Open Access

Systemic immune inflammation index and gastric cancer prognosis: A systematic review and meta‑analysis

  • Authors:
    • Xiaomao Yang
    • Chen Wu
  • View Affiliations

  • Published online on: February 1, 2024     https://doi.org/10.3892/etm.2024.12410
  • Article Number: 122
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to pool the available data on the associations between the systemic immune inflammation index (SII) and overall survival (OS) or recurrence‑free survival (RFS) in patients with gastric cancer (GC). A systematic search was conducted in the PubMed, EMBASE and Scopus databases for observational studies, and a random effects model was used to conduct the statistical analysis. Pooled effect sizes were reported as hazard ratios (HRs) with corresponding 95% confidence intervals (CI). Data from 30 studies (24 conducted in China) with follow‑ups ranging between 15.5 and 65.6 months were analyzed. Patients with GC and high SII levels had poor OS (HR, 1.53; 95% CI, 1.34‑1.75) and recurrence free survival (HR, 1.41; 95% CI, 1.17‑1.70). These increased risks were present irrespective of the treatment strategy (surgical or non‑surgical management), the sample size (<500 and ≥500) and the cut‑off used to define high and low SII (<600 and ≥600 x109 cells/l). The results of this meta‑analysis suggest that high pretreatment SII levels were associated with poor OS and RFS in patients with GC.

Introduction

Gastric cancer (GC), a significant global public health burden, is among the top-ranked cancers for causing significant levels of mortality and disability (1). Globally, as per the estimated data for the year 2019, GC is the fifth most diagnosed cancer, fourth leading cause of cancer-associated mortalities and contributes to 1.7 million disability-adjusted life years (2-4). GC can be difficult to detect in its initial stages due to mild or absent symptoms, and is usually diagnosed at the advanced disease stage (5). GC requires a multidisciplinary approach to treatment, involving gastroenterologists, surgeons, medical oncologists and radiation oncologists (6,7). Advances in surgical techniques, chemotherapy, targeted therapy and immunotherapy have improved GC treatment outcomes, but early detection and timely treatment remain critical targets to improve the prognosis of the disease (8,9).

Despite advances in the diagnosis and treatment of GC, patients with advanced disease stages face a poor prognosis with a 5-year overall survival (OS) rate of <5% (10,11). This highlights the need for improved prognostic indices to guide clinical decision-making and improve patient outcomes. Systemic inflammatory responses contribute to the tumor microenvironment, promoting angiogenesis, tumor development and metastasis (12,13). Neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, lymphocyte-monocyte ratio and systemic immune-inflammation index (SII) have shown promise as prognostic markers in patients with specific types of cancer, such as metastatic non-small cell lung cancer, testicular germ cell tumor and rectal cancer (14-16). Moreover, the levels of these markers can be measured from routine blood tests, making them easily accessible and relatively inexpensive.

The SII has demonstrated its prognostic value in various types of tumor, such as urological cancers, including prostate cancer, small cell lung cancer and esophageal cancer (17-21), and is used to assess and quantify the systemic inflammatory response. It is a composite index that takes into account blood-based markers, such as neutrophil count, lymphocyte count and platelet count [SII=(platelet count x neutrophil count)/lymphocyte count]. It can be easily and inexpensively measured using blood samples and, therefore, has the potential to be adopted in everyday clinical practice for personalized treatment planning. It may also be used in combination with other clinical and pathological variables, such as tumour size, differentiation, clinical stage, vascular or lymphatic invasion, distant metastasis or abnormal carcinoembryonic antigen (CEA), to improve prognostic accuracy and guide treatment decisions for patients with GC. To the best of our knowledge, only two meta-analyses have focused on SII: One including eight studies and the other including 11 studies (22,23). The meta-analysis by Qiu et al showed that a high pretreatment SII is associated with poorer OS, but not poor disease-free survival (DFS) in patients with GC (22). By contrast, the analysis by Fu et al showed that higher SII levels are associated with poorer OS and DFS (23). The present study was designed to update the analysis with the data from new publications and evaluate the association of SII with OS or RFS in patients with GC.

Materials and methods

Search for eligible studies

Electronic databases (PubMed (https://pubmed.ncbi.nlm.nih.gov/), Embase (https://www.elsevier.com/products/embase) and Scopus (https://www.scopus.com/home.uri) were searched for relevant studies from the inception of database up to the 15th of April, 2023. The search strategy comprised the following terms: (Systemic immune-inflammation index OR SII OR immunonutritional biomarker OR platelet count OR neutrophil count OR lymphocyte count) AND (stomach tumor OR gastric tumor OR gastric neoplasm OR gastric malignancy OR gastric carcinoma OR gastric adenocarcinoma) AND (clinical outcome OR mortality OR survival OR death OR disease-free survival). The present study also manually reviewed the reference lists of pertinent articles and systematic reviews to identify additional studies that satisfied the inclusion criteria.

Screening and selection of studies

Subsequently, two study authors (XY and CW) independently screened all identified studies for inclusion based on pre-established eligibility criteria. The inclusion criteria were: i) Studies examining the association between pre-treatment SII and OS, DFS or recurrence-free survival (RFS) in patients with GC; ii) studies on adult patients with histologically-confirmed GC; iii) studies providing sufficient data on the association between pre-treatment SII and survival outcomes, including odds ratios/relative risks/hazard ratios (HRs) and 95% confidence intervals (CI); iv) studies published in English. The exclusion criteria were: i) Studies published as conference abstracts, case reports or letters to the editor; ii) studies conducted on animal models or cell lines; iii) studies that did not consider pre-treatment SII levels as an exposure of interest; iv) studies that lacked sufficient data or methodological quality (Newcastle Ottawa scale score <5) (24).

