Abstract
The neutrophil‐to‐lymphocyte ratio (NLR) has been extensively studied in patients with gastric cancer (GC) treated with immune checkpoint inhibitors (ICIs), although the results are controversial. Therefore, we performed the present meta‐analysis to systematically assess the correlation between NLR and prognosis and clinicopathological factors in GC patients undergoing treatment with ICIs. Among the electronic databases, PubMed, Web of Science, Embase, and Cochrane Library were thoroughly searched. To estimate the prognostic value of NLR for progression‐free survival (PFS) and overall survival (OS), hazard ratios (HRs) were calculated, and their 95% confidence intervals were calculated. This meta‐analysis included 10 studies with 830 patients. Based on the pooled data, a high NLR was associated with poor OS in GC patients receiving ICIs (HR = 2.13, 95% confidence interval [CI] = 1.70–2.66, p < 0.001). Elevated NLR was a significant biomarker for worse PFS in GC patients treated with ICIs (HR = 1.74, 95% CI = 1.22–2.48, p = 0.002). There was, however, no significant correlation between NLR and sex, age, and Eastern Cooperative Oncology Group Performance Status (PS) of the ECOG, differentiation, human epidermal growth factor receptor 2 (HER2) status, or PD‐L1 status in GC patients treated with ICIs. High NLR before treatment was associated with poor OS and PFS in GC patients treated with ICIs, according to our meta‐analysis. NLR is an effective biomarker for evaluating the prognosis of GC patients receiving ICIs and provide more valuable information for treatment decisions in the era of immunotherapy for GC.
Keywords: gastric cancer, immune checkpoint inhibitors, meta‐analysis, PD‐1/PD‐L1, survival
Abbreviations
- CI
confidence interval
- ECOG
Eastern Cooperative Oncology Group
- GC
gastric cancer
- GEJ
gastroesophageal junction
- HER2
human epidermal growth factor receptor 2
- HR
hazard ratio
- ICIs
immune checkpoint inhibitors
- LMR
lymphocyte‐to‐monocyte ratio
- NLR
neutrophil‐to‐lymphocyte ratio
- NOS
Newcastle–Ottawa scale
- OR
odds ratio
- OS
overall survival
- PD‐1
programmed cell death‐1
- PD‐L1
programmed cell death ligand‐1
- PFS
progression‐free survival
- PLR
platelet‐to‐lymphocyte ratio
- PRISMA
Preferred Reporting Items for Systematic Reviews and Meta‐Analyses
- PS
performance status
- ROC
receiver‐operating characteristic
- SII
systemic immune‐inflammation index
- TILs
tumor infiltrating lymphocytes
- TME
tumor microenvironment
1. INTRODUCTION
Globally, gastric cancer (GC) is the sixth most prevalent cancer and the third most common cause of cancer‐related deaths. 1 In 2020, an estimated 1,089,103 new cases of GC and 768,793 deaths due to GC occurred worldwide. 1 GC treatment includes surgery, chemotherapy, targeted therapy, and immunotherapy. 2 , 3 In recent years, immune checkpoint inhibitors (ICIs), such as monoclonal antibodies against programmed cell death‐1 (PD‐1) and programmed cell death ligand‐1 (PD‐L1), have improved survival outcomes in patients with advanced GC (AGC). 4 , 5 However, because of the lack of reliable and novel biomarkers to predict the efficacy and survival of GC patients receiving ICIs, there is still a poor prognosis for patients with AGC. The 5‐year survival of GC is ~20%–30% in most countries around the world, except in Japan and Korea. 6 Therefore, the identification of efficient prognostic markers for GC patients treated with ICIs is urgently needed.
Growing evidence has shown that immune responses play a pivotal role in cancer progression and development. 7 A series of blood‐derived parameters have been demonstrated to be effective prognostic markers for solid tumors. 8 These indices include the platelet‐to‐lymphocyte ratio, 9 systemic immune‐inflammation index, 10 lymphocyte‐to‐monocyte ratio, 11 and neutrophil‐to‐lymphocyte ratio (NLR). 12 NLR is calculated as the neutrophil count divided by lymphocyte count in peripheral blood. Previous studies have explored the prognostic effect of NLR in GC patients receiving ICIs; however, the results were controversial. 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 For example, some studies identified high NLR as a significant biomarker for GC patients receiving ICIs, 13 , 19 whereas other researchers denied this association. 14 , 21 , 22 Therefore, we conducted this meta‐analysis to evaluate the prognostic significance of NLR in GC patients treated with ICIs. Furthermore, we also investigated the correlation between NLR and clinicopathological characteristics in GC patients.
2. MATERIALS AND METHODS
2.1. Search strategy
Meta‐analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta‐Analyses (PRISMA) guidelines. 23 A thorough search was conducted in PubMed, Web of Science, Embase, and Cochrane Library. The search items were as follows: (“neutrophil‐to‐lymphocyte ratio” OR “NLR” OR “neutrophil to lymphocyte ratio”) AND (“gastric cancer” OR “gastric carcinoma” OR “gastric neoplasms”) AND (“immune checkpoint inhibitor” OR “CTLA‐4” OR “CTLA4” OR “ICIs” OR “immunotherapy” OR “programmed death ligand‐1 inhibitor” OR “PD‐L1 inhibitor” OR “programmed death‐1 inhibitor” OR “PD‐1 inhibitor” OR “atezolizumab” OR “avelumab” OR “cemiplimab” OR “durvalumab” OR “ipilimumab” OR “nivolumab” OR “pembrolizumab”). The search timeline was from inception to July 5, 2022. The publication search was limited to English language. A manual search of the references of retrieved studies was conducted to identify possible inclusions.
