Skip to main content
Lippincott Open Access logoLink to Lippincott Open Access
. 2025 Jul 10;9:e2400907. doi: 10.1200/PO-24-00907

Immune Checkpoint Inhibitors in Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor–Resistant Chemotherapy-Naïve Advanced Non–Small Cell Lung Cancer: A Meta-Analysis Based on Eight Randomized Trials

Letian Huang 1, Shuling Zhang 1, Li Sun 1, Jietao Ma 1, Chengbo Han 1,
PMCID: PMC12262128  PMID: 40638873

Abstract

PURPOSE

The efficacy and safety of combination strategies involving immune checkpoint inhibitors (ICIs) in patients with advanced epidermal growth factor receptor (EGFR)–mutant non–small cell lung cancer (NSCLC) who have developed resistance to EGFR-tyrosine kinase inhibitors (TKIs) remains uncertain.

METHODS

We conducted a systematic review and meta-analysis of randomized controlled trials (RCTs) comparing ICIs combined with chemotherapy with or without antiangiogenic therapy (C/A) versus C/A alone in the treatment of advanced NSCLC after resistance to EGFR-TKIs. We searched databases, including PubMed, Cochrane Library, Embase, Web of Science, and meeting abstracts. Hazard ratios (HRs) and 95% CI for median overall survival (OS) and median progression-free survival (PFS) were calculated. Risk ratios (RRs) and 95% CI were used as indicators of objective response rate (ORR) and adverse events (AEs).

RESULTS

Eight RCTs involving 10 cohorts and 2,269 patients were included. Adding ICIs to C/A significantly improved PFS (HR, 0.67 [95% CI, 0.57 to 0.80]; P < .001), OS (HR, 0.89 [95% CI, 0.79 to 0.99]; P = .031), and ORR (RR, 0.80 [95% CI, 0.74 to 0.88]; P < .001) comparedwith C/A alone. Subgroup analyses showed that the benefits were more pronounced in patients with PD-L1 expression ≥50%, specific EGFR mutations (Leu858Arg), absence of Thr790Met mutation, and treatment with pemetrexed-platinum. No significant increase in grade 3 or higher AEs was observed, but rates of discontinuation and specific AEs (rash, hypothyroidism, and hypertension) were significantly higher in the ICI+C/A group.

CONCLUSION

This meta-analysis suggests that the addition of ICIs to C/A may improve survival outcomes in patients with advanced NSCLC after resistance to EGFR-TKIs, particularly in selected subpopulations such as those with high PD-L1 expression or specific EGFR mutations. However, careful monitoring for specific AEs is warranted.

INTRODUCTION

Lung cancer is the leading cause of cancer-related mortality worldwide.1,2 Non–small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases, with a 5-year survival rate of <10% in advanced stages.3,4 Targeted therapy with epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) has become a cornerstone treatment for NSCLC patients with EGFR-activating mutations. EGFR mutation is present in approximately 60%-80% of actionable genetic alterations (AGAs) found in patients with NSCLC.5-7 However, resistance to EGFR-TKIs typically develops within 1-2 years. At this stage, the standard of care (SOC)—platinum-based doublet chemotherapy with or without antivascular endothelial growth factor (VEGF) agents—shows limited efficacy, thereby underscoring the urgent need for more effective therapeutic strategies.8,9

In recent years, immune checkpoint inhibitors (ICIs) targeting PD-1 or its PD-L1, and cytotoxic T-lymphocyte antigen 4 have become the SOC for patients with advanced NSCLC, particularly for those without AGAs. These therapies have significantly prolonged the survival of this patient population.10,11 However, the efficacy of ICIs in patients with AGAs is generally limited, especially in patients with EGFR-mutant NSCLC during initial treatment.12 Notably, resistance to TKIs can alter the tumor immune microenvironment (TIME),13 potentially enhancing the efficacy of ICIs by increasing PD-L1 expression and tumor-infiltrating lymphocyte density.14-16 However, evidences regarding the impact of TKI resistance on ICI efficacy remains inconclusive.

Some randomized controlled trials (RCTs) have not shown a clear advantage of ICI monotherapy over chemotherapy in patients who have developed resistance to TKIs.14,17-19 However, the Checkmate 722 and KEYNOTE-789 studies suggested a trend toward improved progression-free survival (PFS) with ICI plus chemotherapy (chemoimmunotherapy) compared with chemotherapy alone in patients with EGFR-TKI–resistant NSCLC.20,21 The IMpower150 trial, which combined ICI with chemotherapy and anti-VEGF agents (ICI+CA), showed promising results for PFS and overall survival (OS) in patients with EGFR-TKI–resistant NSCLC.22,23 Similar benefits were observed in the ORIENT-31 trial.24 Recent clinical trials, including IMpower151, ATTLAS, and HARMONi-A, have shown mixed results regarding the efficacy of ICIs in combination regimens.25-27 ATTLAS and HARMONi-A demonstrated the beneficial effects of simultaneous blockade of immune checkpoints and VEGF pathways, whether used in combination or as two-in-one bispecific antibodies. However, IMpower151 was unable to replicate these findings.

Given these discrepancies, a meta-analysis focusing on prospective RCTs is warranted to assess the benefits of adding ICIs or ICIs plus anti-VEGF to the initial treatment after resistance to TKIs. This analysis aimed to identify subgroups of patients most likely to benefit, refine combination therapy strategies, and evaluate safety profiles to optimize treatment approaches for patients with EGFR-TKI–resistant NSCLC.