The present study specifically focused on observational studies exploring the association between pretreatment SII and survival outcomes in patients with GC. The inclusion criteria were restricted to studies published during the preceding decade, between 2013 and 2023, to ensure that the findings are based on up-to-date literature reflecting contemporary evidence.

Full texts of potentially relevant studies were screened to determine their final eligibility. As this research involved analyzing previously published studies through a systematic review and meta-analysis, the need for ethical approval was waived. However, clear and thorough reporting of the methods and findings were ensured in the present study by following the PRISMA guidelines (25). The present study was prospectively registered at PROSPERO, with number CRD42023424804.

Data extraction, quality assessment and analysis

Next, two independent reviewers performed data extraction. The risk of bias of the observational studies was evaluated by calculating the Newcastle-Ottawa Scale values for each study (24). In cases of discrepancies in data extraction and bias assessments, a consensus was reached after discussion. For each outcome of interest, the present study performed a random-effects meta-analysis to calculate the pooled effect sizes along with their corresponding 95% CIs. I2 statistic was used to assess statistical heterogeneity. Egger's test was used for detecting publication bias (26). Subgroup analysis was also conducted based on the primary treatment modality, sample size, cut-off used for SII and location of conduct of study. P<0.05 was considered to indicate a statistically significant difference.

Results

The present study included data from 30 studies in this analysis (27-56). Fig. 1 presents the process of the study selection. The majority of studies (n=24) were conducted in China (Table I); two studies were conducted in Japan and four in Turkey. Except for one study that had a prospective cohort design, all studies had a retrospective cohort design. In 24 studies, the main GC management strategy was gastrectomy, whereas in the remaining six studies, non-surgical management strategies included immune checkpoint inhibitors, combination of chemotherapy and radiotherapy, anti-programmed death 1 treatment and a combined immune- and chemo-therapy (Table I). The study sample sizes ranged from 45 to 2,257 participants, with 19 studies having <500 participants and 11 studies having ≥500 participants. The follow-up periods varied from 15.5 to 65.6 months. Quality scores on the NOS ranged from 6 to 9, with a mean score of 7.53, indicating overall acceptable study quality (Table I).

Table I

Characteristics of the studies included in the present meta-analysis.

Table I

Characteristics of the studies included in the present meta-analysis.

