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

Open Access 01.12.2023 | Research

Novel prognostic indicator combining inflammatory indicators and tumor markers for gastric cancer

verfasst von: Liang Yu, Runben Jiang, Wanjing Chen, Yanwei Liu, Gui Wang, Xin Gong, Yong Wang

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

Abstract

Background

Gastric cancer (GC) is one of the most common malignant tumors worldwide, and we hope to identify an economical but practical prognostic indicator. It has been reported that inflammatory indicators and tumor markers are associated with GC progression and are widely used to predict prognosis. However, existing prognostic models do not comprehensively analyze these predictors.

Methods

This study retrospectively reviewed 893 consecutive patients who underwent curative gastrectomy from January 1, 2012, to December 31, 2015, in the Second Hospital of Anhui Medical University. Prognostic factors predicting overall survival (OS) were analyzed using univariate and multivariate Cox regression analyses. Nomograms including independent prognostic factors were plotted for predicting survival.

Results

Ultimately, 425 patients were enrolled in this study. Multivariate analyses demonstrated that the neutrophil-to-lymphocyte ratio (NLR, total neutrophil count/lymphocyte count × 100%) and CA19-9 were independent prognostic factors for OS (p=0.001, p=0.016). The NLR-CA19-9 score (NCS) is constructed as the combination of the NLR and CA19-9. We defined NLR<2.46 and CA19-9≤37 U/ml as an NCS of 0, NLR≥2.46 or CA19-9>37 U/ml as an NCS 1, and NLR≥2.46 and CA19-9>37 U/ml as an NCS of 2. The results showed that higher NCS was significantly associated with worse clinicopathological characteristics and OS (p<0.05). Multivariate analyses revealed that the NCS was an independent prognostic factor for OS (NCS1: p<0.001, HR=3.172, 95% CI=2.120–4.745; NCS2: p<0.001, HR=3.052, 95% CI=1.928–4.832). Compared with traditional predictive indices, the NCS had the highest AUC for a 12-month survival, a 36-month survival, a 60-month survival, and OS (AUC= 0.654, 0.730, 0.811, 0.803, respectively). The nomogram had a higher Harrell’s C-index than the TNM stage alone (0.788 vs. 0.743).

Conclusions

The NCS provides more accurate predictions of the prognosis of GC patients, and its predictive value is significantly better than that of traditional inflammatory indicators or tumor markers. It is an effective complement to existing GC assessment systems.
Hinweise

Publisher’s Note

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

Introduction

Gastric cancer (GC) is the fifth-most common malignancy in humans and ranks third in a tumor-related mortality according to the latest epidemiologic data [1]. Radical resection combined with chemotherapy has consistently been the core method for curing GC. Unfortunately, due to the highly aggressive nature of GC, almost 50% of patients suffer from tumor recurrence or metastasis after curative resection, and the 5-year survival rate remains less than 30% [2, 3]. Currently, the most common criteria used to predict GC patients’ long-term outcomes include the TNM staging system, tumor markers, and inflammation indicators, but clinical outcomes can vary in patients who have the same stages and similar treatment regimens [46], indicating that these systems provide incomplete prognostic information.
In 1863, Virchow first discovered the relationship between inflammation and cancer [7]; subsequently, an increasing number of related studies were carried out. A growing number of studies have proven the relationship between malignant tumors and inflammation [812]. Many scholars believe that immune status is closely related to survival in patients with various malignancies, including GC [6, 1315]. The neutrophil-to-lymphocyte ratio (NLR) is a significant prognostic indicator of gastric cancer. Current researchers not only believe that gastric cancer patients with higher NLRs have a poorer prognosis [9, 16] but also indicate to a certain extent that the incidence of complications after surgery, such as anastomotic leakage, has increased [16, 17]. The NLR has been adopted for prognostic evaluation in many cancers, as well as the platelet-to-lymphocyte ratio (PLR) and lymphocyte-to-monocyte ratio (LMR).
The relationship between GC and tumor markers has basically been clarified, and a series of studies have explored the value of tumor markers in the diagnosis and prognosis of gastric cancer [1820]. At the same time, tumor markers, such as carcinoembryonic antigen (CEA) and CA19-9, have been used to determine prognosis and monitor the therapeutic effects of treatments. The level of CEA may be increased in gastric carcinoma, lung carcinoma, and especially colorectal carcinoma, while CA19-9 is used mainly as a specific marker for pancreatic cancer.
However, these indicators remain controversial. For both inflammation indicators and tumor markers, the specificity and sensitivity in predicting the long-term outcome of GC patients alone are poor, and thus, there is an urgent need for a new, easy method to predict GC more accurately and perform targeted follow-up treatment and observation for patients who may have a poorer prognosis. Therefore, we attempted to combine inflammation indicators and tumor markers with a high predictive value to predict the prognosis of GC patients more accurately. In addition, we used prospective clinical data to investigate whether the new indicator could effectively predict postoperative outcomes of GC and compared its predictive value with other traditional indices.

Materials and methods

Patients

Between January 1, 2012, and December 31, 2015, a total of 893 consecutive patients admitted to the Second Hospital of Anhui Medical University were recruited for the trial. Patient eligibility criteria of this study included the following: (1) all patients who underwent gastrectomy with curative R0 resection; (2) postoperative pathology confirmed GC; (3) no active inflammatory, chronic infection, or autoimmune rheumatic diseases; and (4) no other malignancies. Patients who met the following criteria were excluded from this study: (1) serious complications or death that occurred within 15 days after operative, (2) treatment with neoadjuvant chemotherapy, (3) lack of inflammation and tumor marker data, and (4) acute complications such as perforation or bleeding. All patients, except for those with pTNM stage I, received 6–8 cycles of postoperative chemotherapy based on fluorouracil combined with platinum. This study was reviewed and approved by the Ethical Review Committee of the Second Hospital of Anhui Medical University, and the approved number is YX2021-138(f1). The patients were informed orally or in writing about the relevant matters of the study. All patients expressed their complete understanding of the study and signed an informed consent form.

