Skip to main content
Discover Oncology logoLink to Discover Oncology
. 2025 Mar 12;16:307. doi: 10.1007/s12672-025-02089-y

The role of PD-L1 in EGFR-mutant non-small cell lung cancer

Wentao Gao 1, Lingling Wang 1, Yanyan Zhao 1, Lucheng Zhu 2,
PMCID: PMC11904073  PMID: 40072720

Abstract

Lung cancer remains the leading cause of cancer-related deaths globally. In China, nearly half of non-small cell lung cancer (NSCLC) patients carry epidermal growth factor receptor (EGFR) mutations. EGFR tyrosine kinase inhibitors (EGFR-TKIs) have significantly improved the prognosis for patients with EGFR mutations and are considered the preferred treatment for these individuals. Programmed death-ligand 1 (PD-L1) expression levels are now widely used as a biomarker to predict the effectiveness of PD-1/PD-L1 immunotherapy in NSCLC. Additionally, the impact of PD-L1 expression on the efficacy of EGFR-TKIs has garnered considerable attention from researchers. This review explores recent studies on the epidemiology and clinical outcomes associated with PD-L1 in NSCLC patients harboring driver gene mutations.

Keywords: Non-small cell lung carcinoma, Molecular targeted therapy, PD-L1

Introduction

Programmed death-ligand 1 (PD-L1) is a crucial predictive marker for immunotherapy in non-small-cell lung cancer (NSCLC). Multiple studies have demonstrated that PD-L1-positive NSCLC patients benefit from PD-1/PD-L1 inhibitors, either alone or in combination with chemotherapy [14]. For patients with low or negative PD-L1 expression, the KEYNOTE-189 and KEYNOTE-407 trials revealed that combining chemotherapy with PD-1/PD-L1 inhibitors offers additional survival advantages [2, 3].

However, the situation is more complex in NSCLC patients with epidermal growth factor receptor (EGFR) mutations. EGFR tyrosine kinase inhibitors (TKIs) are the standard treatment for advanced NSCLC patients with EGFR-sensitive mutations, irrespective of PD-L1 expression. Several studies have indicated that EGFR-mutant patients generally do not benefit from immunotherapy [57]. Nevertheless, the ORIENT-31 and HARMONi-A studies have shown that immunotherapy can provide significant benefits for NSCLC patients who have developed resistance to EGFR-TKIs [8, 9].

A substantial proportion of EGFR-mutant NSCLC patients also express PD-L1. The effect of PD-L1 expression on the efficacy of EGFR-TKIs has attracted our attention. This article reviews the literature on the role of PD-L1 in EGFR-mutant NSCLC to enhance our understanding of the value of PD-L1 in this patient population.

PD-L1 expression in EGFR-mutant NSCLC

PD-L1 is an immune checkpoint protein expressed on both tumors and tumor-infiltrating immune cells. PD-L1 expression is typically classified as negative (< 1%), low (1–49%), and high (≥ 50%). As shown in Fig. 1, EGFR mutation as well as the use of EGFR-TKIs impacted PD-L1 expression [1012]. A global EXPRESS study analyzed PD-L1 expression levels in NSCLC patients, using tumor samples from 2368 patients across 18 countries. The study found that 52% of patients had a PD-L1 tumor proportion score (TPS) ≥ 1%, and 22% had a PD-L1 TPS ≥ 50% [10, 12]. A separate study on PD-L1 expression in advanced NSCLC patients in China revealed that 51.8% had a PD-L1 TPS ≥ 1%, and 21.5% had a PD-L1 TPS ≥ 50% [13]. These findings suggest that PD-L1 expression patterns are similar between global and Chinese populations.

Fig. 1.

Fig. 1

PD-L1 expression pattern in whole, EGFR-mutated, and TKIs resistant NSCLC patients

Among EGFR-mutant patients, 44% had a PD-L1 TPS ≥ 1%, while 13% had a PD-L1 TPS ≥ 50% [10]. Another study in China showed that 29.5% of NSCLC patients with EGFR mutations had a PD-L1 TPS between 1–49%, and 14.3% (30 out of 210) had a PD-L1 TPS ≥ 50% [13]. These data indicate that high PD-L1 expression is lower in patients with EGFR mutations compared to those with wild-type EGFR. Furthermore, several studies have demonstrated that NSCLC with uncommon EGFR mutations tends to show higher PD-L1 expression compared to those with common EGFR mutations (e.g., 19 DEL and 21 L858R mutations) [14, 15].