Author (year)Study designCountryPatients characteristicsSample sizeSII cut-off (x109 cells/l)Newcastle Ottawa quality score(Refs.)
Gu (2020)Prospective cohortChinaWith advanced GC; underwent radical resection (distal gastrectomy in majority); mean age, 63 years; men, 68%; T3-T4, 57% and >N0, 59%; follow-up, at least 5 years598≥556.08(27)
Demircan (2023)Retrospective cohortTurkeyPatients undergoing neoadjuvant chemotherapy; median age, 60 years; men, 68%; poorly differentiated, 36%; median follow-up, 22.5 months140≥741.08(28)
Inoue (2021)Retrospective cohortJapanPatients with gastric adenocarcinoma undergoing curative gastrectomy; median age, 67 years; men, 65%; median BMI, 22 kg/m2; differentiated tumour, 52%; follow-up, at least 5 years447≥395.08(29)
Wei (2022)Retrospective cohortChinaWith advanced GC; underwent gastrectomy; median age, 59 years and men, 65%; T4a/T4b, 75%; N3a/N3b, 70%; poorly differentiated, 89%; median follow-up, 15.5 months218≥1185.28(30)
Zhaojun (2022)Retrospective cohortChinaWith advanced GC; stage2/3, 82%; underwent gastrectomy; aged ≤60 years, 56%; men, 78%; poorly differentiated, 65%; median follow-up, 46 months354≥489.57(31)
Wang (2022)Retrospective cohortChinaPatients undergoing radical D2 gastrectomy followed by adjuvant chemotherapy; median age, 59 years; men, 77%; pT4, 70%; pN3a/pN3b, 56%; median follow-up, 29.1 months89>369.28(32)
Qu (2022)Retrospective cohortChinaPatients with advanced GC undergoing anti-PD-1 therapy; men, 68%; age >65 years, 27%; poor differentiation, 60%; lymph node metastasis, 72%; median follow-up, 17.5 months106>1140.99(33)
Wan (2022)Retrospective cohortChinaPatients with advanced GC receiving immunotherapy combined with chemotherapy; age <60 years, 76%; men, 78%; stage IV, 80%; poor differentiation, 39%; median follow-up, 27.3 months45≥1154.77(34)
Yekeduz (2022)Retrospective cohortTurkeyPatients with advanced GC undergoing gastric resection surgery; median age, 53 years; men, 64%; T3/T4, 79%; N2/N3, 48%; median follow-up, 25.5 months120>708.07(35)
Liu (2023)Retrospective cohortChinaPatients with GC undergoing gastric resection surgery, 52% or chemotherapy/ radiotherapy, 40%; mean age, 59 years; men, 72%; T3/T4, 65%1,133>712.67(36)
Uzunoglu (2023)Retrospective cohortTurkeyPatients undergoing gastrectomy; mean age, 64 years; male, 65%; mean follow-up time, 33.4 months152>892.06(37)
Wang (2023)Retrospective cohortChinaPatients undergoing radical gastrectomy; median age, ~58 years; male, 75%; stage 2/3, 90%; poor differentiation, 40%542>489.97(38)
He (2022)Retrospective cohortChinaPatients undergoing radical gastrectomy; median age, ~61 years; male, 79%; stage 1, 65%; negative lympho-vascular invasion, 87%; mean follow-up, 65 months548>508.38(39)
Chen (2021)Retrospective cohortChinaPatients undergoing treatment with immune checkpoint inhibitor; median age, 60 years; male, 75%; stage 3/4, 100%; poor differentiation, 75%; median follow-up, 23.8 months139≥665.39(40)
Shi (2022)Retrospective cohortChinaPatients undergoing radical surgery; age ≤60 years, 68%; male, 67%; stage 2/3, 76%; poor differentiation, 34%; median follow-up, 57 months496>315.08(41)
Wang (2021)Retrospective cohortChinaPatients undergoing radical surgery; median age, 61 years; male, 76%; stage2/3, 80%; median follow-up 56 months608≥372.88(42)
Huang (2016)Prospective cohortChinaPatients undergoing radical surgery; age >50 years, 71%; male, 67%; stage 3, 50%; median follow-up, 655 days455≥572.07(43)
Chen (2017)Retrospective cohortChinaPatients undergoing neoadjuvant chemotherapy or radical surgery; median age, 57 years; male, 71%; stage 2/3, 100%292≥600.08(44)
Wang (2017)Retrospective cohortChinaPatients undergoing radical surgery; age <60 years, 56%; male, 63%; TNM stage 3/4, 68%444≥660.07(45)
Guo (2018)Retrospective cohortChinaPatients undergoing radical surgery; age >65 years, 31%; male, 67%; TNM stage 3, 57%; poor differentiation, 62%; median follow-up, 35 months1,058≥521.68(46)
Shi (2018)Retrospective cohortChinaPatients undergoing radical surgery; age >60 years, 33%; male, 69%; TNM stage 2/3, 73%; poor differentiation, 47%; median follow-up, 36 months688>320.07(47)
Wang (2019)Retrospective cohortChinaPatients undergoing radical surgery; age >60 years, 37%; male, 73%; TNM stage 3, 100%; poor differentiation, 61%182≥600.08(48)
Yilmaz (2020)Retrospective cohortTurkeyPatients undergoing radical surgery; median age, 59 years; male, 63%; TNM stage 3, 43%; median follow-up of 30 months85≥802.06(49)
Hirahara (2021)Retrospective cohortJapanPatients undergoing radical surgery; median age, 73 years; male, 73%; TNM stage 2/3, 58%; follow-up >60 months212≥661.97(50)
Lin (2021)Retrospective cohortChinaPatients undergoing radical surgery; mean age, 70 years; male, 75%; TNM stage 2/3, 70%; median follow-up, 65.6 months2,257≥569.98(51)
Zhou (2016)Retrospective cohortChinaPatients undergoing radical surgery; median age, 63 years; male, 81%; median follow-up, 48 months192>543.97(52)
Liu (2015)Retrospective cohortChinaPatients undergoing radical surgery; age >60 years, 45%; male, 69%; TNM stage 3, 65%; median follow-up, 25 months455≥660.08(53)
Fang (2022)Retrospective cohortChinaPatients undergoing gastrectomy; age >60 years, 50%; male, 75%; TNM stage 2/3, 65%; poor differentiation, 45%; minimum follow-up, 24 months755Change in SII (Δ SII)7(54)
Yin (2020)Retrospective cohortChinaPatients undergoing gastrectomy along with chemotherapy; age >60 years, 30%; male, 74%; TNM stage 2/3, 89%; poor differentiation, 65%; follow-up, 60 months576Change in SII (Δ SII)8(55)
Zhu (2020)Retrospective cohortChinaPatients undergoing gastrectomy; age <55 years, 48%; male, 65%; TNM stage 3, 57%512≥527.07(56)

[i] GC, gastric cancer; TNM, tumor node metastasis; BMI, body mass index.

OS

Patients with high SII levels had poor OS (HR, 1.53; 95% CI, 1.34-1.75; n=27; I2=72.4%), compared with patients with low SII levels (Fig. 2). Egger's test (P=0.01) and funnel plots (Fig. S1) indicated the presence of publication bias. Patients with high SII levels had poorer OS, irrespective of whether they had received surgical or non-surgical management, whether they were part of sample sizes <500 or >500, or the cut-offs used to define high and low SII (<600 and ≥600 x109 cells/l) (Table II). Notably, the elevated risk of poorer OS associated with a high SII level was statistically significant in studies conducted in China (HR, 1.53; 95% CI, 1.34-1.75; n=21; I2=73.7%), but not in studies conducted in other settings (Table II).

Table II

Association between SII and overall survival as well as recurrence free survival, within various subgroups.

Table II

Association between SII and overall survival as well as recurrence free survival, within various subgroups.

