Impact of RBM10 and PD-L1 expression on the prognosis of pathologic N1–N2 epidermal growth factor receptor mutant lung adenocarcinoma
Original Article

Impact of RBM10 and PD-L1 expression on the prognosis of pathologic N1–N2 epidermal growth factor receptor mutant lung adenocarcinoma

Tetsuya Isaka1,2^, Yohei Miyagi3, Tomoyuki Yokose4, Haruhiro Saito5, Rika Kasajima3, Kozue Watabe1, Naoko Shigeta1, Noritake Kikunishi1, Shunsuke Shigefuku1, Kotaro Murakami1, Hiroyuki Adachi1, Takuya Nagashima1, Hiroyuki Ito1

1Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan; 2Department of Surgery, Yokohama City University, Yokohama, Japan; 3Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan; 4Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan; 5Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan

Contributions: (I) Conception and design: T Isaka, Y Miyagi, R Kasajima; (II) Administrative support: T Isaka, Y Miyagi; (III) Provision of study materials or patients: T Isaka, Y Miyagi, T Yokose, H Saito, K Watabe, N Shigeta, N Kikunishi, S Shigefuku, K Murakami, H Adachi, T Nagashima, H Ito; (IV) Collection and assembly of data: T Isaka, Y Miyagi, T Yokose, H Saito, K Watabe, N Shigeta, N Kikunishi, S Shigefuku, K Murakami, H Adachi, T Nagashima, H Ito; (V) Data analysis and interpretation: T Isaka, Y Miyagi, T Yokose, H Saito, H Adachi, T Nagashima, H Ito; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

^ORCID: 0000-0002-3302-7133.

Correspondence to: Tetsuya Isaka, MD, PhD. Department of Thoracic Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi, Yokohama, Kanagawa 241-8515, Japan; Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa, Yokohama, Kanagawa 236-0004, Japan. Email: t-isaka@kcch.jp.

Background: Impact of RNA-binding motif protein 10 (RBM10) and programmed death-ligand 1 (PD-L1) on the postoperative prognosis of patients with epidermal growth factor receptor gene mutation (EGFR-Mt) lung adenocarcinoma with pathological lymph node metastasis is still unclear.

Methods: Patients who underwent curative surgery for pN1–N2 EGFR-Mt lung adenocarcinoma (n=129) harboring the EGFR exon 19 deletion mutation (Ex19) (n=66) or EGFR exon 21 L858R mutation (Ex21) (n=63) between January 2010 and December 2020 were included in this retrospective study. The prognoses of patients with low/high cytoplasmic RBM10 expression and PD-L1 negativity/positivity based on immunohistochemistry (IHC) of resected specimens were compared using the log-rank test. The effects of RBM10 and PD-L1 expression on overall survival (OS) were examined via multivariable analysis using the Cox proportional hazards regression model. The effects of RBM10 and PD-L1 expression on progression-free survival (PFS) of EGFR-tyrosine kinase inhibitors (TKIs) therapy among patients with recurrent pN1–N2 EGFR-Mt lung adenocarcinoma (n=67) were examined using log-rank tests.

Results: The RBM10 low expression group showed significantly better 5-year OS than the RBM10 high expression group (89.4% vs. 71.5%, P=0.020), and the PD-L1 negative group tended to have longer 5-year OS than the PD-L1 positive group (86.4% vs. 68.4%, P=0.050). Multivariable analysis showed that high RBM10 expression [hazard ratio (HR), 3.12; 95% confidence interval (CI): 1.19–8.17; P=0.021] and PD-L1 positivity (HR, 3.80; 95% CI: 1.64–8.84; P=0.002) were independent poor prognostic factors for OS. PFS of patients with relapse and first-line EGFR-TKI treatment was significantly better in the PD-L1-negative group than in the PD-L1-positive group (34.5 vs. 12.1 months, P=0.045). PFS of patients with Ex21 relapse and first-line EGFR-TKI treatment was significantly better in the RBM10 low expression group than in the RBM10 high expression group (25.5 vs. 13.0 months, P=0.025).

Conclusions: High RBM10 expression and PD-L1 positivity are poor prognostic factors for OS in patients with pN1–N2 EGFR-Mt lung adenocarcinoma after curative surgery. In patients with recurrent pN1–N2 EGFR-Mt lung adenocarcinoma, PD-L1 and RBM10 expression may influence response to EGFR-TKIs.

Keywords: Epidermal growth factor receptor gene mutation (EGFR-Mt); overall survival (OS); programmed death-ligand 1 (PD-L1); primary lung adenocarcinoma; RNA-binding motif protein 10 (RBM10)


Submitted Jun 01, 2023. Accepted for publication Aug 29, 2023. Published online Sep 25, 2023.

doi: 10.21037/tlcr-23-355


Highlight box

Key findings

• High cytoplasmic RNA-binding motif protein 10 (RBM10) expression and programmed death-ligand 1 (PD-L1) positivity were poor prognostic factors for overall survival in patients with pN1–N2 epidermal growth factor receptor gene mutation (EGFR-Mt) lung adenocarcinoma after curative surgery. In patients with recurrent pN1–N2 EGFR-Mt, PD-L1 and RBM10 expression may influence response to EGFR-tyrosine kinase inhibitors (TKIs).

What is known and what is new?

• RBM10 participates in the regulation of mRNA splicing, apoptosis induction, angiogenesis, and cell growth inhibition. The expression of PD-L1 in tumors is associated with the response to immune checkpoint inhibitor therapy.

• This study examined the relationship between RBM10/PD-L1 expression (measured via immunohistochemistry) and both the prognosis of pN1–N2 EGFR-Mt after resection and the efficacy of EGFR-TKI therapy after recurrence.

What is the implication, and what should change now?

• Evaluation of RBM10 and PD-L1 expression could be useful in considering postoperative and post-relapse treatment of patients with pN1–N2 EGFR-Mt. Large prospective studies are necessary to evaluate the usefulness of RBM10 and PD-L1 expression in predicting prognosis in patients with pN1–N2 EGFR-Mt, and determining the efficacy of EGFR-TKI after relapse.


