)
 
10 (8.85%)
44 (10.84%)
 
Tumor size
  
0.0372#
  
0.014#
 ≤ 3
39 (33.05%)
591 (42.92%)
 
39 (34.51%)
193 (47.54%)
 
 > 3
79 (66.95%)
786 (57.08%)
 
74 (65.49%)
213 (52.46%)
 
Mean of LN Harvested
  
 < 0.0001*
  
 < 0.0001*
 
13.13 ± 9.58
19.57 ± 5.59
 
13.34 ± 9.67
19.35 ± 5.40
 
pT stage
  
0.0103
  
0.484
T1
28 (23.73%)
212 (15.40%)
 
28 (24.78%)
93 (22.91%)
 
T2
12 (10.17%)
412 (29.92%)
 
12 (10.62%)
49 (12.07%)
 
T3
47 (39.83%)
644 (46.77%)
 
42 (37.17%)
156 (38.42%)
 
T4
31 (26.27%)
109 (7.92%)
 
31 (27.43%)
108 (26.60%)
 
pN stage
  
 < 0.001
  
0.294
N0
51 (43.22%)
453 (32.90%)
 
51 (45.13%)
174 (42.86%)
 
N1
27 (22.88%)
256 (18.59%)
 
25 (22.12)
79 (19.46%)
 
N2
17 (14.41%)
285 (20.70%)
 
17 (15.04)
64 (15.76)
 
N3
23 (19.49%)
383 (27.81%)
 
20 (17.70%)
89 (21.92%)
 
TNM stage
  
0.5478
  
0.161
Stage I
39 (33.05%)
350 (25.42%)
 
39 (34.51%)
150 (36.95%)
 
Stage II
22 (18.64%)
457 (33.19%)
 
20 (17.70%)
110 (27.09%)
 
Stage III
57 (48.31%)
570 (41.39%)
 
54 (47.79%)
146 (35.96%)
 
pLNR
  
 < 0.0001#
  
 < 0.0001#
0
51 (43.22%)
453 (32.90%)
 
51 (45.13%)
174 (42.86%)
 
0〜0.5
32 (27.12%)
592 (42.99%)
 
28 (24.78%)
184 (45.32%)
 
 ≥ 0.5
35 (29.66%)
332 (24.11%)
 
34 (30.09%)
48 (11.82%)
 
RGC remnant gastric cancer, GC gastric cancer, LN lymph nodes, T tumor, N node, M metastasis, LNR lymph nodes ratio
#chi-square test
*t-test
Cochran-Mantel–Haenszel test

Survival analysis of RGC

The overall survival rate was 46.61% for RGC patients compared to 55.08% for control groups (P < 0.01), and the mean overall survival time of RGC patients was 40.23 ± 32.27 months, compared with 55.06 ± 34.29 months in the control group (P = 0.023 after matching) (Fig. 1). As shown in the OS curves, the OS rates of RGC patients with TNM stage I (92.31%) and stage III (14.81%) were almost similar to GC patients (91.33% and 20.51%) (Fig. 2A-E). But the OS rate of RGC patients with TNM stage II was 45.00%, which was much worse than the control group (75.45%) (Fig. 2F, G). Another interesting finding showed that there was no significant difference in OS rates among RGC patients with TNM stage IIb, GC patients with TNM stage IIIa (P = 0.736), and IIIb (P = 0.105) (Fig. 3).