 Overall survivalRecurrence-free survival
SubgroupsPooled HR (95% CI)nHeterogeneity I2 (%)Pooled HR (95% CI)nHeterogeneity I2 (%)
Primary treatment      
     Surgery (gastrectomy)1.54 (1.31, 1.80)2177.71.35 (1.06, 1.85)663.0
     Non-surgical1.54 (1.33, 1.79)60.01.51 (1.23, 1.86)50.0
Sample size of the included studies      
     <5001.67 (1.42, 1.97)981.81.47 (1.17, 1.85)1041.9
     ≥5001.36 (1.12, 1.65)1852.71.23 (1.08, 1.40)1-
Cut-off for SII used in the included studies      
     <6001.54 (1.26, 1.88)1381.81.68 (1.02, 2.78)365.4
     ≥6001.54 (1.30, 1.82)1448.21.36 (1.07, 1.73)841.1
Location of study      
     China1.53 (1.34, 1.75)2173.71.43 (1.23, 1.66)719.9
     Other than China1.70 (0.92, 3.13)672.71.18 (0.65, 2.13)469

[i] SII, systemic immune inflammation index; HR, hazard ratio.

RFS

Patients with high SII levels had poorer RFS (HR, 1.41; 95% CI, 1.17-1.70; n=11; I2=45.2%) compared with those with low SII levels (Fig. 3). Egger's test (P=0.591) and funnel plot inspection suggested a lack of publication bias (Fig. S2). Subgroup analyses showed that regardless of sample size, type of treatment received or the cut-off used to define high and low levels of SII, individuals with high SII levels had poorer RFS. However, the association between high SII levels and elevated risk of poor RFS was statistically significant in studies conducted in China (HR, 1.43; 95% CI, 1.23-1.66; n=7; I2=19.9%), but not in studies conducted in other locations (Table II).

Discussion

The present meta-analysis revealed that high SII levels were associated with poor OS and RFS in patients with GC irrespective of the sample size, treatment received or cut-offs used to define high and low SII levels.

A meta-analysis by Qiu et al demonstrated a significant correlation between high SII levels and unfavorable OS outcomes (22). However, this study revealed no significant associations with the RFS (22). Another review by Fu et al that included 11 studies with ~7,000 patients with GC revealed that a higher SII is associated with an ~53% increase in the risk of death and a 57% increase in the risk of disease recurrence or progression (23). Studies have also shown that a high SII is associated with unfavorable survival outcomes in patients with solid tumors, hepatocellular cancer, urological cancers, small cell lung cancer and esophageal squamous cell cancer (19,57-59). The findings from these meta-analyses indicate that SII may serve as a reliable marker of prognosis in various cancer types and could provide valuable information for clinical decision-making and patient management.

Systemic inflammatory responses are involved in cancer progression (12,13,60,61). SII is a commonly used systemic inflammation marker. A high SII score has been associated with poor prognosis in different types of cancers, such as hepatocellular, prostate, renal cell and non-small cell lung cancers (19,58). One potential explanation for the observed association may be the involvement of lymphocytes, specifically tumor-infiltrating lymphocytes, which inhibit the increases in the number of cancer cells (62,63). Thus, low lymphocyte counts, contributing to high SII scores, may indicate a weakened immune response favoring cancer cell survival and growth (64,65). Another hypothesis involves the role of neutrophils, which are capable of secreting various growth factors and interleukins that stimulate tumor cell growth. These neutrophils could enhance tumor progression (66,67) by promoting tumor angiogenesis and invasion, and releasing proteases that degrade the extracellular matrix and facilitate cancer cell migration (67). Thus, high neutrophil counts, which contribute to high SII scores, may be indicative of inflammatory environments supporting tumor growth and metastases (68,69). Finally, elevated platelet counts have been shown to increase SII scores and may be indicative of tumor microenvironments that support the survival and spread of cancer cells (70,71). Taken together, increased SII scores may reflect the presence of prevailing pro-tumor microenvironments, which could contribute to poor prognosis for patients with GC.

The findings of the present study may be used for improving clinical practice, since they support the use of SII as a potentially valuable prognostic tool that can lead to more personalized treatment strategies. By modifying prognostic criteria to incorporate SII, clinicians could improve the prediction of outcomes and tailor treatment plans, ultimately leading to improved patient care and timely monitoring. Incorporation of SII in the panel of prognostic indicators for GC may foster multidisciplinary collaboration among healthcare professionals, particularly oncologists, hematologists and immunologists. The present study also provided incentive to conduct further research into the underlying mechanisms connecting high SII levels to adverse outcomes.

The present meta-analysis has some limitations. First, all included studies were conducted in Asian countries, mostly in China, and this may complicate the generalizability of the findings. The significant association of SII with poor OS and RFS in the Chinese studies and the lack thereof in studies conducted outside of China may be attributed to the considerably larger number of studies from China, which could have increased the statistical power of the analysis. Conversely, the limited number of studies from non-Chinese countries may have underpowered the analysis. As a result, statistical significance may have remained undetected, even if it genuinely existed. Second, the selected studies used diverse thresholds to categorize patients into high and low SII groups, leading to discrepancies in the interpretation of SII levels and subsequent outcomes. Third, most of the included studies were retrospective in nature, which may have introduced various selection and misclassification biases. Fourth, the heterogeneity among the included studies was significant, and the specific reasons for this heterogeneity remain unclear. Finally, the present study found evidence of publication bias in the analysis for the overall survival outcome, which may have influenced the results.

