EGFR mutations in patients with lung adenocarcinoma and malignant pleural effusion: a propensity score-matched analysis of a single-center database
Original Article

EGFR mutations in patients with lung adenocarcinoma and malignant pleural effusion: a propensity score-matched analysis of a single-center database

Qiwei Yang1, Ziyi Wang1, Qiang Fu1, Xiaohai Hu1, Liang Chen1, Weiyang Chen1, Ling Lv1, Zhenghua Liu1, Wanfu Men1, Danni Li2, Wenya Li1

1Department of Thorax, The First Hospital of China Medical University, Shenyang, China; 2Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China

Contributions: (I) Conception and design: W Li, D Li, Q Yang; (II) Administrative support: W Li; (III) Provision of study materials or patients: W Li, D Li; (IV) Collection and assembly of data: Q Yang, Z Wang, Q Fu, X Hu, L Chen, W Chen; (V) Data analysis and interpretation: Q Yang, L Lv, Z Liu, W Men; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Wenya Li, PhD. Department of Thorax, The First Hospital of China Medical University, No. 210 Baita 1st Street, Hunnan District, Shenyang 110001, China. Email: saint5288@hotmail.com; Danni Li, PhD, MD. Department of Medical Oncology, The First Hospital of China Medical University, No. 210 Baita 1st Street, Hunnan District, Shenyang 110001, China. Email: dnli@cmu.edu.cn.

Background: Malignant pleural effusion (MPE) is associated with poor prognosis in patients with advanced lung adenocarcinoma (LUAD), and abnormal activation of epidermal growth factor receptor (EGFR) plays a crucial role in the development of LUAD. This study aimed to investigate the correlation between EGFR mutations and the occurrence of MPE in patients with LUAD and evaluate the effect of EGFR mutations on the prognosis of patients with LUAD with MPE.

Methods: A case-control study design was adopted that included patients pathologically diagnosed with LUAD. Clinical data were collected, and patients were divided into the MPE group and the non-MPE (N-MPE) group based on the presence of MPE. Propensity score matching (PSM) was used to control for confounding factors. The correlation between EGFR mutations and the occurrence of MPE in LUAD was initially examined. Additionally, various factors affecting the overall survival (OS) of patients with LUAD and MPE were evaluated.

Results: A total of 849 patients were included in the study. After 1:2 PSM, there were 180 patients in the MPE group and 360 in the N-MPE group. The EGFR mutation rate was significantly higher in the MPE group compared to the N-MPE group [62.7% vs. 50.2%; odds ratio (OR) =1.668; P=0.006]. This difference was primarily attributed to the T790M mutation (8.3% vs. 1.3%; OR =8.015; P<0.001), but no significant differences observed in other mutation sites between the groups. Further evaluation of factors affecting OS in patients with LUAD and MPE revealed that EGFR mutation was an independent protective factor for OS [hazard ratio (HR) 0.662, 95% CI: 0.456–0.962; P=0.03]. For patients with LUAD, MPE, and EGFR mutations, treatment with third-generation EGFR-tyrosine kinase inhibitors (TKIs) alone (HR 0.466, 95% CI: 0.233–0.930; P=0.03) or sequential first- and third-generation EGFR-TKIs (HR 0.385, 95% CI: 0.219–0.676; P=0.001) was associated with better median OS compared to first-generation EGFR-TKIs alone (first-generation EGFR-TKIs: 35 months, 95% CI: 28.4–41.6; third-generation EGFR-TKIs: 50 months, 95% CI: 37.3–62.7; sequential first- and third-generation EGFR-TKIs: 51 months, 95% CI: 45.6–56.4; P<0.001).

Conclusions: This study found there to be a positive correlation between EGFR mutations, particularly the T790M mutation, and MPE in patients with LUAD. EGFR mutation was associated with improved OS in patients with LUAD and MPE. For patients with LUAD, MPE, and EGFR mutations, sequential treatment with first- and third-generation EGFR-TKIs or third-generation EGFR-TKIs alone is recommended, as these regimens provide significant benefit to OS.

Keywords: Lung adenocarcinoma (LUAD); malignant pleural effusion (MPE); EGFR mutation; T790M mutation


Submitted Aug 25, 2024. Accepted for publication Sep 18, 2024. Published online Sep 27, 2024.

doi: 10.21037/tlcr-24-757


Highlight box

Key findings

• Epidermal growth factor receptor (EGFR) mutations, particularly the T790M mutation, are associated with the occurrence of malignant pleural effusion (MPE) in lung adenocarcinoma (LUAD).

What is known and what is new?

EGFR mutations promote tumor cell proliferation and migration. The application of EGFR-tyrosine kinase inhibitors (TKIs) can improve the prognosis of patients with LUAD and EGFR mutations.

EGFR mutations, particularly the T790M mutation, were associated with MPE in LUAD. For patients with MPE and LUAD, EGFR mutation was associated with longer overall survival (OS). It is especially recommended to use a regimen of sequential first- and third-generation EGFR-TKIs or third-generation EGFR-TKIs alone for patients with LUAD, MPE, and EGFR mutations to get better survival benefit.

What is the implication, and what should change now?

• The conclusion that EGFR mutations, particularly the T790M mutation are associated with MPE can provide guidance and direction for research on the mechanisms of lung cancer metastasis.