Association between primary disease and clinicopathological characteristics

To find the relationship between primary disease and clinicopathological characteristics in RGC, all RGC patients were divided into two sub-groups (RGC-B for primary benign disease vs. RGC-M for primary malignant disease). Primary benign disease meant benign ulcer, while primary malignant disease referred to gastric adenocarcinoma. In all 118 cases, 61 (51.69%) RGC patients had primary GC in the first operation. The time interval, number of LN harvested, and pN stages were statistically different between the two groups (Table 2). Theoretically speaking, the primary disease was an important personal factor so it may influence the prognosis, however, our study showed there were no obvious survival differences attributed to it (40.99 ± 33.50 months vs. 39.52 ± 31.34 months; P = 0.26).
Table 2
Association between primary disease and clinicopathological features
Clinical features
RGC-B
(n = 57)
RGC-M
(n = 61)
P-value
Gender
  
0.271#
female
9 (15.79%)
13 (21.31%)
 
male
48 (84.21%)
48 (78.69%)
 
Age (year)
  
0.062#
 ≤ 60
21 (36.84%)
28 (45.90%)
 
 > 60
36 (63.16%)
33 (54.10%)
 
Time interval(month)
  
 < 0.001*
 
151.16 ± 76.30
68.05 ± 32.05
 
Tumor location
  
0.445#
anastomosis
29 (50.88%)
31 (50.82%)
 
non-anastomosis
28 (49.12%)
30 (49.18%)
 
Tumor Grade
  
0.122
poorly
37 (64.91%)
33 (54.10%)
 
moderately
17 (29.82%)
21 (34.43%)
 
well
3 (5.26%)
7 (11.48%)
 
Tumor size
  
0.251#
 ≤ 3
35 (61.40%)
32 (52.46%)
 
 > 3
22 (38.60%)
29 (47.54%)
 
Mean of LN Harvested
  
 < 0.001*
 
19.86 ± 7.46
6.84 ± 6.61
 
pT* stage
  
0.189
T1
13 (22.81%)
15 (24.59%)
 
T2
7 (12.28%)
5 (8.20%)
 
T3
24 (42.11%)
23 (37.70%)
 
T4
13 (22.81%)
18 (29.51%)
 
pN* stage
  
0.028
N0
23 (40.35%)
28 (45.90%)
 
N1
9 (15.79%)
18 (29.51%)
 
N2
10 (17.54%)
7 (11.48%)
 
N3
15 (26.32%)
8 (13.11%)
 
TNM stage
  
0.387
Stage 1
19 (33.33%)
20 (32.79%)
 
Stage 2
10 (17.54%)
12 (19.67%)
 
Stage 3
28 (49.12%)
29 (47.54%)
 
RGC-B remnant gastric cancer primary benign disease, RGC-M primary malignant disease, RGC remnant gastric cancer, GC gastric cancer, LN lymph nodes, T tumor, N node, M metastasis, LNR lymph nodes ratio
#chi-square test
*t-test
Wilcoxon rank sum test

The effects of Lymph Nodes Ratio (LNR)

There was an interesting finding that the OS of RGC patients with TNM stages II varied much from each other (Fig. 2A, B). The OS of patients with stage IIb was much worse than IIa (P < 0.001) and similar to IIIa (P = 0.463, Fig. 3A) and IIIb (P = 0.014, Fig. 3B), which indicated that the current TNM classification (7th edition) might not be suitable to the patients with RGC. Further, we deemed it might be caused by the number of lymph nodes harvested in RGC patients being much less than in GC patients (P < 0.001), which might affect the results of TNM classification evaluated by the number of lymph nodes status (Fig. 4).
Univariate log-rank test analysis revealed that tumor size and pLNR were significant prognostic factors for RGC. Multivariate Cox proportional hazards model analysis revealed that pLNR (HR: 2.405, P = 0.028; Table 3) was an independent prognostic factor to OS in RGC. Further, the correlation analysis also showed there was a significant correlation between the number of metastatic lymph nodes and the number of lymph nodes harvested in RGC patients (r = 0.79, P < 0.001). So we deemed the TNM classification evaluated by LNR status might be more suitable for RGC patients.
Table 3
Multivariate Cox proportional hazards regression analysis of OS in patients with RGC
Variables
HR (95% CI)
P-value
Gender
 Male
Reference
 