Data collection

The collection of clinical indicators included basic demographic information (age, sex), routine blood tests (total peripheral neutrophils, lymphocyte count, monocyte count, platelet count, CEA, CA19-9), and tumor-related information (size, depth, differentiation, Borrmann type, lymph node, distant metastasis, pathological stage). All blood test data were collected 1 week before surgery. The NLR was calculated by dividing the neutrophil count by the lymphocyte count. The PLR was calculated by dividing the platelet count by the lymphocyte count. The LMR was calculated by dividing the lymphocyte count by the monocyte count. The clinical stage of GC was determined following the eighth American Joint Committee on Cancer (AJCC) guidelines. The optimal cutoff values of CEA (5 ng/ml) and CA19-9 (37 U/ml) were determined by the standards of our hospital.

Follow-up

After surgery, all patients were followed by radiology and laboratory tests every 3 months for 2 years and every 6 months for 2–5 years. In addition, examinations, including chest radiography, endoscopy, or abdominal and pelvic CT, were performed at least once per year. The follow-up period lasted 5 years after surgery or to the date of death. The overall survival (OS) was defined as the time interval from surgery to the last follow-up or to death from any cause.

Statistical analysis

Data are presented as mean ± SD for normally distributed data and median (interquartile range) for data not-normally distributed. Receiver operating characteristic (ROC) curves were generated. The optimal cutoff values of the NLR, PLR, and LMR were obtained through the ROC curves by calculating the Youden indices corresponding to different cutoff values of each inflammatory index in the ROC curve, and the corresponding cutoff values of the maximum value of the Youden index were used to divide the patients into two groups. A Cox proportional hazards regression model was used to identify the independent predictors associated with OS, and variables with a value of p< 0.05 in the univariate analysis were subsequently included in a multivariate analysis. OS was assessed using the Kaplan-Meier method. Differences between the areas under the curve (AUCs) of each inflammatory and tumor marker were compared to determine the predictive value of each index for OS and the postoperative setting time points.
Models with independent prognostic factors were selected to plot the nomogram. The predictive values for survival were determined by Harrell’s C-index. In theory, the higher the C-index is the more precise the prognosis prediction. The decision curve analysis (DCA) plot can be used to represent the model with the greatest net benefits that had the highest clinical use, and it has been widely used to estimate whether the clinical use of diagnostic tests and prediction models would do more good than harm [21]. In the current study, DCA was conducted to evaluate the clinical use of the nomogram by quantifying the net benefits over the AJCC staging system. The calibration curve, the best method to visually compare the accordance between the predicted risk and the actual absolute risk, was used to evaluate the consistency of the model. If the calibration curve of the prediction model was closer to the standard curve, the consistency of the nomogram was better [22].
All statistical analyses were performed using SPSS v.22.0 for Windows (SPSS Inc., Chicago, IL, USA). The nomogram, DCA, and calibration curves were plotted with R Studio (version 1.1.463, with packages “rms,” “survival,” “hmisc,” and “rmda”). p values less than 0.05 were considered statistically significant.

Results

Patient clinicopathological characteristics

Overall, 425 patients were included in the study (Fig. 1). There were 311 (73.18%) males and 114 (26.82%) females. According to the eighth edition of the AJCC staging system, a total of 68 patients (16.00%) had TNM stage I, 162 patients (38.12%) had TNM stage II, 163 patients (38.35%) had TNM stage III, and 32 patients (7.53%) had TNM stage IV. All patients in TNM stage IV were GC patients with isolated hepatic metastases. According to the standard of surgical treatment of gastric cancer, we performed radical resection of the primary and metastatic lesions in these patients. Regarding Borrmann types, 230 patients (54.12%) had Borrmann type I-II, and 195 patients (45.88%) had Borrmann type III-IV. In addition, there were 168 patients (39.53%) with tumor sizes greater than 5 cm and 184 patients (43.29%) with poor differentiation. The characteristics are detailed in Table 1.
Table 1
Patients clinicopathological characteristics
Variables
n (%) or median (IQR)
Sex
 Female
114 (26.82%)
 Male
311 (73.18%)
Age
 <65years
216 (50.82%)
 ≥65years
209 (49.18%)
Tumor size
 <5cm
257 (60.47%)
 ≥5cm
168 (39.53%)
Differentiation
 Moderate/well
241 (56.71%)
 Poor
184 (43.29%)
Borrmann type
 I–II
230 (54.12%)
 III–IV
195 (45.88%)
Tumor depth
 T1
45 (10.59%)
 T2
47 (11.06%)
 T3
230 (54.12%)
 T4
103 (24.24%)
Lymph node
 N0
100 (23.53%)
 N1
181 (42.59%)
 N2
103 (24.24%)
 N3
41 (9.65%)
Distant metastasis
 M0
393 (92.47%)
 M1
32 (7.53%)
pTNM stage
 I
68 (16.00%)
 II
162 (38.12%)
 III
163 (38.35%)
 IV
32 (7.53%)
CEA
 ≤5ng/ml
276 (64.94%)
 >5ng/ml
149 (35.06%)
CA19-9
 ≤37U/ml
317 (74.59%)
 >37U/ml
108 (25.41%)
NLR
2.71 (1.73–4.46)
PLR
136.8 (94.0–207.6)
LMR
4.46 (2.87–6.27)
NLR the neutrophil-to-lymphocyte ratio, PLR the platelet-to-lymphocyte ratio, LMR the lymphocyte-to-monocyte ratio

Survival analysis

In this study, 24 patients were lost during follow-up, with 12-, 36-, and 60-month OS rates of 87.3%, 53.9%, and 43.4%, respectively. The median survival time was 42 months. The optimal cutoff points for preoperative NLR, PLR, and LMR for postoperative survivals were obtained by calculating the maximum Youden index of the ROC curves, which were 2.46, 127.8, and 4.93, respectively (Fig. 2, Table 2), and based on these values, the entire sample was divided into two groups to identify the independent predictors associated with survival. The results of univariate analysis for the whole sample revealed that age, tumor size, differentiation, Borrmann type, tumor depth, lymph node involvement, distant metastasis, pTNM, CEA, CA19-9, NLR, PLR, and LMR were related to OS. Further multivariate analyses revealed that age, tumor depth, pTNM, CA19-9, and NLR were independent risk factors for OS (p=0.038, 0.009, <0.001, 0.016, 0.001, respectively) (Table 3). Kaplan–Meier survival curves comparing the OS of each hematological parameter showed that elevated CEA, CA19-9, NLR, and PLR and decreased LMR were associated with reduced OS (Fig. 3a–e).
Table 2
The optimal cutoff points of NLR, PLR, and LMR
 