Introduction

Epidermal growth factor receptor gene mutations (EGFR-Mt) account for approximately half of the driver gene mutations in lung adenocarcinoma in Asian populations, with exon 19 deletion mutation (Ex19) and exon 21 L858R mutation (Ex21) accounting for 85–90% of EGFR-Mt (1-4). Lung adenocarcinomas harboring EGFR-Mt recur at a high rate even after complete resection (5), with a poor 5-year disease-free survival rate of 26.2% (5,6). Although EGFR-tyrosine kinase inhibitors (TKIs) are effective after recurrence (7,8), the prognosis of patients with EGFR-Mt lung adenocarcinoma with pathological lymph node metastasis (stage pN1–pN2) after curative surgery is poor, with 5-year overall survival (OS) rates of 65% for patients with pN1–N2 EGFR-Mt lung adenocarcinoma and 48% for patients with pN1–N2 Ex21 lung adenocarcinoma (6,9). Even with EGFR-TKI treatment, 20–30% of patients with advanced or recurrent EGFR-Mt lung adenocarcinoma have a poor prognosis owing to the low initial response (10). Overcoming the low response to EGFR-TKIs will improve OS of patients with EGFR-Mt lung adenocarcinoma after resection. New therapeutic targets that improve the prognosis of patients with pN1–N2 EGFR-Mt lung adenocarcinoma after surgery need to be elucidated.

RNA-binding motif protein 10 (RBM10) is involved in the regulation of mRNA splicing, apoptosis induction, angiogenesis, and cell growth inhibition (11,12). Several studies have reported that RBM10 expression was correlated with various factors related to prognosis in solid tumors, including lung cancer (13,14) and breast cancer (15). Recently, Nanjo et al. (16) reported that genetic inactivation of RBM10 in EGFR-Mt cells diminished EGFR-TKI-mediated apoptosis. However, the functional roles of RBM10 in EGFR-Mt lung adenocarcinoma remain unknown and require further elucidation.

Programmed death-ligand 1 (PD-L1) expression in tumors is associated with the response to immune checkpoint inhibitors, and higher PD-L1 expression is associated with higher therapeutic efficacy (17,18). Recent clinical and preclinical studies have shown that high PD-L1 expression was associated with a reduced response to EGFR-TKIs in patients with unresectable advanced EGFR-Mt lung adenocarcinoma (19-21). However, the impact of PD-L1 expression on prognosis in patients with EGFR-Mt lung adenocarcinoma after surgery remains unknown.

This study examined the relationship between RBM10/PD-L1 expression, as measured using immunohistochemistry (IHC), and both the prognosis of pN1–N2 EGFR-Mt lung adenocarcinoma after resection and the efficacy of EGFR-TKI treatment after recurrence. We present this article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-355/rc).


Methods

Patients

This retrospective study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by institutional review board of Kanagawa Cancer Center (2019 Eki-174) and individual consent for this retrospective analysis was waived.

Of the 321 patients with stage pN1–N2 lung adenocarcinoma who underwent lobectomy or a greater extent of lung resection with lymph node dissection at Kanagawa Cancer Center between January 2010 and December 2020, 129 patients (40.2%) who harbored EGFR mutations in either Ex19 (n=66) or Ex21 (n=63) were included in this study. Patients who received neoadjuvant therapy were excluded. Patients who received adjuvant EGFR-TKI therapy were also excluded.

Pathological diagnosis and detection of EGFR mutation

Pathological diagnosis was based on hematoxylin and eosin (HE) staining of formalin-fixed paraffin-embedded tissue sections. To confirm the diagnosis of adenocarcinoma, IHC for thyroid transcription factor 1 and Alcian blue staining were performed. Synaptophysin, CD56, chromogranin A, p63, and p40 staining procedures were also performed as needed. Blood vessel and pleural invasion were evaluated using EVG staining, and lymphatic vessel invasion was evaluated using D2-40, if necessary. For the EGFR mutation analysis of lung adenocarcinoma, the loop-hybrid mobility shift assay method (22), Cycleave/fragment method (23), or the Cobas®EGFR mutation test v2 (24) was performed on surgically resected specimens.

IHC analyses

Tissue microarray (TMA)

The most representative tumor areas containing tumor cells and excluding necrotic areas were marked on the HE-stained slides. Tumor samples were obtained using a 2-mm-diameter core, embedded in a paraffin acceptor block, and placed in a block to produce a TMA block.

Evaluation of RBM10 expression in the TMA

TMA thin sections were deparaffinized, immersed in 10 mM sodium citrate buffer for 20 min at 121 ℃, and incubated with rabbit polyclonal anti-RBM10 antibody (1:300, cat no. HPA034972; Sigma) for 1 h at room temperature. After confirmation of tumor cells in the HE-stained TMA, RBM10 expression was evaluated by assessing the staining intensity of the tumor cytoplasm and classified as low, moderate, or high (Figure 1). The percentage of stained tumor cells among all tumor cells in the TMA was evaluated in 5% increments. RBM10 expression was considered high if tumor cells in the TMA were stained as high intensity or ≥75% of the tumor cells in the TMA were stained as moderate intensity. RBM10 expression was considered low if tumor cells in the TMA were stained as low intensity or <75% of the tumor cells in the TMA were stained moderate intensity (Figure 1).

Figure 1 RBM10 expression on immunohistochemistry (original magnification ×400). RBM10 expression was considered high if tumor cells in the TMA were stained as high intensity or 75% of the tumor cells in the TMA were stained as moderate intensity. RBM10 expression was considered low if tumor cells in the TMA were stained as low intensity or <75% of the tumor cells in the TMA were stained moderate intensity. RBM10, RNA-binding motif protein 10; TMA, tissue microarray.

In situ hybridization (ISH) of RBM10

ISH to detect RBM10 mRNA sequences in the cytoplasm (25) was performed on the representative sections of pN1–N2 EGFR-Mt lung adenocarcinomas using the RNAscope Probe-Hs-RBM10 (cat no. 419881; Advanced Cell Diagnostics, Hayward, CA, USA) and visualized using the RNAscope 2.5 HD Reagent Kit—RED (cat no. 322350; Advanced Cell Diagnostics) according to the manufacturer’s protocol.

Evaluation of PD-L1 expression

The TMA sections were incubated with a rabbit monoclonal anti-PD-L1 antibody (clone E1L3N, 1:300, cat no. 13684; Cell Signaling Technology) for 1 h at room temperature. The tumor was considered PD-L1 positive if more than 1% of tumor cells in the TMA were stained and was considered negative if less than 1% of tumor cells were stained.