 Female
1.41(0.69–2.90)
0.301
Age
0.98 (0.93–1.02)
0.254
T
 T1
Reference
 
 T2
11.79 (0.89–156.00)
0.061
 T3
7.72 (0.48–124.14)
0.149
 T4
14.73 (0.86–253.18)
0.064
N
 N0
Reference
 
 N1
8.34 (1.59–43.80)
0.012
 N2
12.38 (2.18–70.22)
0.004
 N3a
8.45 (1.31–54.62)
0.025
 N3b
10.35 (1.52–70.23)
0.017
Tumor Grade
 well
Reference
 
 moderately
0.49 (0.05–4.81)
0.537
 poorly
0.65 (0.07–6.36)
0.71
Chemotherapy
 Yes
Reference
 
 No
0.57 (0.21–1.54)
0.264
TNM
3.899 (2.62–5.18)
 < 0.001
LNR
2.40 (1.21–3.59)
0.028
Primary malignant disease
 No
Reference
 
 Yes
1.06 (0.55–2.05)
0.9
RGC remnant gastric cancer, GC gastric cancer, T tumor, N node, M metastasis, LNR lymph nodes ratio, OS overall survival

Discussion

Previous studies have shown that the incidence of RGC is low, accounting for about 1% to 8% of GC [26]. In recent years, due to the popularization of screening and the continuous improvement of inspection methods, the detection rate of RGC has gradually increased, and clinicians and researchers have paid more and more attention to the disease. In this study, we found that RGC had a much worse OS than GC, although the TNM stage was similar in both groups. There was little or no difference in the OS curves of patients with IIa, IIIa, and IIIb RGC, suggesting that current TNM staging systems are unable to assess OS in patients with advanced RGC. LNR can be an independent prognostic factor in patients with RGC.

RGC with a worse prognosis may be caused by the unsuitable classification system

The clinicopathologic characteristics and prognosis of RGC had been investigated in a few studies, which had not reached a consensus yet [10, 27, 28]. According to most reports, RGC was frequently diagnosed at an advanced stage, which caused a relatively low rate of curative resection and poor prognosis, suggesting that RGC may have distinct biological features from primary GC [29]. But some authors have compared RGC with primary GC and discovered no significant difference in survival between RGC and primary GC [30]. In most studies, the lymph node N staging of RGCs still follows the grading criteria of the UICC. However, in patients with first-time GC, postoperative lymph node drainage changes, and the lymph nodes detected by RGC cannot fully define the N stage, especially since RGC occurs after GC. The total number of postoperative lymph node dissections during re-surgery usually does not exceed 10, which is significantly less than the number of lymph nodes dissected by RGC after surgery for benign lesions. This may result in inaccurate staging. Studies have shown that the total lymph node detection rate and peritoneal lymph node metastasis rate of RGC are lower than those of primary proximal gastric cancer (PPGC), and TNM grading may not be suitable for RGC. In our study, we found the OS of RGC was much worse than GC, although the TNM stages of patients in the two groups were similar. But we deemed that this phenomenon might be caused by an unsuitable TNM classification system for RGC, for there was nearly no difference in OS curve among stage IIb, IIIa, and IIIb RGC patients, which indicated the present TNM staging system could not evaluate the OS of advanced RGC patients with lymph nodes metastasis. So our study also lent further support that it is necessary to investigate a new indicator to evaluate the lymph node status in advanced RGC.
In this study, RGC patients were divided into the RGC-B group and RGC-M group according to the nature of the initial disease. The numbers of the two groups were 57 (48.31%) and 61 (51.69%), respectively. The number of cases in the RGC-M group was slightly higher than that in the RGC-B group. This may have a great relationship with the application of proton pump inhibitors and the increase in the rate of radical resection of primary GC in recent years. By comparing the clinicopathological features of patients in the RGC-B group and the RGC-M group, it was interesting to find that the clinicopathological features of RGC-M were similar to those of RGC-B, except for the time interval from the start of the first operation. It indicated the primary disease has no effects on the characteristics of RGC. And there was no difference in tumor location between RGC-M and RGC-B also lends further support to the previous conclusion that the conception of CRS was more reasonable in the definition of RGC [6, 31]. The survival analysis of this study showed that the prognosis of patients in the RGC-B group was better than that of the patients in the RGC-M group, but the difference was not statistically significant. This may be related to the small number of samples included in this study, and there may be some bias.