2.2. Inclusion and exclusion criteria
The inclusion criteria were as follows: (1) pathological evidence confirming the diagnosis of GC; (2) GC patients receiving ICI treatment, including but not limited to PD‐1/PD‐L1, CTLA‐4, or their combined inhibitors; (3) studies reporting hazard ratios (HR) and 95% confidence intervals (CI) for NLR in terms of survival; (4) a cut‐off value dividing high and low NLR; and (5) articles published in English. The exclusion criteria were as follows: (1) reviews, case reports, meeting abstracts, and letter; (2) studies that did not provide survival information; (3) animal studies; and (4) duplicate studies.
2.3. Data extraction and quality assessment
Two investigators (LL and LP) performed independent data extraction from eligible studies. Disagreements were resolved through discussions between the two investigators. In total, we extracted the following information: name of the first author, year, country, sample size, sex, study period, age, study design, treatment, NLR cut‐off value, method of determining NLR, follow‐up, survival outcomes, study center, cancer type, survival analysis type, and HRs and 95% CIs. The Newcastle–Ottawa scale (NOS) quality assessment tool was used for assessing the methodological quality of included studies. 24 The NOS consists of three aspects: selection, comparability, and outcomes. NOS has a maximum score of 9 and studies with an NOS score ≥6 is typically considered high‐quality.
2.4. Statistical analysis
To assess the prognostic value of NLR in GC patients treated with ICIs, we calculated the combined HRs and 95% CIs. Heterogeneity among studies was evaluated using the Cochrane Q test and I 2 test. All meta‐analyses used a random‐effects model because statistical heterogeneity could be easily underpowered, especially for subgroup analysis, where study numbers may be small in each group. Subgroup analysis stratified by various clinicopathological factors was used to detect the source of heterogeneity. The association between NLR and clinical features was evaluated by combining the odds ratios (ORs) and 95% CIs. To verify the stability of the results, a sensitivity analysis was conducted. Tests for publication bias were performed using Begg's and Egger's tests. All analyses were performed using Stata SE 12.0 (StataCorp, College Station, Texas). A value of p < 0.05 was considered statistically significant.
2.5. Ethics statement
The Institutional Review Board approval was not necessary because all the data in the study were retrieved from public databases.
3. RESULTS
3.1. Literature search
The initial literature search enrolled 120 records, and after the removal of duplicate studies, 55 records remained. A total of 39 records were discarded after examining the titles and abstracts, and 16 studies were subsequently evaluated by reading the full text. Six studies were excluded for the following reasons: no use of ICIs (n = 2), no survival data (n = 2), not regarding GC (n = 1), and no cut‐off value for NLR (n = 1). Finally, 10 studies with 830 patients 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 were included in this meta‐analysis (Figure 1).
FIGURE 1.

The PRISMA flowchart of the literature selection process.
3.2. Characteristics of included studies
As shown in Table 1, eligible studies had the following baseline characteristics. Five studies were conducted in Japan, 13 , 14 , 15 , 16 , 20 three studies were performed in China, 17 , 19 , 22 and two studies were performed in South Korea. 18 , 21 Eight studies were retrospective studies 13 , 14 , 15 , 16 , 17 , 18 , 20 , 22 and two were prospective trials. 19 , 21 All the studies were published in English. In this study, the sample size ranged from 24 to 185, with a median value of 74. All the studies used anti‐PD‐L1 antibodies (Table 1). The cut‐off values of NLR ranged from 2.5 to 5, with a median value of 2.95. The methods used to determine the cut‐off value of NLR were receiver operating characteristic (ROC) analysis (n = 6), 15 , 16 , 17 , 19 , 20 , 22 median value (n = 2), 14 , 21 and literature (n = 2). 13 , 18 The survival analysis types were multivariate (n = 6) 15 , 16 , 17 , 18 , 19 , 21 and univariate (n = 4). 13 , 14 , 20 , 22 Seven studies were conducted in a single center 14 , 15 , 16 , 17 , 18 , 20 , 22 and three studies were carried out in a multicenter. 13 , 19 , 21 The association between NLR and overall survival (OS) has been reported in 10 studies 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 in GC patients receiving ICIs and eight studies 13 , 14 , 15 , 16 , 17 , 19 , 21 , 22 reported progression‐free survival (PFS). NOS scores ranged from 7 to 9, indicating that all studies were of high quality (Table 1).
TABLE 1.
