Retrospective Study Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 28, 2024; 30(24): 3059-3075
Published online Jun 28, 2024. doi: 10.3748/wjg.v30.i24.3059
Development and validation of a prognostic immunoinflammatory index for patients with gastric cancer
Zhi-Chang Ba, Yuan-Zhou Li, Department of Radiology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
Zhi-Chang Ba, Xi-Qing Zhu, Zhi-Chang Ba and Xi-Qing Zhu.
Xi-Qing Zhu, Zhi-Guo Li, Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
ORCID number: Zhi-Chang Ba (0009-0002-5743-0258); Xi-Qing Zhu (0009-0005-0414-9974); Yuan-Zhou Li (0009-0004-1792-2332).
Author contributions: Ba ZX and Zhu XQ contributed equally to this work; Ba ZC contributed to writing of the original draft and review & editing of the manuscript; Ba ZC and Zhu XQ contributed to data curation and investigation; Zhu XQ contributed to methodology and supervision; Li ZG and Li YZ contributed to resources, funding acquisition, and project administration.
Supported by Clinical Research Foundation of Jie-Ping Wu Medical Foundation, No. 320.6750.2022-07-13.
Institutional review board statement: This study was approved by the Ethics Committee of Harbin Medical University Cancer Hospital (No. 2019-57-IIT).
Informed consent statement: All study participants, or their legal guardian, provided informed written consent prior to study enrollment.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: The material supporting the conclusion of this article has been included within the article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: Https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Yuan-Zhou Li, PhD, Professor, Department of Radiology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang Province, China. 830667@hrbmu.edu.cn
Received: April 5, 2024
Revised: May 22, 2024
Accepted: May 29, 2024
Published online: June 28, 2024
Processing time: 81 Days and 8.3 Hours

Abstract
BACKGROUND

Studies have demonstrated the influence of immunity and inflammation on the development of tumors. Although single biomarkers of immunity and inflammation have been shown to be clinically predictive, the use of biomarkers integrating both to predict prognosis in patients with gastric cancer remains to be investigated.

AIM

To investigate the prognostic and clinical significance of inflammatory biomarkers and lymphocytes in patients undergoing surgical treatment for gastric cancer.

METHODS

Univariate COX regression analysis was performed to identify potential prognostic factors for patients with gastric cancer undergoing surgical treatment. Least absolute shrinkage and selection operator-COX (LASSO-COX) regression analysis was performed to integrate these factors and formulate a new prognostic immunoinflammatory index (PII). The correlation between PII and clinical characteristics was statistically analyzed. Nomograms incorporating the PII score were devised and validated based on the time-dependent area under the curve and decision curve analysis.

RESULTS

Patients exhibiting elevated neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and systemic immune inflammatory index displayed inferior progression-free survival (PFS) and overall survival (OS). Conversely, low levels of CD3(+), CD3(+) CD8(+), CD4(+)CD8(+), and CD3(+)CD16(+)CD56(+) T lymphocytes were associated with improved PFS and OS, while high CD19(+) T lymphocyte levels were linked to worse PFS and OS. The PII score demonstrated associations with tumor characteristics (primary tumor site and tumor size), establishing itself as an independent prognostic factor for both PFS and OS. Time-dependent area under the curve and decision curve analysis affirmed the effectiveness of the PII-based nomogram as a robust prognostic predictive model.

CONCLUSION

PII may be a reliable predictor of prognosis in patients with gastric cancer undergoing surgical treatment, and it offers insights into cancer-related immune-inflammatory responses, with potential significance in clinical practice.

Key Words: Gastric cancer, Immunity, Inflammation, Prognosis, Nomogram

Core Tip: We conducted a retrospective study in which data for lymphocyte subsets and blood inflammatory markers were collected from 291 patients who underwent surgical treatment for gastric cancer. Least absolute shrinkage and selection operator-COX regression analysis was used to construct a new immune-inflammatory biomarker. Internal validation divided into training and validation sets confirmed that patients with higher prognostic immunoinflammatory index scores had a worse prognosis.



INTRODUCTION

Gastric cancer is the fifth most prevalent malignancy globally, accounting for > 1 million new cases annually and posing a persistent public health challenge[1]. Investigations on the incidence and mortality trends of gastric cancer underscore a consistent decline worldwide[2-4]. Despite this trend, East Asia remains disproportionately burdened with the highest rates of gastric cancer incidence and mortality among geographical regions[5,6]. Advances in multimodal treatment strategies for gastric cancer have been substantiated by scientific evidence[7,8]. Surgical resection is currently the primary therapeutic option for resectable gastric cancer, while cases with advanced or inoperable disease benefit from che-motherapy, immunotherapy, and targeted therapies[9-11]. However, patients with advanced metastases face a grim prognosis, with a median survival time of approximately 1 year[12]. Traditional classification tools provide limited prognostic information for patients with gastric cancer, necessitating the exploration of convenient and effective clinical biomarkers to guide treatment and assess prognosis[13].

Cancer is recognized as a chronic inflammatory condition, in which neutrophils and monocytes infiltrate adipose tissue and other tissues, thereby contributing to nutrient depletion and disease progression[14]. Inflammation emerges as a pivotal risk factor for tumor advancement[15]. Peripheral blood biomarkers, such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and the systemic immune inflammatory index (SII) which is responsive to systemic inflammation, have been established as prognostic assessment tools for various types of cancer[16-19]. Beyond assessing the degree of inflammation, consideration of the body's immune status is imperative. Peripheral blood immune responses to cancer cells constitute a highly sensitive defense mechanism, outperforming conventional tumor markers in the early diagnosis of gastric cancer[20]. Furthermore, cancer cells within the tumor microenvironment activate immune cells upon entering the bloodstream. This establishes a correlation between immune cells in the tumor microenvironment and the patient’s systemic inflammatory response[21,22]. Lymphocyte subsets, serving as monitors of patient immunity, have emerged as critical biomarkers for evaluating patient prognosis[23,24].