In conclusion, high pretreatment SII levels were associated with poor OS and RFS in patients with GC. SII levels, therefore, may serve as a potential prognostic marker. However, the present study has limitations, such as the lack of diversity in patient ethnicity, the variability in cut-off values and the reliability on retrospective studies. Thus, larger studies with a prospective design are needed to confirm the findings.

Supplementary Material

Forest plot for overall survival.
Forest plot for recurrence free survival.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XY conceived and designed the study. XY and CW collected the data and performed the literature search. XY was involved in the writing of the manuscript. XY and CW confirm the authenticity of all the raw data All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patients consent to participate

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

World Health Organization: Cancer Fact Sheets: Stomach Cancer. WHO, Geneva, 2020.

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

3 

Song Y, Liu X, Cheng W, Li H and Zhang D: The global, regional and national burden of stomach cancer and its attributable risk factors from 1990 to 2019. Sci Rep. 12(11542)2022.PubMed/NCBI View Article : Google Scholar

4 

GBD 2017 Stomach Cancer Collaborators. The global, regional, and national burden of stomach cancer in 195 countries, 1990-2017: A systematic analysis for the global burden of disease study 2017. Lancet Gastroenterol Hepatol. 5:42–54. 2020.PubMed/NCBI View Article : Google Scholar

5 

Maconi G, Manes G and Porro GB: Role of symptoms in diagnosis and outcome of gastric cancer. World J Gastroenterol. 14:1149–1155. 2008.PubMed/NCBI View Article : Google Scholar

6 

Sitarz R, Skierucha M, Mielko J, Offerhaus GJA, Maciejewski R and Polkowski WP: Gastric cancer: Epidemiology, prevention, classification, and treatment. Cancer Manag Res. 10:239–248. 2018.PubMed/NCBI View Article : Google Scholar

7 

Boniface MM, Wani SB, Schefter TE, Koo PJ, Meguid C, Leong S, Kaplan JB, Wingrove LJ and McCarter MD: Multidisciplinary management for esophageal and gastric cancer. Cancer Manag Res. 8:39–44. 2016.PubMed/NCBI View Article : Google Scholar

8 

Guan WL, He Y and Xu RH: Gastric cancer treatment: Recent progress and future perspectives. J Hematol Oncol. 16(57)2023.PubMed/NCBI View Article : Google Scholar

9 

Song Z, Wu Y, Yang J, Yang D and Fang X: Progress in the treatment of advanced gastric cancer. Tumour Biol. 39(1010428317714626)2017.PubMed/NCBI View Article : Google Scholar

10 

Shah MA and Ajani JA: Gastric cancer-an enigmatic and heterogeneous disease. JAMA. 303:1753–1754. 2010.PubMed/NCBI View Article : Google Scholar

11 

Ajani JA, Bentrem DJ, Besh S, D'Amico TA, Das P, Denlinger C, Fakih MG, Fuchs CS, Gerdes H, Glasgow RE, et al: Gastric cancer, version 2.2013: Featured updates to the NCCN guidelines. J Natl Compr Cancer Netw. 11:531–546. 2013.PubMed/NCBI View Article : Google Scholar

12 

Liu J, Lin PC and Zhou BP: Inflammation fuels tumor progress and metastasis. Curr Pharm Des. 21:3032–3040. 2015.PubMed/NCBI View Article : Google Scholar

13 

Greten FR and Grivennikov SI: Inflammation and cancer: Triggers, mechanisms, and consequences. Immunity. 51:27–41. 2019.PubMed/NCBI View Article : Google Scholar

14 

Liu J, Li S, Zhang S, Liu Y, Ma L, Zhu J, Xin Y, Wang Y, Yang C and Cheng Y: Systemic immune-inflammation index, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio can predict clinical outcomes in patients with metastatic non-small-cell lung cancer treated with nivolumab. J Clin Lab Anal. 33(e22964)2019.PubMed/NCBI View Article : Google Scholar

15 

Wang S, Yang X, Yu Z, Du P, Cao Y, Ji Y, Ma J and Yang Y: The values of systemic immune-inflammation index and neutrophil-lymphocyte ratio in predicting testicular germ cell tumors: A retrospective clinical study. Front Oncol. 12(893877)2022.PubMed/NCBI View Article : Google Scholar

16 

Xia LJ, Li W, Zhai JC, Yan CW, Chen JB and Yang H: Significance of neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, lymphocyte-to-monocyte ratio and prognostic nutritional index for predicting clinical outcomes in T1-2 rectal cancer. BMC Cancer. 20(208)2020.PubMed/NCBI View Article : Google Scholar

17 

Zhu S, Cheng Z, Hu Y, Chen Z, Zhang J, Ke C, Yang Q, Lin F, Chen Y and Wang J: Prognostic value of the systemic immune-inflammation index and prognostic nutritional index in patients with medulloblastoma undergoing surgical resection. Front Nutr. 8(754958)2021.PubMed/NCBI View Article : Google Scholar

18 

Huang Y, Gao Y, Wu Y and Lin H: Prognostic value of systemic immune-inflammation index in patients with urologic cancers: A meta-analysis. Cancer Cell Int. 20(499)2020.PubMed/NCBI View Article : Google Scholar

19 

Yang R, Chang Q, Meng X, Gao N and Wang W: Prognostic value of systemic immune-inflammation index in cancer: A meta-analysis. J Cancer. 9:3295–3302. 2018.PubMed/NCBI View Article : Google Scholar