• The identification of factors related to OS and a comparison of different EGFR-TKI treatment regimens are beneficial for clinical decision-making in patients with LUAD and MPE.


Introduction

Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related death globally, and among the histological subtypes of NSCLC, lung adenocarcinoma (LUAD) is the most prevalent (1). In recent years, significant progress has been made in understanding the signaling pathways involved in the development and progression of LUAD. The abnormal activation of the epidermal growth factor receptor (EGFR) plays a crucial role in LUAD. EGFR is a member of the receptor tyrosine kinase ErbB family. Ligands binding to its extracellular domain induces conformational changes, leading to receptor dimerization, tyrosine phosphorylation, and activation of downstream signaling pathways, including RAS-MAPK, PI3K-AKT, and JAK-STAT, promoting cell growth and survival (2). EGFR tyrosine kinase inhibitors (TKIs) inhibit these pathways, thereby constraining tumor cell proliferation (3-5). The widespread clinical application of various EGFR-TKIs has significantly improved the prognosis of patients with LUAD and EGFR mutations (3,5).

Many studies have shown that EGFR mutations are closely related to various tumor metastases (6,7). In the field of lung cancer, previous research has focused more on the correlation between EGFR mutations and brain metastases (8,9), with the results indicating that patients with NSCLC and brain metastases have a higher frequency of EGFR mutations compared to those without brain metastases (52.0% vs. 22.0%; P<0.001) (10). The pleura is a common site for LUAD metastasis, which mainly manifests as pleural nodules and malignant pleural effusion (MPE). About 50% of patients with advanced NSCLC develop pleural effusion, which is more common in LUAD (11). Despite advances in multidisciplinary treatment improving overall care, the prognosis for patients with MPE remains poor, with a 5-year survival rate of only 6.4% (12,13). Two studies investigated the correlation between EGFR mutations and MPE in LUAD, but these studies were mostly based on nonpaired case-control designs with small sample sizes, resulting in potential bias for which further validation may be necessary (14,15).

Therefore, through a retrospective study, we aimed to determine the correlation between EGFR mutations and MPE in LUAD and evaluate the potential survival impact benefit of therapies based on EGFR mutations for patients with LUAD and MPE. We present this article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-757/rc).


Methods

Patients

This retrospective study included patients with LUAD who visited the Thoracic Surgery Department and Oncology Department of the First Hospital of China Medical University between January 2013 and December 2020. The inclusion criteria were as follows: (I) pathologically confirmed LUAD and (II) no history of other malignant tumors. A total of 4,461 patients met these criteria. Patients with incomplete clinical data, loss to follow-up, or death due to non-LUAD-related causes were excluded, resulting in a final sample size of 849 patients. Various clinical data were collected, including baseline information at admission, imaging findings, pathological results, genetic testing results, treatment regimens, and survival times. Tumor diameter and the number of lymph node metastases were restaged according to the ninth edition of the tumor-node-metastasis (TNM) staging system (16). Age was categorized into two groups: <60 and ≥60 years.

MPE was defined as the presence of pleural effusion on computed tomography (CT) imaging, confirmed by cytopathological examination showing tumor cells in the effusion (12). Patients were divided into MPE and non-MPE groups based on the presence of MPE at initial diagnosis. MPE group was categorized as stage IV according to the TNM staging system due to the presence of MPE, while the non-MPE group included patients ranging from stage I to stage IV. To control for confounding factors, propensity score matching (PSM) was performed between the two groups. After matching, the correlation between EGFR mutation and MPE in LUAD was analyzed. Furthermore, univariate and multivariate analyses were conducted to investigate factors associated with overall survival (OS) in patients with LUAD and MPE (Figure 1).

Figure 1 Flowchart of study. PSM, propensity score matching; MPE, malignant pleural effusion; N-MPE, non-malignant pleural effusion; EGFR, epidermal growth factor receptor.

This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the ethics committee of the First Hospital of China Medical University (No. 2023-54). The requirement for individual consent was waived due to the retrospective nature of the analysis.

EGFR mutation analysis

This study collected mutation information from patients diagnosed with LUAD who had undergone genetic testing. Testing methods included fluorescence polymerase chain reaction (PCR), amplification refractory mutation system PCR, and next-generation sequencing. All genetic test reports for patients were obtained from accredited genetic testing companies, such as Geneseeq Genetic Testing Company. The mutations in EGFR exons 18 to 21 were detected, including G719X, exon 19 deletion (19 Del), exon 20 insertion (20 Ins), S768I, T790M, L858R, and L681Q, among others.

Study endpoints

The primary follow-up endpoint of this study was OS, which was defined as the time from the initial diagnosis of LUAD until death or the follow-up cutoff date of September 30, 2023. During the follow-up period, deaths due to LUAD-related causes were recorded as positive outcomes, while data from patients still alive at the end of the follow-up period were considered censored to calculate cumulative survival rates. Follow-up information was primarily obtained through hospital medical records and telephone communications with patients or their relatives.