Primary/secondary outcomes of this study and word definitions

The primary outcome of this study was to compare the OS of patients with EGFR-Mt lung adenocarcinoma in RBM10 low/high and PD-L1 negative/positive expression groups. The second outcome was to compare the progression-free survival (PFS) of patients with recurrent EGFR-Mt lung adenocarcinoma in the RBM10 low/high and PD-L1 negative/positive expression groups.

Intrathoracic recurrence included cervicothoracic lymph node recurrence, lung recurrence, and pleural dissemination. Distant recurrence included central nervous system, abdominal organ, and bone metastases. The EGFR-TKIs administered included first-generation (gefitinib or erlotinib), second-generation (afatinib), and third-generation (osimertinib) EGFR-TKIs and administration of EGFR-TKIs after relapse was based on the decision of respiratory physician. EGFR-TKIs were administered unless the patient did not wish to receive the treatment or was intolerant to the treatment due to a systemic condition. The time interval from surgery to all-cause death was defined as OS, and it was censored if the patients were event-free at the last follow-up. The time interval from the administration of the first-line EGFR-TKI to progression or death was defined as PFS.

Statistical analysis

Continuous variables between the two groups were compared using the Mann-Whitney U test or Student’s t-test based on the results of normality testing. Categorical variables between the two groups were compared using Fisher’s exact test. The OS curve of patients with pN1–N2 EGFR-Mt lung adenocarcinoma and the PFS of patients with recurrent pN1–N2 EGFR-Mt lung adenocarcinoma with low/high RBM10 expression and PD-L1 negativity/positivity were created using the Kaplan-Meier method. The OS curves of the two groups were compared using the log-rank test. To estimate the median follow-up period, the reverse Kaplan-Meier method was performed. The Cox proportional hazards regression model was applied for the univariable and multivariable analyses for the following variables: age (<65/≥65 years), sex (male/female), carcinoembryonic antigen (CEA) level (≤10/>10 ng/mL), tumor size by computed tomography (CT) (≤3.0/>3.0 cm), blood vessel invasion (−/+), lymphatic vessel invasion (−/+), pleural invasion (−/+), pathological lymph node metastasis (−/+), EGFR-TKI administration (−/+), EGFR mutation subtype (Ex19/Ex21), RBM10 expression (low/high), and PD-L1 expression (negative/positive). Multivariable analysis was performed with the significant variables (P<0.10) identified in the univariable analysis. P value <0.05 was considered statistical significance. EZR on R commander version 1.61 was used for all statistical analyses (26).


Results

RBM10 mRNA expression using RNAscope

RBM10 mRNA expression, as evaluated using RNAscope in representative RBM10 cytoplasmic high and low expression tumors, was prominent in the RBM10 high expression group as measured by using IHC, while RBM10 mRNA expression was low in the RBM10 low expression group (Figure 2).

Figure 2 RBM10 mRNA expression was evaluated by RNA in situ hybridization using RNAscope® technology on the semiserial sections used for the immunohistochemistry of RBM10 protein that demonstrated representative cytoplasmic or nuclear staining. Hybridization probe, Probe-Hs-RBM10 and HybEZTM Hybridization System was used according to the manufacturer’s protocol (see Methods). RBM10 mRNA was visualized as cytoplasmic red dots (original magnification, ×400). RBM10, RNA-binding motif protein 10.

Comparison of the clinicopathological features of pN1–N2 EGFR-Mt lung adenocarcinoma with low/high RBM10 expression and negative/positive PD-L1 expression

Among the 129 patients with EGFR-Mt lung adenocarcinoma, 44 (34.1%) and 21 (16.3%) showed high RBM10 expression and positive PD-L1 expression, respectively (Table 1). Ex21 lung adenocarcinoma was observed in 44 patients (51.8%) in the low RBM10 expression group and 19 patients (43.2%) in the high RBM10 expression group (P=0.458) (Table 1). Pathological ipsilateral mediastinal lymph node involvement (pN2) was more frequent in patients with low RBM10 expression group than in patients with high RBM10 expression group (70.6% vs. 40.9%, P=0.001; Table 1). Ex21 lung adenocarcinoma was observed in 51 patients (47.2%) in the PD-L1-negative group and in 12 patients (57.1%) in the PD-L1-positive group (P=0.478) (Table 1). The solid adenocarcinoma subtype was more frequent in the PD-L1 positive group than in the PD-L1 negative group (38.1% vs. 8.3%, P=0.007; Table 1). Moreover, the PD-L1 negative group had a lower maximum standardized uptake value than the PD-L1 positive group (7.1 vs. 11.2, P=0.005; Table 1).

Table 1

Comparison of the clinicopathological features of pN1–N2 EGFR mutant lung adenocarcinomas (n=129) with low/high RBM10 expression and negative/positive PD-L1 expression

