Background: PD-1 inhibitors have been approved for the first-line treatment of patients with advanced gastric cancer, gastroesophageal junction cancer, or esophageal adenocarcinoma. However, the results of several clinical trials are not entirely consistent, and the dominant population of first-line immunotherapy for advanced gastric/gastroesophageal junction cancer still needs to be precisely determined. Objective: This objective of this study is to evaluate the efficacy of anti-PD-1/PD-L1 therapy in advanced gastric/gastroesophageal junction adenocarcinoma patients through a systematic review and meta-analysis of relevant clinical trials. Method: The PubMed, Embase, and Cochrane Library electronic databases were searched up to August 1, 2022, for clinical trials of anti-PD-1/PD-L1 immunotherapy for the first-line treatment of advanced gastroesophageal cancer. Hazard ratios and 95% confidence intervals for overall survival, progression-free survival, and objective response rates were extracted and pooled for meta-analysis. Prespecified subgroups included the following: agent type, PD-L1 expression, and high microsatellite instability. Results: This study analyzed 5 RCTs involving 3,355 patients. Compared with the chemotherapy group, the combined immunotherapy group had a significantly higher objective response rate (OR = 0.63, 95% CI: 0.55–0.72, p < 0.00001) and prolonged overall survival (HR = 0.82, 95% CI: 0.76–0.88, p < 0.00001) and progression-free survival (HR = 0.75, 95% CI: 0.69–0.82, p < 0.00001). The combination of immunotherapy and chemotherapy prolonged OS in both MSI-H (HR = 0.38, p = 0.002) and MSS (HR = 0.78, p < 0.00001) populations, but there was a significant difference between groups (p = 0.02). However, in improving ORR, the benefit of ICI combined with chemotherapy in the MSS group and MSI-H group was not significantly different between groups (p = 0.52). Combination therapy with ICIs was more effective than chemotherapy alone in prolonging OS in the subgroup with a high CPS, regardless of the CPS cutoff for PD-L1. However, when the cutoff of CPS was 1, the difference between subgroups did not reach statistical significance (p = 0.12), while the benefit ratio of the MSI-H group was higher when the cutoff was 10 (p = 0.004) than when the cutoff value was 5 (p = 0.002). Conclusions: For first-line treatment of advanced gastroesophageal cancer, an ICI combination strategy is more effective than chemotherapy. The subgroup of patients with a CPS ≥10 has a more significant benefit, and CPS ≥10 has the potential to be used as an accurate marker of the dominant population of immuno-combined therapy.

Gastric cancer is one of the malignant tumors with the highest morbidity and mortality worldwide [1]. Most patients with gastric cancer are in the advanced stage of the tumor when they are first diagnosed [2]. With the continuous exploration of molecular genetics by researchers, the roles of various molecular targets, such as VEGF and HER2, in the progression of gastric cancer have gradually emerged, and the treatment of gastric cancer has continued to improve [3‒5]. The treatment strategy for advanced gastric cancer has evolved from chemotherapy monotherapy, such as cisplatin and anthracycline, to a 5-FU-based regimen and platinum-containing regimen and then developed from a combination regimen containing various new chemotherapy drugs to a combination of chemotherapy and targeted therapy [6‒8]. Over the past 40 years, the median overall survival (OS) of advanced gastric cancer has effectively improved from 4 months to 13.8 months under the continuous development of chemotherapy regimens. Unfortunately, the development of new targeted drugs frequently fails, and the median OS stagnates at the 1-year mark. The innovation of gastric cancer treatment is stuck in a bottleneck, and a breakthrough is urgently needed.

The emergence of immunotherapy brings new hope for the treatment of advanced gastric cancer. Due to the lack of antigen expression in tumor cells and the existence of an immunosuppressive environment, malignant tumors often exhibit immune evasion. Although gastrointestinal tumors are not as highly immunogenic as melanoma and nonsmall cell lung cancer, the infiltration of lymphocytes around tumors is closely related to the efficacy of tumor immunotherapy and long-term prognosis [9, 10]. In recent years, immune checkpoint inhibitors targeting CTLA-4, PD-1/PD-L1, and other immune checkpoints have achieved promising results in various advanced malignant tumors, including melanoma and nonsmall cell lung cancer [11‒14].

Thus, immunotherapy has been initially explored in gastric cancer, from the back line to the front line and from monotherapy to the combination strategy. A number of clinical trials of advanced gastric and gastroesophageal junction adenocarcinoma (G/GEJAC) suggest that ICI therapy can improve the efficacy with controllable toxicity and side effects [15‒18]. The KEYNOTE-059 trial confirmed that the use of pembrolizumab in recurrently advanced gastric cancer resulted in a higher response rate in patients with a positive combined positive score (CPS) of PD-L1 [19, 20]. Based on this result, pembrolizumab was approved by the US Food and Drug Administration (FDA) for advanced G/GEJAC patients with a CPS ≥1. Due to the affected functions, such as eating and digestion, patients with advanced gastric cancer are often too weak to suffer drug stacking. In addition to being unable to tolerate multiple rounds of treatment, their immune systems are also more damaged. Therefore, first-line therapy may be a better time to treat. After moving the number of treatment lines forward, the phase III randomized first-line study CHECKMATE-649 found that ICI-combined chemotherapy can significantly improve the survival benefit for HER2-negative advanced G/GEJAC patients compared with chemotherapy alone [21]. On April 16, 2021, the FDA first approved nivolumab combined with chemotherapy for the first-line treatment of all advanced G/GEJAC based on the results of CHECKMATE-649.