Statistical methods

PSM analysis was performed using R version 4.2.1 (The R Foundation of Statistical Computing), Factors included in PSM were age, gender, smoking history, T-stage and N-stage, and the matched ratio of MPE group to non-MPE group was 1 to 2 with a caliper value of 0.02. Data processing and statistical analyses were conducted using SPSS 26.0 software (IBM Corp., Armonk, NY, USA). Categorical variables were analyzed using the Chi-squared test or Fisher exact test. Correlation analysis was conducted using binary logistic regression. Univariate analysis was performed using the Cox proportional hazards model, and factors with P values less than 0.05 were included in the multivariate analysis to identify those factors associated with OS. Kaplan-Meier curves were plotted using GraphPad Prism 10 software (GraphPad Software, Inc., La Jolla, CA, USA), and the log-rank test was used to compare survival curves across different subgroups for statistical differences. All statistical tests were two-sided, and P values less than 0.05 were considered statistically significant.


Results

Patient characteristics in the MPE and N-MPE groups

A total of 849 patients were included in the study, with 180 in the MPE group and 669 in the N-MPE group. To control for the influence of confounding factors, PSM analysis was performed between the MPE and N-MPE groups. After PSM, there were 180 patients in the MPE group and 360 in the N-MPE group. The distribution of the aforementioned covariates across the two groups was balanced (P>0.05) (Table 1).

Table 1

Patient characteristics in the MPE and N-MPE groups (n=540)

Characteristic MPE (N=180),
n (%)
N-MPE (N=360), n (%) P value
Age 0.69
   ≥60 years 128 (71.1) 250 (69.4)
   <60 years 52 (28.8) 110 (30.5)
Gender 0.95
   Male 77 (42.7) 155 (43.0)
   Female 103 (57.2) 205 (56.9)
Smoking 0.42
   Yes 21 (11.6) 51 (14.1)
   No 159 (88.3) 309 (85.8)
Stage at diagnosis
   T 0.28
    T1 26 (14.4) 52 (14.4)
    T2 48 (26.6) 102 (28.3)
    T3 24 (13.3) 68 (18.8)
    T4 82 (45.5) 138 (38.3)
   N 0.56
    N0 25 (13.8) 36 (10.0)
    N1 17 (9.4) 37 (10.2)
    N2 68 (37.7) 134 (37.2)
    N3 70 (38.8) 153 (42.5)

MPE, malignant pleural effusion; N-MPE, non-malignant pleural effusion.

Correlation analysis of EGFR mutation and MPE

The types of EGFR mutations included in this study were exon 18 G719X mutations, 19 Del, 20 Ins, exon 20 S768I mutations, T790M mutations, and exon 21 L858R and L681Q mutations. Among the 540 patients who remained after PSM, 294 had EGFR mutations. The EGFR mutation rate was significantly higher in patients with LUAD and MPE compared to those without MPE, indicating a positive correlation between EGFR mutation and MPE occurrence [62.7% vs. 50.2%; odds ratio (OR) =1.668; P=0.006]. Notably, there was a significant difference in T790M mutation rates between the two groups (8.3% vs. 1.3%; OR =8.015; P<0.001), while no significant differences were observed for other mutation sites (Table 2).

Table 2

Correlation analysis of EGFR mutation with MPE in 540 patients

Variable MPE (N=180), n (%) N-MPE (N=360), n (%) OR (95% CI) P value
EGFR status
   EGFR WT 67 (37.2) 179 (49.7) Reference
   EGFR MT 113 (62.7) 181 (50.2) 1.668 (1.157–2.405) 0.006
EGFR mutation sites
   EGFR WT 67 (37.2) 179 (49.7) Reference
   G719X 4 (2.2) 8 (2.2) 1.336 (0.389–4.582) 0.65
   19 Del 49 (27.2) 84 (23.3) 1.558 (0.993–2.445) 0.054
   20 Ins 5 (2.7) 9 (2.5) 1.484 (0.480–4.589) 0.49
   S768I 0 (0.0) 2 (0.5)
   T790M 15 (8.3) 5 (1.3) 8.015 (2.804–22.911) <0.001
   L858R 38 (21.1) 63 (17.5) 1.611 (0.986–2.632) 0.057
   L681Q 2 (1.1) 10 (2.7) 0.534 (0.114–2.502) 0.43

MPE, malignant pleural effusion; N-MPE, non-malignant pleural effusion; OR, odds ratio; CI, confidence interval; EGFR, epidermal growth factor receptor; WT, wild type; MT, mutant type.

Patient characteristics in the MPE group

Among the 180 patients with LUAD and MPE, 113 (62.8%) patients were EGFR mutant type and 67 (37.2%) patients were EGFR wild type. Therefore, the 180 patients were divided into two groups (EGFR MT group and EGFR WT group), and their characteristics are presented in Table 3. There were 77 male patients (42.7%), and 132 patients (73.3%) had an Eastern Cooperative Oncology Group (ECOG) score of 0 or 1. 117 patients (65%) were receiving EGFR-TKI treatment, while 63 patients (35%) were receiving non-EGFR-TKI treatment. A total of 77 patients (42.7%) received systemic antiangiogenic targeted therapy, mainly consisting of bevacizumab. Moreover, 8 patients (4.4%) had mutations other than EGFR and received corresponding targeted drug therapy. These included four patients with ALK mutations treated with crizotinib, two patients treated with the selective MET kinase inhibitor savolitinib, one patient treated with the selective RET inhibitor pralsetinib, and one patient with a BRAF V600 mutation treated with a combination of dabrafenib and trametinib. Due to the small number of cases, these were collectively classified as “other targeted drug treatments”. A total of 141 patients (78.3%) received systemic chemotherapy, mainly platinum-based doublet chemotherapy, 44 patients (24.4%) received immunotherapy with anti-PD-1/PD-L1 drugs, and 51 patients (28.3%) received intrapleural perfusion therapy, which involved indwelling chest catheters and infusion of cisplatin or cisplatin combined with bevacizumab or recombinant human endostatin. Local treatment was administered to 31 patients (17.2%), with 6 patients receiving interventional therapy and 29 patients receiving radiotherapy for primary or metastatic lesions.

