• Open Access
    Review

    It might be a dead end: immune checkpoint inhibitor therapy in EGFR-mutated NSCLC

    Ken Akao
    Yuko Oya *
    Takaya Sato
    Aki Ikeda
    Tomoya Horiguchi
    Yasuhiro Goto
    Naozumi Hashimoto
    Masashi Kondo
    Kazuyoshi Imaizumi

    Explor Target Antitumor Ther. 2024;5:826–840 DOI: https://doi.org/10.37349/etat.2024.00251

    Received: December 05, 2023 Accepted: April 24, 2024 Published: July 19, 2024

    Academic Editor: Nicola Normanno, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Italy

    This article belongs to the special issue Novel Strategies and Targets for Immunotherapy of Cancer

    Abstract

    Despite innovative advances in molecular targeted therapy, treatment strategies using immune checkpoint inhibitors (ICIs) for epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) have not progressed significantly. Accumulating evidence suggests that ICI chemotherapy is inadequate in this population. Biomarkers of ICI therapy, such as programmed cell death ligand 1 (PD-L1) and tumor-infiltrating lymphocytes (TILs), are not biomarkers in patients with EGFR mutations, and the specificity of the tumor microenvironment has been suggested as the reason for this. Combination therapy with PD-L1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors is a concern because of its severe toxicity and limited efficacy. However, early-stage NSCLC may differ from advanced-stage NSCLC. In this review, we comprehensively review the current evidence and summarize the potential of ICI therapy in patients with EGFR mutations after acquiring resistance to treatment with EGFR-tyrosine kinase inhibitors (TKIs) with no T790M mutation or whose disease has progressed on osimertinib.

    Keywords

    EGFR, immune checkpoint inhibitor, tumor microenvironment, PD-L1

    Introduction

    Epidermal growth factor receptor (EGFR) mutations are major genetic variants reported in lung adenocarcinomas, with reported incidences of approximately 50% in Asians and 10–15% in Caucasians [1, 2]. Lung cancer patients with EGFR mutations tend to have little or no smoking history. EGFR-tyrosine kinase inhibitors (TKIs) have been successfully developed for EGFR-mutated non-small cell lung cancer (NSCLC) and continue to stand as robust first-line treatments for advanced NSCLC harboring EGFR mutations [36]. Despite their efficacy, resistance to EGFR-TKIs occurs in almost all patients [7]. However, optimal treatment strategies have not yet been established.

    For patients who have developed resistance to EGFR-TKIs, treatment strategies based on resistance mechanisms are currently under investigation. However, at present, for patients with any EGFR mutation who have progressed on EGFR-TKIs with no T790M mutation or whose disease has progressed on osimertinib, treatment based on the nondriver mutation guideline may be offered [8].

    Unlike cytotoxic anticancer drugs, immune checkpoint inhibitor (ICI) therapy is an attractive option due to its potential for a durable response. However, EGFR-mutated NSCLC may be at a disadvantage for immunotherapy because of the lack of smoking history and low tumor mutation burden (TMB). Nevertheless, programmed cell death ligand 1 (PD-L1) expression has been reported to be increased in EGFR-mutant NSCLC [911]. Furthermore, in preclinical models, programmed death 1 (PD-1) antibody blockade improved survival in mice with EGFR-driven adenocarcinoma by enhancing effector T cell function and reducing the levels of tumor-promoting cytokines [12], suggesting that ICI treatment for EGFR-mutated cases may be beneficial. Therefore, many clinical trials have been conducted to investigate the efficacy of ICI therapy in patients with NSCLC with EGFR mutations.

    However, several clinical trials have shown that the efficacy of ICI therapy in patients with EGFR mutations is limited, and there are concerns regarding its toxicity. Pneumonitis has been reported to be enhanced by the concomitant or sequential use of ICIs [1315]. Therefore, ICI therapy for patients with EGFR mutations is challenging. Recent data show a trend toward the addition of vascular endothelial growth factor (VEGF) being effective in EGFR-mutated cases; however, this is not conclusive.

    In addition, ICIs have recently been introduced for locally advanced NSCLC and advanced stages [1618]. However, whether immune-oncology (IO) should be administered to patients with EGFR mutations at this stage needs to be considered. Thus, summarizing and discussing the findings regarding ICI treatment for patients with EGFR mutations may help us to consider whether patients with EGFR-mutated locally advanced NSCLC should be treated with ICI. In this review, we summarize the treatments, including ICI, and consider whether they are necessary for patients or not.