Basic characteristics of included studies in this meta‐analysis.
| Study | Year | Country | Sample size | Gender (M/F) | Study design | Age (years) median (range) | Treatment | Cut‐off value of NLR | Cut‐off determination | Follow‐up (month) median (range) | Survival endpoint | Survival analysis | Study center | Study period | Cancer type | NOS score |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Ogata, T. | 2018 | Japan | 24 | 19/7 | Retrospective | 64 (44–86) | Nivolumab | 5 | Literature | 5.7 | OS, PFS | Univariate | Multicenter | Jun–Dec, 2017 | AGC | 8 |
| Namikawa, T. | 2020 | Japan | 29 | 19/10 | Retrospective | 71 (49–86) | Nivolumab | 2.5 | Median value | 32 | OS, PFS | Univariate | Single center | 2017–2019 | AGC | 7 |
| Ota, Y. | 2020 | Japan | 98 | 68/30 | Retrospective | 66 (33–84) | Nivolumab | 3 | ROC curve | 4.9 (0.5–18.7) | OS, PFS | Multivariate | Single center | 2014–2018 | Advanced/recurrent GC | 8 |
| Yamada, T. | 2020 | Japan | 89 | 42/47 | Retrospective |
<65 years: 27 ≥65 years: 62 |
Nivolumab | 2.5 | ROC curve | 5.8 (0.6–35.6) | OS, PFS | Multivariate | Single center | 2014–2019 | AGC | 7 |
| Gou, M. | 2021 | China | 137 | 98/39 | Retrospective | 59 | Nivolumab/Pembrolizumab/Sintilimab/Toripalimab | 3.23 | ROC curve | 10.9 | OS, PFS | Multivariate | Single center | 2016–2020 | Metastatic GC | 8 |
| Kim, N. | 2021 | South Korea | 185 | 120/65 | Retrospective | 59 (47–70) | Nivolumab/Pembrolizumab | 3 | Literature | 7.3 | OS | Multivariate | Single center | 2016–2019 | AGC | 8 |
| Ruan, D. Y. | 2021 | China | 58 | 41/17 | Prospective | 60 (52–66) | Toripalimab | 2.7 | ROC curve | 4.5 | OS, PFS | Multivariate | Multicenter | 2016–2021 | AGC | 9 |
| Ishido, K. | 2022 | Japan | 59 | 45/14 | Retrospective | 71 (43–86) | Nivolumab | 2.5 | ROC curve | 5.9 (0.6–43.6) | OS | Univariate | Single center | 2017–2020 | AGC | 7 |
| Kim, J. H. | 2022 | South Korea | 45 | 34/11 | Prospective | 60 (23–76) | Nivolumab | 2.9 | Median value | 28.3 | OS, PFS | Multivariate | Multicenter | 2014–2016 | AGC | 9 |
| Qu, Z. | 2022 | China | 106 | 72/34 | Retrospective |
≤65 years: 77 >65 years: 29 |
Nivolumab/Pembrolizumab/Toripalimab/Sintilimab/Camrelizumab | 3.11 | ROC curve | 17.5 | OS, PFS | Univariate | Single center | 2019–2021 | AGC | 8 |
Abbreviations: AGC, advanced gastric cancer; GC, gastric cancer; NLR, neutrophil‐to‐lymphocyte ratio; NOS, Newcastle–Ottawa scale; OS, overall survival; PFS, progression‐free survival; ROC, receiver operating characteristic.
3.3. Association between NLR and OS
Ten studies with a total of 830 patients reported an association between NLR and OS in GC patients receiving ICIs. The pooled results were as follows: HR = 2.13, 95% CI = 1.70–2.66, p < 0.001, suggesting that a high NLR predicted poor OS in GC patients receiving ICIs (Figure 2; Table 2). Subgroup analysis demonstrated that an elevated NLR remained a significant prognostic factor for poor OS, irrespective of country, sample size, cut‐off value, survival analysis, cancer type, study center, or ICI treatment type (all p < 0.05; Table 2).
FIGURE 2.

Forest plots of NLR expression and overall survival risk in gastric cancer patients receiving immune checkpoint inhibitors.
TABLE 2.
Subgroup analysis of the prognostic value of NLR for OS in gastric cancer patients receiving ICIs.
| Subgroups | No. of studies | No. of patients | Effects model | HR (95% CI) | p | Heterogeneity | |
|---|---|---|---|---|---|---|---|
| I 2 (%) | p value | ||||||
| Total | 10 | 830 | Random | 2.13 (1.70–2.66) | <0.001 | 13.2 | 0.322 |
| Country | |||||||
| Japan | 5 | 299 | Random | 1.95 (1.47–2.60) | <0.001 | 0 | 0.485 |
| China | 3 | 301 | Random | 2.85 (1.44–5.64) | 0.003 | 52.4 | 0.122 |
| South Korea | 2 | 230 | Random | 1.85 (1.16–2.94) | 0.009 | 0 | 0.701 |
| Sample size | |||||||
| <75 | 5 | 215 | Random | 2.27 (1.31–3.95) | 0.004 | 44.0 | 0.126 |
| ≥75 | 5 | 615 | Random | 2.16 (1.69–2.76) | <0.001 | 0 | 0.533 |
| Study design | |||||||
| Retrospective | 8 | 727 | Random | 2.14 (1.73–2.65) | <0.001 | 0 | 0.497 |
| Prospective | 2 | 103 | Random | 3.66 (0.57–23.35) | 0.170 | 74.9 | 0.046 |
| Cut‐off value | |||||||
| <3 | 5 | 280 | Random | 1.90 (1.30–2.78) | 0.001 | 26.6 | 0.244 |
| ≥3 | 5 | 550 | Random | 2.36 (1.81–3.08) | <0.001 | 0 | 0.438 |
| Cut‐off determination | |||||||
| ROC curve | 6 | 547 | Random | 2.23 (1.70–2.94) | <0.001 | 22.6 | 0.264 |
| Literature | 2 | 209 | Random | 2.68 (1.11–6.46) | 0.029 | 38.4 | 0.203 |
| Median value | 2 | 74 | Random | 1.47 (0.83–2.61) | 0.189 | 0 | 0.539 |
| Survival analysis | |||||||
| Multivariate | 6 | 612 | Random | 2.20 (1.65–2.94) | <0.001 | 25.2 | 0.245 |
| Univariate | 4 | 218 | Random | 1.96 (1.29–2.9) | 0.002 | 12.1 | 0.332 |
| Cancer type | |||||||
| AGC | 8 | 595 | Random | 1.96 (1.49–2.57) | <0.001 | 9.0 | 0.360 |
| Metastatic/recurrent GC | 2 | 235 | Random | 2.50 (1.72–3.63) | <0.001 | 20.1 | 0.263 |
| Study center | |||||||
| Single center | 7 | 703 | Random | 2.09 (1.69–2.60) | <0.001 | 0 | 0.590 |
| Multicenter | 3 | 127 | Random | 3.81 (1.18–12.26) | 0.025 | 62.5 | 0.070 |
| Treatment ICIs type | |||||||
| Nivolumab | 6 | 344 | Random | 1.91 (1.47–2.49) | <0.001 | 0 | 0.605 |
| Nivolumab or other ICIs | 4 | 486 | Random | 2.51 (1.59–3.95) | <0.001 | 39.4 | 0.175 |
Abbreviations: AGC, advanced gastric cancer; ICIs, immune checkpoint inhibitors; NLR, neutrophil‐to‐lymphocyte ratio; OS, overall survival; ROC, receiver operating characteristic.