However, not all first-line clinical trials have yielded completely consistent results. The ATTRACTION-4 results suggested that the combined strategy of ICI did not significantly prolong OS compared with chemotherapy alone [22, 23]. Not all gastric cancer patients have a clear response to ICIs, and the screening of ICI-predominant populations is still difficult. In gastric cancer tissues, the expression level of PD-L1 is distinct and associated with many clinicopathological features with poor prognosis. KEYNOTE-062, which only enrolled patients with a CPS ≥1, confirmed that pembrolizumab-combined chemotherapy improved objective response rate (ORR) [24]. However, OS (CPS ≥1 or CPS ≥10) and progression-free survival (PFS) (CPS ≥1) in the pembrolizumab-combined group were not superior to those in the chemotherapy group. The distinctive role of a CPS ≥1 in patients undergoing ICI-combined chemotherapy for G/GEJAC remains unclear. Although microsatellite instability (MSI) has a good predictive effect on the efficacy of ICIs in many solid tumors, it has not been clearly indicated in G/GEJAC. Therefore, it is very important to explore the efficacy of ICI-combined chemotherapy in different subgroups and to identify biomarkers for the population benefiting from ICI therapy. This article aims to evaluate the efficacy of anti-PD-1/PD-L1 therapy in the whole population and subgroups of patients through a meta-analysis of clinical trials for first-line treatment in advanced G/GEJAC.

Search Strategy

Clinical trials and randomized controlled trials published in PubMed, Embase, Cochrane database, and Clinical Trials (www.ClinicalTrials.gov) before January 2023 were retrieved for primary screening. The search strategy was utilized as follows: (((advanced gastric cancer [MeSH Terms]) OR gastroesophageal cancer [MeSH Terms]) OR esophageal cancer [MeSH Terms]) AND ((immunotherapy OR PD-1 inhibitor OR PD-L1 inhibitor OR anti-PD-1 OR anti-PD-L1 OR pembrolizumab OR nivolumab OR opdivo OR keytruda OR atezolizumab OR tecentriq OR toripalimab OR JS001 OR sintilimab OR tislelizumab OR immune checkpoint)). To avoid missing the latest research progress in some clinical trials, we also searched conference proceedings in the American Society of Clinical Oncology (ASCO), European Society of Medical Oncology (ESMO), ASCO Gastrointestinal, and ESMO Gastrointestinal during the past 5 years (from 2018 to 2022). Duplicate articles were eliminated from our initial screening by reading titles and abstracts. For the rest of the literature, we intensively read the full text to evaluate the content of the articles and determine whether they met the inclusion and exclusion criteria. In addition, we also searched relevant references manually to find and supplement other potentially relevant reports.

Inclusion and Exclusion Criteria

The included literature met the following criteria: (1) randomized clinical trials; (2) metastatic or advanced adenocarcinoma of the gastro-esophagus; (3) comparison of the therapeutic effect and long-term outcome of anti-PD-1/PD-L1 immune checkpoint therapy with standard therapy; (4) first-line therapy; and (5) specific data on OS, PFS, and ORR, including hazard ratios (HRs), survival curves, and the number of clinical remission and the total number of patients in each group. The exclusion criteria were as follows: (1) retrospective phase I study or single-arm phase II study; (2) early-stage gastroesophageal cancer; (3) first-line maintenance therapy, second-line and third-line therapy; (4) lack of specific data on survival and clinical efficacy.

Data Extraction

All data used in this study were obtained from publications, relevant online supplementary materials, and presentations or abstracts of conference proceedings. Data were independently reviewed and extracted by two reviewers (Jun Wang and Jing Chen). If there was a disagreement, a consensus solution was obtained through consultation with the third reviewer Wenxuan Wu. When multiple papers or literature referred to the same data, the latest data corresponding to the desired study endpoint were included in the review, and these data were cross-checked with information mentioned in other literature.

The comparative data of long-term survival and short-term clinical efficacy between standard treatment and ICI-combined treatment were collected, including HRs and 95% confidence intervals (CIs) of median OS and median PFS, ORR, and number of patients in clinical remission. In addition, OS, PFS, and ORR data for populations with MSI or various PD-L1 expression levels were collected. We also extracted the NCT number, ICI type and agents, number of patients included in each group, treatment regimen given to the experimental and control groups, endpoint of the study, cutoff time and median follow-up time.