In this study, we performed a retrospective analysis involving 291 patients with gastric cancer who underwent surgical interventions. Comprehensive datasets comprising laboratory indexes for inflammatory biomarkers and lymphocyte subsets were obtained from these patients. By employing the least absolute shrinkage and selection operator-COX (LASSO-COX) regression analysis, we amalgamated the identified biomarkers to formulate a novel immune-inflammatory biomarker denoted as the prognostic immunoinflammatory index (PII). The primary objective of this study was to analyze the predictive capabilities of the PII specifically within the context of patients with gastric cancer who underwent surgical treatment.

MATERIALS AND METHODS
Patients

This study included patients with gastric cancer who underwent surgical treatment at Harbin Medical University Cancer Hospital between January 2016 and December 2019. The inclusion criteria were: (1) Histologically confirmed diagnosis of gastric cancer; (2) absence of extrahepatic metastasis; and (3) completion of lymphocyte subset testing. The exclusion criteria were: (1) Having received alternative treatments prior to surgery; (2) history of other malignant tumors; (3) insufficient clinical data or lack of follow-up information; and (4) presence of certain diseases or conditions affecting hematological parameters. The enrolled patients were randomly allocated based on a 7:3 ratio, into a training set (n = 204) and a validation set (n = 87). Due to the retrospective nature of this study, the requirement for informed consent was waived by the Ethics Committee of the Affiliated Cancer Hospital of Harbin Medical University.

Data collection and definition of variables

Comprehensive data were systematically obtained from all participants encompassing clinical characteristics, tumor attributes, laboratory-related examinations, and lymphocyte subset analyses. This study specifically focused on preoperative immune-inflammatory biomarkers and lymphocyte subsets, including NLR, PLR, SII, and CD3(+), CD3(+)CD4(+), CD3(+)CD8(+), CD4(+)CD8(+), CD3(+)CD16(+)CD8(+), CD3(-)CD16(+)CD56(+), and CD3(+)CD16(+)CD56(+) lymphocyte subsets. The calculation of biomarkers was performed using the following formulas: NLR = neutrophils/lymphocytes; PLR = platelets/lymphocytes; SII = neutrophils/lymphocytes × platelets; and systemic inflammation response index (SIRI) = (neutrophils × monocytes)/lymphocytes.

The primary endpoints for assessment were overall survival (OS), defined as the period from the date of surgery to that of death or last follow-up, and progression-free survival (PFS), defined as the interval between the date of surgery and that of disease recurrence or last follow-up.

PII score construction

Univariate COX regression analysis was conducted for all immune-inflammatory biomarkers and lymphocyte subsets. Variables exhibiting significance based on a threshold of P < 0.05 were retained for subsequent analyses. The prognostic significance of these retained biomarkers was evaluated using the LASSO-COX regression analysis. Ultimately, PII scores were computed based on variables that did not possess zero coefficients in the LASSO-COX regression analysis.

Statistical analysis

For categorical variables, analyses were conducted using the Pearson's chi-squared test or Fisher's test. For continuous variables, the Student’s t-test was used. Kaplan-Meier curves were employed to illustrate and compare the differences in OS and PFS utilizing the log-rank test. Nomograms representing OS and PFS were generated based on the results derived from COX multivariate regression analysis, employing the inverse stepwise selection method to minimize Akaike information criterion values. To validate the performance and clinical benefit of the nomograms in the training cohort, time-dependent area under the curve (AUC) and decision curve analysis (DCA) were utilized.

RESULTS
Baseline characteristics and PII score

A total of 291 patients participated in this study (male: 69.8%, female: 30.2%; mean age: 59.05 years). Radical surgery was performed in 274 individuals (94.1% of the study population). Demographic and clinical characteristics of the training (n = 204) and validation (n = 87) sets are shown in Table 1.

Table 1 Clinicopathological characteristics, n (%).
Variable
All patients
Training set
Validation set
Age (years), mean (± SD)59.05 (10.45)58.45 (10.38)60.48 (10.54)
Sex,
    Male203 (69.8)144 (70.6)59 (67.8)
    Female88 (30.2)60 (29.4)28 (32.2)
Body mass index (kg/m2), mean (SD)22.86 (3.22)22.95 (3.04)22.65 (3.63)
Radical resection
    Yes274 (94.2)195 (95.6)79 (90.8)
    No17 (5.8)9 (4.4)8 (9.2)
Primary tumor site
    Upper 1/311 (3.7)8 (3.9)3 (3.4)
    Middle 1/338 (13.1)28 (13.7)10 (11.5)
    Low 1/3208 (71.5)142 (69.7)66 (75.9)
    Whole34 (11.7)26 (12.7)8 (9.2)
Borrmann type
        Ⅰ32 (11.0)22 (10.8)10 (11.5)
        Ⅱ87 (29.9)63 (30.9)24 (27.6)
        Ⅲ153 (52.6)109 (53.4)44 (50.6)
        Ⅳ19 (6.5)10 (4.9)9 (10.3)
Lymph node positivity
        Yes138 (47.4)99 (48.5)39 (44.8)
        No153 (52.6)105 (51.5)48 (55.2)
Tumor size
        < 20 mm29 (10.0)18 (8.8)11 (12.6)
        20–50 mm124 (42.6)89 (43.6)35 (40.2)
        > 50 mm138 (47.4)97 (47.6)41 (47.1)
Differentiation
        Poor101 (34.7)72 (35.3)29 (33.3)
        Moderate149 (51.2)106 (52.0)43 (49.4)
        Well26 (8.9)19 (9.3)7 (8.0)
        Unknown15 (5.2)7 (3.4)8 (9.3)
Lauren type
        Intestinal143 (49.1)105 (51.5)38 (43.7)
        Diffuse51 (17.5)35 (17.2)16 (18.4)
        Mixed84 (28.9)58 (28.4)26 (29.9)
        Unknown13 (4.5)6 (2.9)7 (8.0)
TNM stage
        Ⅰ117 (40.2)81 (39.7)36 (41.4)
        Ⅱ71 (24.4)53 (26.0)18 (20.7)
        Ⅲ89 (30.6)62 (30.4)27 (31.0)
        Ⅳ14 (4.8)8 (3.9)6 (6.9)
Carcinoembryonic antigen
        < 1.97 ng/mL144 (49.5)107 (52.5)37 (42.5)
        ≥ 1.97 ng/mL147 (50.9)97 (47.5)50 (57.5)
CA199
        < 10.19 U/L145 (49.8)103 (50.5)42 (48.3)
        ≥ 10.19 U/L146 (50.2)101 (49.5)45 (51.7)
CA724
        < 2.17 U/L145 (49.8)106 (52.0)39 (44.8)
        ≥ 2.17 U/L146 (50.2)98 (48.0)48 (55.2)
CA125II
        < 10.21 U/L145 (49.8)102 (50.0)43 (49.4)
        ≥ 10.21 U/L146 (50.2)102 (50.0)44 (50.6)
NLR, mean (SD)2.63 (2.98)2.62 (3.03)2.66 (2.87)
PLR, mean (SD)148.10 (7.47)149.64 (78.22)144.50 (68.87)
SII, mean (SD)668.08 (768.43)683.94 (796.04)630.89 (702.44)
SIRI, mean (SD)70.69 (29.36)70.05 (24.26)72.18 (38.93)
CD3(+) (%), mean (SD)70.91 (38.40)69.00 (9.31)75.41 (68.84)
CD3(+)CD4(+) (%), mean (SD)40.82 (8.67)40.79 (8.22)40.88 (9.69)
CD3(+)CD8(+) (%), mean (SD)23.42 (8.07)23.49 (8.02)23.24 (8.25)
CD4(+)/CD8(+), mean (SD)2.04 (1.08)1.99 (0.92)2.17 (1.37)
CD3(+)CD4(+)CD8(+) (%), mean (SD)0.54 (1.23)0.61 (1.44)0.36 (0.37)
CD19(+) (%), mean (SD)11.32 (4.71)11.68 (4.76)10.48 (4.51)
CD3(-)CD16(+)CD56(+) (%), mean (SD)16.72 (9.55)16.25 (8.34)17.83 (11.02)
CD3(+)CD16(+)CD56(+) (%), mean (SD)3.13 (4.34)3.26 (4.89)2.83 (2.63)