Cutoff value
AUC (95%CI)
maximal Youden index
p
NLR
2.46
0.778 (0.734–0.823)
0.508
<0.001
PLR
127.8
0.635 (0.582–0.688)
0.234
<0.001
LMR
4.93
0.287 (0.238–0.336)
0.361
<0.001
NLR the neutrophil-to-lymphocyte ratio, PLR the platelet-to-lymphocyte ratio, LMR the lymphocyte-to-monocyte ratio
Table 3
Univariate and multivariate analyses of variables associated with OS
Variables
Univariate analysis
Multivariate analysis
Hazard ratio (95%CI)
p
Hazard ratio (95%CI)
p
Sex
 
0.728
  
 Female
1
   
 Male
1.052 (0.790–1.403)
   
Age
 
<0.001
 
0.038
 <65 years
1
 
1
 
 ≥65 years
1.963 (1.516–2.541)
 
1.342 (1.017–1.771)
 
Tumor size
 
<0.001
  
 <5cm
1
   
 ≥5cm
1.666 (1.294–2.147)
   
Differentiation
 
0.008
  
 Moderate/well
1
   
 Poor
1.409 (1.094–1.814)
   
Borrmann type
 
<0.001
  
 I–II
1
   
 III–IV
1.614 (1.253–2.079)
   
Tumor depth
 
<0.001
 
0.009
 T1
1
 
1
 
 T2
4.305 (1.215–15.258)
0.024
1.218 (0.202–7.341)
0.829
 T3
11.662 (3.710–36.658)
<0.001
1.200 (0.176–8.203)
0.852
 T4
41.409 (13.073–131.166)
<0.001
2.228 (0.317–15.649)
0.421
Lymph node
 
<0.001
  
 N0
1
   
 N1
5.937 (3.398–10.375)
<0.001
  
 N2
10.133 (5.736–17.902)
<0.001
  
 N3
15.816 (8.540–29.291)
<0.001
  
Distant metastasis
 
<0.001
  
 M0
1
   
 M1
4.458 (3.022–6.577)
   
pTNM stage
 
<0.001
 
<0.001
 I
1
   
 II
6.519 (2.623–16.200)
<0.001
2.673 (0.501–14.263)
0.250
 III
25.415 (10.389–62.172)
<0.001
5.570 (0.915–33.897)
0.062
 IV
51.590 (19.885–133.844)
<0.001
11.700 (1.921–71.263)
0.008
CEA
 
<0.001
  
 ≤5ng/ml
1
   
 >5ng/ml
2.098 (1.626–2.706)
   
CA19-9
 
<0.001
 
0.016
 ≤37U/ml
1
 
1
 
 >37U/ml
2.937 (2.256–3.823)
 
1.442 (1.072–1.939)
 
NLR
 
<0.001
 
0.001
 ≤2.46
1
 
1
 
 >2.46
4.468 (3.303–6.045)
 
1.772 (1.245–2.522)
 
PLR
 
<0.001
  
 ≤127.8
1
   
 >127.8
1.979 (1.518–2.580)
   
LMR
 
<0.001
  
 ≤4.93
1
   
 >4.93
0.365 (0.276–0.484)
   
(1) NLR the neutrophil-to-lymphocyte ratio, PLR the platelet-to-lymphocyte ratio, LMR the lymphocyte-to-monocyte ratio. (2) OS overall survival

Novel prognosis score factor

According to the results of multivariate analysis, a novel prognostic prediction system involving the NLR-CA19-9 score (NCS), which combines the CA19-9 level and the NLR, was established. Based on this, the scoring standard of the comprehensive index was obtained, which ranged from 0 to 2. We defined NLR<2.46 and CA19-9≤37 U/ml as an NCS of 0, NLR≥2.46 or CA19-9>37 U/ml as an NCS 1, and NLR≥2.46 and CA19-9>37 U/ml as an NCS of 2. The NCS was 0 for 168 (39.53%) patients, 1 for 175 (41.18%) patients, and 2 for 82 (19.29%) patients. The association between the NCS and the clinicopathological characteristics of patients with GC is demonstrated in Table 4. A higher NCS was significantly associated with worse clinicopathological characteristics, such as tumor depth, lymph node involvement, pTNM stage, CEA, PLR, and LMR. We continued to analyze the statistical relationship of the NCS and other clinicopathological characteristics with survival. The results showed that the NCS was also an independent prognostic factor for postoperative OS in GC patients, in addition to age, tumor depth, and pTNM (p<0.001, 0.024, 0.018, and <0.001, respectively) (Table 5). Similarly, we performed Kaplan-Meier survival curves based on the NCS, and the results showed that the higher the NCS was, the shorter the survival period, which indicated a worse prognosis (Fig. 4).
Table 4
Statistical relationship between the NCS and other variables
Variables
NCS
p
0 n (%)
1 n (%)
2 n (%)
Sex
0.585
 Female
44 (26.19%)
51 (29.14%)
19 (23.17%)
 Male
124 (73.81%)
124 (70.86%)
63 (76.83%)
Age
<0.001
 <65years
106 (63.10%)
85 (48.57%)
25 (30.49%)
 ≥65years
62 (36.90%)
90 (51.43%)
57 (69.51%)
Tumor size
0.001
 <5cm
120 (71.43%)
97 (55.43%)
40 (48.78%)
 ≥5cm
48 (28.57%)
78 (44.57%)
42 (51.22%)
Differentiation
0.252
 Moderate/well
100 (9.52%)
101 (57.71%)
40 (48.78%)
 Poor
68 (40.48%)
74 (42.29%)
42 (51.22%)
Borrmann type
0.001
 I–II
109 (64.88%)
87 (49.71%)
34 (41.46%)
 III–IV
59 (35.12%)
88 (50.29%)
48 (58.54%)
Tumor depth
<0.001
 T1
35 (20.83%)
10 (5.71%)
0 (0)
 T2
30 (17.86%)
15 (8.57%)
2 (2.44%)
 T3
90 (53.57%)
100 (57.14%)
40 (48.78%)
 T4
13 (7.74%)
50 (28.57%)
40 (48.78%)
Lymph node
<0.001
 N0
70 (41.67%)
24 (13.71%)
6 (7.32%)
 N1
63 (37.50%)
81 (46.29%)
37 (45.12%)
 N2
30 (17.86%)
48 (27.43%)
25 (30.49%)
 N3
5 (2.98%)
22 (12.57%)
14 (17.07%)
Distant metastasis
<0.001
 M0
166 (98.81%)
157 (89.71%)
70 (85.37%)
 M1
2 (1.19%)
18 (10.29%)
12 (14.63%)
pTNM stage
<0.001
 I
53 (31.55%)
14 (8.00%)
1 (1.22%)
 II
75 (44.64%)
69 (39.43%)
18 (21.95%)
 III
38 (22.62%)
74 (42.29%)
51 (62.20%)
 IV
2 (1.19%)
18 (10.29%)
12 (14.63%)
CEA
<0.001
 ≤5ng/ml
130 (77.38%)
117 (66.86%)
29 (35.37%)
 >5ng/ml
38 (22.62%)
58 (33.14%)
53 (64.63%)
PLR
<0.001
 ≤127.8
112 (66.67%)
62 (35.43%)
20 (24.39%)
 >127.8
56 (33.33%)
113 (64.57%)
62 (75.61%)
LMR
<0.001
 ≤4.93
52 (30.95%)
114 (65.14%)
71 (86.59%)
 >4.93
116 (69.05%)
61 (34.86%)
11 (13.41%)
(1) PLR the platelet-to-lymphocyte ratio, LMR the lymphocyte-to-monocyte ratio. (2) NCS the NLR-CA19-9 score
Table 5
Univariate and multivariate analyses incorporating the NCS
Variables
Univariate analysis
Multivariate analysis
Hazard ratio (95%CI)
p
Hazard ratio (95%CI)
p
Sex
 