LNR was a better factor to evaluate the lymph node's status in RGC

Lymph node status is an established prognostic factor in both GC and RGC [12, 32]. But the Japanese classification of gastric carcinoma nor the TNM classification raised a special lymph node classification system for RGC till now. At present, most staging system (UICC/AJCC) stratifies the N value simply on the basis of the count of metastatic lymph nodes. But the total number of lymph nodes harvested in RGC was much less than in GC, which might affect the results of the N value based on the count of metastatic lymph nodes. Accumulated evidence showed the lymph nodes ratio (LNR) could reflect both qualitative and quantitative lymph node spread as well as the extent of disease, which might serve as an additional prognostic indicator to better evaluate the outcome of patients with advanced RGC [3335]. In particular, we focused on the prognostic value and staging accuracy of the metastatic lymph nodes ratio (LNR) in patients with RGC undergoing curative resection, and we also found that LNR was an independent prognostic factor in RGC patients in multivariate analyses, for LNR was less influenced by the total number of lymph nodes harvested in operation. Since Okusa firstly demonstrated the impact of the “frequency of the metastases” on patients’ outcomes, there were few articles focusing on LNR in RGC to our knowledge [36]. In the present study, we reported that LNR might consider a simple, reproducible, and highly reliable prognostic factor in RGC patients. But the threshold of LNR in the TNM staging system of RGC needs a larger sample size to confirm.
There still existed some limitations to this study. First, the major limitation was its retrospective design in a single hospital. Secondly, we were unable to collect some information on the study patients, such as lifestyles, dietary preferences, and environmental features for statistical analysis, which might be important influencing factors. Further prospective studies were necessary to clarify the pathophysiology and development of RGC.

Conclusion

In this study, we revealed important characteristics of RGC in detail. The OS of RGC patients with lymph node metastasis was much worse than GC with similar TNM stages, and LNR might consider a highly reliable indicator to evaluate the prognostic in RGC. Although the time interval from the first operation to RGC in RGC-B patients was much longer than that in RGC-M patients, the primary disease did not affect overall survival. In summary, after familiarizing the clinical-pathological characteristics and prognostic factors of RGC patients, clinicians can more accurately predict postoperative recurrence and metastasis, and better provide individualized and integrated treatment options for RGC patients.

Acknowledgements

Not applicable.

Declarations

All procedures performed in the study were by the ethical standards of the Institutional and National Research Committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. This study was approved by the Medical Ethics Committee of Changhai Hospital of the Second Military Medical University. The informed consent requirement was exempted.
Not applicable.