Survival analysis employing the Kaplan-Meier method demonstrated that, among the inflammatory biomarkers, NLR, PLR, SII, and SIRI were identified as prognostic risk factors. Elevated NLR, PLR, SII, and SIRI were associated with diminished OS. In the analysis of lymphocyte subsets, lower CD3(+), CD3(+)CD8(+), CD4(+)CD8(+), and CD3(+)CD16(+)CD56(+) lymphocyte subsets were linked to improved OS. CD19(+) lymphocytes conferred a protective effect, indicating that higher CD19(+) lymphocyte levels were associated with reduced OS (Figure 1). Kaplan-Meier analyses of the relationship between inflammatory biomarkers, lymphocyte subsets, and PFS consistently supported these findings (Figure 2). The Kaplan-Meier analysis identified all eight immune and inflammatory biomarkers as prognostic factors for OS. To isolate independent prognostic factors, all eight biomarkers were subjected to the LASSO-COX regression analysis, revealing six biomarkers with non-zero coefficients. The subsequent equation was established based on their coefficients: PII score = 0.608 × NLR + 0.277 × PLR + 0.059 × SIRI + 0.480 × CD3(+) + 0.176 × CD3(+)CD16(+)CD56(+) - 0.459 × CD4(+)CD8(+) - 0.501 × CD19(+) (Figure 3A and B). The intercorrelations between the eight immune-inflammatory biomarkers are depicted in Figure 3C. In the training set, a high PII score was associated with inferior PFS and OS (Figure 3D and E).

Figure 1
Figure 1 Kaplan–Meier curves for progression-free survival. A: Progression-free survival (PFS) by CD3(+) lymphocytes; B: PFS by CD3(+)CD8(+) lymphocytes; C: PFS by CD4(+)CD8(+) lymphocytes; D: PFS by CD3(+)CD16(+)CD56(+) lymphocytes; E: PFS by CD19(+) lymphocytes; F: PFS by platelet-to-lymphocyte ratio; G: PFS by neutrophil-to-lymphocyte ratio; H: PFS by systemic immune inflammatory index; I: PFS by systemic inflammation response index. PLR: Platelet-to-lymphocyte ratio; NLR: Neutrophil-to-lymphocyte ratio; SII: Systemic immune inflammatory index; SIRI: Systemic inflammation response index; PFS: Progression-free survival.
Figure 2
Figure 2 Kaplan–Meier curves for overall survival. A: Overall survival by CD3(+) lymphocytes; B: OS by CD3(+)CD8(+) lymphocytes; C: OS by CD4(+)CD8(+) lymphocytes; D: OS by CD3(+)CD16(+)CD56(+) lymphocytes; E: OS by CD19(+) lymphocytes; F: OS by platelet-to-lymphocyte ratio; G: OS by neutrophil-to-lymphocyte ratio; H: OS by systemic immune inflammatory index; I: OS by systemic inflammation response index. PLR: Platelet-to-lymphocyte ratio; NLR: Neutrophil-to-lymphocyte ratio; SII: Systemic immune inflammatory index; SIRI: Systemic inflammation response index; PFS: Progression-free survival; OS: Overall survival.
Figure 3
Figure 3 Construction of PII score using the least absolute shrinkage and selection operator-COX regression model. A: Partial likelihood deviance for least absolute shrinkage and selection operator (LASSO) coefficient profiles; B: LASSO coefficient profiles of eight immune and inflammatory related biomarkers; C: Heatmap of correlations of the immune and inflammatory related biomarkers; D: Progression-free survival by prognostic immunoinflammatory index (PII); E: Overall survival by PII. PII: Prognostic immunoinflammatory index; PFS: Progression-free survival; OS: Overall survival.
Association between PII score and clinical characteristics

The PII score construction involved determining the optimal cutoff value through receiver operating characteristic curve analysis (i.e., 70.69). Subsequently, patients were stratified into high- and low-value groups based on this cutoff value. Exploration of potential differences in clinical characteristics among distinct PII score groups ensued. Utilizing the Pearson's chi-squared test or Fisher's test, we conducted a comprehensive analysis of the clinical characteristics among different PII score groups. The outcomes revealed statistically significant variations in body mass index, primary tumor site, tumor size, carbohydrate antigen 199 (CA199), NLR, PLR, SII, SIRI, and CD3(+), CD3(+)CD8(+), and CD19(+) lymphocyte subsets (P < 0.05 for all), as depicted in Table 2.