Table 3

Characteristics of patients in the MPE group (n=180)

Characteristic EGFR MT (n=113), n (%) EGFR WT (n=67), n (%) P value
Age 0.42
   ≥60 years 78 (69.0) 50 (74.6)
   <60 years 35 (30.9) 17 (25.3)
Gender 0.009
   Male 40 (35.3) 37 (55.2)
   Female 73 (64.6) 30 (44.7)
Smoking 0.93
   No 100 (88.4) 59 (88.0)
   Yes 13 (11.5) 8 (11.9)
ECOG 0.46
   0–1 85 (75.2) 47 (70.1)
   ≥2 28 (24.7) 20 (29.8)
Stage at diagnosis
   T 0.65
    T1 16 (14.1) 10 (14.9)
    T2 32 (28.3) 16 (23.8)
    T3 17 (15.0) 7 (10.4)
    T4 48 (42.4) 34 (50.7)
   N 0.14
    N0 14 (12.3) 11 (16.4)
    N1 10 (8.8) 7 (10.4)
    N2 50 (44.2) 18 (26.8)
    N3 39 (34.5) 31 (46.2)
EGFR-TKIs treatment <000.1
   No 2 (1.7) 61 (91.0)
   Yes 111 (98.2) 6 (8.9)
Antiangiogenic therapy 0.61
   No 63 (55.7) 40 (59.7)
   Yes 50 (44.2) 27 (40.2)
Other targeted therapy 0.053
   No 111 (98.2) 61 (91.0)
   Yes 2 (1.7) 6 (8.9)
Systemic chemotherapy 0.19
   No 28 (24.7) 11 (16.4)
   Yes 85 (75.2) 56 (83.5)
Immunotherapy 0.23
   No 82 (72.5) 54 (80.5)
   Yes 31 (27.4) 13 (19.4)
Intrapleural perfusion 0.09
   No 76 (67.2) 53 (79.1)
   Yes 37 (32.7) 14 (20.8)
Local treatment 0.30
   No 91 (80.5) 58 (86.5)
   Yes 22 (19.4) 9 (13.4)

MPE, malignant pleural effusion; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; WT, wild type; MT, mutant type; ECOG, Eastern Cooperative Oncology Group.

Prognostic factors analysis in patients with MPE

We followed up with 180 patients with LUAD and MPE with a median follow-up duration of 61 months and plotted survival curves (Figure 2). As of the follow-up cutoff date, 55 patients (30.5%) were still alive, representing a median OS of 42 months (95% CI: 35.7–44.3) and a 5-year survival rate of 25.5% (Figure 2A). Using the Cox regression model, we conducted univariate and multivariate analyses to identify the factors associated with OS in the 180 patients with LUAD and MPE. In the univariate analysis, an ECOG score ≥2 was associated with shorter OS. Conversely, EGFR mutation, antiangiogenic therapy, and intrapleural perfusion therapy were associated with a longer OS (Figure 3). Multivariate analysis showed that EGFR mutation was an independent prognostic factor for prolonged OS [hazard ratio (HR) 0.662, 95% CI: 0.456–0.962; P=0.03]. Additionally, the factors independently associated with longer OS were antiangiogenic therapy (HR 0.684, 95% CI: 0.474–0.987; P=0.043) and intrapleural perfusion therapy (HR 0.622, 95% CI: 0.441–0.992; P=0.046), Conversely, ECOG scores ≥2 were independently associated with shorter OS (HR 1.911, 95% CI: 1.226–2.886; P=0.002) (Figure 4).

Figure 2 Kaplan-Meier curves for overall survival in different populations. (A) Survival curves for patients with LUAD and MPE. (B) Survival curves for patients with LUAD and MPE stratified by the EGFR mutant type and EGFR wild type. (C) Survival curves for patients with LUAD, MPE, and EGFR mutations stratified by different EGFR-TKI treatment regimens. MPE, malignant pleural effusion; OS, overall survival; CI, confidence interval; EGFR, epidermal growth factor receptor; WT, wild type; MT, mutant type; TKI, tyrosine kinase inhibitor; gen, generation; LUAD, lung adenocarcinoma.
Figure 3 Univariate analyses for OS in patients with LUAD and MPE. HR, hazard ratio; CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; WT, wild type; MT, mutant type; OS, overall survival; LUAD, lung adenocarcinoma; MPE, malignant pleural effusion.
Figure 4 Multivariate analyses for OS in patients with LUAD and MPE. HR, hazard ratio; CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; WT, wild type; MT, mutant type; OS, overall survival; LUAD, lung adenocarcinoma; MPE, malignant pleural effusion.