METHODS

Search Strategy

This meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidance (Data Supplement, Table S1). We conducted a comprehensive electronic search of the PubMed, Cochrane Library, Embase, and Web of Science databases to identify trials published on or before June 30, 2024. In addition, we reviewed conference abstracts from the World Conference on Lung Cancer, American Society of Clinical Oncology, and European Society for Medical Oncology. The search strategy used the following terms: “non-small cell lung cancer,” “NSCLC,” “epidermal growth factor receptor,” “EGFR,” “immune checkpoint inhibitors,” “ICI,” “PD-1,” “PD-L1,” “tyrosine kinase inhibitor,” “TKI,” “randomized controlled trial,” and “RCT.” Two authors independently screened titles, abstracts, and full-text articles.

Eligibility Criteria

The inclusion criteria for eligible trials were as follows: (1) RCT design; (2) enrollment of advanced NSCLC patients with positive EGFR mutation status who had previously received EGFR-TKI treatment and experienced disease progression; (3) treatment of the intervention arm with any PD-1/PD-L1 inhibitor in combination with other antitumor agents; (4) no treatment with PD-1/PD-L1 inhibitor in the control arm; (5) chemotherapy-naïve status and no administration of other antitumor therapies after disease progression on EGFR-TKI; and (6) inclusion of clinical outcomes such as PFS, OS, objective response rate (ORR), or adverse events (AEs). Exclusion criteria included review articles, case reports, opinions, editorials, duplicate publications, studies not related to the research topic, and studies with insufficient data for meta-analysis.

Data Collection and Quality Assessment

The study characteristics extracted included authors, year of publication, country, patient demographics, sample size, and end points. The end points included PFS, OS, ORR, and safety. The quality of the studies was assessed using the Cochrane Collaboration's tool to evaluate the risk of bias.28 Quality assessment criteria included the generation of the allocation sequence, concealment of allocation, blinding, use of intention-to-treat analysis, and the proportion of patients lost to follow-up. Two independent reviewers screened the studies, extracted the data, and performed the quality evaluations. Disagreements were resolved by discussion and consensus among the participating researchers.

Statistical Analysis

PFS and OS are expressed as hazard ratios (HRs) with 95% CIs. ORR and AEs were expressed as risk ratios (RRs) with 95% CIs. Publication bias was assessed using funnel plots, Begg's test, and Egger's test. All statistical tests were two-sided, and statistical significance was defined as P < .05. Heterogeneity between studies was evaluated using the Q test and the I2 statistic. An I2 statistic >50% or a P value <.10 indicated significant heterogeneity. A fixed-effects model was used when there was no significant heterogeneity between studies. In cases where significant heterogeneity was found, a random-effects model was used. The meta-analysis was performed using STATA software, version 12.0 (StataCorp, College Station, TX).

RESULTS

Study Selection and Characteristics

A total of 1,059 records were screened from databases and conference abstracts. After titles and abstracts review, 32 articles were identified for further evaluation. Finally, eight studies, comprising 10 cohorts and a total of 2,269 patients, met the eligibility criteria and were included in the meta-analysis (Fig 1). Among them, six cohorts compared the addition of ICI to SOC (platinum-based doublet chemotherapy ± anti-VEGF agents; ICI+SOC) versus SOC alone. Three cohorts compared the combination of ICI, anti-VEGF agents, and SOC (ICI + anti-VEGF + SOC) versus SOC alone. Anti-VEGF therapies were administered in five trials, including one trial that used ivonescimab, a bispecific antibody targeting both PD-1 and VEGF. Chemotherapy regimens administered included pemetrexed in five trials and paclitaxel in three trials. Detailed characteristics of the included trials are summarized in Table 1.

FIG 1.

FIG 1.

PRISMA flowchart outlining the process of study selection. PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses.

TABLE 1.

Characteristics of Included Studies

Study Authors Recruitment Period Sample Size Intervention Arm (n) Control Arm (n) East Asian (%) Age, Years (median, range) Female (%) Smoker (%) Median PFS (months) Median OS (months) ORR (%)
Checkmate 72220 Mok et al 2017-2020 294 Nivolumab + pemetrexed + platinum (144) Placebo + pemetrexed + platinum (150) 94.40 64 (29-80) 60.20 38.10 5.6
5.4
19.4
15.9
31%
27%
KEYNOTE-78921 Yang et al 2018-2023 492 Pembrolizumab + pemetrexed + platinum (245) Placebo + pemetrexed + platinum (247) 60.90 63 (34-87) 61.60 33.90 5.6
5.5
15.9
14.7
29%
27.1%
ORIENT-3124 Lu et al 2019-2022 476 (A) Sintilimab + IBI305 + pemetrexed + cisplatin (158) (C) Placebo + pemetrexed + cisplatin (160) 100 57 (51-65) 59.24 29.80 (A) 7.2
(B) 5.5
(C) 4.3
(A) 21.1
(B) 20.5
(C) 19.2
(A) 48.1%
(B) 34.8%
(C) 29.4%
(B) Sintilimab + pemetrexed + cisplatin (158)
IMpower15023 Reck et al 2015-2016 78 (A) Atezolizumab + bevacizumab + paclitaxel + carboplatin (22) (C) Placebo + bevacizumab + paclitaxel + carboplatin (28) 40.30 63 (31-82) 54.83 44.35 (A) 9.7
(B) 5.7
(C) 6.1
(A) 27.8
(B) 14.9
(C) 18.1
(A) 71%
(B) 36%
(C) 42%
(B) Atezolizumab + paclitaxel + carboplatin (28)
IMpower15125 Zhou et al 2020-2023 163 Atezolizumab + bevacizumab + pemetrexed (98%)/paclitaxel (2%) + carboplatin (81) Placebo + bevacizumab + pemetrexed (98%)/paclitaxel + carboplatin (82) 100 NA NA NA 8.5
8.3
NA NA
ATTLAS26 Park et al 2019-2022 212 Atezolizumab + bevacizumab + paclitaxel + carboplatin (144) Pemetrexed + platinum (68) 100 62 (31-90) 56.16 37.28 8.7
5.6
20.6
20.3
69.5%
41.9%
IMpower13029 West et al 2015-2017 44 Atezolizumab + nab-paclitaxel + carboplatin (32) Nab-paclitaxel + carboplatin (12) NA NA NA NA 7.0
6.0
14.4
10.0
NA
HARMONi-A27 Zhang et al 2022-2022 322 Ivonescimab + pemetrexed + carboplatin (161) Placebo + pemetrexed + carboplatin (161) 100 59 (32-75) 51.55 29.50 7.0
4.8
17.1
14.5
50.6%
35.4%