PD-L1 expression in NSCLC patients after developing resistance to TKIs remains complex and controversial [16]. Lin et al. reported that gefitinib reduced PD-L1 expression by inhibiting NF-κB signaling [17], while Jiang et al. found that osimertinib inhibited PD-L1 mRNA expression and induced PD-L1 protein degradation [18]. However, Isomoto et al. observed that among 134 EGFR-TKI-resistant patients, the proportion of patients with high PD-L1 expression increased from 14% to 28% following treatment [11]. The discrepancy of PD-L1 expression after osimertinib treatment might cause by several reasons. Firstly, initial osimertinib treatment might cause PD-L1 expression degradation, while sustained osimertinib cause drug resistance and activation of bypass pathway, inducing PD-L1 expression. Secondly, human tumor tissue is a complex environment, in vitro cannot fully simulate TME. PD-L1 is regulation by not only tumor cells by also tumor microenvironment [19]. Overall, changes in PD-L1 expression after EGFR-TKI treatment in NSCLC patients with EGFR mutations present a multifaceted issue, requiring further clinical and laboratory studies for comprehensive understanding.

Correlation between PD-L1 expression and TKI efficacy

PD-L1 is a critical biomarker for immunotherapy, but its significance in patients receiving TKIs (Tyrosine Kinase Inhibitors) remains unclear. A retrospective study analyzed 186 EGFR-mutant lung adenocarcinoma patients treated with gefitinib, erlotinib, or afatinib [20]. The results indicated that patients with high PD-L1 expression had significantly shorter median progression-free survival (PFS) (6.6 vs. 13.0 months, p < 0.0001) and overall survival (OS) (11.5 vs. 32.9 months, p < 0.0001) compared to those with low or negative PD-L1 expression. Additionally, the objective response rate (ORR) was lower in patients with high PD-L1 expression (57% vs. 76%).

Another retrospective study reinforced this finding, showing that high PD-L1 expression was associated with a significantly shorter median PFS in response to EGFR-TKIs (6.0 vs. 9.5 vs. 3.8 months, p < 0.001) [21]. Multiple studies have consistently demonstrated a correlation between high PD-L1 expression and poor response to EGFR-TKIs (Table 1) [10, 2225].

Table 1.

Literature on the correlation between PD-L1 expression and clinical efficacy of EGFR-TKIs

Study Numbers EGFR-TKIs PD-L1 Antibody Cutoff Line of therapy Clinical outcomes in PD-L1 high expression
D'Incecco 2015 95 Gefitinib, erlotinib AB58810 (Abcam) 5% staining ≥ 1 Higher ORR and better TTP [32]
Lin 2015 56 Gefitinib, erlotinib AB58810 (Abcam) H score:mean 1 Better PFS and higher DCR [13]
Tang 2015 99 Most 1-generation TKIs E1L3N (CST) H score: 5 Not mentioned No impact on ORR and PFS [33]
Soo 2017 90 1st line EGFR TKI SP142 (Ventana) H score: 109 1 Poor PFS [34]
Yoneshima 2018 71 Not mentioned 22C3 (Dako) 1% of TPS 1 Poor PFS [35]
TC3/IC3: > 50%/10%
Su 2018 101 Most 1-generation TKIs SP142 (Ventana) TC1-2/IC1-2:5–49%/5–9% 1 Significantly lower ORR and PFS [21]
TC0/IC0: < 5%/5%
Hsu 2019 123 Gefitinib, erlotinib, afatinib SP263 (Ventana) 1%, 25%, 50% TPS 1 Shorter PFS and higher primary resistance rate [36]
Yang 2020 153 Gefitinib, erlotinib, afatinib 22C3 (Dako) 1%, 50% TPS 1, > 2 Lower ORR and shorter PFS [37]
Inomata 2020 47 Gefitinib, erlotinib, afatinib, osimertinib 22C3 (Dako) 1% of TPS 1 Poor PFS [22]
Yoon 2020 131 Gefitinib, erlotinib, afatinib 22C3 (Dako) 1%, 50% TPS 1 Poor PFS,OS and lower acquired T790M mutations rate [10]
Brown 2020 54 Osimertinib SP263 (Ventana) 1%, 25%, 50% TC 1 No difference in ORR and DoR [26]
Liu 2021 186 Gefitinib, erlotinib, afatinib SP263 (Ventana) 1%, 50% TPS 1 Poor PFS, OS [20]
Shiozawa 2022 158 Osimertinib Not available 1%, 50% TPS 1 Poor PFS [38]
Yang 2024 134 Osimertinib SP263 (Ventana) 1%, 50% TC  ≥ 2 No difference in ORR, DCR, PFS, and OS [25]