0.728
  
 Female
1
   
 Male
1.052 (0.790–1.403)
   
Age
 
<0.001
 
0.024
 <65years
1
 
1
 
 ≥65years
1.963 (1.516–2.541)
 
1.375 (1.042–1.813)
 
Tumor size
 
<0.001
  
 <5cm
1
   
 ≥5cm
1.666 (1.294–2.147)
   
Differentiation
 
0.008
  
 Moderate/well
1
   
 Poor
1.409 (1.094–1.814)
   
Borrmann type
 
<0.001
  
 I–II
1
   
 III–IV
1.614 (1.253–2.079)
   
Tumor depth
 
<0.001
 
0.018
 T1
1
 
1
 
 T2
4.305 (1.215–15.258)
0.024
1.332 (0.221–8.038)
0.755
 T3
11.662 (3.710–36.658)
<0.001
1.240 (0.181–8.488)
0.827
 T4
41.409 (13.073–131.166)
<0.001
2.202 (0.313–15.492)
0.428
Lymph node
 
<0.001
  
 N0
1
   
 N1
5.937 (3.398–10.375)
<0.001
  
 N2
10.133 (5.736–17.902)
<0.001
  
 N3
15.816 (8.540–29.291)
<0.001
  
Distant metastasis
 
<0.001
  
 M0
1
   
 M1
4.458 (3.022–6.577)
   
pTNM stage
 
<0.001
 
<0.001
 I
1
 
1
 
 II
6.519 (2.623–16.200)
<0.001
2.396 (0.446–12.859)
0.308
 III
25.415 (10.389–62.172)
<0.001
5.445 (0.889–33.357)
0.067
 IV
51.590 (19.885–133.844)
<0.001
10.274 (1.677–62.953)
0.012
CEA
 
<0.001
  
 ≤5ng/ml
1
   
 >5ng/ml
2.098 (1.626–2.706)
   
PLR
 
<0.001
  
 ≤127.8
1
   
 >127.8
1.979 (1.518–2.580)
   
LMR
 
<0.001
  
 ≤4.93
1
   
 >4.93
0.365 (0.276–0.484)
   
NCS
 
<0.001
 
<0.001
 0
1
 
1
 
 1
5.874 (4.048–8.524)
<0.001
3.172 (2.120–4.745)
<0.001
 2
8.710 (5.795–13.093)
<0.001
3.052 (1.928–4.832)
<0.001
(1) PLR the platelet-to-lymphocyte ratio, LMR the lymphocyte-to-monocyte ratio. (2) NCS the NLR-CA19-9 score

Predictive value

To compare the predictive power of the NCS with other hematological parameters for OS and 12-, 36-, and 60-month survival, we compared the AUC of each inflammatory and tumor marker with that of the NCS. It was confirmed that the NCS had the highest AUC (0.803, 0.763, 0.811, respectively) for OS and 36- and 60-month survival, respectively, and the differences were all statistically significant (p<0.05) (Fig. 5a–d, Table 6). The higher AUC further confirmed the favorable sensitivity and specificity of the NCS.
Table 6
Comparison of the AUC between inflammatory indicators and tumor markers
Variables
OS
12 months
36 months
60mo
AUC (95%CI)
z statistic
p*
AUC (95%CI)
z statistic
p*
AUC (95%CI)
Z statistic
p*
AUC (95%CI)
z statistic
p*
CEA
0.637 (0.589–0.682)
5.962
<0.001
0.586 (0.537–0.633)
1.768
0.077
0.603 (0.554–0.649)
5.734
<0.001
0.636 (0.589–0.682)
6.270
<0.001
CA19-9
0.652 (0.605–0.697)
8.562
<0.001
0.598 (0.550–0.645)
2.138
0.033
0.644 (0.597–0.690)
6.726
<0.001
0.655 (0.608–0.700)
8.841
<0.001
NLR
0.754 (0.710–0.794)
4.039
<0.001
0.613 (0.565–0.660)
1.707
0.088
0.713 (0.668–0.756)
3.676
<0.001
0.761 (0.717–0.801)
4.089
<0.001
PLR
0.620 (0.572–0.666)
6.818
<0.001
0.581 (0.533–0.629)
1.651
0.099
0.619 (0.570–0.665)
5.311
<0.001
0.613 (0.565–0.659)
7.419
<0.001
LMR
0.680 (0.634–0.724)
4.711
<0.001
0.573 (0.534–0.621)
2.385
0.017
0.653 (0.605–0.698)
4.406
<0.001
0.683 (0.636–0.727)
4.936
<0.001
NCS
0.803 (0.762–0.840)
-
-
0.654 (0.607–0.699)
-
-
0.763 (0.720–0.803)
-
-
0.811 (0.770–0.847)
-
-
(1) NLR the neutrophil-to-lymphocyte ratio, PLR the platelet-to-lymphocyte ratio, LMR the lymphocyte-to-monocyte ratio. (2) NCS the NLR-CA19-9 score
*Comparison of AUC between the NLR-CA19-9 score and other variables