Table 2 Association between prognostic immunoinflammatory index score and clinical characteristics, n (%).
Variable
PII score
P value
Group 1, n = 130
Group 2, n = 74
Age (years), mean (SD)58.03 (10.52)59.18 (10.17)0.398
Sex0.301
        Male95 (73.1)49 (66.2)
        Female35 (26.9)25 (33.8)
Body mass index (kg/m2), mean (SD)23.38 (3.13)22.19 (2.73)0.004
Radical resection0.075
        Yes127 (97.7)68 (91.9)
        No3 (2.3)6 (8.1)
Primary tumor site0.016
        Upper 1/34 (3.1)4 (5.4)
        Middle 1/317 (13.1)11 (14.9)
        Low 1/399 (76.1)43 (58.1)
        Whole10 (7.7)16 (21.6)
Borrmann type0.275
        Ⅰ16 (12.3)6 (8.1)
        Ⅱ43 (33.1)20 (27.0)
        Ⅲ67 (51.5)42 (56.8)
        Ⅳ4 (3.1)6 (8.1)
Lymph node positivity
        Yes67 (47.7)37 (50.0)
        No68 (52.3)37 (50.0)
Tumor size0.031
        < 20 mm14 (10.8)4 (5.4)
        20–50 mm63 (48.5)26 (35.1)
        > 50 mm53 (40.7)44 (59.5)
Differentiation0.100
        Poor43 (33.2)29 (39.2)
        Moderate70 (53.8)36 (48.6)
        Well15 (11.5)4 (5.4)
        Unknown2 (1.5)5 (6.8)
Lauren type0.125
        Intestinal67 (51.5)38 (51.4)
        Diffuse23 (17.7)12 (16.2)
        Mixed39 (30.0)19 (25.6)
        Unknown1 (0.8)5 (6.8)
TNM stage0.155
        Ⅰ52 (40.0)29 (39.2)
        Ⅱ34 (26.2)19 (25.7)
        Ⅲ42 (32.3)20 (27.0)
        Ⅳ2 (1.5)6 (8.1)
Carcinoembryonic antigen0.729
        < 1.97 ng/mL67 (51.5)40 (54.1)
        ≥1.97 ng/mL63 (48.5)34 (45.9)
CA1990.032
        < 10.19 U/L73 (56.2)30 (40.5)
        ≥ 10.19 U/L57 (43.8)44 (59.5)
CA7240.056
        < 2.17 U/L61 (46.9)45 (60.8)
        ≥ 2.17 U/L69 (53.1)29 (39.2)
CA125II0.244
        < 10.21 U/L61 (46.9)41 (55.4)
        ≥ 10.21 U/L69 (53.1)33 (44.6)
NLR, mean (SD)1.69 (0.67)4.24 (4.54)< 0.001
PLR, mean (SD)107.02 (31.74)224.36 (79.72)< 0.001
SII, mean (SD)392.19 (180.21)1196.46 (1134.42)< 0.001
SIRI, mean (SD)0.88 (0.48)2.37 (3.27)< 0.001
CD3(+) (%), mean (SD)67.21 (9.87)72.14 (7.30)< 0.001
CD3(+)CD4(+) (%), mean (SD)40.46 (8.14)41.34 (8.38)0.445
CD3(+)CD8(+) (%), mean (SD)22.36 (7.60)25.47 (8.40)0.005
CD4(+)/CD8(+), mean (SD)2.07 (0.94)1.84 (0.87)0.069
CD3(+)CD4(+)CD8(+) (%), mean (SD)0.53 (0.73)0.76 (2.20)0.657
CD19(+) (%), mean (SD)12.58 (4.89)10.10 (4.09)< 0.001
CD3(-)CD16(+)CD56(+) (%), mean (SD)16.99 (9.71)14.94 (6.92)0.282
CD3(+)CD16(+)CD56(+) (%), mean (SD)3.17 (5.48)3.41 (3.67)0.202
COX regression analysis and nomogram construction

Variables exhibiting statistical significance (i.e., P < 0.05) in the univariate COX regression analysis were incorporated into the subsequent multivariate regression analysis. Given the inclusion of the PII score in the multivariate analysis, individual biomarkers constituting the PII score were intentionally omitted to avoid statistical redundancy. The outcomes of the multivariate COX regression analysis identified tumor-node-metastasis (TNM) stage, Borrmann type, and PII score as independent prognostic factors for PFS and OS, as shown in Table 3. Nomograms for PFS and OS were subsequently constructed, encompassing TNM stage, Borrmann type, and PII score (Figure 4). The predictive models exhibited substantial accuracy, with a concordance index of 0.798 and 0.792 for PFS and OS, respectively.