IHC, Immunohistochemistry; H score, histological score; TPS, tumour proportion score; TC, Tumor cell; ORR, objective response rate; TTP, time to progression; PFS, progression-free survival; DCR, disease-control rate; OS, overall survival; PD-L1, programmed death ligand-1

However, some studies present contradictory findings. For example, Lin et al. reported that PD-L1 expression was correlated with longer PFS and OS in advanced lung adenocarcinoma patients with EGFR mutations following EGFR-TKI treatment [13]. Similarly, the FLAURA study showed no significant difference in PFS between PD-L1 positive (≥ 1%) and PD-L1 negative patients [26].

These inconsistencies could be attributed to several factors. First, different studies employed various PD-L1 antibodies and cutoff values. Notably, studies utilizing the approved 22C3, SP142, and SP263 antibodies generally support the association between high PD-L1 expression and poor TKI efficacy in NSCLC. Second, first- and second-generation TKIs yielded consistent results regarding poor outcomes in patients with high PD-L1 expression, whereas osimertinib showed mixed results. For instance, the FLAURA study found similar efficacy for osimertinib in PD-L1 positive and negative patients, albeit with a small sample size. Conversely, a larger retrospective study demonstrated poor efficacy of osimertinib in PD-L1 positive NSCLC patients [27]. Third, PD-L1 is a dynamic biomarker, influenced by prior treatments. For patients previously treated with EGFR-TKIs, PD-L1 expression levels may fluctuate [11, 18].

Taken together, most studies suggest that high PD-L1 expression negatively impacts the efficacy of EGFR-TKIs, especially first- and second-generation EGFR-TKIs.

Possible mechanism between PD-L1 and EGFR-TKIs resistance

Li et al. demonstrated that PD-L1 promotes cell proliferation by regulating the cell cycle and inhibiting apoptosis [28]. The study also revealed that PD-L1 induces autophagy through the mitogen-activated protein kinase (MAPK) signaling pathway, leading to primary resistance to gefitinib. Chen et al. found that EGFR mutations induce PD-L1 expression via the p-ERK1/2/p–c-Jun pathway, and increased PD-L1 expression leads to T-cell apoptosis through the PD-L1/PD-1 axis [29]. Additionally, another EGFR-TKI resistance mechanism, MET activation, was shown to promote PD-L1 expression [30]. Despite these findings, the precise mechanisms by which PD-L1 regulates resistance to EGFR-TKIs remain unclear.

In EGFR-TKI naïve patients, high PD-L1 expression correlates with activation of alternative oncogenic which confers primary resistance [31]. While in patients with acquired T790M resistance, PD-L1 expression was often the consequence of acquired resistance, which had limited impact on subsequent EGFR-TKIs.

Conclusion

PD-L1 expression levels can serve as a biomarker for predicting and evaluating the efficacy of EGFR-TKIs in NSCLC patients with EGFR mutations. Studies indicate that high PD-L1 expression is associated with a poor prognosis in patients receiving EGFR-TKI treatment.

Acknowledgements

Not applicable.

Author contributions

Writing of the original draft and editing: WTG, and LCZ; methodology and data acquisition: LLW; data analyses and interpretation: LCZ and YYZ; study concept and design, project administration: LCZ; manuscript revision: WTG and LLW. All the authors have read and approved the final manuscript.

Funding

This study was supported by grants from the Youth Science and Technology Innovation Training Project of Hangzhou Cancer Hospital (HZCH2021QN01) and National Natural Science Foundation of China (82373889). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Data availability

No datasets were generated or analysed during the current study.

Declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.de Castro G, Kudaba I, Wu YL, Lopes G, Kowalski DM, Turna HZ, Caglevic C, Zhang L, Karaszewska B, Laktionov KK, et al. Five-year outcomes with pembrolizumab versus chemotherapy as first-line therapy in patients with non-small-cell lung cancer and programmed death ligand-1 tumor proportion score ≥ 1% in the KEYNOTE-042 study. J Clin Oncol. 2023;41(11):1986–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gandhi L, Rodríguez-Abreu D, Gadgeel S, Esteban E, Felip E, De Angelis F, Domine M, Clingan P, Hochmair MJ, Powell SF, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med. 2018;378(22):2078–92. [DOI] [PubMed] [Google Scholar]
  • 3.Paz-Ares L, Luft A, Vicente D, Tafreshi A, Gümüş M, Mazières J, Hermes B, Çay Şenler F, Csőszi T, Fülöp A, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med. 2018;379(21):2040–51. [DOI] [PubMed] [Google Scholar]
  • 4.Zhou C, Wang Z, Sun Y, Cao L, Ma Z, Wu R, Yu Y, Yao W, Chang J, Chen J, et al. Sugemalimab versus placebo, in combination with platinum-based chemotherapy, as first-line treatment of metastatic non-small-cell lung cancer (GEMSTONE-302): interim and final analyses of a double-blind, randomised, phase 3 clinical trial. Lancet Oncol. 2022;23(2):220–33. [DOI] [PubMed] [Google Scholar]
  • 5.Lee CK, Man J, Lord S, Cooper W, Links M, Gebski V, Herbst RS, Gralla RJ, Mok T, Yang JC. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):210–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lisberg A, Cummings A, Goldman JW, Bornazyan K, Reese N, Wang T, Coluzzi P, Ledezma B, Mendenhall M, Hunt J, et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naïve patients with advanced NSCLC. J Thorac Oncol. 2018;13(8):1138–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T, Kowalski DM, Dols MC, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389(10066):255–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lu S, Wu L, Jian H, Cheng Y, Wang Q, Fang J, Wang Z, Hu Y, Han L, Sun M, et al. Sintilimab plus chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer with disease progression after EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): second interim analysis from a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Respir Med. 2023;11(7):624–36. [DOI] [PubMed] [Google Scholar]
  • 9.Fang W, Zhao Y, Luo Y, Yang R, Huang Y, He Z, Zhao H, Li M, Li K, Song Q et al. Ivonescimab plus chemotherapy in non-small cell lung cancer with EGFR variant: a randomized clinical trial. Jama. 2024. [DOI] [PMC free article] [PubMed]
  • 10.Yoon BW, Chang B, Lee SH. High PD-L1 expression is associated with unfavorable clinical outcome in EGFR-mutated lung adenocarcinomas treated with targeted therapy. Onco Targets Ther. 2020;13:8273–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Isomoto K, Haratani K, Hayashi H, Shimizu S, Tomida S, Niwa T, Yokoyama T, Fukuda Y, Chiba Y, Kato R, et al. Impact of EGFR-TKI treatment on the tumor immune microenvironment in EGFR mutation-positive non-small cell lung cancer. Clin Cancer Res. 2020;26(8):2037–46. [DOI] [PubMed] [Google Scholar]
  • 12.Dietel M, Savelov N, Salanova R, Micke P, Bigras G, Hida T, Antunez J, Guldhammer Skov B, Hutarew G, Sua LF, et al. Real-world prevalence of programmed death ligand 1 expression in locally advanced or metastatic non-small-cell lung cancer: the global, multicenter EXPRESS study. Lung Cancer. 2019;134:174–9. [DOI] [PubMed] [Google Scholar]