Variables RBM10 PD-L1
Low†1 (n=85) High†2 (n=44) P value§ Negative‡1 (n=108) Positive‡2 (n=21) P value§
Age (years), mean (SD) 67.0 (11.1) 65.0 (12.3) 0.344 65.6 (11.2) 69.7 (12.8) 0.144
Male, n (%) 42 (49.4) 23 (52.3) 0.853 53 (49.1) 12 (57.1) 0.634
Brinkman index, mean (SD) 239 (375.3) 272 (301.6) 0.174// 254 (363.1) 231 (287.5) 0.834//
CEA (ng/mL), mean (SD) 8.4 (12.9) 7.6 (10.1) 0.779// 7.5 (10.0) 11.2 (19.3) 0.655//
CT tumor size (cm), mean (SD) 3.4 (1.4) 3.7 (1.7) 0.300 3.5 (1.4) 3.6 (1.9) 0.784
PET maxSUV, mean (SD) 7.7 (4.8) 7.9 (5.1) 0.795// 7.1 (4.4) 11.2 (5.8) 0.005//
Lymphatic vessel invasion +, n (%) 50 (58.8) 24 (54.5) 0.709 63 (58.3) 11 (52.4) 0.637
Blood vessel invasion +, n (%) 65 (76.5) 32 (72.7) 0.671 84 (77.8) 13 (61.9) 0.166
Pleural invasion +, n (%) 41 (48.2) 23 (52.3) 0.713 58 (53.7) 6 (28.6) 0.055
pN2 +, n (%) 60 (70.6) 18 (40.9) 0.001 67 (62.0) 11 (52.4) 0.468
Subtype of adenocarcinoma, n (%)
   Lepidic 2 (2.4) 2 (4.5) 3 (2.8) 1 (4.8)
   Papillary 30 (35.3) 20 (45.5) 43 (39.8) 7 (33.3)
   Acinar 35 (41.1) 16 (36.4) 47 (43.5) 4 (19.0)
   Micropapillary 4 (4.7) 3 (6.8) 6 (5.6) 1 (4.8)
   Solid 14 (16.5) 3 (6.8) 0.417 9 (8.3) 8 (38.1) 0.007
Recurrence +, n (%) 58 (68.2) 33 (75.0) 0.542 77 (71.3) 14 (66.7) 0.794
Initial site of recurrence, n (%*)
   Intrathoracic only 29 (50.0*) 13 (39.4*) 39 (50.6*) 3 (21.4*)
   Distant 29 (50.0*) 20 (60.6*) 0.385 38 (49.4*) 11 (78.6*) 0.078
1st line EGFR-TKI, n (%*) 44 (75.9*) 23 (69.7*) 0.622 57 (74.0*) 10 (71.4*) 1.000
   First-generation 17 (29.3*) 9 (27.3*) 21 (27.3*) 5 (35.7*)
   Second-generation 2 (3.5*) 3 (9.1*) 4 (5.2*) 1 (7.1*)
   Third-generation 25 (43.1*) 11 (33.3*) 0.447 32 (41.5*) 4 (28.6*) 0.479
EGFR mutation, n (%)
   Exon 19 deletion mutation 41 (48.2) 25 (56.8) 57 (52.8) 9 (42.9)
   Exon 21 L858R point mutation 44 (51.8) 19 (43.2) 0.458 51 (47.2) 12 (57.1) 0.478

†1, RBM10 low expression: tumor cells stained as low intensity or <75% of the tumor cells stained moderate intensity; †2, RBM10 high expression: tumor cells stained as high intensity or ≥75% of the tumor cells stained as moderate intensity; ‡1, PD-L1 negative expression: less than 1% of tumor cells stained; ‡2, PD-L1 positive expression: more than 1% of tumor cells stained; *, percentage in recurrent cases; §, Fisher’s exact test; , Student’s t-test; //, Mann-Whitney U test. EGFR, epidermal growth factor receptor; RBM10, RNA-binding motif protein 10; PD-L1, programmed death-ligand 1; SD, standard deviation; CEA, carcinoembryonic antigen; CT, computed tomography; PET, positron emission tomography; SUV, standardized uptake value; TKI, tyrosine kinase inhibitor.

Impact of RBM10 and PD-L1 expression on the OS of patients with pN1–N2 EGFR-Mt lung adenocarcinoma

The median follow-up period was 54.7 months. The 5-year OS rate was significantly lower in the RBM10 high expression group than in the RBM10 low expression group (89.4% vs. 71.5%, P=0.020; Figure 3A) and tended to be worse in the PD-L1 positive group than in the PD-L1 negative group (86.4% vs. 68.4%; P=0.050; Figure 3B).

Figure 3 Comparison of overall survival between the low and high RBM10 expression groups (A) and PD-L1 negative/positive groups (B) among patients with stage pN1–N2 EGFR mutant lung adenocarcinoma. The solid line describes the probability of survival, and the dotted lines represent the 95% confidence interval. RBM10, RNA-binding motif protein 10; OS, overall survival; PD-L1, programmed death-ligand 1; EGFR, epidermal growth factor receptor.

In the multivariable analysis, high RBM10 expression [hazard ratio (HR), 3.12; 95% confidence interval (CI), 1.19–8.17; P=0.021], PD-L1 positivity (HR, 3.80; 95% CI: 1.64–8.84; P=0.002), and pN2 (HR, 4.19; 95% CI: 1.57–11.2; P=0.004) were independent prognostic factors for OS (Table 2).

Table 2

Univariable and multivariable analyses of the overall survival of patients with pN1–N2 EGFR-Mt lung adenocarcinoma (n=129)

Variables Univariable analysis Multivariable analysis
HR 95% CI P value HR 95% CI P value
Age (≥65 years) 1.68 0.69–4.05 0.250
Sex (male) 1.64 0.72–3.76 0.243
CEA (>10 ng/mL) 0.40 0.09–1.70 0.214
CT tumor size (>3.0 cm) 1.69 0.72–3.95 0.226
Lymphatic vessel invasion (+) 0.84 0.38–1.89 0.679
Blood vessel invasion (+) 1.11 0.44–2.80 0.830
Pleural invasion (+) 0.87 0.39–1.95 0.741
pN2 (vs. pN1) 2.29 0.91–5.78 0.079 4.19 1.57–11.2 0.004
EGFR-TKI administration 0.76 0.34–1.71 0.513
EGFR exon 21 L858R 1.93 0.85–4.37 0.114
RBM10 high expression 2.51 1.12–5.60 0.025 3.12 1.19–8.17 0.021
PD-L1 positive expression 2.46 0.97–6.23 0.058 3.80 1.64–8.84 0.002

EGFR-Mt, epidermal growth factor receptor gene mutation; HR, hazards ratio; CI, confidence interval; CEA, carcinoembryonic antigen; CT, computed tomography; EGFR, epidermal growth factor receptor; TKI, tyrosine kinase inhibitor; RBM10, RNA-binding motif protein 10; PD-L1, programmed death-ligand 1.

Impact of RBM10 and PD-L1 expression on the OS of patients with Ex21 and Ex19 lung adenocarcinoma

Figure 4 shows an analysis of OS according to EGFR subtype. Among patients with Ex21, those in the RBM10 high expression group had poorer OS than those in the RBM10 low expression group (83.9% vs. 60.0%, P=0.012; Figure 4A). There was no significant difference between the RBM10 high and low expression groups among patients with Ex19 lung adenocarcinoma (Figure 4B). There was no difference in the OS of patients with the different EGFR mutation subtypes according to PD-L1 positivity (Figure 4C,4D).