Nomogram

To make individualized predictions of the survival probability in all GC patients, we combined all independent prognostic factors, including age, TNM stage, tumor depth, and NCS, as described in detail in Table 4, and established a nomogram for the entire cohort (Fig. 6).
Discrimination and calibration are both important features of model performance. In this study, Harrell’s C-index of our nomogram (0.788, 95% CI 0.813–0.762) was higher than that of the TNM stage alone (0.743, 95% CI 0.770–0.717), indicating a more accurate and robust performance estimate. In addition, the DCA of the novel nomogram revealed a superior net clinical benefit over the 8th AJCC TNM staging system alone and exhibited higher clinical use than the AJCC staging system in OS prediction (Fig. 7). Furthermore, the calibration plots showed super agreement between the actual and predicted survival (Fig. 8a–c). All the above results show the better predictive capability of the established nomogram over the existing AJCC staging system.

Discussion

Advances in modern medicine have allowed scholars to gradually turned their attention to the early treatment and follow-up of various malignant tumors, which puts forward extremely high requirements on how to distinguish patients with a poor prognosis. To date, researchers have established several scoring systems that reflect inflammation or tumor status [2325]. Our study enrolled 425 GC patients and found that the NLR and CA19-9 were independent prognostic factors for postoperative OS. Based on this, we developed a novel index combining these inflammatory and tumor markers and confirmed that the new index could provide better prognostic value than either the NLR or CA19-9 alone.
Inflammation is an important characteristic of the tumor microenvironment and is associated with the promotion, progression, and metastasis of tumors [8]. Tumor cells produce cancer-related inflammatory mediators, resulting in relative neutrophilia, thrombocytosis, and lymphocytopenia. These phenomena cause an elevated NLR and PLR [26] and further affect the occurrence, progression, and metastasis of tumors [27]. Neutrophils are currently believed to promote cancer cell proliferation and metastasis by producing proangiogenic chemokines and vascular endothelial growth factor [2830], and lymphocytes are antitumor factors involved in cytotoxic activity [31]. Numerous studies have proven the relationship between a high NLR and poor outcomes in various malignancies, such as gastric cancer [9, 16], colon cancer [13], and pancreatic cancer [32]. The present study revealed the NLR as an independent factor with a cutoff value of 2.46, and its AUC was higher than that of other hematological parameters.
Carbohydrate antigen (CA) 19-9 is one of the most common tumor markers of gastric cancer, and positivity is frequently related to tumor stage, poor prognosis, recurrence, and metastasis [1820]. Jing et al. [19] found that surgery can significantly reduce the level of CA19-9; if the level returns to normal after surgery, the prognosis is not significantly different from that of patients with normal CA19-9 before surgery. As confirmed in our study, the prognosis of GC patients with low levels of CA19-9 before surgery was significantly better than that of patients with high levels. Of course, we consider the NLR and CA19-9 to represent not only simple changes in several indicators but also the balance of tumor and antitumor status in the body. When this balance is broken, tumor promotion is prioritized, leading to a poor prognosis.
Unfortunately, previous conclusions on these indicators have not always been consistent. Some scholars have insisted that there was no significant correlation between the preoperative NLR and survival time in GC patients [3335]. In addition, controversy about CA19-9 also exists. Researchers have reported a low positive rate of CA19-9 in GC patients [36, 37]. In addition, some scholars believe that CA19-9 cannot accurately predict the prognosis of patients with GC [18], especially early GC [37]. This means that using an indicator alone to predict prognosis may lose some potential information, resulting in limited predictive value. In this study, we discussed the predictive significance of various inflammatory indicators and tumor markers in the prognosis of GC in detail and innovatively introduced a new index, the NLR-CA19-9 score (NCS). We found that the NCS not only could serve as an independent prognostic factor but also had a higher AUC than other inflammatory and tumor markers for OS, 12-, 36-, and 60-month survival, which means that the NCS has a stable prognostic ability that was better than that of other indicators. It was worth noting that there were no significant differences in AUC between the NCS and NLR, PLR, or CEA at 12 months after surgery. We believed that the survival of patients at 12 months after surgery was relatively higher (87.3%), resulting in no statistically significant difference. With the progress of follow-up, the survival of patients had declined, and the differences had gradually become apparent. Therefore, the differences were all statistically significant for OS and 36- and 60-month survival (p<0.05). Moreover, a model including the NCS (C-index, 0.788) was superior to that constructed without it (C-index, 0.743) in predicting OS after radical gastrectomy. Importantly, the novel index in our model could be conveniently attained from routine laboratory inspection, which was advantageous because it avoids the potential unavailability of other predictors.
The results of this study that the NCS, as a novel scoring system, has favorable sensitivity and specificity in predicting the long-term postoperative outcomes of GC patients and could provide more precise and informative prognostic value than other indicators. This new indicator may have vital use in predicting recurrence, therapeutic intervention, and surveillance strategies, especially when incorporating the TNM stage. For example, patients with a higher NCS could be strongly recommended to receive postoperative multimodal treatment, such as chemotherapy, immunotherapy, or targeted therapy and should receive a more rigorous follow-up schedule, with comprehensive medical examinations at least every 3 months for 5 years, such as gastrointestinal endoscopy, CT with contrast, and serum tumor markers, which may provide a survival benefit for GC patients.
Several limitations remain in our present study. First, the main drawback of our study is its retrospective nature, and bias in the process of patient selection cannot be avoided. Second, because the specific time of tumor recurrence in some patients cannot be completely determined, we did not study the relationship between various indicators and postoperative progression-free survival (PFS). Third, the pathological stages of the patients in our study are relatively broad, so the conclusion may not apply to patients with a particular TNM stage. But we are still expanding the sample size to verify whether our conclusion are applicable to similar stages.