Statistical Analysis

Meta-analyses were performed using Review Manager 5.4.1 software (the Cochrane Collaboration, Copenhagen, Denmark). Figures including forest plots and funnel plots were automatically drawn by Review Manager 5.4.1 software. All reported p values are two sided, with p values <0.05 defined as statistically significant. We used I2 to assess heterogeneity across studies. Statistically significant heterogeneity was defined when I2 was greater than 50%, while the statistical analysis was considered to have no significant heterogeneity when I2 was less than 50%. When the heterogeneity was significant, the random-effects model was used for analysis; when there was acceptable heterogeneity, the fixed-effects model was used for analysis.

For long-term outcomes, HRs from Cox proportional-hazards models were performed in clinical trials to compare survival differences between standard chemotherapy and ICI-combined treatment. We extracted HRs and 95% CIs for OS and PFS assessments in the overall population of the included studies. Meta-analyses were performed to obtain pooled HRs, 95% CIs, and p values via the generic inverse variance method, comparing the effects of different treatments on OS and PFS. For short-term clinical efficacy, clinical trials usually perform imaging assessments of tumor regression in patients according to RECIST criteria. We extracted ORR for the overall population in the included studies, as well as the total number and the number of tumor regressions in each arm. Odds ratios (ORs), 95% CIs, and p values were meta-analyzed to assess whether there were significant differences in clinical efficacy among different arms. For further subgroup analysis, we additionally collected the above OS, PFS, and ORR information in the MSI and PD-L1 subgroups.

Basic Characteristics of the Included Clinical Trials

The process of literature selection is shown in the flowchart (Fig. 1). We retrieved a total of 197 results from online databases, while manual searches identified 35 studies from other sources. After a rough screening to eliminate duplicates, 127 studies were left for further retrieval. By completely reading the titles and abstracts of the above studies, 92 records were excluded due to inconsistencies in research purposes, cancer types, treatment regimens, or lack of comparable data. Then, full-text articles of the remaining 35 studies were thoroughly assessed. Consequently, 30 studies were excluded due to noncompliant therapeutic lines, phases of trials, treatment regimens, or unreported efficacy, and outcome data. Ultimately, 5 randomized clinical trials were included in this study.

Fig. 1.

Flowchart of literature search and screening.

Fig. 1.

Flowchart of literature search and screening.

Close modal

The basic characteristics of the included clinical trials are shown in Table 1. The five clinical trials finally included are all for first-line treatment of advanced G/GEJAC. Three arms were set in the KEYNOTE-062 trial, including a chemotherapy group, pembrolizumab monotherapy, and chemotherapy combined with pembrolizumab. To compare the difference in efficacy between ICI-combined and single chemotherapy, this study only included data from the control arm of chemotherapy and the experimental arm of pembrolizumab-combined therapy. The KEYNOTE-062 researchers published a paper in 2020 releasing ORR and partial survival data [25] and updated the final analysis results at the ASCO meeting in 2022 [26]. Therefore, ORR data for this study were derived from papers published in 2020, while survival data were extracted from results presented at the 2022 ASCO meeting.

Table 1.

Characteristics of first-line trials included in the meta-analysis

Author [Ref], publication yearTrialClinicalTrials.gov numberPhaseCancer typeHER2PD-1/PD-L1 inhibitorCutoff timePatients, N
Shitara et al. [24], 2020 KEYNOTE-062 NCT02494583 III aGC/GEJC Negative Pembrolizumab 19 Apr 2021 507 
Janjigian et al. [21], 2021 CHECKMATE-649 NCT02872116 III aGC/GEJC Negative Nivolumab 27 May 2021 1,581 
Kang et al. [22, 23], 2022 ATTRACTION-4 part 2 NCT02746796 III aGC/GEJC Negative Nivolumab 31 Oct 2018 724 
Janjigian et al. [26], 2021 KEYNOTE-811-cohort global NCT03615326 III aGC/GEJC Positive Pembrolizumab 17 Jun 2020 264 
Xu et al. [27], 2021 ORIENT-16 NCT03745170 III aGC/GEJC Negative Sintilimab 20 Jun 2021 650 
Author [Ref], publication yearTrialClinicalTrials.gov numberPhaseCancer typeHER2PD-1/PD-L1 inhibitorCutoff timePatients, N
Shitara et al. [24], 2020 KEYNOTE-062 NCT02494583 III aGC/GEJC Negative Pembrolizumab 19 Apr 2021 507 
Janjigian et al. [21], 2021 CHECKMATE-649 NCT02872116 III aGC/GEJC Negative Nivolumab 27 May 2021 1,581 
Kang et al. [22, 23], 2022 ATTRACTION-4 part 2 NCT02746796 III aGC/GEJC Negative Nivolumab 31 Oct 2018 724 
Janjigian et al. [26], 2021 KEYNOTE-811-cohort global NCT03615326 III aGC/GEJC Positive Pembrolizumab 17 Jun 2020 264 
Xu et al. [27], 2021 ORIENT-16 NCT03745170 III aGC/GEJC Negative Sintilimab 20 Jun 2021 650 