Figure 4 Comparison of overall survival among patients with low and high RBM10 expressing Ex21 lung adenocarcinoma (A) and Ex19 lung adenocarcinoma (B). Comparison of overall survival in patients with PD-L1-positive and -negative Ex21 (C) and Ex19 lung adenocarcinoma (D). The solid line describes the probability of survival, and the dotted lines represent the 95% confidence interval. RBM10, RNA-binding motif protein 10; OS, overall survival; PD-L1, programmed death-ligand 1; Ex21, exon 21 L858R mutation; Ex19; exon 19 deletion mutation.

Impact of RBM10 and PD-L1 expression on the PFS of patients with recurrent EGFR-Mt lung adenocarcinoma who were treated with first-line EGFR-TKIs

Among the patients treated with first-line EGFR-TKIs after recurrence (n=67), PFS was not significantly different between the RBM10 high and low expression groups (Figure 5A). PFS was better in the PD-L1 negative group than in the PD-L1 positive group (median, 34.5 vs. 12.1 months, P=0.045; Figure 5B). Among the patients with Ex21 who were treated with first-line EGFR-TKIs, the RBM10 low expression group had significantly better PFS than the high expression group (median PFS, 25.5 vs. 13.0 months, P=0.025; Figure 5C). In the multivariable analysis, high RBM10 expression in Ex21 (HR, 3.16; 95% CI: 1.17–8.53; P=0.023) were independent prognostic factors for PFS (Table S1).

Figure 5 Comparison of PFS of patients with recurrent EGFR mutant lung adenocarcinoma who were treated with first-line EGFR-TKIs between the RBM10 low/high expression (A) and PD-L1 negative/positive groups (B). Comparison of PFS among patients with recurrent Ex21 lung adenocarcinoma who were treated with first-line EGFR-TKIs in the RBM10 low/high expression groups (C). The solid line describes the probability of survival, and the dotted lines represent the 95% confidence interval. RBM10, RNA-binding motif protein 10; EGFR-Mt, epidermal growth factor receptor gene mutation; PD-L1, programmed death-ligand 1; Ex21, exon 21 L858R mutation; PFS, progression-free survival; TKIs, tyrosine kinase inhibitors.

Discussion

This is the first study to demonstrate that high RBM10 protein expression in the cytoplasm and PD-L1 positivity, as measured by IHC, are independent poor prognostic factors in patients with pN1–N2 EGFR-Mt lung adenocarcinoma after curative surgery. Among patients with recurrent EGFR-Mt lung adenocarcinoma, the PD-L1-positive group showed a poor response to first-line EGFR-TKI treatment. In addition, among patients with recurrent Ex21 lung adenocarcinoma, the RBM10 high expression group also showed a poor response to first-line EGFR-TKI treatment. Thus, the response to first-line EGFR-TKIs in patients with recurrent EGFR-Mt lung adenocarcinoma could be predicted based on cytoplasmic RBM10 and PD-L1 expression. RBM10 and PD-L1 may be key biomolecular targets that could improve the prognosis of patients with pN1–N2 EGFR-Mt lung adenocarcinoma. RBM10 shows particular promise for pN1–N2 Ex21 lung adenocarcinoma after surgery.

Little is known about the relationship between RBM10 expression and prognosis in patients with solid tumors. Sun et al. (13) compared the prognosis of 90 patients with pN0–N2 primary lung adenocarcinomas based on nuclear and cytoplasmic RBM10 expression and reported that pN1–N2 was more frequent in patients with high RBM10 expression and that OS was poorer in the RBM10 high expression group than in the RBM10 low expression group. In contrast, Guan et al. (14) reported that low RBM10 expression was associated with advanced stage in 41 primary lung adenocarcinomas. Rintala-Maki et al. (15) reported that RBM10 v2, a variant of RBM10, was associated with poor prognosis in 61 patients with breast cancer. The present study demonstrated that pN2 was more frequent in the RBM10 low expression group, indicating that low RBM10 expression was associated with advanced stage in pN1–N2 EGFR-Mt lung adenocarcinoma, similar to the findings of Guan et al. (14). However, the RBM10 low expression group showed better OS due to the higher efficacy of EGFR-TKI treatment in patients with low RBM10 expression than in those with high RBM10 expression.

As shown in Table 1, high RBM10 expression was observed in 44 (34.1%) patients. To date, few studies have evaluated the IHC expression of RBM10 in primary lung cancer tissues using the RBM10 antibody No. HPA034972 (Sigma); therefore, there are no criteria for its cut-off value for RBM10 expression. Zhang et al. classified RBM10 staining intensity into four levels (strong, moderate, faint, and no staining) and defined RBM10-positive as 10% of tumor cells or more being stained at a strong or moderate intensity, while the rest were defined as RBM10-negative; 63 out of 87 lung cancer patients (72.4%) were RBM10-positive (27). In the present study, 34.1% of pN1–N2 EGFR-Mt lung adenocarcinomas showed high RBM10 expression, which was lower than that reported by Zhang et al. This may be attributable to the fact that our study population consisted of patients with pN1–N2 or advanced lung cancer with a higher RBM10 expression threshold than that reported by Zhang et al.

A previous study showed that RBM10 overexpression inhibited various malignant behaviors of lung adenocarcinoma, including cell cycle progression, cell viability and proliferation, and colony formation (11,14,28,29). RBM10 suppressed lung adenocarcinoma cell proliferation via the RAP1/AKT/CREB signaling pathway, independent of the MAPK/ERK and P38/MAPK signaling pathways (28). RBM10 promoted apoptosis via the AKT signaling pathway and by activating p53 (28,30,31). These functional roles of RBM10 support the finding of a higher frequency of pN2 in the RBM10 low expression group than in the high RBM10 expression group. Moreover, our findings were consistent with those of a study by Lengel et al. that showed that mutation of the RBM10 gene, which is considered a tumor suppressor gene, is often associated with non-metastatic early stage lung adenocarcinoma (32). However, other studies have shown contradictory results: upregulation of RBM10 expression stimulated proliferative signaling pathways in lung adenocarcinoma cells (12,13) and inhibited the stimulation of apoptotic signaling pathways in lung adenocarcinoma cells (13,33). Therefore, further elucidation of the molecular role of RBM10 in primary lung adenocarcinoma is required.