Competing interests

The authors declare no competing interests.
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/​. 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.
Literatur
2.
Zurück zum Zitat Kung CY, Fang WL, Wang RF, et al. Prognosis and clinicopathologic features in patients with gastric stump cancer after curative surgery. Curr Oncol. 2020;27(3):e259–64.CrossRefPubMedPubMedCentral Kung CY, Fang WL, Wang RF, et al. Prognosis and clinicopathologic features in patients with gastric stump cancer after curative surgery. Curr Oncol. 2020;27(3):e259–64.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Mak TK, Guan B, Peng J, et al. Prevalence and characteristics of gastric remnant cancer: A systematic review and meta-analysis. Asian J Surg. 2021;44(1):11–7.CrossRefPubMed Mak TK, Guan B, Peng J, et al. Prevalence and characteristics of gastric remnant cancer: A systematic review and meta-analysis. Asian J Surg. 2021;44(1):11–7.CrossRefPubMed
4.
Zurück zum Zitat Kidokoro T, Hayashida Y, Urabe M. Long-term surgical results of carcinoma of the gastric remnant: a statistical analysis of 613 patients from 98 institutions. World J Surg. 1985;9(6):966–71.CrossRefPubMed Kidokoro T, Hayashida Y, Urabe M. Long-term surgical results of carcinoma of the gastric remnant: a statistical analysis of 613 patients from 98 institutions. World J Surg. 1985;9(6):966–71.CrossRefPubMed
5.
Zurück zum Zitat Japanese Gastric Cancer A. Japanese Classification of Gastric Carcinoma - 2nd English Edition. Gastric Cancer. 1998;1(1):10–24. Japanese Gastric Cancer A. Japanese Classification of Gastric Carcinoma - 2nd English Edition. Gastric Cancer. 1998;1(1):10–24.
6.
Zurück zum Zitat Nozaki I, Hato S, Kobatake T, et al. Incidence of metachronous gastric cancer in the remnant stomach after synchronous multiple cancer surgery. Gastric Cancer. 2014;17(1):61–6.CrossRefPubMed Nozaki I, Hato S, Kobatake T, et al. Incidence of metachronous gastric cancer in the remnant stomach after synchronous multiple cancer surgery. Gastric Cancer. 2014;17(1):61–6.CrossRefPubMed
7.
Zurück zum Zitat Gao Z, Jiang K, Ye Y, et al. Interpretation on Chinese surgeons’ consensus opinion for the definition of gastric stump cancer (version 2018). Zhonghua Wei Chang Wai Ke Za Zhi. 2018;21(5):486–90.PubMed Gao Z, Jiang K, Ye Y, et al. Interpretation on Chinese surgeons’ consensus opinion for the definition of gastric stump cancer (version 2018). Zhonghua Wei Chang Wai Ke Za Zhi. 2018;21(5):486–90.PubMed
8.
Zurück zum Zitat Hanyu T, Wakai A, Ishikawa T, et al. Carcinoma in the Remnant Stomach During Long-Term Follow-up After Distal Gastrectomy for Gastric Cancer: Analysis of Cumulative Incidence and Associated Risk Factors. World J Surg. 2018;42(3):782–7.CrossRefPubMed Hanyu T, Wakai A, Ishikawa T, et al. Carcinoma in the Remnant Stomach During Long-Term Follow-up After Distal Gastrectomy for Gastric Cancer: Analysis of Cumulative Incidence and Associated Risk Factors. World J Surg. 2018;42(3):782–7.CrossRefPubMed
10.
Zurück zum Zitat Song XH, Liu K, Sun LF, et al. Clinicopathological characteristics and prognostic factors of remnant gastric cancer: A single-center retrospective analysis of 90 patients. Int J Surg. 2018;51:97–103. Song XH, Liu K, Sun LF, et al. Clinicopathological characteristics and prognostic factors of remnant gastric cancer: A single-center retrospective analysis of 90 patients. Int J Surg. 2018;51:97–103.
11.