CHECKMATE-649 lays the foundation for nivolumab combined with chemotherapy as a first-line treatment for advanced HER-2-negative gastric cancer. It also has three treatment arms, including chemotherapy, nivolumab combined with chemotherapy, and nivolumab combined with ipilimumab [24]. This study only extracted the latest efficacy and survival data of the chemotherapy arm and nivolumab-combined chemotherapy arm. The phase III portion of the ATTACTION-4 trial compared the efficacy and safety of nivolumab plus chemotherapy or placebo plus chemotherapy as first-line therapy in Asian patients with HER2-negative advanced G/GEJAC. Efficacy and survival data for treatment groups in ATTRACTION-4 were extracted for this meta-analysis. The KEYNOTE-811 global cohort compared the efficacy and safety of patients receiving pembrolizumab or placebo in combination with trastuzumab and chemotherapy as standard first-line therapy for HER2-positive advanced G/GEJAC. The latest published results of interim analysis for KEYNOTE-811 provided ORR-related data for this study. The ORIENT-16 trial is the first phase III study to report the efficacy and safety of sintilimab-combined chemotherapy versus placebo-combined chemotherapy in the first-line treatment of advanced G/GEJAC. This meta-analysis included the latest ORR and survival results of ORIENT-16 presented in an oral presentation at the ESMO Congress 2021. All trials included were randomized, prospective, and multicenter clinical trials. We assessed the risk of bias and found that all included studies were of acceptable quality (online suppl. Fig. 1, 2; for all online suppl. material, see https://doi.org/10.1159/000531457).

Long-Term Outcome and Short-Term Response of Patients with ICIs

In the intention-to-treat cohort, as Figure 2a shows, treatment with ICIs prolonged the median OS overall compared with chemotherapy alone (HR = 0.82, 95% CI: 0.76–0.88, p < 0.00001). Integrated analysis of 4 trials (ATTRACTION-4, CHECKMATE-649, KEYNOTE-062, and ORIENT-16) supported that median PFS acquired the same benefit as median OS (Fig. 2b). Compared to the chemotherapy group, the median PFS was significantly longer in the ICI-based group (HR = 0.75, 95% CI: 0.69–0.82, p < 0.00001). Collecting the ORR data, we evaluated the short-term clinical responses of all included trials (n = 5), as shown in Figure 2c. Meta-analysis showed that when combined with ICIs, the objective response significantly improved compared to accepting chemotherapy alone (OR = 0.63, 95% CI: 0.55–0.72, p < 0.00001).

Fig. 2.

Relationship of endpoint and therapeutic strategy. a OS. b PFS. c ORR.

Fig. 2.

Relationship of endpoint and therapeutic strategy. a OS. b PFS. c ORR.

Close modal

Effect of ICIs on Patients by Subgroups

Microsatellite Instability

OS was first assessed to evaluate different MSIs (Fig. 3a). As the forest plot shows, the median OS was prolonged in the ICI-combined group compared with the control group in both MSI-H patients (HR = 0.38, 95% CI: 0.20–0.70, p = 0.002) and MSS patients (HR = 0.78, 95% CI: 0.70–0.87, p < 0.00001). Although subgroup differences met statistical significance (p = 0.02), high heterogeneity existed in the subgroups (I2 = 80.7%). Regarding the ORR as the endpoint of short-term response, we assessed the cumulative HRs for the ORR of the MSI-H and MSS subgroups (Fig. 3b). Similarly, ICI-combined treatment did not improve ORR significantly when compared with chemotherapy alone in MSI-H patients (OR = 0.44, 95% CI: 0.18–1.08, p = 0.07). However, in MSS patients, ICI-combined treatment was related to a higher ORR than in the control group (OR = 0.60, 95% CI: 0.48–0.74, p < 0.00001).

Fig. 3.

Relationship of endpoint and therapeutic strategy stratified by MSI. a OS. b ORR.

Fig. 3.

Relationship of endpoint and therapeutic strategy stratified by MSI. a OS. b ORR.

Close modal

Combined Positive Score

CPS was frequently discussed as the most promising biomarker for ICI-based treatment. Thus, OS, PFS, and ORR data were sorted for the subgroup analysis. To accurately analyze the subpopulation who acquire benefits most from ICI-based strategies, 1, 5, and 10 were most commonly set as the cutoff value of CPS. As the main endpoint of survival, the median OS data of various subgroups were adequate (Fig. 4). Analysis of the survival data in the CPS ≥1 subgroup suggested that ICI-combined treatment prolonged the median OS compared with chemotherapy (HR = 0.76, 95% CI: 0.70–0.84, p < 0.00001), while the median OS did not significantly differ between the ICI-combined arm and chemotherapy arm in the CPS <1 subgroup (HR = 0.95, 95% CI: 0.73–1.24, p = 0.70). Similarly, the HR for the median OS benefit with the ICI-combined arm compared to the chemotherapy arm was 0.69 (95% CI: 0.62–0.78, p < 0.00001) in the CPS ≥5 subgroup, while the HR in the CPS <5 subgroup was 0.94 (95% CI: 0.79–1.12, p = 0.49). The subgroup difference (p = 0.002) between CPS ≥10 (HR = 0.66, 95% CI: 0.58–0.75, p < 0.00001) and CPS <10 (HR = 0.91, 95% CI: 0.78–1.06, p = 0.23) patients was similar to that of subgroups when the CPS cutoff was 5 (p = 0.004).