Little is known about the role of RBM10 in EGFR-Mt lung adenocarcinoma. A previous preclinical study reported that Src family kinases downstream of EGFR directly phosphorylate the tyrosine residues of RBM10 and promote transfer of RBM10 from the nucleus to the cytoplasm to regulate FilGAP, which is associated with cancer cell invasion and metastasis (34,35). Recently, Nanjo et al. (16) reported that a RBM10 mutation resulting in the loss of RBM10 expression limited the initial response to EGFR-TKI due to suppression of mitochondria-mediated apoptosis in response to EGFR-TKI in EGFR-Mt tumor cells caused by a decreased ratio of Bcl-xS to Bcl-xL. They also compared the PFS of 70 patients with stage IIIB/IV advanced EGFR-Mt lung adenocarcinoma and showed that the RBM10 mutation-positive group (n=13) had a lower response to EGFR-TKI treatment than the RBM10 mutation-negative group (n=57) (median PFS, 5.7 vs. 13.4 months; P<0.0001) (16). However, the present study showed that high RBM10 expression in the cytoplasm was associated with lower efficacy of EGFR-TKI treatment. One possible reason for this paradoxical result was that the polyclonal anti-RBM10 antibodies used in our study (cat no., HPA034972) recognize the N-terminus of RBM10, whiles the ones used in Nanjo’s study (cat no., A301-006A) recognize the C-terminus of RMB10. These two polyclonal antibodies probably did not completely recognize the same molecular variants of RBM10. A second possible reason was that we evaluated cytoplasmic staining of RBM10, while Nanjo et al. evaluated nuclear staining of RBM10 using IHC. The present study findings indicated high RBM10 mRNA expression in tumors with high cytoplasmic RBM10 IHC staining (Figure 2), possibly indicating that cytoplasmic RBM10 staining was specific. The abundance of nuclear and cytoplasmic RBM10 could have different functional meanings. A third possibility is that we assessed surgically resected specimens of pN1–N2 EGFR-Mt, whereas Nanjo et al. assessed unresectable EGFR-Mt lung adenocarcinoma. Considering these reasons, further IHC analyses using a highly specific monoclonal antibody against RBM10 are necessary.

The present study showed that RBM10 expression affected the prognosis of patients with pN1–N2 EGFR-Mt lung adenocarcinoma after curative resection because of the low response to first-line EGFR-TKIs, especially in patients with Ex21 lung adenocarcinoma. The response rate to EGFR-TKIs has been reported to be 75–80% (36,37). However, in some patients, the duration of response to EGFR-TKIs is short, and the prognosis is poor (10,38). Previous studies have reported a lower response to EGFR-TKIs and shorter PFS in patients with Ex21 lung adenocarcinoma than in patients with Ex19 lung adenocarcinoma (39,40). The higher frequency of compound mutations and lower frequency of EGFR variant alleles in Ex21 lung adenocarcinoma than in Ex19 lung adenocarcinoma were reported to be associated with the lower response to EGFR-TKIs in Ex21 lung adenocarcinoma (41,42). Nanjo et al. (16) reported a lower response to EGFR-TKIs in patients with Ex21 lung adenocarcinoma, which was associated with the higher frequency of RBM10 co-mutations in Ex21 than in Ex19 (3% vs. 15%) (16). The results of the present study suggest that RBM10 may be a novel target associated with EGFR-TKI sensitivity and prognosis after curative surgery in patients with pN1–N2 Ex21 lung adenocarcinoma. However, further biomolecular studies aimed at elucidating the functional role of RBM10 in pN1–N2 Ex21 lung adenocarcinoma are needed.

In the present study, PD-L1 expression was an independent prognostic factor for OS in patients with pN1–N2 EGFR-Mt lung adenocarcinoma after surgery. PFS was worse in the PD-L1 positive group than in PD-L1 negative group in patients with recurrent disease treated with first-line EGFR-TKIs, which was consistent with previous reports showing that PD-L1 positivity was associated with worse PFS and a lower objective response rate among patients with stage IV unresectable EGFR-Mt lung adenocarcinoma (19-21,43). A previous in vitro study reported that PD-L1 contributes to primary resistance to EGFR-TKIs in EGFR-Mt cells by inducing epithelial-mesenchymal transition via activation of the TGF-β/Smad pathway (20).

In a large-scale meta-analysis, PD-L1 was shown to be associated with worse OS in patients with resectable lung cancer (44); however, there are only a few reports on the association between prognosis and PD-L1 expression in EGFR-Mt lung adenocarcinoma after resection. Bai et al. (45) analyzed PD-L1 expression in 73 patients with p-stage I–IV EGFR-Mt lung adenocarcinoma using PD-L1 antibody clone E1L3N and reported that 19 patients were positive for PD-L1 (26.0%) and that PD-L1 was a poor prognostic factor for OS after resection. Takamochi et al. (46) analyzed PD-L1 expression in 438 patients with p-stage I–IV EGFR-Mt lung adenocarcinoma using PD-L1 antibody 22C3 and reported that 89 patients who were PD-L1 positive (20.8%) had poor RFS, although there was no difference in OS after surgery. The present study was the first to analyze PD-L1 expression in pN1–N2 EGFR-Mt lung adenocarcinoma and show that PD-L1 positivity is a poor prognostic factor for OS in patients with pN1–N2 EGFR-Mt lung adenocarcinoma.

This study has several limitations. First, this is a single-center retrospective study with a small sample size, and a large prospective study is warranted to draw firm conclusions. Second, because RBM10 and PD-L1 expression was analyzed in TMAs, errors may have been caused due to tumor heterogeneity. Third, as this study included patients with pN1–N2 EGFR-Mt lung adenocarcinoma, it is unclear whether the results apply to pathological lymph node-negative or unresectable stage IV EGFR-Mt lung adenocarcinoma. Fourth, this study did not analyze the correlation between RBM10 expression and RBM10 mutations. RBM10 mutations, including missense and frame shift mutations, were identified in 8–22% of lung adenocarcinomas (47-49). Thus, further studies are necessary to evaluate the correlation between cytoplasmic expression of RBM10 and RBM10 mutations in EGFR-Mt lung adenocarcinoma and the biomolecular function of cytoplasmic RBM10, especially in Ex21 lung adenocarcinoma.