To visually present the association of EGFR mutation with OS in patients with LUAD and MPE, we plotted survival curves. The median OS for EGFR-mutant patients was 47 months (95% CI: 41.5–52.5), which was significantly better than the 31 months in those with the EGFR wild type (95% CI: 25.9–36.1; P=0.03) (Figure 2B).

Prognostic factor analysis of patients with EGFR mutations and MPE

To overcome the inevitable resistance to EGFR-TKIs, new EGFR-TKIs drugs continue to be developed, providing various options and strategies for clinical applications. To clarify the impact of different EGFR-TKIs treatment regimens on OS in patients with LUAD, MPE, and EGFR mutations, we further analyzed these patients. Among the 111 patients with EGFR mutations and MPE who received EGFR-TKI treatment, 28 received first-generation EGFR-TKIs alone, 51 received sequential first- and third-generation EGFR-TKIs, and 27 received third-generation EGFR-TKIs alone (5 patients who received second-generation EGFR-TKIs were excluded from the analysis due to the small sample size). The Cox regression model was used for univariate and multivariate analyses. The results showed that the independent prognostic factors for prolonged OS were administration of third-generation EGFR-TKIs alone (HR 0.466, 95% CI: 0.233–0.930; P=0.03) and sequential first- and third-generation EGFR-TKIs (HR 0.385, 95% CI: 0.219–0.676; P=0.001) but not administration of first-generation EGFR-TKIs alone. Additionally, intrapleural perfusion therapy (HR 0.557, 95% CI: 0.327–0.951; P=0.03) was also independently associated with a longer OS (Figures 5,6).

Figure 5 Univariate analyses for OS in patients with LUAD, MPE, and EGFR mutations. HR, hazard ratio; CI, confidence interval; ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; gen, generation; OS, overall survival; LUAD, lung adenocarcinoma; MPE, malignant pleural effusion.
Figure 6 Multivariate analyses for OS in patients with LUAD, MPE, and EGFR mutations. EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; gen, generation; HR, hazard ratio; CI, confidence interval; OS, overall survival; LUAD, lung adenocarcinoma; MPE, malignant pleural effusion.

Survival curves were plotted to compare the survival benefits of the three main EGFR-TKI treatment regimens for patients with LUAD and MPE. The results showed that compared to first-generation EGFR-TKIs alone, both third-generation EGFR-TKIs alone and sequential first- and third-generation EGFR-TKIs had better median OS (first-generation EGFR-TKIs: 35 months, 95% CI: 28.4–41.6; third-generation EGFR-TKIs: 50 months, 95% CI: 37.3–62.7; sequential first- and third-generation EGFR-TKIs: 51 months, 95% CI: 45.6–56.4; P<0.001) (Figure 2C).


Discussion

This study demonstrated that EGFR mutations, especially the T790M mutation, are associated with the occurrence of MPE in LUAD. For patients with LUAD and MPE, EGFR mutations independently associated with prolonged OS.

Previous studies have shown that EGFR activation, triggered by ligand binding, activates EGFR intrinsic tyrosine kinase activity and a series of downstream signals, thereby mediating invasion and metastasis in various tumors (7,17,18). In NSCLC, EGFR mutation is most commonly associated with brain metastasis (19-21). Recently, the correlation between EGFR mutation and MPE has also been reported: Zou et al. collected data from 136 East Asian patients with LUAD and found a significant correlation between MPE occurrence and EGFR mutation (P=0.03) (14). Smits et al. analyzed 394 patients with LUAD from 13 hospitals in Western Europe and found that EGFR mutation frequency was significantly higher in patients with MPE (P=0.02) (22). The specific EGFR mutation sites and the precise targeting of metastatic lesions remain a focus of interest for researchers worldwide. Notably, the T790M mutation is particularly relevant due to its association with EGFR-TKI resistance in lung cancer (23-25). A real-world clinical study indicated a positive correlation between T790M mutation and NSCLC brain metastasis (P=0.03) (26). Chen et al. conducted a retrospective study on 365 patients with EGFR-mutant NSCLC and found that the T790M mutation carriers were more prone to metastasis, including brain, bone, liver, and intrapulmonary metastases (P<0.001) (24).

Previous studies on the correlation between EGFR mutation and MPE in NSCLC included relatively small sample sizes, affecting the accuracy of results (14,15). Moreover, limited by gene detection technologies and sample sizes, these studies often only analyzed the presence or absence of EGFR mutations or focused solely on classic mutations such L858R and 19 Dels, neglecting T790M mutation rates in MPE. In our study, the EGFR mutation rate in patients with LUAD and MPE was 62.7% compared to 50.2% in those without MPE (OR =1.668; P=0.006), suggesting a potential association between EGFR mutation and MPE occurrence. Additionally, this study, being the first to examine the correlation between specific EGFR mutation sites and MPE, demonstrated a significant positive correlation between T790M mutation and MPE occurrence (8.3% vs. 1.3%; OR =8.015; P<0.001).