Abbreviations: NA, not available; ORR, objective response rate; OS, overall survival; PFS, progression-free survival.

PFS Comparison Between ICI+C/A and C/A

Ten cohorts from eight trials, comprising 2,269 patients, were analyzed to compare PFS between ICI+C/A and C/A. The ICI+C/A group showed a significant PFS benefit over the C/A group, with a pooled HR of 0.67 (95% CI, 0.57 to 0.80, P < .001; Figs 2A and 2B). Significant heterogeneity was observed among the studies (I2 = 61.2%, P = .006; Figs 2A and 2B).

FIG 2.

FIG 2.

Forest plots of HR for PFS between ICI+C/A and C/A: (A) stratified by the addition of ICI or ICI+anti-VEGF; and (B) stratified by chemotherapeutic agents. Forest plots of HR for OS between ICI+C/A and C/A: (C) stratified by the addition of ICI or ICI+anti-VEGF; and (D) stratified by chemotherapeutic agents. Forest plots of RR for ORR between ICI+C/A and C/A: (E) stratified by the addition of ICI or ICI+anti-VEGF; and (F) stratified by chemotherapeutic agents. C/A, chemotherapy and/or antiangiogenic therapy; DL, DerSimonian-Laird; HR, hazard ratio; ICIs, immune checkpoint inhibitors; IV, Inverse-Variance; MH, Mantel-Haenszel; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; RR, risk ratio; SOC, standard of care; VEGF, vascular endothelial growth factor.

PFS Benefit of ICI+SOC Subgroup and ICI+Anti-VEGF+SOC Subgroup

In subgroup analyses, the ICI+SOC group demonstrated a significant PFS benefit over the SOC group, with a pooled HR of 0.76 (95% CI, 0.67 to 0.86, P < .001; Fig 2A), and the ICI+anti-VEGF+SOC group also demonstrated a significant PFS benefit over the SOC group, with a pooled HR of 0.51 (95% CI, 0.43 to 0.61, P < .001; Fig 2A).

PFS Benefit of ICI With Different Chemotherapeutic Agents

In the pemetrexed-platinum subgroup, the ICI+C/A group showed a significant PFS benefit compared with the C/A group, with a pooled HR of 0.67 (95% CI, 0.55 to 0.81, P < .001; Fig 2B). By contrast, no significant differences were observed between the groups in the paclitaxel-platinum subgroup, with a pooled HR of 0.70 (95% CI, 0.46 to 1.06, P = .094; Fig 2B). Notably, the point estimates for both chemotherapy types were comparable, and the interaction test for subgroup differences was nonsignificant (P = .73). Additionally, heterogeneity across studies was moderate (I2 = 61.2%).

PFS Benefit by Patient Characteristics

On the basis of different clinical characteristics, we performed additional subgroup analyses, including age, sex, Eastern Cooperative Oncology Group performance status (ECOG PS), smoking history, presence of brain or liver metastases, type of EGFR mutation, presence of the T790M mutation, PD-L1 expression levels, and previous lines of EGFR-TKI therapy. We found that ICI combination therapy provided a significant PFS benefit over non-ICI treatment, regardless of age, sex, ECOG PS, smoking status, or brain metastases. However, in subgroups with PD-L1 expression <1%, liver metastases, EGFR exon 19 deletion, presence of T790M mutation, and two previous lines of EGFR-TKIs, the addition of ICI did not confer a PFS benefit. By contrast, in subgroups with PD-L1 expression ≥50%, EGFR Leu858Arg mutation, absence of T790M mutation, and one previous line of EGFR-TKIs, a significant PFS benefit was observed with ICI+C/A compared with C/A (Fig 3).

FIG 3.

FIG 3.

Subgroup forest plots of HR for PFS between ICI+C/A and C/A groups. C/A, chemotherapy and/or anti-angiogenic therapy; ECOG PS, Eastern Cooperative Oncology Group performance status; EGFR, epidermal growth factor receptor; HR, hazard ratio; ICI, immune checkpoint inhibitors; PFS, progression-free survival; TKI, tyrosine kinase inhibitor.

OS Comparison Between ICI+C/A and C/A

Nine cohorts from seven trials, comprising 2,106 patients, were analyzed to compare OS between ICI+C/A and C/A. The ICI+C/A group showed a significant OS benefit over the C/A group, with a pooled HR of 0.89 (95% CI, 0.79 to 0.99, P = .031; Figs 2C and 2D). No significant heterogeneity was observed between the studies (I2 = 0.00%, P = .900; Figs 2C and 2D).