Conclusions

High expression of RBM10 and PD-L1 positivity are poor prognostic factors for OS in patients with pN1–N2 EGFR-Mt lung adenocarcinoma after curative surgery. The response to EGFR-TKIs can be predicted based on PD-L1 expression in patients with recurrent EGFR-Mt lung adenocarcinoma and RBM10 expression in patients with recurrent Ex21 lung adenocarcinoma.


Acknowledgments

Mr. Nakamura Yoshiyasu and Ms. Yoshihara Mitsuyo, Kanagawa Cancer Center Research Institute are acknowledged for their excellent technical assistance.

Funding: This study was funded by a grant from Kanagawa Cancer Foundation and JSPS KAKENHI (Grant Number JP21K16519).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-355/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-355/dss

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-355/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-23-355/coif). TI reports receiving Grant from Kanagawa Cancer Foundation and JSPS KAKENHI (Grant Number JP21K16519). 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. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by institutional review board of Kanagawa Cancer Center (2019 Eki-174) and individual consent for this retrospective analysis was waived.

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. Kawaguchi T, Koh Y, Ando M, et al. Prospective Analysis of Oncogenic Driver Mutations and Environmental Factors: Japan Molecular Epidemiology for Lung Cancer Study. J Clin Oncol 2016;34:2247-57. [Crossref] [PubMed]
  2. Kohno T, Tsuta K, Tsuchihara K, et al. RET fusion gene: translation to personalized lung cancer therapy. Cancer Sci 2013;104:1396-400. [Crossref] [PubMed]
  3. Isaka T, Yokose T, Ito H, et al. Correlations Between the EGFR Mutation Status and Clinicopathological Features of Clinical Stage I Lung Adenocarcinoma. Medicine (Baltimore) 2015;94:e1784. [Crossref] [PubMed]
  4. Zhang YL, Yuan JQ, Wang KF, et al. The prevalence of EGFR mutation in patients with non-small cell lung cancer: a systematic review and meta-analysis. Oncotarget 2016;7:78985-93. [Crossref] [PubMed]
  5. Isaka T, Ito H, Yokose T, et al. Prognostic factors for relapse-free survival in stage IB-IIIA primary lung adenocarcinoma by epidermal growth factor receptor mutation status. BMC Cancer 2022;22:966. [Crossref] [PubMed]
  6. Isaka T, Nakayama H, Yokose T, et al. Epidermal Growth Factor Receptor Mutations and Prognosis in Pathologic N1-N2 Pulmonary Adenocarcinoma. Ann Thorac Surg 2016;102:1821-8. [Crossref] [PubMed]
  7. 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 2010;11:121-8. [Crossref] [PubMed]
  8. Yokoyama Y, Sonobe M, Yamada T, et al. Gefitinib treatment in patients with postoperative recurrent non-small-cell lung cancer harboring epidermal growth factor receptor gene mutations. Int J Clin Oncol 2015;20:1122-9. [Crossref] [PubMed]
  9. Isaka T, Ito H, Nakayama H, et al. Effect of epidermal growth factor receptor gene mutation on the prognosis of pathological stage II-IIIA (8th edition TNM classification) primary lung cancer after curative surgery. Lung Cancer 2021;162:128-34.
  10. Wang J, Wang B, Chu H, et al. Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFR mutations. Onco Targets Ther 2016;9:3711-26. [Crossref] [PubMed]
  11. Bechara EG, Sebestyén E, Bernardis I, et al. RBM5, 6, and 10 differentially regulate NUMB alternative splicing to control cancer cell proliferation. Mol Cell 2013;52:720-33. [Crossref] [PubMed]
  12. Loiselle JJ, Roy JG, Sutherland LC. RBM10 promotes transformation-associated processes in small cell lung cancer and is directly regulated by RBM5. PLoS One 2017;12:e0180258. [Crossref] [PubMed]
  13. Sun X, Jia M, Sun W, et al. Functional role of RBM10 in lung adenocarcinoma proliferation. Int J Oncol 2019;54:467-78. [Crossref] [PubMed]
  14. Guan G, Li R, Tang W, et al. Expression of RNA-binding motif 10 is associated with advanced tumor stage and malignant behaviors of lung adenocarcinoma cancer cells. Tumour Biol 2017;39:1010428317691740. [Crossref] [PubMed]
  15. Rintala-Maki ND, Goard CA, Langdon CE, et al. Expression of RBM5-related factors in primary breast tissue. J Cell Biochem 2007;100:1440-58. [Crossref] [PubMed]
  16. Nanjo S, Wu W, Karachaliou N, et al. Deficiency of the splicing factor RBM10 limits EGFR inhibitor response in EGFR-mutant lung cancer. J Clin Invest 2022;132:e145099. [Crossref] [PubMed]
  17. 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 2016;387:1540-50. [Crossref] [PubMed]
  18. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018-28. [Crossref] [PubMed]
  19. Lee CC, Soon YY, Leong CN, et al. Impact of programmed death-ligand 1 expression on the patients of stage IV non-small cell lung cancer harboring epidermal growth factor receptor mutation: a systematic review and meta-analysis. Acta Oncol 2020;59:1430-7. [Crossref] [PubMed]
  20. Zhang Y, Zeng Y, Liu T, et al. The canonical TGF-β/Smad signalling pathway is involved in PD-L1-induced primary resistance to EGFR-TKIs in EGFR-mutant non-small-cell lung cancer. Respir Res 2019;20:164. [Crossref] [PubMed]
  21. Yang CY, Liao WY, Ho CC, et al. Association between programmed death-ligand 1 expression, immune microenvironments, and clinical outcomes in epidermal growth factor receptor mutant lung adenocarcinoma patients treated with tyrosine kinase inhibitors. Eur J Cancer 2020;124:110-22. [Crossref] [PubMed]
  22. Matsukuma S, Yoshihara M, Kasai F, et al. Rapid and simple detection of hot spot point mutations of epidermal growth factor receptor, BRAF, and NRAS in cancers using the loop-hybrid mobility shift assay. J Mol Diagn 2006;8:504-12. [Crossref] [PubMed]