Conclusion

In conclusion, to the best of our knowledge, the NCS is an independent prognostic factor for OS in GC patients and provides great value in predicting postoperative overall survival. It is an effective complement to existing GC assessment systems. Future studies should consider combining the NCS into the current TNM system to more specifically predict the prognosis of GC patients, who will be likely to benefit from a rigorous follow-up strategy.

Acknowledgements

The authors are grateful for the kind assistance from their colleagues.

Declarations

This study was reviewed and approved by the Ethical Review Committee of the Second Hospital of Anhui Medical University. All patients expressed their complete understanding of the study and signed an informed consent form. We confirmed that all methods were performed in accordance with the relevant guidelines and regulations.
Not applicable.

Competing interests

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRefPubMed Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRefPubMed
2.
Zurück zum Zitat Duarte HO, Gomes J, Machado JC, Reis CA. Gastric cancer: basic aspects. Helicobacter. 2018;23(Suppl 1):e12523.CrossRefPubMed Duarte HO, Gomes J, Machado JC, Reis CA. Gastric cancer: basic aspects. Helicobacter. 2018;23(Suppl 1):e12523.CrossRefPubMed
3.
Zurück zum Zitat Li S, Cong X, Gao H, Lan X, Li Z, Wang W, et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 2019;38(1):6.CrossRefPubMedPubMedCentral Li S, Cong X, Gao H, Lan X, Li Z, Wang W, et al. Tumor-associated neutrophils induce EMT by IL-17a to promote migration and invasion in gastric cancer cells. J Exp Clin Cancer Res. 2019;38(1):6.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Sasako M, Inoue M, Lin JT, Khor C, Yang HK, Ohtsu A. Gastric Cancer Working Group report. Jpn J Clin Oncol. 2010;40(Suppl 1):i28–37.CrossRefPubMed Sasako M, Inoue M, Lin JT, Khor C, Yang HK, Ohtsu A. Gastric Cancer Working Group report. Jpn J Clin Oncol. 2010;40(Suppl 1):i28–37.CrossRefPubMed
5.
Zurück zum Zitat Noh SH, Park SR, Yang HK, Chung HC, Chung IJ, Kim SW, et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15(12):1389–96.CrossRefPubMed Noh SH, Park SR, Yang HK, Chung HC, Chung IJ, Kim SW, et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15(12):1389–96.CrossRefPubMed
6.
Zurück zum Zitat Wang H, Ding Y, Li N, Wu L, Gao Y, Xiao C, et al. Prognostic value of neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, and combined neutrophil-lymphocyte ratio and platelet-lymphocyte ratio in stage IV advanced gastric cancer. Front Oncol. 2020;10:841.CrossRefPubMedPubMedCentral Wang H, Ding Y, Li N, Wu L, Gao Y, Xiao C, et al. Prognostic value of neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, and combined neutrophil-lymphocyte ratio and platelet-lymphocyte ratio in stage IV advanced gastric cancer. Front Oncol. 2020;10:841.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–45.CrossRefPubMed Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet. 2001;357(9255):539–45.CrossRefPubMed
8.
Zurück zum Zitat Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.CrossRefPubMed Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.CrossRefPubMed
9.
Zurück zum Zitat Fang T, Wang Y, Yin X, Zhai Z, Zhang Y, Yang Y, et al. Diagnostic sensitivity of NLR and PLR in early diagnosis of gastric cancer. J Immunol Res. 2020;2020:9146042.CrossRefPubMedPubMedCentral Fang T, Wang Y, Yin X, Zhai Z, Zhang Y, Yang Y, et al. Diagnostic sensitivity of NLR and PLR in early diagnosis of gastric cancer. J Immunol Res. 2020;2020:9146042.CrossRefPubMedPubMedCentral
10.
11.
Zurück zum Zitat Hou J, Karin M, Sun B. Targeting cancer-promoting inflammation - have anti-inflammatory therapies come of age? Nat Rev Clin Oncol. 2021;18(5):261–79.CrossRefPubMedPubMedCentral Hou J, Karin M, Sun B. Targeting cancer-promoting inflammation - have anti-inflammatory therapies come of age? Nat Rev Clin Oncol. 2021;18(5):261–79.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Inoue H, Kosuga T, Kubota T, Konishi H, Shiozaki A, Okamoto K, et al. Significance of a preoperative systemic immune-inflammation index as a predictor of postoperative survival outcomes in gastric cancer. World J Surg Oncol. 2021;19(1):173.CrossRefPubMedPubMedCentral Inoue H, Kosuga T, Kubota T, Konishi H, Shiozaki A, Okamoto K, et al. Significance of a preoperative systemic immune-inflammation index as a predictor of postoperative survival outcomes in gastric cancer. World J Surg Oncol. 2021;19(1):173.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Dolan RD, McSorley ST, Park JH, Watt DG, Roxburgh CS, Horgan PG, et al. The prognostic value of systemic inflammation in patients undergoing surgery for colon cancer: comparison of composite ratios and cumulative scores. Br J Cancer. 2018;119(1):40–51.CrossRefPubMedPubMedCentral Dolan RD, McSorley ST, Park JH, Watt DG, Roxburgh CS, Horgan PG, et al. The prognostic value of systemic inflammation in patients undergoing surgery for colon cancer: comparison of composite ratios and cumulative scores. Br J Cancer. 2018;119(1):40–51.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Nozoe T, Iguchi T, Egashira A, Adachi E, Matsukuma A, Ezaki T. Significance of modified Glasgow prognostic score as a useful indicator for prognosis of patients with gastric carcinoma. Am J Surg. 2011;201(2):186–91.CrossRefPubMed Nozoe T, Iguchi T, Egashira A, Adachi E, Matsukuma A, Ezaki T. Significance of modified Glasgow prognostic score as a useful indicator for prognosis of patients with gastric carcinoma. Am J Surg. 2011;201(2):186–91.CrossRefPubMed