OS Benefit of ICI+SOC Subgroup and ICI+Anti-VEGF+SOC Subgroup

In subgroup analyses, the ICI+SOC group demonstrated a significant OS benefit over the SOC alone group, with a pooled HR of 0.86 (95% CI, 0.75 to 0.99, P = .032; Fig 2C). By contrast, the ICI+anti-VEGF+SOC group showed a trend toward improved OS over the SOC group, but the difference was not statistically significant (HR, 0.91 [95% CI, 0.75 to 1.10]; P = .315; Fig 2C).

OS Benefit of ICI With Different Chemotherapeutic Agents

In the pemetrexed subgroup, the ICI+C/A group showed significantly better OS than the C/A group, with a pooled HR of 0.87 (95% CI, 0.77 to 0.98, P = .021; Fig 2D). By contrast, there was no significant difference between the two groups in the paclitaxel subgroup, with a pooled HR of 0.99 (95% CI, 0.75 to 1.30, P = .917; Fig 2D).

ORR Comparison Between ICI+C/A and C/A

Eight cohorts from six trials, comprising 2,062 patients, were analyzed to compare the ORR between ICI+C/A and C/A. The ICI+C/A group showed significantly better ORR than the C/A group, with a pooled risk ratio (RR) of 0.80 (95% CI, 0.74 to 0.88, P < .001; Figs 2E and 2F). No heterogeneity was observed between the studies (I2 = 47.6%, P = .064; Figs 2E and 2F).

ORR Benefit of ICI+SOC Subgroup and ICI+Anti-VEGF+SOC Subgroup

In subgroup analyses, the ICI+SOC group demonstrated no statistically significant ORR benefit over the SOC group, with a pooled HR of 0.89 (95% CI, 0.79 to 1.01, P = .075; Fig 2E), while the ICI+anti-VEGF+SOC group demonstrated a significant ORR benefit over the SOC group, with a pooled HR of 0.70 (95% CI, 0.62 to 0.79, P < .001; Fig 2E).

ORR Benefit of ICI With Different Chemotherapeutic Agents

In the pemetrexed subgroup, the ICI+C/A group showed a significantly higher ORR than the C/A group, with a pooled RR of 0.82 (95% CI, 0.72 to 0.94, P = .004; Fig 2F). By contrast, in the paclitaxel subgroup, there was no statistically significant difference in ORR between the two groups, with a pooled RR of 0.73 (95% CI, 0.49 to 1.11, P = .140; Fig 2F).

Safety

To evaluate safety profiles, treatment-emergent AEs (TEAEs), treatment-related AEs (TRAEs), grade ≥3 TEAEs or TRAEs, death-related TEAEs or TRAEs, TEAEs or TRAEs leading to discontinuation, and specific AEs (anemia, rash, hypothyroidism, and hypertension) were included. The risks of specific AEs were significantly higher in the ICI+C/A group compared with the C/A group, including rash (RR, 1.41 [95% CI, 1.23 to 1.62]; P < .001), hypothyroidism (RR, 1.62 [95% CI, 1.46 to 1.80]; P < .001), hypertension (RR, 1.57 [95% CI, 1.30 to 1.90]; P < .001), TEAEs leading to discontinuation (RR, 1.42 [95% CI, 1.20 to 1.69]; P < .001), and TRAEs leading to discontinuation (RR, 1.45 [95% CI, 1.31 to 1.61]; P < .001). No significant differences were observed between the two groups for all TEAEs (P = .763), grade ≥3 TEAEs (P = .484), death-related TEAEs (P = .843), all TRAEs (P = .174), grade ≥3 TRAEs (P = .089), death-related TRAEs (P = .559), or anemia (P = .463; Table 2).

TABLE 2.

Meta-Analysis of Adverse Events

Adverse Events ICI+C/A v C/A
No. of Studies No. of Patients Main Effect Heterogeneity
ICI+C/A C/A RR (95% CI) P I2 (%) P Model
All TEAEs 5 874 801 1.10 (0.60 to 2.02) .763 69.10 .012 Random
TEAEs ≥grade 3 5 874 801 1.06 (0.89 to 1.27) .484 73.00 .005 Random
TEAEs leading to discontinuation 3 475 481 1.42 (1.20 to 1.69) <.001 0.00 .540 Fixed
TEAEs related to death 4 720 727 1.04 (0.73 to 1.46) .843 39.60 .174 Random
All TRAEs 5 854 783 1.24 (0.91 to 1.71) .174 50.60 .088 Random
TRAEs ≥grade 3 5 854 783 1.15 (0.98 to 1.36) .089 70.20 .009 Random
TRAEs leading to discontinuation 5 854 783 1.45 (1.31 to 1.61) <.001 49.20 .069 Fixed
TRAEs related to death 5 854 783 1.13 (0.75 to 1.69) .559 0.00 .648 Fixed
Rash 3 468 394 1.41 (1.23 to 1.62) <.001 0.00 .948 Fixed
Hypothyroidism 4 713 640 1.62 (1.46 to 1.80) <.001 0.00 .571 Fixed
Anemia 5 854 783 1.04 (0.94 to 1.16) .408 0.00 .591 Fixed
Hypertension 3 468 394 1.57 (1.30 to 1.90) <.001 62.30 .07 Random

Abbreviations: C/A, chemotherapy with or without antiangiogenic therapy; ICIs, immune checkpoint inhibitors; RR, risk ratio; TEAEs, treatment-emergent adverse events; TRAEs, treatment-related adverse events.