Figure 4
Figure 4 Nomograms for prediction of survival. A: Nomogram for progression-free survival; B: Nomogram for overall survival. TNM: Tumor-node-metastasis; PII: Prognostic immunoinflammatory index.
Table 3 COX regression analysis of overall survival and progression-free survival.
Parameter
OS
PFS
Univariate analysis
Multivariate analysis
Univariate analysis
Multivariate analysis
HR (95%CI)
P value
HR (95%CI)
P value
HR (95%CI)
P value
HR (95%CI)
P value
Age (years)2.035 (1.325-3.061)0.0011.541 (0.982-2.417)0.0511.978 (1.302-3.007)0.0011.488 (0.948-2.335)0.084
Sex
        Male1 (Ref.)1 (Ref.)
        Female0.912 (0.597-1.392)0.6690.918 (0.601-1.401)0.692
Body mass index (kg/m2)0.527 (0.342-0.811)0.0040.853 (0.536-1.357)0.5020.527 (0.342-0.812)0.0040.838 (0.526-1.334)0.456
CD3(+) (%)2.051 (1.387-3.033)< 0.0011.995 (1.350-2.949)0.001
CD3(+)CD4(+) (%)1.526 (0.961-2.424)0.0731.528 (0.963-2.426)0.072
CD3(+)CD8(+) (%)1.516 (1.034-2.221)0.0291.513 (1.003-2.217)0.030
CD4(+)/CD8(+)0523 (0.341-0.802)0.0020.517 (0.337-0.792)0.002
CD3(+)CD4(+)CD8(+) (%)1.516 (0.940-2.445)0.0881.504 (0.933-2.425)0.094
CD19(+) (%)0.351 (0.171-0.721)0.0030.358 (0.174-0.735)0.004
CD3(-)CD16(+)CD56(+) (%)0.703 (0.480-1.030)0.0700.712 (0.486-1.043)0.082
CD3(+)CD16(+)CD56(+) (%)1.880 (1.207-2.931)0.0051.865 (1.197-2.905)0.005
PLR
        < 2.811 (Ref.)1 (Ref.)
        ≥ 2.811.913 (1.241-2.949)0.0031.898 (1.231-2.926)0.003
NLR
        < 115.221 (Ref.)1 (Ref.)
        ≥ 155.222.390 (1.588)< 0.0012.338 (1.554-3.518)< 0.001
SII
        < 533.411 (Ref.)1 (Ref.)
        ≥ 533.411.514 (1.035-2.217)0.0311.515 (1.035-2.217)0.032
SIRI
        < 1.511 (Ref.)1 (Ref.)
        ≥ 1.511.899 (1.263-2.857)0.0021.881 (1.251-2.830)0.002
Radical resection
        Yes1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        No4.356 (2.431-7.807)< 0.0011.909 (0.789-4.615)0.1514.182 (2.335-7.492)< 0.0011.513 (0.595-3.846)0.384
Borrmann type
        Ⅰ1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        Ⅱ6.025 (1.435-25.300)0.0142.237 (0.477-10.497)0.3076.081 (1.448-25.533)0.0142.278 (0.484-10.729)0.298
        Ⅲ8.012 (1.958-32.780)0.0042.350 (0.515-10.715)0.2708.088 (1.977-33.091)0.0042.435 (0.533-11.124)0.251
        Ⅳ27.087 (6.171-118.891)< 0.0015.407 (1.063-27.496)0.04228.997 (6.605-127.294)< 0.0015.301 (1.043-26.930)0.044
Lymph node positivity
        No1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        Yes3.445 (2.264-5.242)< 0.0011.132 (0.545-2.351)0.7403.537 (2.324-5.383)< 0.0011.010 (0.485-2.103)0.979
Tumor size
        < 20 mm1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        20–50 mm2.710 (0.829-8.858)0.0990.708 (0.192-2.607)0.6032.715 (0.831-8.870)0.0980.686 (0.185-2.544)0.573
        > 50 mm6.917 (2.176-21.988)0.0010.666 (0.178-2.494)0.5466.883 (2.165-21.878)0.0010.622 (0.164-2.361)0.485
TNM stage
        Ⅰ1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        Ⅱ3.833 (1.585-7.910)< 0.0013.566 (1.545-8.233)0.0033.875 (1.878-7.996)< 0.0013.433 (1.475-7.989)0.004
        Ⅲ11.441 (5.999-21.819)< 0.0019.085 (3.382-24.404)< 0.00111.807 (6.187-22.533)< 0.0018.541 (3.152-23.143)< 0.001
        Ⅳ35.899 (15.895-81.079)< 0.00111.978 (3.812-37.641)< 0.00145.844 (20.022-104.969)< 0.00116.297 (4.934-53.825)< 0.001
Carcinoembryonic antigen
        < 1.97 ng/mL1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        ≥ 1.97 ng/mL1.586 (1.078-2.335)0.0191.425 (0.926-2.195)0.1071.543 (1.048-2.271)0.0281.328 (0.867-2.034)0.192
CA199
        < 10.19 U/L1 (Ref.)1 (Ref.)
        ≥ 10.19 U/L1.355 (0.923-1.989)0.1201.347 (0.918-1.977)0.128
CA724
        < 2.17 U/L1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        ≥ 2.17 U/L2.159 (1.451-3.213)< 0.0011.510 (0.992-2.297)0.0542.154 (1.448-3.205)< 0.0011.456 (0.957-2.214)0.079
CA125II
        < 10.21 U/L1 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        ≥ 10.21 U/L1.849 (1.250-2.734)0.0021.198 (0.791-1.813)0.3941.837 (1.242-2.716)0.0021.171 (0.770-1.781)0.460
PII
        < 70.691 (Ref.)1 (Ref.)1 (Ref.)1 (Ref.)
        ≥ 70.691.888 (1.289-2.766)0.0011.670 (1.115-2.500)0.0131.881 (1.284-2.755)0.0011.639 (1.096-2.450)0.016

The assessment of predictive performance involved the generation of time-dependent AUC and DCA. The 1-, 3-, and 5-year AUC values of the nomogram for PFS were 0.834, 0.841, and 0.863, respectively (Figure 5A). Similarly, the 1-, 3-, and 5-year AUC values of the nomogram for OS were 0.830, 0.821, and 0.850, respectively (Figure 5B). DCA demonstrated that the PII-nomograms of PFS and OS were slightly better than the TNM staging system in terms of predictive power (Figure 6). DCA further affirmed the clinical utility and patient benefit of the nomogram prediction model constructed using the PII, as illustrated in Figure 7.