  • 13.Lin C, Chen X, Li M, Liu J, Qi X, Yang W, Zhang H, Cai Z, Dai Y, Ouyang X. Programmed death-ligand 1 expression predicts tyrosine kinase inhibitor response and better prognosis in a cohort of patients with epidermal growth factor receptor mutation-positive lung adenocarcinoma. Clin Lung Cancer. 2015;16(5):e25-35. [DOI] [PubMed] [Google Scholar]
  • 14.Evans M, O’Sullivan B, Hughes F, Mullis T, Smith M, Trim N, Taniere P. The clinicopathological and molecular associations of PD-L1 expression in non-small cell lung cancer: analysis of a series of 10,005 cases tested with the 22C3 assay. Pathol Oncol Res. 2020;26(1):79–89. [DOI] [PubMed] [Google Scholar]
  • 15.Cho JH, Zhou W, Choi YL, Sun JM, Choi H, Kim TE, Dolled-Filhart M, Emancipator K, Rutkowski MA, Kim J. Retrospective molecular epidemiology study of PD-L1 expression in patients with EGFR-mutant non-small cell lung cancer. Cancer Res Treat. 2018;50(1):95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chen Q, Xia L, Wang J, Zhu S, Wang J, Li X, Yu Y, Li Z, Wang Y, Zhu G, et al. EGFR-mutant NSCLC may remodel TME from non-inflamed to inflamed through acquiring resistance to EGFR-TKI treatment. Lung Cancer. 2024;192: 107815. [DOI] [PubMed] [Google Scholar]
  • 17.Lin K, Cheng J, Yang T, Li Y, Zhu B. EGFR-TKI down-regulates PD-L1 in EGFR mutant NSCLC through inhibiting NF-κB. Biochem Biophys Res Commun. 2015;463(1–2):95–101. [DOI] [PubMed] [Google Scholar]
  • 18.Jiang XM, Xu YL, Huang MY, Zhang LL, Su MX, Chen X, Lu JJ. Osimertinib (AZD9291) decreases programmed death ligand-1 in EGFR-mutated non-small cell lung cancer cells. Acta Pharmacol Sin. 2017;38(11):1512–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Inoue C, Miki Y, Saito R, Hata S, Abe J, Sato I, Okada Y, Sasano H: PD-L1 induction by cancer-associated fibroblast-derived factors in lung adenocarcinoma cells. Cancers (Basel). 2019;11(9):1257. [DOI] [PMC free article] [PubMed]
  • 20.Liu J, Itchins M, Nagrial A, Cooper WA, De Silva M, Barnet M, Varikatt W, Sivasubramaniam V, Davis A, Gill AJ, et al. Relationship between PD-L1 expression and outcome in EGFR-mutant lung cancer patients treated with EGFR tyrosine kinase inhibitors. Lung Cancer. 2021;155:28–33. [DOI] [PubMed] [Google Scholar]
  • 21.Su S, Dong ZY, Xie Z, Yan LX, Li YF, Su J, Liu SY, Yin K, Chen RL, Huang SM, et al. Strong programmed death ligand 1 expression predicts poor response and de novo resistance to EGFR tyrosine kinase inhibitors among NSCLC patients with EGFR mutation. J Thorac Oncol. 2018;13(11):1668–75. [DOI] [PubMed] [Google Scholar]
  • 22.Inomata M, Azechi K, Takata N, Hayashi K, Tokui K, Taka C, Okazawa S, Kambara K, Imanishi S, Miwa T et al. Association of tumor PD-L1 expression with the T790M mutation and progression-free survival in patients with egfr-mutant non-small cell lung cancer receiving EGFR-TKI therapy. Diagnostics (Basel). 2020;10(12). [DOI] [PMC free article] [PubMed]
  • 23.Hsu KH, Tseng JS, Yang TY, Chen KC, Su KY, Yu SL, Chen JJW, Huang YH, Chang GC. PD-L1 strong expressions affect the clinical outcomes of osimertinib in treatment naïve advanced EGFR-mutant non-small cell lung cancer patients. Sci Rep. 2022;12(1):9753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Papazyan T, Denis MG, Sagan C, Raimbourg J, Herbreteau G, Pons-Tostivint E. Impact of PD-L1 expression on the overall survival of caucasian patients with advanced EGFR-mutant NSCLC treated with frontline osimertinib. Target Oncol. 2024;19(4):611–21. [DOI] [PubMed] [Google Scholar]
  • 25.Yang CY, Liao WY, Ho CC, Chen KY, Tsai TH, Hsu CL, Su KY, Chang YL, Wu CT, Hsu CC, et al. PD-L1 expression and immune profiling cannot predict osimertinib efficacy in lung cancer with EGFR T790 M mutation: a translational study. J Formos Med Assoc. 2024. [DOI] [PubMed]