Study Quality and Publication Bias

The risk of bias assessment for the included RCTs was low, indicating that all studies were of high quality (Data Supplement, Table S2). Sensitivity analyses for PFS and OS were performed to assess heterogeneity, and no significant heterogeneity was detected in PFS or OS across the studies (Data Supplement, Figs S1 and S2). To minimize publication bias, strict inclusion criteria were applied to the selected studies, and several methods were used to detect publication bias. Visual inspection of funnel plots did not reveal any significant asymmetry. Additionally, both the Egger linear regression test and Begg rank-correlation test showed no evidence of publication bias (Data Supplement, Figs S3 and S4).

DISCUSSION

To our knowledge, this comprehensive meta-analysis provides the first large-scale evidence demonstrating potential survival benefits of ICI-based regimens in patients with EGFR-mutant NSCLC who have developed resistance to EGFR-TKIs. Unlike previous meta-analyses,30,31 which included a broader range of study designs, our analysis strictly focused on RCTs that specifically evaluated initial treatment strategies after resistance to EGFR-TKIs. The majority of trials included in this analysis provided updated data from the past 2 years, ensuring the relevance and timeliness of our findings. Our results demonstrate that the addition of ICIs to chemotherapy (with or without antiangiogenic agents) significantly improves PFS and OS in EGFR-TKI–resistant NSCLC, despite inconsistent OS benefits in individual RCTs.

The optimal combination strategy for ICIs was investigated in this meta-analysis. Typically, ICI monotherapy was associated with inferior PFS, OS, and ORR compared with chemotherapy in EGFR-mutant NSCLC, regardless of previous EGFR-TKI exposure.30,32,33 Therefore, ICI monotherapy is not recommended for these patients. Notably, even in EGFR-mutant patients with high PD-L1 expression, the efficacy of ICI monotherapy is often suboptimal, likely because of poor infiltration of CD8+ T cells and low tumor mutation burden,34 highlighting the limited predictive value of PD-L1 in this population. Chemoimmunotherapy appears effective in overcoming this limitation, with our analysis demonstrating superior efficacy over chemotherapy alone. Preclinical studies suggest this synergy arises from mechanisms such as TIME remodeling, PD-L1 upregulation, immunogenic cell death, neoantigen load increase, and CD8+ T-cell functional restoration.35

In trials like ORIENT-31 and IMpower151, adding antiangiogenic agents to chemoimmunotherapy prolonged PFS. In the subgroup analysis of our meta-analysis, ICI + antiVEGF + SOC demonstrated superior efficacy compared with SOC alone, with significantly improved PFS (HR, 0.51 v HR, 0.76 for ICI + SOC) and ORR benefits. By contrast, ICI + SOC showed no significant ORR advantage over SOC. Our findings suggest that the combination of anti-VEGF agents with ICIs exerts a more pronounced effect on tumor response. Notably, the phase III APPLE trial observed a PFS benefit (9.7 v 5.8 months, HR, 0.67) and a trend toward OS improvement with adding bevacizumab to chemoimmunotherapy in EGFR-TKI-resistant NSCLC.36 Mechanistically, antiangiogenic agents facilitate the enhancement of antigen-specific T-cell trafficking into the tumor microenvironment through the inhibition of VEGF. This modulation not only reduces the presence of immunosuppressive macrophages and monocytes but also potentiates T-cell–mediated cytotoxicity, thereby enhancing the ability of the immune system to target and eliminate cancer cells.37 These findings suggest that, for patients with EGFR-TKI–resistant NSCLC, the synergistic use of ICIs in combination with chemotherapy and antiangiogenic therapies is a particularly promising strategy. This approach appears to be particularly beneficial in scenarios where the tumor burden is significantly elevated or requires rapid reduction.

The choice of chemotherapy backbone (pemetrexed v paclitaxel) in combination with ICIs remains a topic of clinical interest. Although pemetrexed-based regimens showed statistically significant improvements in PFS, OS, and ORR compared with C/A alone, the apparent advantage over paclitaxel should be interpreted cautiously. The HR point estimates for both chemotherapy types were similar (pemetrexed HR, 0.67 v paclitaxel HR, 0.70), and the overlapping confidence intervals, coupled with a nonsignificant interaction test (P = .73), indicate no conclusive evidence of superiority for either regimen. The nonsignificant result for paclitaxel may reflect limited statistical power because of smaller sample sizes in this subgroup (three trials, n = 228 v five trials, n = 1,252 for pemetrexed) rather than a true lack of efficacy.

These findings align with the established role of pemetrexed in lung adenocarcinoma—the predominant histology for EGFR-mutant NSCLC—where its mechanism of action and toxicity profile may synergize better with immunotherapy.38,39 However, no definitive conclusions can be drawn regarding preferential use of pemetrexed over paclitaxel without direct comparative trials. Future studies should prioritize head-to-head comparisons of chemotherapy backbones in ICI-based regimens, with standardized stratification for PD-L1 expression, EGFR mutation subtypes, and previous therapy lines to minimize confounding.