Figure 5
Figure 5 Time-dependent receiver operating characteristic curve analysis of the nomogram fo prediction of survival. A: 1-, 3-, and 5-year area under the curves for progression-free survival; B: 1-, 3-, and 5-year area under the curves for overall survival. AUC: Area under the receiver operating characteristic curve.
Figure 6
Figure 6 Decision curve analysis of the nomogram for prediction of survival. A: Decision curve analysis for progression-free survival; B: Decision curve analysis for overall survival.
Figure 7
Figure 7 Decision curve analysis of the nomogram for prediction of 12-, 36-, and 60-mo survival. A: 12-, 36-, and 60-mo decision curve analysis for progression-free survival; B: 12-, 36-, and 60-mo decision curve analysis for overall survival.
DISCUSSION

Studies have revealed the intricate connection between immunity, inflammation, and cancer[25,26]. Inflammation assumes a pivotal role across tumorigenesis, growth stages, and developmental phases, exerting a lasting impact on subsequent treatment modalities and patient survival[27]. The carcinogenic potential of Helicobacter pylori, a recognized risk factor for gastric cancer, is closely associated with its induction of chronic inflammation, thereby promoting tumor development[28]. This symbiotic relationship between gastric cancer and inflammation underscores chronic inflammation as a predisposing factor for gastric cancer, with tumor cell heterogeneity reciprocally enhancing the expression of inflammatory cytokines and chemokines[29]. Notably, the role of inflammation extends to tumor metastasis, as evidenced by studies indicating a significant impact on cancer-related mortality, particularly in gastrointestinal cancers and distant metastases; prolonged aspirin use contributes to reduced overall mortality[30,31]. In light of these considerations, the identification of convenient and effective biomarkers for assessing the degree of inflammation in patients remains crucial.

Numerous clinical studies have investigated the prognostic implications of inflammatory biomarkers and lymphoid subgroups in patients with gastric cancer. For instance, Miyamoto et al[32] conducted a study involving 154 consecutive patients with gastric cancer, demonstrating that a high NLR correlated with poorer clinical outcomes. Zurlo et al[33] corroborated these findings in their assessment of NLR as a prognostic factor in patients with locally advanced gastric cancer. PLR has also emerged as an independent predictor of mortality and morbidity in gastric cancer[34]. Comparative studies on NLR and PLR as prognostic factors for gastric cancer, such as the retrospective study performed by Kim et al[35] involving 2000 patients who underwent radical surgery, concluded that NLR exhibited greater prognostic power than PLR. The SII, encompassing lymphocytes, platelet count, and neutrophils, provides a comprehensive estimate of the body's immune response and inflammatory status[36,37]. Zhang et al[38] and He et al[39] proposed SII as a good biomarker for diagnosing gastric cancer compared with NLR and PLR, highlighting its significant correlation with OS in this setting. It has been shown that SIRI predicts patient prognosis in a variety of cancer types[40,41]. In our hospital, the study of lymphocyte subpopulations further affirmed their role as predictive biomarkers for gastric cancer, surpassing the predictive capability of inflammatory biomarkers[42]. Additionally, lymphocyte subsets, in conjunction with the nutritional indicator prognostic nutritional index, emerged as prognostic biomarkers, reflecting the immune and nutritional status of patients[43].

While serum immune indicators are easily detectable in clinical settings, individual immune and inflammatory biomarkers exhibit limitations in prognostic risk analysis. Therefore, the integration of indicators is imperative to establish a more comprehensive biomarker for assessing the immune and inflammatory status of patients. In our study, the initial dichotomization of collected immune and inflammatory biomarkers, followed by univariate COX regression analysis, revealed potential prognostic factors associated with the prognosis of gastric cancer. These factors were subjected to the LASSO-COX regression analysis, culminating in the derivation of the PII score, with non-zero coefficients serving as the basis for the formula. The PII, encompassing a broad spectrum of patient immune-inflammatory levels, demonstrated a significant correlation with OS in the training set, with high PII scores indicting poorer OS. Notably, PII scores exhibited correlations with tumor characteristics, including primary tumor site and tumor size. The 1-, 3-, and 5-year AUC values for the nomograms of PFS and OS, constructed by incorporating TNM stage, Borrmann type, and PII, exhibited remarkable performance. DCA further confirmed the beneficial impact of the PII score on patients with gastric cancer.

The immune response protects the body from the development of tumors; however, other immune responses (e.g., chronic inflammation) also play a role in this process[44]. The present study focused on inflammatory biomarkers and lymphoid subpopulations because altering the tumor microenvironment (including cytokines and chemokines) may alter the immune cells in patients receiving conventional treatment for gastric cancer[45]. Since inflammatory markers take into account absolute lymphocyte and neutrophil counts, the number of these immune cells may be affected by the cytokine environment. Detection of blood lymphocyte subsets also reflects the immune cell status. Therefore, the combination of inflammatory biomarkers and lymphocyte subsets utilized in this study to predict the prognosis of patients with gastric cancer is also of interest.

Despite the robust predictive accuracy of our model, certain limitations warrant consideration. The wide distribution of patients with gastric cancer, particularly across diverse ethnicities[46], highlights the need for multicenter studies in the future to validate the present findings. Additionally, the impact of Helicobacter pylori on serum immune-inflammatory factors was not investigated in this study. Furthermore, the retrospective nature of this study may have led to data omissions, potentially reducing the number of patients included in the analysis.

CONCLUSION

The PII may provide a comprehensive assessment of the immune-inflammatory status of the body, offering valuable insights for individualized prognostic evaluations in patients with gastric cancer. Elevated PII scores were associated with poorer clinical outcomes.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade A, Grade B

Novelty: Grade A, Grade B

Creativity or Innovation: Grade A, Grade B

Scientific Significance: Grade A, Grade B

P-Reviewer: Gao W, China; Verma V, United States S-Editor: Li L L-Editor: Wang TQ P-Editor: Chen YX