3.4. Association between NLR and PFS
A total of eight studies with 586 patients showed that NLR was a significant prognostic factor for PFS in GC patients receiving ICIs. The combined data showed that a high NLR was a significant biomarker for worse PFS in GC patients treated with ICIs: HR = 1.74, 95% CI = 1.22–2.48, p = 0.002 (Table 3; Figure 3). Subgroup analysis indicated that the study design did not affect the prognosis of PFS based on NLR (all p < 0.05; Table 3).
TABLE 3.
Subgroup analysis of the prognostic value of NLR for PFS in gastric cancer patients receiving ICIs.
| Subgroups | No. of studies | No. of patients | Effects model | HR (95% CI) | p | Heterogeneity | |
|---|---|---|---|---|---|---|---|
| I 2 (%) | p value | ||||||
| Total | 8 | 586 | Random | 1.74 (1.22–2.48) | 0.002 | 63.8 | 0.007 |
| Country | |||||||
| Japan | 4 | 240 | Random | 1.25 (0.88–1.77) | 0.206 | 18.6 | 0.297 |
| China | 3 | 301 | Random | 2.41 (1.56–3.72) | <0.001 | 48.9 | 0.141 |
| South Korea | 1 | 45 | — | 2.17 (1.05–4.52) | 0.037 | — | — |
| Sample size | |||||||
| <75 | 4 | 156 | Random | 1.81 (1.13–2.90) | 0.014 | 29.6 | 0.235 |
| ≥75 | 4 | 430 | Random | 1.71 (0.99–2.93) | 0.053 | 80.1 | 0.002 |
| Study design | |||||||
| Retrospective | 6 | 483 | Random | 1.61 (1.01–2.56) | 0.045 | 73.3 | 0.002 |
| Prospective | 2 | 103 | Random | 2.15 (1.35–3.43) | 0.001 | 0 | 0.974 |
| Cut‐off value | |||||||
| <3 | 4 | 221 | Random | 1.43 (0.88–2.32) | 0.148 | 52.4 | 0.098 |
| ≥3 | 4 | 365 | Random | 2.07 (1.27–3.38) | 0.003 | 66.3 | 0.031 |
| Cut‐off determination | |||||||
| ROC curve | 5 | 288 | Random | 1.78 (1.15–2.77) | 0.010 | 73.8 | 0.004 |
| Literature | 1 | 24 | — | 2.43 (0.92–6.42) | 0.073 | — | — |
| Median value | 2 | 74 | Random | 1.31 (0.45–3.88) | 0.620 | 68.1 | 0.077 |
| Survival analysis | |||||||
| Multivariate | 5 | 427 | Random | 1.87 (1.19–2.93) | 0.006 | 73.7 | 0.004 |
| Univariate | 3 | 159 | Random | 1.46 (0.79–2.69) | 0.232 | 36.6 | 0.207 |
| Cancer type | |||||||
| AGC | 6 | 351 | Random | 1.55 (1.10–2.19) | 0.013 | 32.6 | 0.192 |
| Metastatic/recurrent GC | 2 | 235 | Random | 2.17 (0.93–5.07) | 0.073 | 87.4 | 0.005 |
| Study center | |||||||
| Single center | 5 | 459 | Random | 1.51 (0.90–2.55) | 0.118 | 78.1 | 0.001 |
| Multicenter | 3 | 127 | Random | 2.20 (1.45–3.35) | <0.001 | 0 | 0.976 |
| Treatment ICIs type | |||||||
| Nivolumab | 5 | 285 | Random | 1.38 (0.97–1.94) | 0.070 | 28.2 | 0.233 |
| Nivolumab or other ICIs | 3 | 301 | Random | 2.41 (1.56–3.72) | <0.001 | 48.9 | 0.141 |
Abbreviations: AGC, advanced gastric cancer; ICIs, immune checkpoint inhibitors; NLR, neutrophil‐to‐lymphocyte ratio; PFS, progression‐free survival; ROC, receiver operating characteristic.