The efficacy of ICIs is modulated by several factors, including PD-L1 expression level, age, ECOG performance status, metastatic sites, and EGFR mutation status.40 In our meta-analysis, stratified assessments of baseline characteristics revealed a notable PFS benefit associated with ICI supplementation, independent of age, sex, ECOG performance status, smoking history, or the presence of brain metastases. However, subgroups that did not show additional benefits from ICIs included those with PD-L1 expression below 1%, those with EGFR exon 19 deletions, T790M mutations, or those who had received two previous EGFR-TKI therapies. Interestingly, patients harboring EGFR L858R mutations refractory to EGFR-TKIs benefited from the ICI+C/A regimen, in contrast to the lack of benefit observed in patients with EGFR exon 19 deletions, suggesting potential differences in the TIME influenced by different EGFR mutations. In addition, as the number of EGFR-TKI lines increases, the subsequent efficacy of ICIs may decrease, potentially explaining the divergent results in relevant RCTs, such as ATTLAS, ORIENT-31, and IMpower151, all of which evaluated the efficacy of ICI+CA. Although ATTLAS and ORIENT-31 showed significant improvement in PFS, IMpower151 did not show comparable results. In particular, none of the three studies showed a PFS benefit in the subgroup of patients who received sequential first- or second-generation EGFR-TKIs, followed by third-generation EGFR-TKI therapy. Conversely, patients treated with single-agent EGFR-TKIs showed a PFS benefit in ATTLAS and ORIENT-31. The difference in the proportion of patients receiving single-line EGFR-TKI therapy (46.1% in IMpower151 v 65.3% in ATTLAS and 73.8% in ORIENT-31) may have contributed to the divergent results of IMpower151.23,25,26 However, the underlying mechanisms remain unclear and require further investigation.

In terms of safety profiles, the integration of ICIs did not significantly increase the overall TEAEs/TRAEs incidence, grade ≥3 TEAEs/TRAEs, or mortality. However, the ICI+C/A cohort showed higher rates of treatment discontinuation, hypertension, immune-mediated dermatologic reactions, and hypothyroidism, aligning with previous ICI RCTs. Hypertension may be closely associated with concomitant use of antiangiogenic agents. Therefore, it is imperative to evaluate the safety profile of ICIs in combination with antiangiogenic therapies, considering the unique characteristics of patients with resistance to EGFR-TKIs. Previous studies have found that the combination of EGFR-TKIs and ICIs increases the incidence of certain AEs, such as liver injury and interstitial lung disease (ILD).41 Although sequential EGFR-TKI and ICI use in included studies did not elevate ILD incidence (eg, ORIENT-31: 1%-3% across arms), limited reporting precluded pooled ILD analysis.

Despite the focus of our study on evaluating the efficacy of ICI-based therapeutic approaches in patients with EGFR-TKI–resistant NSCLC, management of these patients remains a major clinical challenge. The emergence of novel agents and evolving treatment paradigms in this setting is encouraging. A recent clinical trial evaluating the combination of amivantamab with chemotherapy, either alone or in combination with lazertinib, showed promising results, demonstrating a significant improvement in PFS compared with chemotherapy alone.42 However, this regimen was associated with a significantly increased incidence of TRAEs, particularly grade ≥3 TRAEs.42 In addition, antibody-drug conjugates targeting TROP-2 and HER-3 have shown encouraging potential in early clinical trials.43,44 The resulting challenge for health care providers is to refine and tailor treatment strategies with these innovative agents and ICIs to individual patient characteristics to maximize therapeutic benefits while mitigating AEs.

This study has several limitations. Primarily, efficacy within the PD-L1 expression range of 1%-50% remains equivocal because of insufficient stratified RCT data. Further investigation of factors, such as tumor mutational burden, microsatellite instability, TP53 comutations, and KRAS mutation status, is required. Second, our meta-analysis had a low representation of patients with NSCLC treated with third-generation EGFR-TKIs as the first-line therapy, a scenario that has become increasingly common in recent years. Among the RCTs reviewed, the HARMONi-A trial uniquely evaluated osimertinib and showed tentative PFS benefits and a suggestive OS improvement with the addition of ICI after osimertinib resistance. However, the OS data from this trial remain immature, and updated RCTs are needed to confirm the benefits of ICIs in this subgroup. Finally, different trials used different ICIs, including multiple PD-L1 and PD-1 inhibitors, increasing the possibility of data bias and heterogeneity in the results.

In conclusion, this meta-analysis suggests that the addition of ICIs to C/A may improve survival outcomes in patients with advanced EGFR-mutant NSCLC who have developed resistance to EGFR-TKIs. The observed benefits were particularly pronounced in specific subgroups, including patients with PD-L1 expression ≥50%, EGFR L858R mutation, absence of T790M mutation, and limited previous EGFR-TKI exposure (≤1 line). Regarding the choice of chemotherapy regimens, pemetrexed-based treatments appeared to show improvements compared with C/A alone, but there is no conclusive evidence of superiority over paclitaxel-based regimens because of similar overall effects and potential limitations in sample size. Despite these advances, limitations such as patient selection bias, tumor heterogeneity, and variability in PD-1/PD-L1 inhibitors may influence outcomes. Future studies must validate these findings in diverse populations. Additionally, mechanistic investigations are needed to elucidate how EGFR mutations modulate immunotherapy responsiveness.

Notably, the decision to use ICI + C/A as the next line of therapy should be carefully considered. Subsequent rebiopsy and sequencing after EGFR-TKI therapy often reveals a heterogeneous molecular phenotype, including MET amplification, new KRAS mutations, and SCLC transformation, which may respond better to specific targeted therapies or other treatment strategies. Therefore, it is essential to perform comprehensive genomic profiling after progression on EGFR-TKIs to identify potential targetable mutations or transformations.

In summary, although this meta-analysis supports the potential benefit of adding ICIs to C/A particularly in certain subpopulations of EGFR-TKI–resistant patients, it does not provide sufficient evidence to recommend this combination as a preferred or priority strategy for all patients after EGFR-TKI resistance. Personalized treatment strategies on the basis of detailed molecular characterization and biomarker assessment are crucial for optimizing outcomes in these patients.