The T790M mutation is a well-established mechanism of secondary resistance to EGFR-TKI therapy. Numerous studies have shown that approximately 50% of NSCLC patients who initially respond to EGFR-TKI eventually develop secondary resistance due to the emergence of the T790M mutation (27,28). Chen et al. have suggested that a significant proportion of untreated NSCLC cases may harbor the T790M mutation from the outset. In this scenario, while first- or second-generation TKIs target and eliminate the initial sensitive mutation sites, T790M-resistant clones can proliferate and become predominant (29). Consequently, the disease progression associated with primary T790M mutation may occur earlier than that driven by acquired T790M mutation, leading to a more rapid disease progression in cases with primary mutations. In our study, we also compared the differences in OS between T790M mutation and common mutations. However, due to the limited sample size, we did not observe significant differences between the two groups.

Reports suggest that the prevalence of primary T790M mutations is quite low, ranging from 1-3%, which is consistent with our study’s finding (overall T790M mutation rate was 3.7%, and 1.3% in the non-MPE group) (30,31). However, we observed a higher rate of 8.3% for T790M mutations in the MPE group, suggesting a potential association between the presence of T790M mutation and MPE. Currently, clinical research has predominantly focused on managing resistance due to secondary T790M mutations after treatment, with limited studies investigating the role of T790M in regulating tumor biology. Our findings suggest that primary T790M mutations may facilitate invasion and metastasis in LUAD. This highlights the need for further research into the underlying signaling pathways and potential therapeutic targets related to T790M mutations. We further sought to identify prognostic factors in patients with LUAD and MPE, particularly the association between EGFR mutation and the OS. ECOG score is a recognized prognostic factor for patients with MPE (32), with higher scores indicating poorer prognosis. We found that an ECOG score ≥2, as compared to lower ECOG scores, was associated with an increase in mortality rate of 91.1% (HR 1.911, 95% CI: 1.226–2.886; P=0.002). Angiogenesis is crucial to tumor growth, proliferation, and metastasis, and numerous preclinical and clinical studies have examined antiangiogenic therapies for inhibiting tumor growth (33,34). In our study, antiangiogenic targeted therapy was associated with better OS. In NSCLC, bevacizumab, U.S. Food and Drug Administration (FDA)-approved antiangiogenic inhibitor, combined with platinum-based chemotherapy is recommended as first-line treatment for patients with advanced LUAD (35). Additionally, intrapleural perfusion therapy is a primary method for treating MPE. Injecting drugs into the pleural cavity can kill metastatic tumor cells, stimulate pleural inflammation, and reduce pleural effusion. A previous study has shown that intravenous cisplatin (100 mg/m2) results in a maximum blood concentration of 6 µg/mL (36), while intrapleural infusion of cisplatin (80 mg/m2) yields a maximum blood concentration of only 0.66±0.31 µg/mL, increasing intrapleural drug concentration and reducing adverse systemic effects (37).

Previous studies have shown that EGFR-mutant patients treated with EGFR-TKIs have better survival than do those with the EGFR wild type (38-40). Similarly, we found that the median OS of EGFR-mutant patients was longer than that of those with the EGFR wild type (EGFR mutant: 47 months, 95% CI: 41.5–52.5; EGFR wild-type: 31 months, 95% CI: 25.9–36.1; P=0.03). Another finding of our study was that among patients with LUAD, EGFR mutations, and MPE, sequential first- and third-generation EGFR-TKIs or third-generation EGFR-TKIs alone were associated with longer OS compared to first-generation EGFR-TKIs alone. First-generation EGFR-TKIs lead to good initial treatment response in patients with classic EGFR mutations; however, most patients develop resistance after 9–14 months, with the T790M mutation being the most common mechanism of resistance(40,41). After resistance, a switch to third-generation EGFR-TKIs, which inhibit the T790M mutation, can significantly improve prognosis as compared to continuation on first-generation EGFR-TKIs or a switch to chemotherapy (40). Therefore, patients receiving sequential first- and third-generation EGFR-TKIs or third-generation EGFR-TKIs alone have a longer median OS, likely due to prolonged inhibition of the EGFR pathway, whereas patients receiving first-generation EGFR-TKIs alone do not benefit from new EGFR inhibitors after the emergence of resistance, resulting in a shorter median OS. This suggests that continuous inhibition of the EGFR pathway is beneficial for OS in patients with LUAD, MPE, and EGFR mutations.

This study involved certain limitations which should be acknowledged. First, although we attempted to reduce bias through PSM analysis and multivariable regression models, there were confounding factors not included in the models, and selection bias could not be completely excluded. Second, grouping certain variables might have simplified certain types of information, such as age, tumor diameter, and the number of lymph node metastases. Finally, as we employed a single-center, retrospective design, this study may be limited by a small sample size and a lack of regional differences and thus may not be more broadly generalizable.


Conclusions

Patients with LUAD with MPE exhibiteda higher incidence of EGFR mutations, including T790M mutations, compared to those without MPE. Specifically, EGFR mutations, especially the T790M mutations, were positively associated with the development of MPE in LUAD. Among patients with LUAD and MPE, EGFR mutations were linked to improved survival. For those with LUAD, MPE, and EGFR mutations, the use of sequential first-generation and third-generation EGFR-TKIs, or third-generation EGFR-TKIs alone, may lead to an improved OS benefit.