FIGURE 3.

Forest plots of NLR expression and progression‐free survival risk in gastric cancer patients receiving immune checkpoint inhibitors.
3.5. Correlation between NLR and clinicopathologic parameters
We further investigated the association between NLR and clinicopathological features in GC patients receiving ICIs. The pooled results demonstrated that there was no significant correlation between NLR and sex (OR = 0.99, 95% CI = 0.65–1.53, p = 0.973), age (OR = 0.77, 95% CI = 0.32–1.85, p = 0.971), Eastern Cooperative Oncology Group Performance Status (ECOG‐PS; OR = 1.41, 95% CI = 0.42–4.75, p = 0.579), differentiation (OR = 1.05, 95% CI = 0.44–2.53, p = 0.909), human epidermal growth factor receptor 2 (HER2) status (OR = 0.88, 95% CI = 0.46–1.69, p = 0.710), or PD‐L1 status (OR = 1.04, 95% CI = 0.48–2.27, p = 0.914) in GC patients treated with ICIs (Table 4).
TABLE 4.
The association between NLR and clinicopathological factors in gastric cancer patients receiving ICIs.
| Variables | No. of studies | No. of patients | Effects model | OR (95% CI) | p | Heterogeneity | |
|---|---|---|---|---|---|---|---|
| I 2 (%) | p value | ||||||
| Sex (male vs. female) | 4 | 390 | Random | 0.99 (0.65–1.53) | 0.973 | 85.1 | <0.001 |
| Age (years) (≥65 vs. <65) | 4 | 390 | Random | 0.77 (0.32–1.85) | 0.971 | 0 | 0.564 |
| ECOG PS (≥2 vs. 0–1) | 4 | 390 | Random | 1.41 (0.42–4.75) | 0.579 | 60.1 | 0.057 |
| Differentiation (poor vs. moderate or well) | 3 | 332 | Random | 1.05 (0.44–2.53) | 0.909 | 72.5 | 0.026 |
| HER2 status (positive vs. negative) | 2 | 195 | Random | 0.88 (0.46–1.69) | 0.710 | 0 | 0.862 |
| PD‐L1 status (positive vs. negative) | 2 | 195 | Random | 1.04 (0.48–2.27) | 0.914 | 0 | 0.577 |
3.6. Sensitivity analysis
Sensitivity analysis was conducted by removing each included study one at a time to assess the impact of each study on the overall results. The OS and PFS results demonstrated that the outcomes were credible and reliable (Figure 4).
FIGURE 4.

Sensitivity analysis for the pooled results in this meta‐analysis. (A) OS; (B) PFS.
3.7. Publication bias assessment
Begg's and Egger's tests were used to detect publication bias in this meta‐analysis. In Begg's and Egger's tests for OS, the p‐values were 0.858 and 0.543, respectively (Figure 5). For PFS, the results were as follows: Begg's test, p = 0.902 and Egger's test, p = 0.477 (Figure 5). These results indicate that there was no significant publication bias in this meta‐analysis.
FIGURE 5.

Publication bias test. (A) Begg's test for OS, p = 0.858; (B) Egger's test for OS, p = 0.543; (C) Begg's test for PFS, p = 0.902; and (D) Egger's test for PFS, p = 0.477.
4. DISCUSSION
The prognostic value of NLR for GC patients receiving treatment with ICIs has been controversial thus far. In the current meta‐analysis, we synthesized data from 10 eligible studies that included 830 patients. The pooled results indicated that an elevated NLR was associated with poor OS and PFS in GC patients using ICIs. In contrast, there was no significant association between NLR and clinicopathological factors in GC patients receiving ICIs. Based on the publication bias test, we concluded that our results are reliable. To the best of our knowledge, this is the first meta‐analysis investigating the prognostic significance of NLR in GC patients treated with ICIs. Our meta‐analysis has implications for the clinical management of GC patients using PD‐L1 antibodies. GC patients with a high pretreatment NLR (≥3) may experience progression or recurrence after receiving ICIs and should be closely monitored and followed‐up.
In the past decade, immunotherapy, including ICIs, has emerged as a breakthrough for GC patients. 25 The ATTRACTION‐2 trial showed that nivolumab improved OS compared with placebo as a third‐ or later‐line treatment for GC. 5 Several anti‐PD‐1 antibodies also show promising activity as a first‐line treatment for GC patients. 26 ATTRACTION‐04, a recent Asian phase II–III trial, found that nivolumab in combination with chemotherapy (CAPOX or S‐1 plus oxaliplatin) significantly improved PFS compared with nivolumab alone in HER2‐negative gastroesophageal junction (GEJ) cancer or GC patients, irrespective of PD‐L1 expression, whereas OS was not significantly improved. 27 Prognostic markers are important to predict the efficacy of ICIs in GC patients. In this meta‐analysis, we report that a high NLR is a significant prognostic biomarker for GC patients receiving ICIs. The NLR is a parameter reflecting inflammatory responses that can promote tumor development and metastasis. In spite of the fact that the precise mechanisms have not yet been fully elucidated, the following factors contribute to its explanation. First, neutrophils are critical components of the tumor microenvironment (TME) 28 that can directly target tumor cells and thereby drive tumor development. Neutrophils can secrete a series of cytokines and chemokines, including transforming growth factor beta (TGF‐β), vascular endothelial growth factor (VEGF), interleukin (IL)‐6, IL‐8, and IL‐12, which can enhance metastasis and angiogenesis in the TME. 29 Second, lymphocytes play a pivotal role in cancer immune surveillance and in preventing the development of malignancy. Tumor‐infiltrating lymphocytes (TILs) secreting interferon‐γ can induce PD‐L1 expression and promote the progression of ovarian cancer. 30 Low lymphocyte counts may result in insufficient immune responses, leading to poor prognosis for solid tumor survivors. 31 Therefore, a high NLR, which is the result of high neutrophil and/or low lymphocyte counts, is a reasonable and reliable prognostic marker for patients with cancer.