Fig. 4.

Relationship of OS and therapeutic strategy stratified by CPS. a Cutoff = 1. b Cutoff = 5. c Cutoff = 10.

Fig. 4.

Relationship of OS and therapeutic strategy stratified by CPS. a Cutoff = 1. b Cutoff = 5. c Cutoff = 10.

Close modal

As another survival endpoint, evaluation of PFS demonstrated similar results as OS (Fig. 5). In the CPS ≥1 subgroup, patients who received ICI-combined therapy had longer PFS than those who received chemotherapy alone (HR = 0.75, 95% CI: 0.68–0.84, p < 0.00001). In the CPS <1 subgroup, patients who received chemotherapy or ICI-combined therapy had indistinctive PFS (HR = 0.93, 95% CI: 0.69–1.25, p = 0.63). However, the subgroup difference did not approach statistical significance (p = 0.19). When the cutoff was set as 5, the magnitude of benefit for ICI-combined therapy was higher compared to chemotherapy alone in the CPS ≥5 subgroup (HR = 0.68, 95% CI: 0.61–0.77, p < 0.00001) versus in the CPS <5 subgroup (HR = 0.93, 95% CI: 0.76–1.14, p = 0.48) and met statistical significance with p = 0.01 for subgroup difference.

Fig. 5.

Relationship of PFS and therapeutic strategy stratified by CPS. a Cutoff = 1. b Cutoff = 5.

Fig. 5.

Relationship of PFS and therapeutic strategy stratified by CPS. a Cutoff = 1. b Cutoff = 5.

Close modal

For short-term response, we calculated the ORs for ORR differences in patients under ICI-combined therapy and chemotherapy in the CPS ≥1 subgroup (OR = 0.55, 95% CI: 0.45–0.66, p < 0.00001) and CPS <1 (OR = 0.82, 95% CI: 0.21–3.22, p = 0.78) (Fig. 6a, subgroup differences p = 0.56, I2 = 0%). When the subgroups were divided by a cutoff of 5, ICI-combined treatment benefitted more than chemotherapy alone in the CPS-high subgroup (OR = 0.56, 95% CI: 0.45–0.69, p < 0.00001), while it benefited the same in the CPS-low subgroup (OR = 0.82, 95% CI: 0.21–3.22, p = 0.78), with no significance between subgroups (Fig. 6b, p = 0.57, I2 = 0%).

Fig. 6.

Relationship of ORR and therapeutic strategy stratified by CPS. a Cutoff = 1. b Cutoff = 5.

Fig. 6.

Relationship of ORR and therapeutic strategy stratified by CPS. a Cutoff = 1. b Cutoff = 5.

Close modal

Tumor Cells

In the subgroup analysis by PD-L1 expression in tumor cells, only median OS data were fully reported in the CHECKMATE-649 and ATTRACTION-4 trials and sequentially collected for our meta-analysis (Fig. 7a). The median OS was not distinct between the ICI-combined group and the control group in patients with ≥1 PD-L1-positive tumor cell (HR = 0.74, 95% CI: 0.38–1.42, p = 0.36). For patients with PD-L1-positive tumor cells <1, the HR for the median OS benefit in the ICI-combined group compared to the chemotherapy group was 0.85 (95% CI: 0.77–0.93, p = 0.0009). In total, the difference between subgroups was not statistically significant (p = 0.68), even with low heterogeneity (I2 = 0%).

Fig. 7.

Relationship of endpoints and therapeutic strategy by subgroup. a Relationship between OS and PD-L1 expression in tumor cells. b Relationship between ORR and ICI agents.

Fig. 7.

Relationship of endpoints and therapeutic strategy by subgroup. a Relationship between OS and PD-L1 expression in tumor cells. b Relationship between ORR and ICI agents.

Close modal

Agents of ICIs

Since the ICI agents used above were both PD-1 inhibitors, we assessed the clinical response of different agents (Fig. 7b). Subgroup analysis suggested that the combination of pembrolizumab (OR = 0.50, 95% CI: 0.30–0.83, p = 0.007) and nivolumab (OR = 0.64, 95% CI: 0.53–0.76, p < 0.00001) could both improve the ORR compared to the chemotherapy group. However, the effect of sintilimab on ORR was rare, which might be due to the limited number of trials, and only ORIENT-16 was included for assessment (OR = 0.75, 95% CI: 0.55–1.04, p = 0.08). Although the heterogeneity of this subgroup assessment was as low as I2 = 0%, the difference between subgroups of various ICI agents did not approach statistical significance (p = 0.38).