DISCLAIMER

This is a US Government work. There are no restrictions on its use.

SUPPORT

Supported by China Health & Medical Development Foundation, and the award number is CLJH-2400401 and 345 Talent Project of Shengjing Hospital of China Medical University.

*

J.M. and C.H. contributed equally to this work.

DATA SHARING STATEMENT

A data sharing statement provided by the authors is available with this article at DOI https://doi.org/10.1200/PO-24-00907

All data analyzed in this study are available in this published article and supplementary material.

AUTHOR CONTRIBUTIONS

Conception and design: Letian Huang, Jietao Ma, Chengbo Han

Financial support: Letian Huang

Collection and assembly of data: Letian Huang, Shuling Zhang, Li Sun, Chengbo Han

Data analysis and interpretation: All authors

Manuscript writing: All authors

Final approval of manuscript: All authors

Accountable for all aspects of the work: All authors

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.

Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

No potential conflicts of interest were reported.

REFERENCES

  • 1.International Agency for Research on Cancer . Global cancer observatory. Cancer Today [DB/OL]. 2025. http://gco.iarc.who.int/today [Google Scholar]
  • 2.Bray F, Laversanne M, Sung H, et al. : Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 74:229-263, 2024 [DOI] [PubMed] [Google Scholar]
  • 3.Thai AA, Solomon BJ, Sequist LV, et al. : Lung cancer. Lancet 398:535-554, 2021 [DOI] [PubMed] [Google Scholar]
  • 4.Duma N, Santana-Davila R, Molina JR: Non-small cell lung cancer: Epidemiology, screening, diagnosis, and treatment. Mayo Clin Proc 94:1623-1640, 2019 [DOI] [PubMed] [Google Scholar]
  • 5.Mitsudomi T, Morita S, Yatabe Y, et al. : Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): An open label, randomised phase 3 trial. Lancet Oncol 11:121-128, 2010 [DOI] [PubMed] [Google Scholar]
  • 6.Maemondo M, Inoue A, Kobayashi K, et al. : Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 362:2380-2388, 2010 [DOI] [PubMed] [Google Scholar]
  • 7.Ramalingam SS, Vansteenkiste J, Planchard D, et al. : Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med 382:41-50, 2020 [DOI] [PubMed] [Google Scholar]
  • 8.Sequist LV, Waltman BA, Dias-Santagata D, et al. : Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 3:75ra26, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Soria JC, Ohe Y, Vansteenkiste J, et al. : Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med 378:113-125, 2018 [DOI] [PubMed] [Google Scholar]
  • 10.Mok TSK, Wu YL, Kudaba I, et al. : Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 393:1819-1830, 2019 [DOI] [PubMed] [Google Scholar]
  • 11.Diker O, Garassino MC: Pembrolizumab plus chemotherapy in lung cancer. N Engl J Med 379:e18, 2018 [DOI] [PubMed] [Google Scholar]
  • 12.Lee CK, Man J, Lord S, et al. : Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-A meta-analysis. J Thorac Oncol 12:403-407, 2017 [DOI] [PubMed] [Google Scholar]
  • 13.Chen N, Fang W, Zhan J, et al. : Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: Implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol 10:910-923, 2015 [DOI] [PubMed] [Google Scholar]
  • 14.Haratani K, Hayashi H, Tanaka T, et al. : Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann Oncol 28:1532-1539, 2017 [DOI] [PubMed] [Google Scholar]
  • 15.Isomoto K, Haratani K, Hayashi H, et al. : Impact of EGFR-TKI treatment on the tumor immune microenvironment in EGFR mutation-positive non-small cell lung cancer. Clin Cancer Res 26:2037-2046, 2020 [DOI] [PubMed] [Google Scholar]
  • 16.Hata A, Katakami N, Nanjo S, et al. : Programmed death-ligand 1 expression and T790M status in EGFR-mutant non-small cell lung cancer. Lung Cancer 111:182-189, 2017 [DOI] [PubMed] [Google Scholar]
  • 17.Borghaei H, Paz-Ares L, Horn L, et al. : Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 373:1627-1639, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Herbst RS, Baas P, Kim DW, et al. : Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 387:1540-1550, 2016 [DOI] [PubMed] [Google Scholar]
  • 19.Kato R, Hayashi H, Chiba Y, et al. : Propensity score-weighted analysis of chemotherapy after PD-1 inhibitors versus chemotherapy alone in patients with non-small cell lung cancer (WJOG10217L). J Immunother Cancer 8:e000350, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mok T, Nakagawa K, Park K, et al. : Nivolumab plus chemotherapy in epidermal growth factor receptor-mutated metastatic non-small-cell lung cancer after disease progression on epidermal growth factor receptor tyrosine kinase inhibitors: Final results of CheckMate 722. J Clin Oncol 42:1252-1264, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yang JC, Lee DH, Lee JS, et al. : Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: Phase 3 KEYNOTE-789 study. J Clin Oncol 42:4029-4039, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hegde PS, Wallin JJ, Mancao C: Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics. Semin Cancer Biol 52:117-124, 2018 [DOI] [PubMed] [Google Scholar]
  • 23.Nogami N, Barlesi F, Socinski MA, et al. : IMpower150 final exploratory analyses for atezolizumab plus bevacizumab and chemotherapy in key NSCLC patient subgroups with EGFR mutations or metastases in the liver or brain. J Thorac Oncol 17:309-323, 2022 [DOI] [PubMed] [Google Scholar]