References
1.  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;68:394-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53206]  [Cited by in F6Publishing: 51871]  [Article Influence: 8645.2]  [Reference Citation Analysis (122)]
2.  Luo G, Zhang Y, Guo P, Wang L, Huang Y, Li K. Global patterns and trends in stomach cancer incidence: Age, period and birth cohort analysis. Int J Cancer. 2017;141:1333-1344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 128]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
3.  Arnold M, Karim-Kos HE, Coebergh JW, Byrnes G, Antilla A, Ferlay J, Renehan AG, Forman D, Soerjomataram I. Recent trends in incidence of five common cancers in 26 European countries since 1988: Analysis of the European Cancer Observatory. Eur J Cancer. 2015;51:1164-1187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 319]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
4.  Ferro A, Peleteiro B, Malvezzi M, Bosetti C, Bertuccio P, Levi F, Negri E, La Vecchia C, Lunet N. Worldwide trends in gastric cancer mortality (1980-2011), with predictions to 2015, and incidence by subtype. Eur J Cancer. 2014;50:1330-1344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 482]  [Article Influence: 48.2]  [Reference Citation Analysis (0)]
5.  López MJ, Carbajal J, Alfaro AL, Saravia LG, Zanabria D, Araujo JM, Quispe L, Zevallos A, Buleje JL, Cho CE, Sarmiento M, Pinto JA, Fajardo W. Characteristics of gastric cancer around the world. Crit Rev Oncol Hematol. 2023;181:103841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 32]  [Reference Citation Analysis (0)]
6.  Thrift AP, El-Serag HB. Burden of Gastric Cancer. Clin Gastroenterol Hepatol. 2020;18:534-542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 735]  [Article Influence: 183.8]  [Reference Citation Analysis (1)]
7.  Alberts SR, Cervantes A, van de Velde CJ. Gastric cancer: epidemiology, pathology and treatment. Ann Oncol. 2003;14 Suppl 2:ii31-ii36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 270]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
8.  Taieb J, Moehler M, Boku N, Ajani JA, Yañez Ruiz E, Ryu MH, Guenther S, Chand V, Bang YJ. Evolution of checkpoint inhibitors for the treatment of metastatic gastric cancers: Current status and future perspectives. Cancer Treat Rev. 2018;66:104-113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
9.  Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet. 2020;396:635-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1150]  [Cited by in F6Publishing: 1956]  [Article Influence: 489.0]  [Reference Citation Analysis (0)]
10.  Digklia A, Wagner AD. Advanced gastric cancer: Current treatment landscape and future perspectives. World J Gastroenterol. 2016;22:2403-2414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 359]  [Cited by in F6Publishing: 366]  [Article Influence: 45.8]  [Reference Citation Analysis (0)]
11.  Zhao Q, Cao L, Guan L, Bie L, Wang S, Xie B, Chen X, Shen X, Cao F. Immunotherapy for gastric cancer: dilemmas and prospect. Brief Funct Genomics. 2019;18:107-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 86]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
12.  Van Cutsem E, Sagaert X, Topal B, Haustermans K, Prenen H. Gastric cancer. Lancet. 2016;388:2654-2664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1282]  [Cited by in F6Publishing: 1386]  [Article Influence: 173.3]  [Reference Citation Analysis (0)]
13.  Chiarello MM, Fico V, Pepe G, Tropeano G, Adams NJ, Altieri G, Brisinda G. Early gastric cancer: A challenge in Western countries. World J Gastroenterol. 2022;28:693-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
14.  Suzuki K. Chronic Inflammation as an Immunological Abnormality and Effectiveness of Exercise. Biomolecules. 2019;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 146]  [Article Influence: 29.2]  [Reference Citation Analysis (0)]
15.  Andrejeva G, Rathmell JC. Similarities and Distinctions of Cancer and Immune Metabolism in Inflammation and Tumors. Cell Metab. 2017;26:49-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 222]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]
16.  Diem S, Schmid S, Krapf M, Flatz L, Born D, Jochum W, Templeton AJ, Früh M. Neutrophil-to-Lymphocyte ratio (NLR) and Platelet-to-Lymphocyte ratio (PLR) as prognostic markers in patients with non-small cell lung cancer (NSCLC) treated with nivolumab. Lung Cancer. 2017;111:176-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 426]  [Article Influence: 60.9]  [Reference Citation Analysis (0)]
17.  Cupp MA, Cariolou M, Tzoulaki I, Aune D, Evangelou E, Berlanga-Taylor AJ. Neutrophil to lymphocyte ratio and cancer prognosis: an umbrella review of systematic reviews and meta-analyses of observational studies. BMC Med. 2020;18:360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 229]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
18.  Li B, Zhou P, Liu Y, Wei H, Yang X, Chen T, Xiao J. Platelet-to-lymphocyte ratio in advanced Cancer: Review and meta-analysis. Clin Chim Acta. 2018;483:48-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 73]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
19.  Hu B, Yang XR, Xu Y, Sun YF, Sun C, Guo W, Zhang X, Wang WM, Qiu SJ, Zhou J, Fan J. Systemic immune-inflammation index predicts prognosis of patients after curative resection for hepatocellular carcinoma. Clin Cancer Res. 2014;20:6212-6222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 519]  [Cited by in F6Publishing: 945]  [Article Influence: 94.5]  [Reference Citation Analysis (0)]
20.  Fang T, Wang Y, Yin X, Zhai Z, Zhang Y, Yang Y, You Q, Li Z, Ma Y, Li C, Song H, Shi H, Zhang Y, Yu X, Gao H, Sun Y, Xie R, Xue Y. Diagnostic Sensitivity of NLR and PLR in Early Diagnosis of Gastric Cancer. J Immunol Res. 2020;2020:9146042.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 76]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
21.  Choi Y, Kim JW, Nam KH, Han SH, Kim JW, Ahn SH, Park DJ, Lee KW, Lee HS, Kim HH. Systemic inflammation is associated with the density of immune cells in the tumor microenvironment of gastric cancer. Gastric Cancer. 2017;20:602-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 66]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
22.  Suzuki H, Kaneko MK, Kato Y. Roles of Podoplanin in Malignant Progression of Tumor. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 19]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
23.  Wu Y, Ye S, Goswami S, Pei X, Xiang L, Zhang X, Yang H. Clinical significance of peripheral blood and tumor tissue lymphocyte subsets in cervical cancer patients. BMC Cancer. 2020;20:173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