PD-1 is a negative regulatory receptor mainly expressed on the surface of activated T cells. It can inhibit T cells by binding to the ligand PD-L1 expressed on the surface of tumor cells, making it unable to attack tumor cells and escape immune surveillance. At present, a variety of drugs targeting the PD-1/PD-L pathway have been successfully validated in trials and have been approved for clinical applications in a variety of solid tumors [13, 25, 28, 29, 30]. In the field of advanced G/GEJAC, many large multicenter clinical trials of various therapeutic phases are underway, and some have revealed exciting results [21, 23, 24, 26, 27, 31]. CHECKMAKE-649 and ORIENT-16 consolidate the position of ICI-combined chemotherapy in the first-line treatment of advanced G/GEJAC. However, the population that is most sensitive to ICI treatment and acquires benefit is still unclear. As KEYNOTE-811 announced part of the efficacy evaluation data, we are the first to retrospectively analyze five phase III clinical trials of advanced G/GEJAC for first-line treatment. This study conducted a meta-analysis of five randomized controlled trials to clarify the prognostic value of ICI-combined chemotherapy in advanced G/GEJAC for first-line treatment and tried to explore the population who could benefit most from ICI-combined chemotherapy through subgroup analysis.

In this meta-analysis, by analyzing short-term efficacy and long-term survival data in the overall population, we found that the combination of ICI and chemotherapy was effective in improving ORR, OS, and PFS compared with chemotherapy alone. Although ATTRACTION-4 and KEYNOTE-062 did not show superior results in long-term survival data, the results of the meta-analysis affirmed the effect of ICI-combined chemotherapy on optimizing OS (HR = 0.77, p < 0.00001) and PFS (HR = 0.75, p < 0.00001). Among the different agents, pembrolizumab (HR = 0.50, p = 0.007) and nivolumab (HR = 0.50, p < 0.00001) both effectively improved the ORR, while sintilimab nonsignificantly improved the ORR (HR = 0.75, p = 0.08). This may be because ORIENT-16 was the only trial of the sintilimab-combined study that had published results with a limited sample size. This result still needs to be supplemented by more clinical trials with larger sample sizes to provide new agents for ICI combination therapy.

To determine the population that could benefit most from ICI combination therapy, we performed subgroup analysis of common biomarkers. It is well known that a high tumor mutational burden is associated with better ICI efficacy in various solid tumors. Tumors with high microsatellite instability (MSI-H) or mismatch repair deficiency showed higher mutation rates and methylation, leading to enhanced expression of neoantigens. Thus, MSI-H tumors are often accompanied by high infiltration of CD8+ T cells and are more sensitive to ICIs. Through sequencing analysis of primary tumors in G/GEJAC, previous researchers also found that MSI-H tumors were resistant to chemotherapy and were more likely to have durable responses to immunotherapy [44]. This meta-analysis suggested that ICI-combined chemotherapy could prolong OS in both MSI-H (HR = 0.38, p = 0.002) and MSS (HR = 0.78, p < 0.00001) populations, and there was a significant difference between the groups (χ2 = 5.17, p = 0.02). In terms of improving the ORR, ICI-combined chemotherapy also had a benefit in the MSS group (HR = 0.60, p < 0.00001) but not in the MSI-H group (HR = 0.44, p = 0.07). However, there was no significant difference between groups (χ2 = 0.41, p = 0.52). This result suggests that MSI has an inaccurate discriminative ability in distinguishing the efficacy of ICIs in patients with G/GEJAC. In the future, combined evaluation of MSI-H and other clinical or pathological features may be required to improve the sensitivity and accuracy of identifying potentially beneficial populations.

In addition to MSI, the expression level of PD-L1 is also a common marker of immunotherapy. In vivo, tumor cells, immune cells such as lymphocytes, macrophages, and interstitial cells also express PD-L1. The common evaluation criteria for PD-L1 expression level include CPS score, TPS score, IPS score, and so on. Different clinical trials have adopted various evaluation criteria for PD-L1 levels. In addition to evaluating PD-L1 expression only on tumor cells, more studies currently use CPS to comprehensively evaluate PD-L1 expression on tumor cells, immune cells, and interstitial cells in tumor lesions. At present, in different cancer types, the cutoff value of the CPS score is different. In the included trials of this study, CPS and TPS were mainly used for the evaluation. EMA and NCCN guidelines have recommended ICI-combined chemotherapy for advanced G/GEJAC with a CPS ≥5. However, the population that the FDA approved is the whole population regardless of CPS. Therefore, the discriminative role of the PD-L1 expression level in G/GEJAC remains unclear. Our subgroup analysis suggested that combination therapy with ICIs was more effective than chemotherapy alone in prolonging OS in the subgroup with a high CPS, regardless of the CPS cutoff for PD-L1. However, when the cutoff of the CPS was 1, the difference between subgroups did not reach statistical significance (χ2 = 2.36, p = 0.12), while the benefit ratio of the MSI-H group when the cutoff was 10 (χ2 = 8.16, p = 0.004) was higher than when the cutoff value was 5 (χ2 = 9.85, p = 0.002).