15.
Zurück zum Zitat Lu J, Xu BB, Zheng ZF, Xie JW, Wang JB, Lin JX, et al. CRP/prealbumin, a novel inflammatory index for predicting recurrence after radical resection in gastric cancer patients: post hoc analysis of a randomized phase III trial. Gastric Cancer. 2019;22(3):536–45.CrossRefPubMed Lu J, Xu BB, Zheng ZF, Xie JW, Wang JB, Lin JX, et al. CRP/prealbumin, a novel inflammatory index for predicting recurrence after radical resection in gastric cancer patients: post hoc analysis of a randomized phase III trial. Gastric Cancer. 2019;22(3):536–45.CrossRefPubMed
16.
Zurück zum Zitat Huang Z, Liu Y, Yang C, Li X, Pan C, Rao J, et al. Combined neutrophil/platelet/lymphocyte/differentiation score predicts chemosensitivity in advanced gastric cancer. BMC Cancer. 2018;18(1):515.CrossRefPubMedPubMedCentral Huang Z, Liu Y, Yang C, Li X, Pan C, Rao J, et al. Combined neutrophil/platelet/lymphocyte/differentiation score predicts chemosensitivity in advanced gastric cancer. BMC Cancer. 2018;18(1):515.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Radulescu D, Baleanu VD, Padureanu V, Radulescu PM, Bordu S, Patrascu S, et al. Neutrophil/lymphocyte ratio as predictor of anastomotic leak after gastric cancer surgery. Diagnostics (Basel). 2020;10(10):799.CrossRefPubMed Radulescu D, Baleanu VD, Padureanu V, Radulescu PM, Bordu S, Patrascu S, et al. Neutrophil/lymphocyte ratio as predictor of anastomotic leak after gastric cancer surgery. Diagnostics (Basel). 2020;10(10):799.CrossRefPubMed
18.
Zurück zum Zitat Gong X, Zhang H. Diagnostic and prognostic values of anti-helicobacter pylori antibody combined with serum CA724, CA19-9, and CEA for young patients with early gastric cancer. J Clin Lab Anal. 2020;34(7):e23268.CrossRefPubMedPubMedCentral Gong X, Zhang H. Diagnostic and prognostic values of anti-helicobacter pylori antibody combined with serum CA724, CA19-9, and CEA for young patients with early gastric cancer. J Clin Lab Anal. 2020;34(7):e23268.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Kambara Y, Miyake H, Nagai H, Yoshioka Y, Shibata K, Asai S, et al. CA19-9 is a significant prognostic marker of patients with stage III gastric cancer. Eur J Surg Oncol. 2020;46(10 Pt A):1918–24.CrossRefPubMed Kambara Y, Miyake H, Nagai H, Yoshioka Y, Shibata K, Asai S, et al. CA19-9 is a significant prognostic marker of patients with stage III gastric cancer. Eur J Surg Oncol. 2020;46(10 Pt A):1918–24.CrossRefPubMed
21.
Zurück zum Zitat Vickers AJ, Van Calster B, Steyerberg EW. Net benefit approaches to the evaluation of prediction models, molecular markers, and diagnostic tests. BMJ. 2016;352:i6.CrossRefPubMedPubMedCentral Vickers AJ, Van Calster B, Steyerberg EW. Net benefit approaches to the evaluation of prediction models, molecular markers, and diagnostic tests. BMJ. 2016;352:i6.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Guo L, Wang Q, Chen K, Liu HP, Chen X. Prognostic value of combination of inflammatory and tumor markers in resectable gastric cancer. J Gastrointest Surg. 2021;25(10):2470–83.CrossRefPubMed Guo L, Wang Q, Chen K, Liu HP, Chen X. Prognostic value of combination of inflammatory and tumor markers in resectable gastric cancer. J Gastrointest Surg. 2021;25(10):2470–83.CrossRefPubMed
23.
Zurück zum Zitat Luo G, Jin K, Deng S, Cheng H, Fan Z, Gong Y, et al. Roles of CA19-9 in pancreatic cancer: biomarker, predictor and promoter. Biochim Biophys Acta Rev Cancer. 2021;1875(2):188409.CrossRefPubMed Luo G, Jin K, Deng S, Cheng H, Fan Z, Gong Y, et al. Roles of CA19-9 in pancreatic cancer: biomarker, predictor and promoter. Biochim Biophys Acta Rev Cancer. 2021;1875(2):188409.CrossRefPubMed
24.
Zurück zum Zitat Ng K, Stenzl A, Sharma A, Vasdev N. Urinary biomarkers in bladder cancer: a review of the current landscape and future directions. Urol Oncol. 2021;39(1):41–51.CrossRefPubMed Ng K, Stenzl A, Sharma A, Vasdev N. Urinary biomarkers in bladder cancer: a review of the current landscape and future directions. Urol Oncol. 2021;39(1):41–51.CrossRefPubMed
25.
Zurück zum Zitat Horii M, Matsushita T. Regulatory B cells and T cell regulation in cancer. J Mol Biol. 2021;433(1):166685.CrossRefPubMed Horii M, Matsushita T. Regulatory B cells and T cell regulation in cancer. J Mol Biol. 2021;433(1):166685.CrossRefPubMed
26.
Zurück zum Zitat Arigami T, Okumura H, Matsumoto M, Uchikado Y, Uenosono Y, Kita Y, et al. Analysis of the fibrinogen and neutrophil-lymphocyte ratio in esophageal squamous cell carcinoma: a promising blood marker of tumor progression and prognosis. Medicine (Baltimore). 2015;94(42):e1702.CrossRefPubMedPubMedCentral Arigami T, Okumura H, Matsumoto M, Uchikado Y, Uenosono Y, Kita Y, et al. Analysis of the fibrinogen and neutrophil-lymphocyte ratio in esophageal squamous cell carcinoma: a promising blood marker of tumor progression and prognosis. Medicine (Baltimore). 2015;94(42):e1702.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Fankhauser CD, Sander S, Roth L, Gross O, Eberli D, Sulser T, et al. Systemic inflammatory markers have independent prognostic value in patients with metastatic testicular germ cell tumours undergoing first-line chemotherapy. Br J Cancer. 2018;118(6):825–30.CrossRefPubMedPubMedCentral Fankhauser CD, Sander S, Roth L, Gross O, Eberli D, Sulser T, et al. Systemic inflammatory markers have independent prognostic value in patients with metastatic testicular germ cell tumours undergoing first-line chemotherapy. Br J Cancer. 2018;118(6):825–30.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Liang W, Ferrara N. The complex role of neutrophils in tumor angiogenesis and metastasis. Cancer Immunol Res. 2016;4(2):83–91.CrossRefPubMed Liang W, Ferrara N. The complex role of neutrophils in tumor angiogenesis and metastasis. Cancer Immunol Res. 2016;4(2):83–91.CrossRefPubMed