Zurück zum Zitat Galata C, Ronellenfitsch U, Weiss C, et al. Surgery for Gastric Remnant Cancer Results in Similar Overall Survival Rates Compared with Primary Gastric Cancer: A Propensity Score-Matched Analysis. Ann Surg Oncol. 2020;27(11):4196–203.CrossRefPubMedPubMedCentral Galata C, Ronellenfitsch U, Weiss C, et al. Surgery for Gastric Remnant Cancer Results in Similar Overall Survival Rates Compared with Primary Gastric Cancer: A Propensity Score-Matched Analysis. Ann Surg Oncol. 2020;27(11):4196–203.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Irino T, Hiki N, Ohashi M, et al. Characteristics of gastric stump cancer: A single hospital retrospective analysis of 262 patients. Surgery. 2016;159(6):1539–47.CrossRefPubMed Irino T, Hiki N, Ohashi M, et al. Characteristics of gastric stump cancer: A single hospital retrospective analysis of 262 patients. Surgery. 2016;159(6):1539–47.CrossRefPubMed
13.
Zurück zum Zitat Takeno S, Hashimoto T, Maki K, et al. Gastric cancer arising from the remnant stomach after distal gastrectomy: a review. World J Gastroenterol. 2014;20(38):13734–40.CrossRefPubMedPubMedCentral Takeno S, Hashimoto T, Maki K, et al. Gastric cancer arising from the remnant stomach after distal gastrectomy: a review. World J Gastroenterol. 2014;20(38):13734–40.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Ramos M, Pereira MA, de Castria TB, et al. Remnant gastric cancer: a neglected group with high potential for immunotherapy. J Cancer Res Clin Oncol. 2020;146(12):3373–83.CrossRefPubMed Ramos M, Pereira MA, de Castria TB, et al. Remnant gastric cancer: a neglected group with high potential for immunotherapy. J Cancer Res Clin Oncol. 2020;146(12):3373–83.CrossRefPubMed
15.
Zurück zum Zitat Bafutto AAF, Castro OAP, Fukuhara DK, et al. Lymph node distribution in patients with remnant gastric cancer. J Cancer Res Clin Oncol. 2023;149(6):2367–74.CrossRefPubMed Bafutto AAF, Castro OAP, Fukuhara DK, et al. Lymph node distribution in patients with remnant gastric cancer. J Cancer Res Clin Oncol. 2023;149(6):2367–74.CrossRefPubMed
16.
Zurück zum Zitat Kavaliauskas P, Dulskas A, Kavaliauskaite R, et al. Short and long-term outcomes in a patient cohort with gastric stump cancer: A propensity score analysis. Asian J Surg. 2021;44(7):1027–8.CrossRefPubMed Kavaliauskas P, Dulskas A, Kavaliauskaite R, et al. Short and long-term outcomes in a patient cohort with gastric stump cancer: A propensity score analysis. Asian J Surg. 2021;44(7):1027–8.CrossRefPubMed
17.
Zurück zum Zitat Tanigawa N, Nomura E, Lee SW, et al. Current state of gastric stump carcinoma in Japan: based on the results of a nationwide survey. World J Surg. 2010;34(7):1540–7.CrossRefPubMedPubMedCentral Tanigawa N, Nomura E, Lee SW, et al. Current state of gastric stump carcinoma in Japan: based on the results of a nationwide survey. World J Surg. 2010;34(7):1540–7.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Tran TB, Hatzaras I, Worhunsky DJ, et al. Gastric remnant cancer: A distinct entity or simply another proximal gastric cancer? J Surg Oncol. 2015;112(8):877–82.CrossRefPubMed Tran TB, Hatzaras I, Worhunsky DJ, et al. Gastric remnant cancer: A distinct entity or simply another proximal gastric cancer? J Surg Oncol. 2015;112(8):877–82.CrossRefPubMed
19.
Zurück zum Zitat Hu X, Tian DY, Cao L, et al. Progression and prognosis of gastric stump cancer. J Surg Oncol. 2009;100(6):472–6.CrossRefPubMed Hu X, Tian DY, Cao L, et al. Progression and prognosis of gastric stump cancer. J Surg Oncol. 2009;100(6):472–6.CrossRefPubMed
20.