This study also has some limitations. Data on additional biomarkers, such as TMB and TCR diversity, could not be extracted from the included literature due to the paucity of published clinical trial data. It is hoped that a more in-depth stratified analysis can be carried out in the future to determine the groups that could benefit from immunotherapy. Second, heterogeneity existed in the included studies. Thus, this article uses more random-effects models for analysis rather than fixed-effects models.

Overall, this meta-analysis suggests that for the first-line treatment of advanced G/GEJAC, ICI-combined chemotherapy is superior to chemotherapy with better short-term efficacy and higher long-term survival. In addition, the ability to distinguish the best benefit population of MSI is poorer than that of the CPS, and the subgroup of patients with a CPS ≥10 may have more significant benefits. A CPS ≥10 has the potential to become an accurate marker of the dominant population of immuno-combined therapy.

We greatly thank the Department of Gastroenterology Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, for technical advice.

An ethics statement is not applicable because this study is based exclusively on published literature.

All authors state that there is no conflict of interest.

This work was supported by Zhejiang Provincial Key Project of Research and Development (2019C03043), Health Science and Technology Plan of Zhejiang Province (2022RC165), and Clinical Research Fund of Zhejiang Medical Association (2021ZYC-A68).

Conceptualization: Shengqi Fei, Jun Wang, and Yu Lu; data review and extraction: Jun Wang, Jian Chen, and Wenxuan Wu; software: Jing Chen and Jia Qi; original draft preparation: Shengqi Fei and Beidi Wang; review and editing: Yaxuan Han, Kefan Wang, and Jian Chen; and supervision: Xiaying Han, Haiyan Zhou, and Jian Chen. All authors have read and agreed to the published version of the manuscript.

Additional Information

Shengqi Fei and Yu Lu contributed equally to this work.

All data generated or analyzed during this study are included in this article and its online supplementary material files. Further inquiries can be directed to the corresponding author.