24.  Li P, Qin P, Fu X, Zhang G, Yan X, Zhang M, Zhang X, Yang J, Wang H, Ma Z. Associations between peripheral blood lymphocyte subsets and clinical outcomes in patients with lung cancer treated with immune checkpoint inhibitor. Ann Palliat Med. 2021;10:3039-3049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
25.  Zitvogel L, Pietrocola F, Kroemer G. Nutrition, inflammation and cancer. Nat Immunol. 2017;18:843-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 258]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
26.  Shaul ME, Fridlender ZG. Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol. 2019;16:601-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 353]  [Article Influence: 70.6]  [Reference Citation Analysis (0)]
27.  Greten FR, Grivennikov SI. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity. 2019;51:27-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 922]  [Cited by in F6Publishing: 1847]  [Article Influence: 369.4]  [Reference Citation Analysis (0)]
28.  Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014;345:196-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 495]  [Article Influence: 49.5]  [Reference Citation Analysis (1)]
29.  Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2019]  [Cited by in F6Publishing: 3078]  [Article Influence: 513.0]  [Reference Citation Analysis (0)]
30.  Rothwell PM, Wilson M, Price JF, Belch JF, Meade TW, Mehta Z. Effect of daily aspirin on risk of cancer metastasis: a study of incident cancers during randomised controlled trials. Lancet. 2012;379:1591-1601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 698]  [Cited by in F6Publishing: 714]  [Article Influence: 59.5]  [Reference Citation Analysis (0)]
31.  Panigrahy D, Gartung A, Yang J, Yang H, Gilligan MM, Sulciner ML, Bhasin SS, Bielenberg DR, Chang J, Schmidt BA, Piwowarski J, Fishbein A, Soler-Ferran D, Sparks MA, Staffa SJ, Sukhatme V, Hammock BD, Kieran MW, Huang S, Bhasin M, Serhan CN, Sukhatme VP. Preoperative stimulation of resolution and inflammation blockade eradicates micrometastases. J Clin Invest. 2019;129:2964-2979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 80]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
32.  Miyamoto R, Inagawa S, Sano N, Tadano S, Adachi S, Yamamoto M. The neutrophil-to-lymphocyte ratio (NLR) predicts short-term and long-term outcomes in gastric cancer patients. Eur J Surg Oncol. 2018;44:607-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 140]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
33.  Zurlo IV, Schino M, Strippoli A, Calegari MA, Cocomazzi A, Cassano A, Pozzo C, Di Salvatore M, Ricci R, Barone C, Bria E, Tortora G, Larocca LM, Basso M, Martini M. Predictive value of NLR, TILs (CD4+/CD8+) and PD-L1 expression for prognosis and response to preoperative chemotherapy in gastric cancer. Cancer Immunol Immunother. 2022;71:45-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
34.  Mungan İ, Dicle ÇB, Bektaş Ş, Sarı S, Yamanyar S, Çavuş M, Turan S, Bostancı EB. Does the preoperative platelet-to-lymphocyte ratio and neutrophil-to-lymphocyte ratio predict morbidity after gastrectomy for gastric cancer? Mil Med Res. 2020;7:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
35.  Kim EY, Lee JW, Yoo HM, Park CH, Song KY. The Platelet-to-Lymphocyte Ratio Versus Neutrophil-to-Lymphocyte Ratio: Which is Better as a Prognostic Factor in Gastric Cancer? Ann Surg Oncol. 2015;22:4363-4370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 113]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
36.  Nøst TH, Alcala K, Urbarova I, Byrne KS, Guida F, Sandanger TM, Johansson M. Systemic inflammation markers and cancer incidence in the UK Biobank. Eur J Epidemiol. 2021;36:841-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 163]  [Article Influence: 54.3]  [Reference Citation Analysis (0)]
37.  Tsilimigras DI, Moris D, Mehta R, Paredes AZ, Sahara K, Guglielmi A, Aldrighetti L, Weiss M, Bauer TW, Alexandrescu S, Poultsides GA, Maithel SK, Marques HP, Martel G, Pulitano C, Shen F, Soubrane O, Koerkamp BG, Endo I, Pawlik TM. The systemic immune-inflammation index predicts prognosis in intrahepatic cholangiocarcinoma: an international multi-institutional analysis. HPB (Oxford). 2020;22:1667-1674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
38.  Zhang J, Zhang L, Duan S, Li Z, Li G, Yu H. Single and combined use of the platelet-lymphocyte ratio, neutrophil-lymphocyte ratio, and systemic immune-inflammation index in gastric cancer diagnosis. Front Oncol. 2023;13:1143154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
39.  He M, Chen ZF, Zhang L, Gao X, Chong X, Li HS, Shen L, Ji J, Zhang X, Dong B, Li ZY, Lei T. Associations of subcutaneous fat area and Systemic Immune-inflammation Index with survival in patients with advanced gastric cancer receiving dual PD-1 and HER2 blockade. J Immunother Cancer. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
40.  Qi Q, Zhuang L, Shen Y, Geng Y, Yu S, Chen H, Liu L, Meng Z, Wang P, Chen Z. A novel systemic inflammation response index (SIRI) for predicting the survival of patients with pancreatic cancer after chemotherapy. Cancer. 2016;122:2158-2167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 248]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
41.  Cai H, Li J, Chen Y, Zhang Q, Liu Y, Jia H. Preoperative inflammation and nutrition-based comprehensive biomarker for predicting prognosis in resectable colorectal cancer. Front Oncol. 2023;13:1279487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
42.  Fang T, Yin X, Wang Y, Zhang L, Wang Y, Zhang X, Zhao X, Lin X, Xue Y. Lymphocyte subset is more suitable than systemic inflammatory response biomarker and immunoglobulin in constructing prognostic nomogram model for advanced gastric cancer. Heliyon. 2023;9:e14669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
43.  Sun H, Wang H, Pan H, Zuo Y, Zhao R, Huang R, Xue Y, Song H. CD19 (+) B Cell Combined with Prognostic Nutritional Index Predicts the Clinical Outcomes of Patients with Gastric Cancer Who Underwent Surgery. Cancers (Basel). 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
44.  Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6912]  [Cited by in F6Publishing: 7821]  [Article Influence: 488.8]  [Reference Citation Analysis (0)]
45.  Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell. 2015;28:690-714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 963]  [Cited by in F6Publishing: 1085]  [Article Influence: 120.6]  [Reference Citation Analysis (0)]
46.  Li P, Huang CM, Zheng CH, Russo A, Kasbekar P, Brennan MF, Coit DG, Strong VE. Comparison of gastric cancer survival after R0 resection in the US and China. J Surg Oncol. 2018;118:975-982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]