20 

Li X, Zhang S, Lu J, Li C and Li N: The prognostic value of systemic immune-inflammation index in surgical esophageal cancer patients: An updated meta-analysis. Front Surg. 9(922595)2022.PubMed/NCBI View Article : Google Scholar

21 

Hong X, Cui B, Wang M, Yang Z, Wang L and Xu Q: Systemic immune-inflammation index, based on platelet counts and neutrophil-lymphocyte ratio, is useful for predicting prognosis in small cell lung cancer. Tohoku J Exp Med. 236:297–304. 2015.PubMed/NCBI View Article : Google Scholar

22 

Qiu Y, Zhang Z and Chen Y: Prognostic value of pretreatment systemic immune-inflammation index in gastric cancer: A meta-analysis. Front Oncol. 11(537140)2021.PubMed/NCBI View Article : Google Scholar

23 

Fu S, Yan J, Tan Y and Liu D: Prognostic value of systemic immune-inflammatory index in survival outcome in gastric cancer: A meta-analysis. J Gastrointest Oncol. 12:344–354. 2021.PubMed/NCBI View Article : Google Scholar

24 

Wells G, Shea B, O'Connell D, Robertson J, Peterson J, Losos M and Tugwell P: The newcastle-ottawa scale (NOS) for assessing the quality of nonrandomized studies in meta-analysis. https://www.ohri.ca/programs/clinical_epidemiology/oxford.asp. Accessed on April 29, 2023.

25 

Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 372(n71)2021.PubMed/NCBI View Article : Google Scholar

26 

Egger M, Smith GD, Schneider M and Minder C: Bias in meta-analysis detected by a simple, graphical test. BMJ. 315:629–634. 1997.PubMed/NCBI View Article : Google Scholar

27 

Gu L, Wang M, Cui X, Mo J, Yuan L, Mao F, Zhang K, Ng DM, Chen P and Wang D: Clinical significance of peripheral blood-derived inflammation markers in advanced gastric cancer after radical resection. BMC Surg. 20(219)2020.PubMed/NCBI View Article : Google Scholar

28 

Demircan NC, Atcı MM, Demir M, Işık S and Akagündüz B: Dynamic changes in systemic immune-inflammation index predict pathological tumor response and overall survival in patients with gastric or gastroesophageal junction cancer receiving neoadjuvant chemotherapy. Asia Pac J Clin Oncol. 19:104–112. 2023.PubMed/NCBI View Article : Google Scholar

29 

Inoue H, Kosuga T, Kubota T, Konishi H, Shiozaki A, Okamoto K, Fujiwara H and Otsuji E: Significance of a preoperative systemic immune-inflammation index as a predictor of postoperative survival outcomes in gastric cancer. World J Surg Oncol. 19(173)2021.PubMed/NCBI View Article : Google Scholar

30 

Wei C, Li C, Chen X, Chen G, Nie R, Zhao C, Zhou Z and Chen Y: Development and verification of a nomogram for predicting the prognosis of resectable gastric cancer with outlet obstruction. BMC Cancer. 22(1154)2022.PubMed/NCBI View Article : Google Scholar

31 

Zhaojun X, Xiaobin C, Juan A, Jiaqi Y, Shuyun J, Tao L, Baojia C, Cheng W and Xiaoming M: Correlation analysis between preoperative systemic immune inflammation index and prognosis of patients after radical gastric cancer surgery: Based on propensity score matching method. World J Surg Oncol. 20(1)2022.PubMed/NCBI View Article : Google Scholar

32 

Wang SB, Chen JY, Xu C, Cao WG, Cai R, Cao L and Cai G: Evaluation of systemic inflammatory and nutritional indexes in locally advanced gastric cancer treated with adjuvant chemoradiotherapy after D2 dissection. Front Oncol. 12(1040495)2022.PubMed/NCBI View Article : Google Scholar

33 

Qu Z, Wang Q, Wang H, Jiao Y, Li M, Wei W, Lei Y, Zhao Z, Zhang T, Zhang Y and Gu K: The effect of inflammatory markers on the survival of advanced gastric cancer patients who underwent anti-programmed death 1 therapy. Front Oncol. 12(783197)2022.PubMed/NCBI View Article : Google Scholar

34 

Wan M, Ding Y, Mao C, Ma X, Li N, Xiao C, Qian J, Jiang H, Zheng Y, Wu L, et al: Association of inflammatory markers with survival in patients with advanced gastric cancer treated with immune checkpoint inhibitors combined with chemotherapy as first line treatment. Front Oncol. 12(1029960)2022.PubMed/NCBI View Article : Google Scholar

35 

Yekedüz E, Dogan İ, Kaya D, MÖzgür İ, Utkan G, Vatansever S, Demirci S, Akbulut H and Ürün Y: Systemic immune-inflammation index as a prognostic marker of late recurrence in operable gastric cancer: A dual-center study. J Gastrointest Cancer. 53:870–879. 2022.PubMed/NCBI View Article : Google Scholar

36 

Liu YY, Ruan GT, Ge YZ, Li QQ, Zhang Q, Zhang X, Tang M, Song MM, Zhang XW, Li XR, et al: Systemic inflammation with sarcopenia predicts survival in patients with gastric cancer. J Cancer Res Clin Oncol. 149:1249–1259. 2023.PubMed/NCBI View Article : Google Scholar