Zurück zum Zitat An JY, Youn HG, Ha TK, et al. Clinical significance of tumor location in remnant gastric cancers developed after partial gastrectomy for primary gastric cancer. J Gastrointest Surg. 2008;12(4):689–94.CrossRefPubMed An JY, Youn HG, Ha TK, et al. Clinical significance of tumor location in remnant gastric cancers developed after partial gastrectomy for primary gastric cancer. J Gastrointest Surg. 2008;12(4):689–94.CrossRefPubMed
21.
Zurück zum Zitat Zhang DW, Dong B, Li Z, et al. Clinicopathologic features of remnant gastric cancer over time following distal gastrectomy. World J Gastroenterol. 2015;21(19):5972–8.CrossRefPubMedPubMedCentral Zhang DW, Dong B, Li Z, et al. Clinicopathologic features of remnant gastric cancer over time following distal gastrectomy. World J Gastroenterol. 2015;21(19):5972–8.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Namikawa T, Kitagawa H, Iwabu J, et al. Tumors arising at previous anastomotic site may have poor prognosis in patients with gastric stump cancer following gastrectomy. J Gastrointest Surg. 2010;14(12):1923–30.CrossRefPubMed Namikawa T, Kitagawa H, Iwabu J, et al. Tumors arising at previous anastomotic site may have poor prognosis in patients with gastric stump cancer following gastrectomy. J Gastrointest Surg. 2010;14(12):1923–30.CrossRefPubMed
23.
Zurück zum Zitat Takahashi M, Takeuchi H, Tsuwano S, et al. Surgical Resection of Remnant Gastric Cancer Following Distal Gastrectomy: A Retrospective Clinicopathological Study. Ann Surg Oncol. 2016;23(2):511–21.CrossRefPubMed Takahashi M, Takeuchi H, Tsuwano S, et al. Surgical Resection of Remnant Gastric Cancer Following Distal Gastrectomy: A Retrospective Clinicopathological Study. Ann Surg Oncol. 2016;23(2):511–21.CrossRefPubMed
24.
Zurück zum Zitat Nakagawa M, Choi YY, An JY, et al. Staging for Remnant Gastric Cancer: The Metastatic Lymph Node Ratio vs. the UICC 7th Edition System. Ann Surg Oncol. 2016;23(13):4322–31. Nakagawa M, Choi YY, An JY, et al. Staging for Remnant Gastric Cancer: The Metastatic Lymph Node Ratio vs. the UICC 7th Edition System. Ann Surg Oncol. 2016;23(13):4322–31.
25.
Zurück zum Zitat Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17(12):3077–9.CrossRefPubMed Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17(12):3077–9.CrossRefPubMed
26.
Zurück zum Zitat Nienhuser H, Blank S, Sisic L, et al. Gastric stump carcinoma: frequency, treatment, complications and prognosis. Chirurg. 2017;88(4):317–27.PubMed Nienhuser H, Blank S, Sisic L, et al. Gastric stump carcinoma: frequency, treatment, complications and prognosis. Chirurg. 2017;88(4):317–27.PubMed
27.
Zurück zum Zitat Liao G, Wen S, Xie X, et al. Laparoscopic gastrectomy for remnant gastric cancer: Risk factors associated with conversion and a systematic analysis of literature. Int J Surg. 2016;34:17–22. Liao G, Wen S, Xie X, et al. Laparoscopic gastrectomy for remnant gastric cancer: Risk factors associated with conversion and a systematic analysis of literature. Int J Surg. 2016;34:17–22.
28.
Zurück zum Zitat Ohira M, Toyokawa T, Sakurai K, et al. Current status in remnant gastric cancer after distal gastrectomy. World J Gastroenterol. 2016;22(8):2424–33.CrossRefPubMedPubMedCentral Ohira M, Toyokawa T, Sakurai K, et al. Current status in remnant gastric cancer after distal gastrectomy. World J Gastroenterol. 2016;22(8):2424–33.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Mezhir JJ, Gonen M, Ammori JB, et al. Treatment and outcome of patients with gastric remnant cancer after resection for peptic ulcer disease. Ann Surg Oncol. 2011;18(3):670–6.CrossRefPubMed Mezhir JJ, Gonen M, Ammori JB, et al. Treatment and outcome of patients with gastric remnant cancer after resection for peptic ulcer disease. Ann Surg Oncol. 2011;18(3):670–6.CrossRefPubMed
30.