Notably, in this meta‐analysis, we found that a high NLR was associated with poor survival in GC patients undergoing treatment with ICIs. However, our results did not recommend immunotherapy, and ICIs are promising treatment methods for AGC patients. Our results should be interpreted with caution in the following respects: First, all GC patients were diagnosed with AGC, recurrent GC, or metastatic GC (Table 1). The prognosis of AGC patients is extremely poor. In the AGC patient group, median PFS and OS were reported as 4.9–6.0 months and 10.5–14.1 months, respectively. 32 The survival duration is relatively short, and there is limited room for improvement of survival using ICIs. Second, immunotherapy remains an effective therapeutic method for AGC patients. ICI treatment has demonstrated a highly variable objective response rate in GC patients, ranging from 10% to 26%. 33 Third, in HER2 positive AGC patients, a double‐blind, placebo‐controlled Phase III KEYNOTE‐811 study is still ongoing to evaluate the efficacy and safety of pembrolizumab or placebo in combination with trastuzumab and chemotherapy as first‐line treatment. 34 Moreover, in the included studies, ICIs were used as third‐line (or later) treatments for patients with unresectable, recurrent, or metastatic GC.
We investigated the prognostic role of NLR for OS and PFS according to cancer type, as shown in the subgroup analysis in Tables 2 and 3. Cancer type did not affect the prognostic role of NLR for OS (Table 2). A high NLR was associated with poor PFS in AGC patients (Table 3). However, the direct association between NLR and cancer type could not be analyzed based on the data of the included studies. Each study enrolled patients with one cancer type (AGC or metastatic/recurrent GC). Future studies are needed to explore the correlation between NLR and GC type.
Through meta‐analysis, many studies have indicated that NLR has prognostic value in several cancer patients receiving ICIs. 35 , 36 A meta‐analysis involving 27 studies showed that in patients treated with immunotherapy for advanced‐stage cancer, a high pretreatment NLR appears to be a promising prognostic biomarker. 35 In a meta‐analysis of 1700 patients, researchers reported that elevated blood NLR was associated with shorter PFS and OS in non‐small‐cell lung cancer treated with PD‐1/PD‐L1 inhibitors. 37 Chen et al. showed that an elevated NLR was associated with inferior OS and PFS in patients with renal cell carcinoma treated with ICIs, based on a meta‐analysis including 12 studies. 38 In a recent meta‐analysis with 14 studies, Takenaka et al. demonstrated that a higher NLR was associated with poor OS, PFS, response, and disease control in patients with head and neck squamous cell carcinoma treated with ICIs. 39 In a meta‐analysis encompassing 1164 patients, Zhang et al. reported that higher modified Glasgow Prognostic Score (mGPS)/GPS was associated with poor prognosis in patients with advanced cancer undergoing therapy with ICIs. 40 In this meta‐analysis, we pooled data from 830 patients and found a significant prognostic value of NLR for GC patients undergoing treatment with ICIs. Our results are in line with those of previous studies on NLR and prognosis in other cancer types. Furthermore, we are the first to report these findings in patients with GC.
Our meta‐analysis has several limitations. First, all eligible studies were from Asia, and specifically from the following three countries: China, Japan, and South Korea. However, we did not restrict the nationality or region of the eligible studies, and only considered English publications. Therefore, our results may be applicable to Asian patients, and the prognostic role of NLR in non‐Asian GC patients treated with ICIs needs to be verified. Second, the results were pooled based on 10 eligible studies but not for each individual patient. Therefore, selection may exist. Third, the studies included in the analysis varied in their cut‐off values of NLR, ranging from 2.5 to 5. A standard cut‐off value for NLR is needed in future studies to avoid the inherent heterogeneity caused by inconsistent data in this meta‐analysis.
5. CONCLUSION
In summary, our meta‐analysis demonstrated that a high pretreatment NLR was significantly associated with poor OS and PFS in GC patients receiving ICIs. NLR is an effective biomarker for assessing the prognosis of GC patients receiving ICIs and provides valuable information for making treatment decisions in the era of immunotherapy for GC. Due to study limitations, prospective clinical trials including patients of diverse ethnicities are needed to validate our findings.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
Supporting information
Data S1. The PRISMA checklist for this meta‐analysis.