37 

Uzunoglu H and Kaya S: Does systemic immune inflammation index have predictive value in gastric cancer prognosis? North Clin Istanb. 10:24–32. 2023.PubMed/NCBI View Article : Google Scholar

38 

Wang H, Yin X, Ma K, Wang Y, Fang T, Zhang Y and Xue Y: Nomogram based on preoperative fibrinogen and systemic immune-inflammation index predicting recurrence and prognosis of patients with borrmann type III advanced gastric cancer. J Inflamm Res. 16:1059–1075. 2023.PubMed/NCBI View Article : Google Scholar

39 

He K, Si L, Pan X, Sun L, Wang Y, Lu J and Wang X: Preoperative systemic immune-inflammation index (SII) as a superior predictor of long-term survival outcome in patients with stage I-II gastric cancer after radical surgery. Front Oncol. 12(829689)2022.PubMed/NCBI View Article : Google Scholar

40 

Chen Y, Zhang C, Peng Z, Qi C, Gong J, Zhang X, Li J and Shen L: Association of lymphocyte-to-monocyte ratio with survival in advanced gastric cancer patients treated with immune checkpoint inhibitor. Front Oncol. 11(589022)2021.PubMed/NCBI View Article : Google Scholar

41 

Shi H, Wang H, Pan J, Liu Z and Li Z: Comparing prognostic value of preoperative platelet indexes in patients with resectable gastric cancer. Sci Rep. 12(6480)2022.PubMed/NCBI View Article : Google Scholar

42 

Wang PX, Wang HJ, Liu JH, Qiu GL, Lu J, Fan L, Liao XH and Che XM: A nomogram combining plasma fibrinogen and systemic immune-inflammation index predicts survival in patients with resectable gastric cancer. Sci Rep. 11(10301)2021.PubMed/NCBI View Article : Google Scholar

43 

Huang L, Liu S, Lei Y, Wang K, Xu M, Chen Y, Liu B, Chen Y, Fu Q, Zhang P, et al: Systemic immune-inflammation index, thymidine phosphorylase and survival of localized gastric cancer patients after curative resection. Oncotarget. 7:44185–44193. 2016.PubMed/NCBI View Article : Google Scholar

44 

Chen L, Yan Y, Zhu L, Cong X, Li S, Song S, Song H and Xue Y: Systemic immune-inflammation index as a useful prognostic indicator predicts survival in patients with advanced gastric cancer treated with neoadjuvant chemotherapy. Cancer Manag Res. 9:849–867. 2017.PubMed/NCBI View Article : Google Scholar

45 

Wang K, Diao F, Ye Z, Zhang X, Zhai E, Ren H, Li T, Wu H, He Y, Cai S and Chen J: Prognostic value of systemic immune-inflammation index in patients with gastric cancer. Chin J Cancer. 36(75)2017.PubMed/NCBI View Article : Google Scholar

46 

Guo J, Chen S, Chen Y, Li S and Xu D: Combination of CRP and NLR: A better predictor of postoperative survival in patients with gastric cancer. Cancer Manag Res. 10:315–321. 2018.PubMed/NCBI View Article : Google Scholar

47 

Shi H, Jiang Y, Cao H, Zhu H, Chen B and Ji W: Nomogram based on systemic immune-inflammation index to predict overall survival in gastric cancer patients. Dis Markers. 2018(1787424)2018.PubMed/NCBI View Article : Google Scholar

48 

Wang Q and Zhu D: The prognostic value of systemic immune-inflammation index (SII) in patients after radical operation for carcinoma of stomach in gastric cancer. J Gastrointest Oncol. 10:965–978. 2019.PubMed/NCBI View Article : Google Scholar

49 

Yılmaz A, Mirili C, Tekin SB and Bilici M: The ratio of hemoglobin to red cell distribution width predicts survival in patients with gastric cancer treated by neoadjuvant FLOT: A retrospective study. Ir J Med Sci. 189:91–102. 2020.PubMed/NCBI View Article : Google Scholar

50 

Hirahara N, Tajima Y, Matsubara T, Fujii Y, Kaji S, Kawabata Y, Hyakudomi R, Yamamoto T, Uchida Y and Taniura T: Systemic immune-inflammation index predicts overall survival in patients with gastric cancer: A propensity score-matched analysis. J Gastrointest Surg. 25:1124–1133. 2021.PubMed/NCBI View Article : Google Scholar

51 

Lin JX, Wang ZK, Huang YQ, Xie JW, Wang JB, Lu J, Chen QY, Lin M, Tu RH, Huang ZN, et al: Dynamic changes in pre- and postoperative levels of inflammatory markers and their effects on the prognosis of patients with gastric cancer. J Gastrointest Surg. 25:387–396. 2021.PubMed/NCBI View Article : Google Scholar

52 

Zhou W, Wu J, Wang Q, Zhang H, Jiang J and Wu C: Evaluate the correlation between preoperative systemic immune-inflammation index and prognosis of patients after curative resection for cardia gastric cancer. Chin Med Biotechnol. 11:510–514. 2016.(In Chinese).