Acknowledgments

Funding: This study was supported by the Natural Science Foundation of Liaoning Provincial Science and Technology Department (No. 2023-MS-159 to W.L.), Liaoning “Xingliao Yingcai Program” Medical Masters Foundation of Liaoning Provincial Health Commission (No. YXMJ-QN-08 to W.L.), and Post-doctor Research Initiation Project of 2023 Joint Fund Project (No. 2023-BSBA-362 to D.L.).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-757/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-757/dss

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-757/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-757/coif). W.L. has received support from the Natural Science Foundation of Liaoning Provincial Science and Technology Department (No. 2023-MS-159) and Liaoning “Xingliao Yingcai Program” Medical Masters Foundation of Liaoning Provincial Health Commission (No. YXMJ-QN-08). D.L. has received support from Post-doctor Research Initiation Project of 2023 Joint Fund Project (No. 2023-BSBA-362). The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the ethics committee of the First Hospital of China Medical University (No. 2023-54). The requirement for individual consent was waived due to the retrospective nature of the analysis.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424. [Crossref] [PubMed]
  2. Cooper AJ, Sequist LV, Lin JJ. Third-generation EGFR and ALK inhibitors: mechanisms of resistance and management. Nat Rev Clin Oncol 2022;19:499-514. [Crossref] [PubMed]
  3. Hsu WH, Yang JC, Mok TS, et al. Overview of current systemic management of EGFR-mutant NSCLC. Ann Oncol 2018;29:i3-9. [Crossref] [PubMed]
  4. Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011;12:735-42. [Crossref] [PubMed]
  5. Wu YL, Fukuoka M, Mok TS, et al. Tumor response and health-related quality of life in clinically selected patients from Asia with advanced non-small-cell lung cancer treated with first-line gefitinib: post hoc analyses from the IPASS study. Lung Cancer 2013;81:280-7. [Crossref] [PubMed]
  6. Shen H, He M, Lin R, et al. PLEK2 promotes gallbladder cancer invasion and metastasis through EGFR/CCL2 pathway. J Exp Clin Cancer Res 2019;38:247. [Crossref] [PubMed]
  7. Liu A, Li Y, Lu S, et al. Stanniocalcin 1 promotes lung metastasis of breast cancer by enhancing EGFR-ERK-S100A4 signaling. Cell Death Dis 2023;14:395. [Crossref] [PubMed]
  8. Adua SJ, Arnal-Estapé A, Zhao M, et al. Brain metastatic outgrowth and osimertinib resistance are potentiated by RhoA in EGFR-mutant lung cancer. Nat Commun 2022;13:7690. [Crossref] [PubMed]
  9. Rosell R, Karachaliou N. Brain metastases in patients with EGFR-mutant non-small-cell lung cancer. Lancet Respir Med 2017;5:669-71. [Crossref] [PubMed]
  10. Ge M, Zhuang Y, Zhou X, et al. High probability and frequency of EGFR mutations in non-small cell lung cancer with brain metastases. J Neurooncol 2017;135:413-8. [Crossref] [PubMed]
  11. Roberts ME, Neville E, Berrisford RG, et al. Management of a malignant pleural effusion: British Thoracic Society Pleural Disease Guideline 2010. Thorax 2010;65:ii32-40. [Crossref] [PubMed]
  12. Morgensztern D, Waqar S, Subramanian J, et al. Prognostic impact of malignant pleural effusion at presentation in patients with metastatic non-small-cell lung cancer. J Thorac Oncol 2012;7:1485-9. [Crossref] [PubMed]
  13. Kumar A, Xu B, Srinivasan D, et al. Long-Term Survival of American Joint Committee on Cancer 8th Edition Staging Descriptors for Clinical M1a Non-Small Cell Lung Cancer. Chest 2024;165:725-37.
  14. Zou J, Bella AE, Chen Z, et al. Frequency of EGFR mutations in lung adenocarcinoma with malignant pleural effusion: Implication of cancer biological behaviour regulated by EGFR mutation. J Int Med Res 2014;42:1110-7. [Crossref] [PubMed]
  15. Lin JB, Lai FC, Li X, et al. Sequential treatment strategy for malignant pleural effusion in non-small cell lung cancer with the activated epithelial grow factor receptor mutation. J Drug Target 2017;25:119-24. [Crossref] [PubMed]
  16. Rami-Porta R, Nishimura KK, Giroux DJ, et al. The International Association for the Study of Lung Cancer Lung Cancer Staging Project: Proposals for Revision of the TNM Stage Groups in the Forthcoming (Ninth) Edition of the TNM Classification for Lung Cancer. J Thorac Oncol 2024;19:1007-27. [Crossref] [PubMed]
  17. Zhong C, Yang J, Lu Y, et al. Achyranthes bidentata polysaccharide can safely prevent NSCLC metastasis via targeting EGFR and EMT. Signal Transduct Target Ther 2020;5:178. [Crossref] [PubMed]
  18. Zhou Z, Zhang Z, Chen H, et al. SBSN drives bladder cancer metastasis via EGFR/SRC/STAT3 signalling. Br J Cancer 2022;127:211-22. [Crossref] [PubMed]
  19. Hsu F, De Caluwe A, Anderson D, et al. EGFR mutation status on brain metastases from non-small cell lung cancer. Lung Cancer 2016;96:101-7. [Crossref] [PubMed]