29.
Zurück zum Zitat Swierczak A, Mouchemore KA, Hamilton JA, Anderson RL. Neutrophils: important contributors to tumor progression and metastasis. Cancer Metastasis Rev. 2015;34(4):735–51.CrossRefPubMed Swierczak A, Mouchemore KA, Hamilton JA, Anderson RL. Neutrophils: important contributors to tumor progression and metastasis. Cancer Metastasis Rev. 2015;34(4):735–51.CrossRefPubMed
30.
Zurück zum Zitat McCourt M, Wang JH, Sookhai S, Redmond HP. Proinflammatory mediators stimulate neutrophil-directed angiogenesis. Arch Surg. 1999;134(12):1325–32.CrossRefPubMed McCourt M, Wang JH, Sookhai S, Redmond HP. Proinflammatory mediators stimulate neutrophil-directed angiogenesis. Arch Surg. 1999;134(12):1325–32.CrossRefPubMed
31.
Zurück zum Zitat Li Z, Li S, Ying X, Zhang L, Shan F, Jia Y, et al. The clinical value and usage of inflammatory and nutritional markers in survival prediction for gastric cancer patients with neoadjuvant chemotherapy and D2 lymphadenectomy. Gastric Cancer. 2020;23(3):540–9.CrossRefPubMedPubMedCentral Li Z, Li S, Ying X, Zhang L, Shan F, Jia Y, et al. The clinical value and usage of inflammatory and nutritional markers in survival prediction for gastric cancer patients with neoadjuvant chemotherapy and D2 lymphadenectomy. Gastric Cancer. 2020;23(3):540–9.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Bhatti I, Peacock O, Lloyd G, Larvin M, Hall RI. Preoperative hematologic markers as independent predictors of prognosis in resected pancreatic ductal adenocarcinoma: neutrophil-lymphocyte versus platelet-lymphocyte ratio. Am J Surg. 2010;200(2):197–203.CrossRefPubMed Bhatti I, Peacock O, Lloyd G, Larvin M, Hall RI. Preoperative hematologic markers as independent predictors of prognosis in resected pancreatic ductal adenocarcinoma: neutrophil-lymphocyte versus platelet-lymphocyte ratio. Am J Surg. 2010;200(2):197–203.CrossRefPubMed
33.
Zurück zum Zitat Zhu GS, Tian SB, Wang H, Ma MG, Liu Y, Du HS LYP. Preoperative neutrophil lymphocyte ratio and platelet lymphocyte ratio cannot predict lymph node metastasis and prognosis in patients with early gastric cancer: a single institution investigation in China. Curr Med Sci. 2018;38(1):78–84.CrossRefPubMed Zhu GS, Tian SB, Wang H, Ma MG, Liu Y, Du HS LYP. Preoperative neutrophil lymphocyte ratio and platelet lymphocyte ratio cannot predict lymph node metastasis and prognosis in patients with early gastric cancer: a single institution investigation in China. Curr Med Sci. 2018;38(1):78–84.CrossRefPubMed
34.
Zurück zum Zitat Toyokawa T, Muguruma K, Tamura T, Sakurai K, Amano R, Kubo N, et al. Comparison of the prognostic impact and combination of preoperative inflammation-based and/or nutritional markers in patients with stage II gastric cancer. Oncotarget. 2018;9(50):29351–64.CrossRefPubMedPubMedCentral Toyokawa T, Muguruma K, Tamura T, Sakurai K, Amano R, Kubo N, et al. Comparison of the prognostic impact and combination of preoperative inflammation-based and/or nutritional markers in patients with stage II gastric cancer. Oncotarget. 2018;9(50):29351–64.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Sun KY, Xu JB, Chen SL, Yuan YJ, Wu H, Peng JJ, et al. Novel immunological and nutritional-based prognostic index for gastric cancer. World J Gastroenterol. 2015;21(19):5961–71.CrossRefPubMedPubMedCentral Sun KY, Xu JB, Chen SL, Yuan YJ, Wu H, Peng JJ, et al. Novel immunological and nutritional-based prognostic index for gastric cancer. World J Gastroenterol. 2015;21(19):5961–71.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Liang Y, Wang W, Fang C, Raj SS, Hu WM, Li QW, et al. Clinical significance and diagnostic value of serum CEA, CA19-9 and CA72-4 in patients with gastric cancer. Oncotarget. 2016;7(31):49565–73.CrossRefPubMedPubMedCentral Liang Y, Wang W, Fang C, Raj SS, Hu WM, Li QW, et al. Clinical significance and diagnostic value of serum CEA, CA19-9 and CA72-4 in patients with gastric cancer. Oncotarget. 2016;7(31):49565–73.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Yu J, Zheng W. An alternative method for screening gastric cancer based on serum levels of CEA, CA19-9, and CA72-4. J Gastrointest Cancer. 2018;49(1):57–62.CrossRefPubMed Yu J, Zheng W. An alternative method for screening gastric cancer based on serum levels of CEA, CA19-9, and CA72-4. J Gastrointest Cancer. 2018;49(1):57–62.CrossRefPubMed
Metadaten
Titel
Novel prognostic indicator combining inflammatory indicators and tumor markers for gastric cancer
verfasst von
Liang Yu
Runben Jiang
Wanjing Chen
Yanwei Liu
Gui Wang
Xin Gong
Yong Wang
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/s12957-023-02926-w

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.