  23. Yoshida K, Yatabe Y, Park JY, et al. Prospective validation for prediction of gefitinib sensitivity by epidermal growth factor receptor gene mutation in patients with non-small cell lung cancer. J Thorac Oncol 2007;2:22-8.
  24. Malapelle U, Sirera R, Jantus-Lewintre E, et al. Profile of the Roche cobas® EGFR mutation test v2 for non-small cell lung cancer. Expert Rev Mol Diagn 2017;17:209-15. [Crossref] [PubMed]
  25. Atout S, Shurrab S, Loveridge C. Evaluation of the Suitability of RNAscope as a Technique to Measure Gene Expression in Clinical Diagnostics: A Systematic Review. Mol Diagn Ther 2022;26:19-37. [Crossref] [PubMed]
  26. Kanda Y. Investigation of the freely available easy-to-use software 'EZR' for medical statistics. Bone Marrow Transplant 2013;48:452-8. [Crossref] [PubMed]
  27. Zhang S, Bao Y, Shen X, et al. RNA binding motif protein 10 suppresses lung cancer progression by controlling alternative splicing of eukaryotic translation initiation factor 4H. EBioMedicine 2020;61:103067. [Crossref] [PubMed]
  28. Jin X, Di X, Wang R, et al. RBM10 inhibits cell proliferation of lung adenocarcinoma via RAP1/AKT/CREB signalling pathway. J Cell Mol Med 2019;23:3897-904. [Crossref] [PubMed]
  29. Zhao J, Sun Y, Huang Y, et al. Functional analysis reveals that RBM10 mutations contribute to lung adenocarcinoma pathogenesis by deregulating splicing. Sci Rep 2017;7:40488. [Crossref] [PubMed]
  30. Jung JH, Lee H, Cao B, et al. RNA-binding motif protein 10 induces apoptosis and suppresses proliferation by activating p53. Oncogene 2020;39:1031-40. [Crossref] [PubMed]
  31. Wang K, Bacon ML, Tessier JJ, et al. RBM10 Modulates Apoptosis and Influences TNF-α Gene Expression. J Cell Death 2012;5:1-19. [Crossref] [PubMed]
  32. Lengel HB, Mastrogiacomo B, Connolly JG, et al. Genomic mapping of metastatic organotropism in lung adenocarcinoma. Cancer Cell 2023;41:970-985.e3. [Crossref] [PubMed]
  33. Dvinge H, Kim E, Abdel-Wahab O, et al. RNA splicing factors as oncoproteins and tumour suppressors. Nat Rev Cancer 2016;16:413-30. [Crossref] [PubMed]
  34. Cao Y, Di X, Zhang Q, et al. RBM10 Regulates Tumor Apoptosis, Proliferation, and Metastasis. Front Oncol 2021;11:603932. [Crossref] [PubMed]
  35. Yamada H, Tsutsumi K, Nakazawa Y, et al. Src Family Tyrosine Kinase Signaling Regulates FilGAP through Association with RBM10. PLoS One 2016;11:e0146593. [Crossref] [PubMed]
  36. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:113-25. [Crossref] [PubMed]
  37. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947-57. [Crossref] [PubMed]
  38. Santoni-Rugiu E, Melchior LC, Urbanska EM, et al. Intrinsic resistance to EGFR-Tyrosine Kinase Inhibitors in EGFR-Mutant Non-Small Cell Lung Cancer: Differences and Similarities with Acquired Resistance. Cancers (Basel) 2019;11:923. [Crossref] [PubMed]
  39. Yang JC, Wu YL, Schuler M, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015;16:141-51. [Crossref] [PubMed]
  40. Zhang Y, Sheng J, Kang S, et al. Patients with exon 19 deletion were associated with longer progression-free survival compared to those with L858R mutation after first-line EGFR-TKIs for advanced non-small cell lung cancer: a meta-analysis. PLoS One 2014;9:e107161. [Crossref] [PubMed]
  41. Kohsaka S, Nagano M, Ueno T, et al. A method of high-throughput functional evaluation of EGFR gene variants of unknown significance in cancer. Sci Transl Med 2017;9:eaan6566. [Crossref] [PubMed]
  42. Gieszer B, Megyesfalvi Z, Dulai V, et al. EGFR variant allele frequency predicts EGFR-TKI efficacy in lung adenocarcinoma: a multicenter study. Transl Lung Cancer Res 2021;10:662-74. [Crossref] [PubMed]
  43. Lan B, Wang Y, Wu J, et al. The predictive and prognostic effects of PD-L1 expression on TKI treatment and survival of EGFR-mutant NSCLC: A meta-analysis. Medicine (Baltimore) 2021;100:e27038. [Crossref] [PubMed]
  44. Tuminello S, Sikavi D, Veluswamy R, et al. PD-L1 as a prognostic biomarker in surgically resectable non-small cell lung cancer: a meta-analysis. Transl Lung Cancer Res 2020;9:1343-60. [Crossref] [PubMed]
  45. Bai Y, Chen X, Hou L, et al. PD-L1 expression and its effect on clinical outcomes of EGFR-mutant NSCLC patients treated with EGFR-TKIs. Cancer Biol Med 2018;15:434-42. [Crossref] [PubMed]
  46. Takamochi K, Hara K, Hayashi T, et al. Programmed death-ligand 1 expression and its associations with clinicopathological features, prognosis, and driver oncogene alterations in surgically resected lung adenocarcinoma. Lung Cancer 2021;161:163-70. [Crossref] [PubMed]
  47. Yin LL, Wen XM, Li M, et al. A gene mutation in RNA-binding protein 10 is associated with lung adenocarcinoma progression and poor prognosis. Oncol Lett 2018;16:6283-92. [Crossref] [PubMed]
  48. Imielinski M, Berger AH, Hammerman PS, et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 2012;150:1107-20. [Crossref] [PubMed]
  49. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014;511:543-50. Erratum in: Nature 2014;514:262 Rogers, K [corrected to Rodgers, K]. Erratum in: Nature 2018;559:E12.
Cite this article as: Isaka T, Miyagi Y, Yokose T, Saito H, Kasajima R, Watabe K, Shigeta N, Kikunishi N, Shigefuku S, Murakami K, Adachi H, Nagashima T, Ito H. Impact of RBM10 and PD-L1 expression on the prognosis of pathologic N1–N2 epidermal growth factor receptor mutant lung adenocarcinoma. Transl Lung Cancer Res 2023;12(10):2001-2014. doi: 10.21037/tlcr-23-355

Download Citation