Li L‐L, Pan L‐S. Prognostic value of neutrophil‐to‐lymphocyte ratio in gastric cancer patients treated with immune checkpoint inhibitors: A meta‐analysis. Kaohsiung J Med Sci. 2023;39(8):842–852. 10.1002/kjm2.12694
REFERENCES
- 1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49. [DOI] [PubMed] [Google Scholar]
- 2. Seeneevassen L, Bessède E, Mégraud F, Lehours P, Dubus P, Varon C. Gastric cancer: advances in carcinogenesis research and new therapeutic strategies. Int J Mol Sci. 2021;22(7):3418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Li K, Zhang A, Li X, Zhang H, Zhao L. Advances in clinical immunotherapy for gastric cancer. Biochim Biophys Acta Rev Cancer. 2021;1876(2):188615. [DOI] [PubMed] [Google Scholar]
- 4. Fuchs CS, Doi T, Jang RW, Muro K, Satoh T, Machado M, et al. Safety and efficacy of Pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE‐059 trial. JAMA Oncol. 2018;4(5):e180013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Kang YK, Boku N, Satoh T, Ryu MH, Chao Y, Kato K, et al. Nivolumab in patients with advanced gastric or gastro‐oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO‐4538‐12, ATTRACTION‐2): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet. 2017;390(10111):2461–71. [DOI] [PubMed] [Google Scholar]
- 6. Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, et al. Global surveillance of trends in cancer survival 2000‐14 (CONCORD‐3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population‐based registries in 71 countries. Lancet. 2018;391(10125):1023–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer [review]. Cell. 2010;140(6):883–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Jin X, Wang K, Shao X, Huang J. Prognostic implications of the peripheral platelet‐to‐lymphocyte ratio and neutrophil‐to‐lymphocyte ratio in predicting pathologic complete response after neoadjuvant chemotherapy in breast cancer patients. Gland Surg. 2022;11(6):1057–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Jiang N, Deng JY, Liu Y, Ke B, Liu HG, Liang H. The role of preoperative neutrophil‐lymphocyte and platelet‐lymphocyte ratio in patients after radical resection for gastric cancer. Biomarkers. 2014;19(6):444–51. [DOI] [PubMed] [Google Scholar]
- 10. Wang Q, 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. 2019;10(5):965–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hsu JT, Wang CC, Le PH, Chen TH, Kuo CJ, Lin CJ, et al. Lymphocyte‐to‐monocyte ratios predict gastric cancer surgical outcomes. J Surg Res. 2016;202(2):284–90. [DOI] [PubMed] [Google Scholar]
- 12. Li Y, Zhang Z, Hu Y, Yan X, Song Q, Wang G, et al. Pretreatment neutrophil‐to‐lymphocyte ratio (NLR) may predict the outcomes of advanced non‐small‐cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs). Front Oncol. 2020;10:654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Ogata T, Satake H, Ogata M, Hatachi Y, Inoue K, Hamada M, et al. Neutrophil‐to‐lymphocyte ratio as a predictive or prognostic factor for gastric cancer treated with nivolumab: a multicenter retrospective study. Oncotarget. 2018;9(77):34520–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Namikawa T, Yokota K, Tanioka N, Fukudome I, Iwabu J, Munekage M, et al. Systemic inflammatory response and nutritional biomarkers as predictors of nivolumab efficacy for gastric cancer. Surg Today. 2020;50(11):1486–95. [DOI] [PubMed] [Google Scholar]
- 15. Ota Y, Takahari D, Suzuki T, Osumi H, Nakayama I, Oki A, et al. Changes in the neutrophil‐to‐lymphocyte ratio during nivolumab monotherapy are associated with gastric cancer survival. Cancer Chemother Pharmacol. 2020;85(2):265–72. [DOI] [PubMed] [Google Scholar]
- 16. Yamada T, Hayashi T, Inokuchi Y, Hayashi K, Watanabe H, Komori K, et al. Impact of the neutrophil‐to‐lymphocyte ratio on the survival of patients with gastric cancer treated with Nivolumab monotherapy. Target Oncol. 2020;15(3):317–25. [DOI] [PubMed] [Google Scholar]
- 17. Gou M, Qu T, Wang Z, Yan H, Si Y, Zhang Y, et al. Neutrophil‐to‐lymphocyte ratio (NLR) predicts PD‐1 inhibitor survival in patients with metastatic gastric cancer. J Immunol Res. 2021;2021:2549295–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Kim N, Yu JI, Lim DH, Lee J, Kim ST, Hong JY, et al. Prognostic impact of sarcopenia and radiotherapy in patients with advanced gastric cancer treated with anti‐PD‐1 antibody. Front Immunol. 2021;12:701668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Ruan DY, Chen YX, Wei XL, Wang YN, Wang ZX, Wu HX, et al. Elevated peripheral blood neutrophil‐to‐lymphocyte ratio is associated with an immunosuppressive tumour microenvironment and decreased benefit of PD‐1 antibody in advanced gastric cancer. Gastroenterol Rep (Oxf). 2021;9(6):560–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Ishido K, Tanabe S, Katada C, Ishibashi Y, Kitahara G, Onoue M, et al. Evaluation of prognostic factors for unresectable or recurrent gastric cancer treated with Nivolumab. J Gastrointest Cancer. 2022. 10.1007/s12029-022-00823-1 [DOI] [PubMed] [Google Scholar]