1.
Siegel
RL
,
Miller
KD
,
Wagle
NS
,
Jemal
A
.
Cancer statistics, 2023
.
CA Cancer J Clin
.
2023
;
73
(
1
):
17
48
.
2.
Smyth
EC
,
Nilsson
M
,
Grabsch
HI
,
van Grieken
NC
,
Lordick
F
.
Gastric cancer
.
Lancet
.
2020
;
396
(
10251
):
635
48
.
3.
Tan
Z
.
Recent advances in the surgical treatment of advanced gastric cancer: a review
.
Med Sci Monit
.
2019
;
25
:
3537
41
.
4.
Chia
NY
,
Tan
P
.
Molecular classification of gastric cancer
.
Ann Oncol
.
2016
;
27
(
5
):
763
9
.
5.
De Re
V
.
Molecular features distinguish gastric cancer subtypes
.
Int J Mol Sci
.
2018
;
19
(
10
):
3121
.
6.
Zhao
Q
,
Cao
L
,
Guan
L
,
Bie
L
,
Wang
S
,
Xie
B
.
Immunotherapy for gastric cancer: dilemmas and prospect
.
Brief Funct Genomics
.
2019
;
18
(
2
):
107
12
.
7.
Song
Z
,
Wu
Y
,
Yang
J
,
Yang
D
,
Fang
X
.
Progress in the treatment of advanced gastric cancer
.
Tumour Biol
.
2017
;
39
(
7
):
1010428317714626
.
8.
Petryszyn
P
,
Chapelle
N
,
Matysiak-Budnik
T
.
Gastric cancer: where are we heading
.
Dig Dis
.
2020
;
38
(
4
):
280
5
.
9.
Chow
A
,
Perica
K
,
Klebanoff
CA
,
Wolchok
JD
.
Clinical implications of T cell exhaustion for cancer immunotherapy
.
Nat Rev Clin Oncol
.
2022
;
19
(
12
):
775
90
.
10.
Zhang
Y
,
Zhang
Z
.
The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications
.
Cell Mol Immunol
.
2020
;
17
(
8
):
807
21
.
11.
Ayodele
O
,
Razak
ARA
.
Immunotherapy in soft-tissue sarcoma
.
Curr Oncol
.
2020
27
Suppl 1
17
23
.
12.
Kennedy
LB
,
Salama
AKS
.
A review of cancer immunotherapy toxicity
.
CA Cancer J Clin
.
2020
;
70
(
2
):
86
104
.
13.
Jadoon
Y
,
Siddiqui
MA
.
Immunotherapy in multiple myeloma
.
Cancer Treat Res Commun
.
2021
;
29
:
100468
.
14.
Reck
M
,
Remon
J
,
Hellmann
MD
.
First-line immunotherapy for non-small-cell lung cancer
.
J Clin Oncol
.
2022
;
40
(
6
):
586
97
.
15.
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
.
16.
Kole
C
,
Charalampakis
N
,
Tsakatikas
S
,
Kouris
NI
,
Papaxoinis
G
,
Karamouzis
MV
.
Immunotherapy for gastric cancer: a 2021 update
.
Immunotherapy
.
2022
;
14
(
1
):
41
64
.
17.
Kim
ST
,
Cristescu
R
,
Bass
AJ
,
Kim
KM
,
Odegaard
JI
,
Kim
K
.
Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer
.
Nat Med
.
2018
;
24
(
9
):
1449
58
.
18.
Janjigian
YY
,
Maron
SB
,
Chatila
WK
,
Millang
B
,
Chavan
SS
,
Alterman
C
.
First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial
.
Lancet Oncol
.
2020
;
21
(
6
):
821
31
.
19.
Fuchs
CS
,
Doi
T
,
Jang
RW
,
Muro
K
,
Satoh
T
,
Machado
M
.
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
.
20.
Bang
YJ
,
Kang
YK
,
Catenacci
DV
,
Muro
K
,
Fuchs
CS
,
Geva
R
.
Pembrolizumab alone or in combination with chemotherapy as first-line therapy for patients with advanced gastric or gastroesophageal junction adenocarcinoma: results from the phase II nonrandomized KEYNOTE-059 study
.
Gastric Cancer
.
2019
;
22
(
4
):
828
37
.
21.
Janjigian
YY
,
Shitara
K
,
Moehler
M
,
Garrido
M
,
Salman
P
,
Shen
L
.
First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial
.
Lancet
.
2021
;
398
(
10294
):
27
40
.
22.
Boku
N
,
Ryu
MH
,
Kato
K
,
Chung
HC
,
Minashi
K
,
Lee
KW
.
Safety and efficacy of nivolumab in combination with S-1/capecitabine plus oxaliplatin in patients with previously untreated, unresectable, advanced, or recurrent gastric/gastroesophageal junction cancer: interim results of a randomized, phase II trial (ATTRACTION-4)
.
Ann Oncol
.
2019
;
30
(
2
):
250
8
.
23.
Kang
YK
,
Chen
LT
,
Ryu
MH
,
Oh
DY
,
Oh
SC
,
Chung
HC
.
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
.
24.
Shitara
K
,
Van Cutsem
E
,
Bang
YJ
,
Fuchs
C
,
Wyrwicz
L
,
Lee
KW
.
Efficacy and safety of pembrolizumab or pembrolizumab plus chemotherapy versus chemotherapy alone for patients with first-line, advanced gastric cancer: the KEYNOTE-062 phase 3 randomized clinical trial
.
JAMA Oncol
.
2020
;
6
(
10
):
1571
80
.
25.
Weber
JS
,
Schadendorf
D
,
Del Vecchio
M
,
Larkin
J
,
Atkinson
V
,
Schenker
M
.
Adjuvant therapy of nivolumab combined with ipilimumab versus nivolumab alone in patients with resected stage IIIB-D or stage IV melanoma (CheckMate 915)
.
J Clin Oncol
.
2023
;
41
(
3
):
517
27
.
26.
Janjigian
YY
,
Kawazoe
A
,
Yañez
P
,
Li
N
,
Lonardi
S
,
Kolesnik
O
.
The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer
.
Nature
.
2021
;
600
(
7890
):
727
30
.
27.
Xu
J-M
,
Jiang
H
,
Pan
Y
,
Gu
K
,
Cang
S
,
Han
L
.
LBA53 Sintilimab plus chemotherapy (chemo) versus chemo as first-line treatment for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma (ORIENT-16): first results of a randomized, double-blind, phase III study
.
Ann Oncol
.
2021
32
S1331
.
28.
Arance
A
,
de la Cruz-Merino
L
,
Petrella
TM
,
Jamal
R
,
Ny
L
,
Carneiro
A
.
Phase II LEAP-004 study of lenvatinib plus pembrolizumab for melanoma with confirmed progression on a programmed cell death protein-1 or programmed death ligand 1 inhibitor given as monotherapy or in combination
.
J Clin Oncol
.
2023
;
41
(
1
):
75
85
.
29.
Kuo
HY
,
Guo
JC
,
Hsu
CH
.
Embracing anti-PD-1 immunotherapy in the treatment of esophageal cancer: implications of positive phase III trials in advanced and resected esophageal cancer
.
J Formos Med Assoc
.
2023
;
122
(
1
):
4
8
.
30.
Meric-Bernstam
F
,
Sweis
RF
,
Kasper
S
,
Hamid
O
,
Bhatia
S
,
Dummer
R
.
Combination of the STING agonist MIW815 (ADU-S100) and PD-1 inhibitor spartalizumab in advanced/metastatic solid tumors or lymphomas: an open-label, multicenter, phase ib study
.
Clin Cancer Res
.
2023
;
29
(
1
):
110
21
.
31.
Shitara
K
,
Ajani
JA
,
Moehler
M
,
Garrido
M
,
Gallardo
C
,
Shen
L
.
Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer
.
Nature
.
2022
;
603
(
7903
):
942
8
.