Zurück zum Zitat An JY, Choi MG, Noh JH, et al. The outcome of patients with remnant primary gastric cancer compared with those having upper one-third gastric cancer. Am J Surg. 2007;194(2):143–7.CrossRefPubMed An JY, Choi MG, Noh JH, et al. The outcome of patients with remnant primary gastric cancer compared with those having upper one-third gastric cancer. Am J Surg. 2007;194(2):143–7.CrossRefPubMed
31.
Zurück zum Zitat Irino T, Hiki N, Nunobe S, et al. Subtotal gastrectomy with limited lymph node dissection is a feasible treatment option for patients with early gastric stump cancer. J Gastrointest Surg. 2014;18(8):1429–33.CrossRefPubMed Irino T, Hiki N, Nunobe S, et al. Subtotal gastrectomy with limited lymph node dissection is a feasible treatment option for patients with early gastric stump cancer. J Gastrointest Surg. 2014;18(8):1429–33.CrossRefPubMed
32.
Zurück zum Zitat Ahn HS, Lee HJ, Hahn S, et al. Evaluation of the seventh American Joint Committee on Cancer/International Union Against Cancer Classification of gastric adenocarcinoma in comparison with the sixth classification. Cancer. 2010;116(24):5592–8.CrossRefPubMed Ahn HS, Lee HJ, Hahn S, et al. Evaluation of the seventh American Joint Committee on Cancer/International Union Against Cancer Classification of gastric adenocarcinoma in comparison with the sixth classification. Cancer. 2010;116(24):5592–8.CrossRefPubMed
33.
Zurück zum Zitat Ataseven B, Grimm C, Harter P, et al. Prognostic value of lymph node ratio in patients with advanced epithelial ovarian cancer. Gynecol Oncol. 2014;135(3):435–40.CrossRefPubMed Ataseven B, Grimm C, Harter P, et al. Prognostic value of lymph node ratio in patients with advanced epithelial ovarian cancer. Gynecol Oncol. 2014;135(3):435–40.CrossRefPubMed
34.
Zurück zum Zitat Taylor MD, Lapar DJ, Thomas CJ, et al. Lymph node ratio predicts recurrence and survival after R0 resection for non-small cell lung cancer. Ann Thorac Surg. 2013;96(4):1163–70.CrossRefPubMed Taylor MD, Lapar DJ, Thomas CJ, et al. Lymph node ratio predicts recurrence and survival after R0 resection for non-small cell lung cancer. Ann Thorac Surg. 2013;96(4):1163–70.CrossRefPubMed
35.
Zurück zum Zitat Hou X, Wei JC, Xu Y, et al. The positive lymph node ratio predicts long-term survival in patients with operable thoracic esophageal squamous cell carcinoma in China. Ann Surg Oncol. 2013;20(5):1653–9.CrossRefPubMed Hou X, Wei JC, Xu Y, et al. The positive lymph node ratio predicts long-term survival in patients with operable thoracic esophageal squamous cell carcinoma in China. Ann Surg Oncol. 2013;20(5):1653–9.CrossRefPubMed
36.
Zurück zum Zitat Okusa T, Nakane Y, Boku T, et al. Quantitative analysis of nodal involvement with respect to survival rate after curative gastrectomy for carcinoma. Surg Gynecol Obstet. 1990;170(6):488–94.PubMed Okusa T, Nakane Y, Boku T, et al. Quantitative analysis of nodal involvement with respect to survival rate after curative gastrectomy for carcinoma. Surg Gynecol Obstet. 1990;170(6):488–94.PubMed
Metadaten
Titel
The value of lymph nodes ratios in the prognosis of resectable remnant gastric cancer through the retrospective propensity score matching analysis
verfasst von
Biao Yang
Tao Liu
Hangtian Cui
Zhengmao Lu
Guoen Fang
Xuchao Xue
Tianhang Luo
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
World Journal of Surgical Oncology / Ausgabe 1/2023
Elektronische ISSN: 1477-7819
DOI
https://doi.org/10.1186/s12957-023-03137-z

Weitere Artikel der Ausgabe 1/2023

World Journal of Surgical Oncology 1/2023 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.