- 21. Kim JH, Ryu MH, Park YS, Ma J, Lee SY, Kim D, et al. Predictive biomarkers for the efficacy of nivolumab as ≥ 3(rd)‐line therapy in patients with advanced gastric cancer: a subset analysis of ATTRACTION‐2 phase III trial. BMC Cancer. 2022;22(1):378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Qu Z, Wang Q, Wang H, Jiao Y, Li M, Wei W, et al. The effect of inflammatory markers on the survival of advanced gastric cancer patients who underwent anti‐programmed death 1 therapy. Front Oncol. 2022;12:783197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Moher D, Liberati A, Tetzlaff J, Altman DG, Grp P. Preferred reporting items for systematic reviews and meta‐analyses: the PRISMA statement. Ann Intern Med. 2009;151(4):264–W64. [DOI] [PubMed] [Google Scholar]
- 24. Stang A. Critical evaluation of the Newcastle‐Ottawa scale for the assessment of the quality of nonrandomized studies in meta‐analyses. Eur J Epidemiol. 2010;25(9):603–5. [DOI] [PubMed] [Google Scholar]
- 25. Kawazoe A, Shitara K, Boku N, Yoshikawa T, Terashima M. Current status of immunotherapy for advanced gastric cancer. Jpn J Clin Oncol. 2021;51(1):20–7. [DOI] [PubMed] [Google Scholar]
- 26. Xu J, Zhang Y, Jia R, Yue C, Chang L, Liu R, et al. Anti‐PD‐1 antibody SHR‐1210 combined with Apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: an open‐label, dose escalation and expansion study. Clin Cancer Res. 2019;25(2):515–23. [DOI] [PubMed] [Google Scholar]
- 27. Boku N, Ryu MH, Oh DY, Oh SC, Chung HC, Lee KW, et al. Nivolumab plus chemotherapy versus chemotherapy alone in patients with previously untreated advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer: ATTRACTION‐4 (ONO‐4538‐37) study. Ann Oncol. 2020;31:S1192. [Google Scholar]
- 28. Treffers LW, Hiemstra IH, Kuijpers TW, van den Berg TK, Matlung HL. Neutrophils in cancer. Immunol Rev. 2016;273(1):312–28. [DOI] [PubMed] [Google Scholar]
- 29. Templeton AJ, McNamara MG, Šeruga B, Vera‐Badillo FE, Aneja P, Ocaña A, et al. Prognostic role of neutrophil‐to‐lymphocyte ratio in solid tumors: a systematic review and meta‐analysis. J Natl Cancer Inst. 2014;106(6):dju124. [DOI] [PubMed] [Google Scholar]
- 30. Abiko K, Matsumura N, Hamanishi J, Horikawa N, Murakami R, Yamaguchi K, et al. IFN‐γ from lymphocytes induces PD‐L1 expression and promotes progression of ovarian cancer. Br J Cancer. 2015;112(9):1501–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Väyrynen JP, Tuomisto A, Klintrup K, Mäkelä J, Karttunen TJ, Mäkinen MJ. Detailed analysis of inflammatory cell infiltration in colorectal cancer. Br J Cancer. 2013;109(7):1839–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kang YK, Chen LT, Ryu MH, Oh DY, Oh SC, Chung HC, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2‐negative, untreated, unresectable advanced or recurrent gastric or gastro‐oesophageal junction cancer (ATTRACTION‐4): a randomised, multicentre, double‐blind, placebo‐controlled, phase 3 trial. Lancet Oncol. 2022;23(2):234–47. [DOI] [PubMed] [Google Scholar]
- 33. Zeng D, Wu J, Luo H, Li Y, Xiao J, Peng J, et al. Tumor microenvironment evaluation promotes precise checkpoint immunotherapy of advanced gastric cancer. J Immunother Cancer. 2021;9(8):e002467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Chung HC, Bang YJ, Fuchs CS, Qin SK, Satoh T, Shitara K, et al. First‐line pembrolizumab/placebo plus trastuzumab and chemotherapy in HER2‐positive advanced gastric cancer: KEYNOTE‐811. Future Oncol. 2021;17(5):491–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Jiang T, Qiao M, Zhao C, Li X, Gao G, Su C, et al. Pretreatment neutrophil‐to‐lymphocyte ratio is associated with outcome of advanced‐stage cancer patients treated with immunotherapy: a meta‐analysis. Cancer Immunol Immunother. 2018;67(5):713–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Sacdalan DB, Lucero JA, Sacdalan DL. Prognostic utility of baseline neutrophil‐to‐lymphocyte ratio in patients receiving immune checkpoint inhibitors: a review and meta‐analysis. Onco Targets Ther. 2018;11:955–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Jiang T, Bai Y, Zhou F, Li W, Gao G, Su C, et al. Clinical value of neutrophil‐to‐lymphocyte ratio in patients with non‐small‐cell lung cancer treated with PD‐1/PD‐L1 inhibitors. Lung Cancer. 2019;130:76–83. [DOI] [PubMed] [Google Scholar]
- 38. Chen X, Meng F, Jiang R. Neutrophil‐to‐lymphocyte ratio as a prognostic biomarker for patients with metastatic renal cell carcinoma treated with immune checkpoint inhibitors: a systematic review and meta‐analysis. Front Oncol. 2021;11:746976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Takenaka Y, Oya R, Takemoto N, Inohara H. Neutrophil‐to‐lymphocyte ratio as a prognostic marker for head and neck squamous cell carcinoma treated with immune checkpoint inhibitors: meta‐analysis. Head Neck. 2022;44(5):1237–45. [DOI] [PubMed] [Google Scholar]
- 40. Zhang Y, Chen S, Chen H, Li W. A comprehensive analysis of Glasgow Prognostic Score (GPS)/the modified Glasgow Prognostic Score (mGPS) on immune checkpoint inhibitor efficacy among patients with advanced cancer. Cancer Med. 2023;12(1):38–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data S1. The PRISMA checklist for this meta‐analysis.