  • 26.Brown H, Vansteenkiste J, Nakagawa K, Cobo M, John T, Barker C, Kohlmann A, Todd A, Saggese M, Chmielecki J, et al. Programmed cell death ligand 1 expression in untreated EGFR mutated advanced NSCLC and response to osimertinib versus comparator in FLAURA. J Thorac Oncol. 2020;15(1):138–43. [DOI] [PubMed] [Google Scholar]
  • 27.Sakata Y, Sakata S, Oya Y, Tamiya M, Suzuki H, Shibaki R, Okada A, Kobe H, Matsumoto H, Yokoi T, et al. Osimertinib as first-line treatment for advanced epidermal growth factor receptor mutation-positive non-small-cell lung cancer in a real-world setting (OSI-FACT). Eur J Cancer. 2021;159:144–53. [DOI] [PubMed] [Google Scholar]
  • 28.Li N, Zuo R, He Y, Gong W, Wang Y, Chen L, Luo Y, Zhang C, Liu Z, Chen P, et al. PD-L1 induces autophagy and primary resistance to EGFR-TKIs in EGFR-mutant lung adenocarcinoma via the MAPK signaling pathway. Cell Death Dis. 2024;15(8):555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen N, Fang W, Zhan J, Hong S, Tang Y, Kang S, Zhang Y, He X, Zhou T, Qin T, et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol. 2015;10(6):910–23. [DOI] [PubMed] [Google Scholar]
  • 30.Saigi M, Alburquerque-Bejar JJ, Mc Leer-Florin A, Pereira C, Pros E, Romero OA, Baixeras N, Esteve-Codina A, Nadal E, Brambilla E, et al. MET-oncogenic and JAK2-inactivating alterations are independent factors that affect regulation of PD-L1 expression in lung cancer. Clin Cancer Res. 2018;24(18):4579–87. [DOI] [PubMed] [Google Scholar]
  • 31.Hsu PC, Jablons DM, Yang CT, You L. Epidermal growth factor receptor (EGFR) pathway, yes-associated protein (YAP) and the regulation of programmed death-ligand 1 (PD-L1) in non-small cell lung cancer (NSCLC). Int J Mol Sci. 2019;20(15):3821. [DOI] [PMC free article] [PubMed]
  • 32.D’Incecco A, Andreozzi M, Ludovini V, Rossi E, Capodanno A, Landi L, Tibaldi C, Minuti G, Salvini J, Coppi E, et al. PD-1 and PD-L1 expression in molecularly selected non-small-cell lung cancer patients. Br J Cancer. 2015;112(1):95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Tang Y, Fang W, Zhang Y, Hong S, Kang S, Yan Y, Chen N, Zhan J, He X, Qin T, et al. The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Oncotarget. 2015;6(16):14209–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Soo RA, Kim HR, Asuncion BR, Fazreen Z, Omar MFM, Herrera MC, Yun Lim JS, Sia G, Soong R, Cho BC. Significance of immune checkpoint proteins in EGFR-mutant non-small cell lung cancer. Lung Cancer. 2017;105:17–22. [DOI] [PubMed] [Google Scholar]
  • 35.Yoneshima Y, Ijichi K, Anai S, Ota K, Otsubo K, Iwama E, Tanaka K, Oda Y, Nakanishi Y, Okamoto I. PD-L1 expression in lung adenocarcinoma harboring EGFR mutations or ALK rearrangements. Lung Cancer. 2018;118:36–40. [DOI] [PubMed] [Google Scholar]
  • 36.Hsu KH, Huang YH, Tseng JS, Chen KC, Ku WH, Su KY, Chen JJW, Chen HW, Yu SL, Yang TY, et al. High PD-L1 expression correlates with primary resistance to EGFR-TKIs in treatment naïve advanced EGFR-mutant lung adenocarcinoma patients. Lung Cancer. 2019;127:37–43. [DOI] [PubMed] [Google Scholar]
  • 37.Yang CY, Liao WY, Ho CC, Chen KY, Tsai TH, Hsu CL, Su KY, Chang YL, Wu CT, Hsu 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. [DOI] [PubMed] [Google Scholar]
  • 38.Shiozawa T, Numata T, Tamura T, Endo T, Kaburagi T, Yamamoto Y, Yamada H, Kikuchi N, Saito K, Inagaki M, et al. Prognostic implication of PD-L1 expression on osimertinib treatment for EGFR-mutated non-small cell lung cancer. Anticancer Res. 2022;42(5):2583–90. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Discover Oncology are provided here courtesy of Springer

RESOURCES