  20. Li H, Tong F, Meng R, et al. E2F1-mediated repression of WNT5A expression promotes brain metastasis dependent on the ERK1/2 pathway in EGFR-mutant non-small cell lung cancer. Cell Mol Life Sci 2021;78:2877-91. [Crossref] [PubMed]
  21. Shin DY, Na II, Kim CH, et al. EGFR mutation and brain metastasis in pulmonary adenocarcinomas. J Thorac Oncol 2014;9:195-9. [Crossref] [PubMed]
  22. Smits AJ, Kummer JA, Hinrichs JW, et al. EGFR and KRAS mutations in lung carcinomas in the Dutch population: increased EGFR mutation frequency in malignant pleural effusion of lung adenocarcinoma. Cell Oncol (Dordr) 2012;35:189-96. [Crossref] [PubMed]
  23. Chougule A, Chandrani P, Noronha V, et al. Real-World Evidence of EGFR Targeted Therapy in NSCLC- A Brief Report of Decade Long Single Center Experience. JTO Clin Res Rep 2023;4:100566. [Crossref] [PubMed]
  24. Chen Y, Deng J, Liu Y, et al. Analysis of metastases in non-small cell lung cancer patients with epidermal growth factor receptor mutation. Ann Transl Med 2021;9:206. [Crossref] [PubMed]
  25. Li Y, Zhang F, Yuan P, et al. High MAF of EGFR mutations and high ratio of T790M sensitizing mutations in ctDNA predict better third-generation TKI outcomes. Thorac Cancer 2020;11:1503-11. [Crossref] [PubMed]
  26. Wei B, Zhao C, Li J, et al. Combined plasma and tissue genotyping of EGFR T790M benefits NSCLC patients: a real-world clinical example. Mol Oncol 2019;13:1226-34. [Crossref] [PubMed]
  27. Sequist LV, Waltman BA, Dias-Santagata D, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011;3:75ra26. [Crossref] [PubMed]
  28. Campo M, Gerber D, Gainor JF, et al. Acquired Resistance to First-Line Afatinib and the Challenges of Prearranged Progression Biopsies. J Thorac Oncol 2016;11:2022-6. [Crossref] [PubMed]
  29. Chen LY, Molina-Vila MA, Ruan SY, et al. Coexistence of EGFR T790M mutation and common activating mutations in pretreatment non-small cell lung cancer: A systematic review and meta-analysis. Lung Cancer 2016;94:46-53. [Crossref] [PubMed]
  30. Tian P, Wang Y, Wang W, et al. High-throughput sequencing reveals distinct genetic features and clinical implications of NSCLC with de novo and acquired EGFR T790M mutation. Lung Cancer 2018;124:205-10. [Crossref] [PubMed]
  31. Pang LL, Zhuang WT, Huang YH, et al. Uncommon de novo EGFR(T790M)-Mutant NSCLC characterized with unique genetic Features: Clinical response and acquired resistance to the third-generation EGFR-TKIs treatment. Lung Cancer 2024;190:107528. [Crossref] [PubMed]
  32. Bibby AC, Dorn P, Psallidas I, et al. ERS/EACTS statement on the management of malignant pleural effusions. Eur Respir J 2018;52:1800349. [Crossref] [PubMed]
  33. Van der Veldt AA, Lubberink M, Bahce I, et al. Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: implications for scheduling of anti-angiogenic drugs. Cancer Cell 2012;21:82-91. [Crossref] [PubMed]
  34. Huinen ZR, Huijbers EJM, van Beijnum JR, et al. Anti-angiogenic agents - overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Nat Rev Clin Oncol 2021;18:527-40. [Crossref] [PubMed]
  35. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006;355:2542-50. [Crossref] [PubMed]
  36. Go RS, Adjei AA. Review of the comparative pharmacology and clinical activity of cisplatin and carboplatin. J Clin Oncol 1999;17:409-22. [Crossref] [PubMed]
  37. Sakaguchi H, Ishida H, Nitanda H, et al. Pharmacokinetic evaluation of intrapleural perfusion with hyperthermic chemotherapy using cisplatin in patients with malignant pleural effusion. Lung Cancer 2017;104:70-4. [Crossref] [PubMed]
  38. Hsiao SH, Lin HC, Chou YT, et al. Impact of epidermal growth factor receptor mutations on intracranial treatment response and survival after brain metastases in lung adenocarcinoma patients. Lung Cancer 2013;81:455-61. [Crossref] [PubMed]
  39. Zhou C, Wu YL, Chen G, et al. Final overall survival results from a randomised, phase III study of erlotinib versus chemotherapy as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer (OPTIMAL, CTONG-0802). Ann Oncol 2015;26:1877-83. [Crossref] [PubMed]
  40. Mok TS, Wu Y-L, Ahn M-J, et al. Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N Engl J Med 2017;376:629-40. [Crossref] [PubMed]
  41. Le T, Gerber DE. Newer-Generation EGFR Inhibitors in Lung Cancer: How Are They Best Used? Cancers (Basel) 2019;11:366. [Crossref] [PubMed]
Cite this article as: Yang Q, Wang Z, Fu Q, Hu X, Chen L, Chen W, Lv L, Liu Z, Men W, Li D, Li W. EGFR mutations in patients with lung adenocarcinoma and malignant pleural effusion: a propensity score-matched analysis of a single-center database. Transl Lung Cancer Res 2024;13(9):2435-2447. doi: 10.21037/tlcr-24-757

Download Citation