53 

Liu X, Sun X, Liu J, Kong P, Chen S, Zhan Y and Xu D: Preoperative C-reactive protein/albumin ratio predicts prognosis of patients after curative resection for gastric cancer. Transl Oncol. 8:339–345. 2015.PubMed/NCBI View Article : Google Scholar

54 

Fang T, Yin X, Wang Y, Zhang L, Zhang X, Zhao X, Wang Y and Xue Y: Proposed models for prediction of mortality in stage-I and stage-II gastric cancer and 5 years after radical gastrectomy. J Oncol. 2022(4510000)2022.PubMed/NCBI View Article : Google Scholar

55 

Yin X, Fang T, Wang Y, Li C, Wang Y, Zhang D and Xue Y: Efficacy of postoperative FOLFOX versus XELOX chemotherapy for gastric cancer and prognostic value of platelet-lymphocyte ratio in patients receiving XELOX. Front Oncol. 10(584772)2020.PubMed/NCBI View Article : Google Scholar

56 

Zhu Z, Cong X, Li R, Yin X, Li C and Xue Y: Preoperative systemic immune-inflammation index (SII) for predicting the survival of patients with stage I-III gastric cancer with a signet-ring cell (SRC) component. BioMed Res Int. 2020(e5038217)2020.PubMed/NCBI View Article : Google Scholar

57 

Zhong JH, Huang DH and Chen ZY: Prognostic role of systemic immune-inflammation index in solid tumors: A systematic review and meta-analysis. Oncotarget. 8:75381–75388. 2017.PubMed/NCBI View Article : Google Scholar

58 

Wang Y, Li Y, Chen P, Xu W, Wu Y and Che G: Prognostic value of the pretreatment systemic immune-inflammation index (SII) in patients with non-small cell lung cancer: A meta-analysis. Ann Transl Med. 7(433)2019.PubMed/NCBI View Article : Google Scholar

59 

Wang B, Huang Y and Lin T: Prognostic impact of elevated pre-treatment systemic immune-inflammation index (SII) in hepatocellular carcinoma: A meta-analysis. Medicine (Baltimore). 99(e18571)2020.PubMed/NCBI View Article : Google Scholar

60 

Grivennikov SI, Greten FR and Karin M: Immunity, inflammation, and cancer. Cell. 140:883–899. 2010.PubMed/NCBI View Article : Google Scholar

61 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011.PubMed/NCBI View Article : Google Scholar

62 

Santoiemma PP and Powell DJ Jr: Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther. 16:807–820. 2015.PubMed/NCBI View Article : Google Scholar

63 

Whiteside TL: Tumor-infiltrating lymphocytes and their role in solid tumor progression. Exp Suppl. 113:89–106. 2022.PubMed/NCBI View Article : Google Scholar

64 

Zhao J, Huang W, Wu Y, Luo Y, Wu B, Cheng J, Chen J, Liu D and Li C: Prognostic role of pretreatment blood lymphocyte count in patients with solid tumors: A systematic review and meta-analysis. Cancer Cell Int. 20(15)2020.PubMed/NCBI View Article : Google Scholar

65 

Ménétrier-Caux C, Ray-Coquard I, Blay JY and Caux C: Lymphopenia in cancer patients and its effects on response to immunotherapy: An opportunity for combination with Cytokines? J Immunother Cancer. 7(85)2019.PubMed/NCBI View Article : Google Scholar

66 

Koizumi K, Hojo S, Akashi T, Yasumoto K and Saiki I: Chemokine receptors in cancer metastasis and cancer cell-derived chemokines in host immune response. Cancer Sci. 98:1652–1658. 2007.PubMed/NCBI View Article : Google Scholar

67 

Masucci MT, Minopoli M and Carriero MV: Tumor associated neutrophils. Their role in tumorigenesis, metastasis, prognosis and therapy. Front Oncol. 9(1146)2019.PubMed/NCBI View Article : Google Scholar

68 

Xiong S, Dong L and Cheng L: Neutrophils in cancer carcinogenesis and metastasis. J Hematol Oncol. 14(173)2021.PubMed/NCBI View Article : Google Scholar

69 

Uribe-Querol E and Rosales C: Neutrophils in cancer: Two sides of the same coin. J Immunol Res. 2015(983698)2015.PubMed/NCBI View Article : Google Scholar

70 

Palacios-Acedo AL, Mège D, Crescence L, Dignat-George F, Dubois C and Panicot-Dubois L: Platelets, thrombo-inflammation, and cancer: Collaborating with the enemy. Front Immunol. 10(1805)2019.PubMed/NCBI View Article : Google Scholar

71 

Gay LJ and Felding-Habermann B: Contribution of platelets to tumour metastasis. Nat Rev Cancer. 11:123–134. 2011.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2024
Volume 27 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang X and Yang X: Systemic immune inflammation index and gastric cancer prognosis: A systematic review and meta‑analysis. Exp Ther Med 27: 122, 2024
APA
Yang, X., & Yang, X. (2024). Systemic immune inflammation index and gastric cancer prognosis: A systematic review and meta‑analysis. Experimental and Therapeutic Medicine, 27, 122. https://doi.org/10.3892/etm.2024.12410
MLA
Yang, X., Wu, C."Systemic immune inflammation index and gastric cancer prognosis: A systematic review and meta‑analysis". Experimental and Therapeutic Medicine 27.3 (2024): 122.
Chicago
Yang, X., Wu, C."Systemic immune inflammation index and gastric cancer prognosis: A systematic review and meta‑analysis". Experimental and Therapeutic Medicine 27, no. 3 (2024): 122. https://doi.org/10.3892/etm.2024.12410