  • 24.Lu S, Wu L, Jian H, et al. : Sintilimab plus chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer with disease progression after EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): Second interim analysis from a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Respir Med 11:624-636, 2023 [DOI] [PubMed] [Google Scholar]
  • 25.Zhou C, Dong X, Chen G, et al. : OA09.06 IMpower151: Phase III study of atezolizumab + bevacizumab + chemotherapy in 1L metastatic nonsquamous NSCLC. J Thorac Oncol 18:S64-S65, 2023 [Google Scholar]
  • 26.Park S, Kim TM, Han JY, et al. : Phase III, randomized study of atezolizumab plus bevacizumab and chemotherapy in patients with EGFR- or ALK-mutated non-small-cell lung cancer (ATTLAS, KCSG-LU19-04). J Clin Oncol 42:1241-1251, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.HARMONi-A Study Investigators, Fang W, Zhao Y, et al. : Ivonescimab plus chemotherapy in non-small cell lung cancer with EGFR variant: A randomized clinical trial. JAMA 332:561-570, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Higgins J, Green S, et al. : Cochrane Handbook for Systematic Reviews of Interventions. Hoboken, NJ: John Wiley & Sons; 2011 [Google Scholar]
  • 29.West H, McCleod M, Hussein M, et al. : Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 20:924-937, 2019 [DOI] [PubMed] [Google Scholar]
  • 30.Qian X, Guo X, Li T, et al. : Efficacy of immune checkpoint inhibitors in EGFR-mutant NSCLC patients with EGFR-TKI resistance: A systematic review and meta-analysis. Front Pharmacol 13:926890, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Qin BD, Jiao XD, Yuan LY, et al. : Immunotherapy-based regimens for patients with EGFR-mutated non-small cell lung cancer who progressed on EGFR-TKI therapy. J Immunother Cancer 12:e008818, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hayashi H, Sugawara S, Fukuda Y, et al. : A randomized phase II study comparing nivolumab with carboplatin-pemetrexed for EGFR-mutated NSCLC with resistance to EGFR tyrosine kinase inhibitors (WJOG8515L). Clin Cancer Res 28:893-902, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dietel M, Savelov N, Salanova R, et al. : Real-world prevalence of programmed death ligand 1 expression in locally advanced or metastatic non-small-cell lung cancer: The global, multicenter EXPRESS study. Lung Cancer 134:174-179, 2019 [DOI] [PubMed] [Google Scholar]
  • 34.Zhou J, Yu X, Hou L, et al. : Epidermal growth factor receptor tyrosine kinase inhibitor remodels tumor microenvironment by upregulating LAG-3 in advanced non-small-cell lung cancer. Lung Cancer 153:143-149, 2021 [DOI] [PubMed] [Google Scholar]
  • 35.Leonetti A, Wever B, Mazzaschi G, et al. : Molecular basis and rationale for combining immune checkpoint inhibitors with chemotherapy in non-small cell lung cancer. Drug Resist Updat 46:100644, 2019 [DOI] [PubMed] [Google Scholar]
  • 36.Shiraishi Y, Kishimoto J, Sugawara S, et al. : Atezolizumab and platinum plus pemetrexed with or without bevacizumab for metastatic nonsquamous non-small cell lung cancer: A phase 3 randomized clinical trial. JAMA Oncol 10:315-324, 2024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chen DS, Mellman I: Oncology meets immunology: The cancer-immunity cycle. Immunity 39:1-10, 2013 [DOI] [PubMed] [Google Scholar]
  • 38.Grønberg BH, Bremnes RM, Fløtten Ø, et al. : Phase III study by the Norwegian lung cancer study group: Pemetrexed plus carboplatin compared with gemcitabine plus carboplatin as first-line chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 27:3217-3224, 2009 [DOI] [PubMed] [Google Scholar]
  • 39.Chapman AM, Sun KY, Ruestow P, et al. : Lung cancer mutation profile of EGFR, ALK, and KRAS: Meta-analysis and comparison of never and ever smokers. Lung Cancer 102:122-134, 2016 [DOI] [PubMed] [Google Scholar]
  • 40.Xu Y, Wang Q, Xie J, et al. : The predictive value of clinical and molecular characteristics or immunotherapy in non-small cell lung cancer: A meta-analysis of randomized controlled trials. Front Oncol 11:732214, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Latif H, Liu SV: Combining immunotherapy and epidermal growth factor receptor kinase inhibitors: Worth the risk? Ann Transl Med 7:S76, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Passaro A, Wang J, Wang Y, et al. : Amivantamab plus chemotherapy with and without lazertinib in EGFR-mutant advanced NSCLC after disease progression on osimertinib: Primary results from the phase III MARIPOSA-2 study. Ann Oncol 35:77-90, 2024 [DOI] [PubMed] [Google Scholar]
  • 43.Shimizu T, Sands J, Yoh K, et al. : First-in-Human, phase I dose-escalation and dose-expansion study of trophoblast cell-surface antigen 2-directed antibody-drug conjugate datopotamab deruxtecan in non-small-cell lung cancer: TROPION-PanTumor01. J Clin Oncol 41:4678-4687, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jänne PA, Baik C, Su WC, et al. : Efficacy and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated non-small cell lung cancer. Cancer Discov 12:74-89, 2022 [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.

Data Availability Statement

A data sharing statement provided by the authors is available with this article at DOI https://doi.org/10.1200/PO-24-00907

All data analyzed in this study are available in this published article and supplementary material.


Articles from JCO Precision Oncology are provided here courtesy of Wolters Kluwer Health

RESOURCES