    Mechanisms and subsequent strategies for EGFR-TKI resistance

    Although EGFR-TKIs are the first-line treatment for EGFR-mutated NSCLC, due to their impressive clinical efficacy, almost all patients develop resistance to EGFR-TKIs [7, 19]. Several mechanisms have been reported for the acquired resistance to EGFR-TKIs. One example is the T790M second mutation, which is a resistance mechanism against first- and second-generation EGFR-TKIs. T790M is reported in approximately 50% of patients who acquire resistance to first-/second-generation EGFR-TKIs [20, 21]. Osimertinib, a third-generation EGFR-TKI developed to overcome this resistance, has shown efficacy against T790M-positive EGFR-mutated NSCLC and has been approved as a second-line treatment [22]. However, osimertinib was superior to first-generation EGFR-TKIs in the FLAURA trial and has been approved as a first-line therapy in many countries (in certain countries, only as second-line therapy). The mechanisms underlying osimertinib resistance are more diverse than those of the first or second generation [2325]. Typical examples include MET amplification, C797S mutations, and signaling pathways other than EGFR, such as RET [23, 26]. Many attempts have been made to overcome these mutations as a strategy for osimertinib resistance, including the ORCHARD trial, which used an adaptation strategy for each resistance mutation, and patritumab deruxtecan (HER3-Dxd) and amivantamab plus lasertinib combination therapies, which target broad resistance [2729]. Prolonged overall survival (OS) has been reported in patients who received platinum-doublet chemotherapy and EGFR-TKIs [30, 31]. As resistance to targeted therapy is expected to develop at a certain point, cytotoxic chemotherapy will continue to hold a prominent position in the treatment of EGFR-mutated NSCLC.

    Furthermore, it has been reported that cancers generally become more heterogeneous during the disease [32, 33]. Therefore, the need for broader strategies, such as cytotoxic chemotherapy or immunotherapy, may strengthen after the first and second treatments fail.

    ICI monotherapy for EGFR-mutated NSCLC

    The KEYNOTE-001 study was the first attempt to investigate the efficacy of ICIs as a first-line treatment for patients with EGFR-mutated NSCLC. The objective response rate (ORR) to pembrolizumab in four EGFR-TKI-naive patients was 50%, with a median progression free survival (mPFS) of 157.5 days and a median OS (mOS) of 559 days. By contrast, the efficacy was limited in 26 patients previously treated with EGFR-TKIs (ORR, 4%; mPFS, 56 days; mOS, 120 days) [34]. These results led to a phase II trial of pembrolizumab in TKI-naive patients expressing PD-L1. However, the interim analysis did not show the efficacy of pembrolizumab in this population; the ORR in the first 10 patients was 0% and the study was terminated early and considered invalid [35]. CheckMate012 reported that nivolumab monotherapy was less effective in patients with EGFR mutations than those with EGFR wild-type (ORR: 14% vs. 30%; mPFS: 1.8 months vs. 8.8 months; mOS: 18.8 months vs. not reached) [36]. Furthermore, in a meta-analysis of phase II/III trials comparing ICIs (nivolumab, pembrolizumab, and, atezolizumab) with docetaxel in the second-line treatment of NSCLC, the OS of ICIs vs. digital therapeutics (DTX) in patients with EGFR mutations was hazard ratio (HR) 1.11 [95% confidence intervals (CI): 0.80–1.53, P = 0.54], indicating that although efficacy has been demonstrated in the overall population, treatment with ICIs is not superior in patients with EGFR mutation-positive [37].

    Compared to platinum doublet, in a phase II study (WJOG8515L) comparing carboplatin (CBDCA) plus pemetrexed and nivolumab as second-line therapy after EGFR-TKI failure, nivolumab showed a shorter PFS than CBDCA plus pemetrexed and did not show a survival benefit [38]. In a retrospective analysis (an immunotarget study) investigating the efficacy of ICI monotherapy for each driver gene mutation, the ORR of ICI was 12.0% and the PFS was only 2.1 months (95% CI: 1.8–2.7) in patients with EGFR mutations [39]. The BIRCH and ATRANTIC trials investigated ICI monotherapy in patients with EGFR-mutated PD-L1. In both trials, ICI monotherapy was less effective in EGFR-mutated cases than in wild-type NSCLC [40, 41].

    These results indicate that treatment with ICIs is effective in the overall population, but not in patients with EGFR mutations.

    The data from these trials (Table 1) suggests that EGFR-mutant NSCLC is less effective than ICI monotherapy. In addition, in a retrospective study, we showed that high PD-L1 expression might not be a predictor of response in patients with EGFR/anaplastic lymphoma kinase (ALK) mutations [42]. A few reports indicate that EGFR is immunologically “cold”, and that the tumor microenvironment (TME) is unfavorable to ICI therapy. Tumors that do not elicit a strong immune response and do not usually respond to immunotherapy are called “cold” tumors. These tumors tend to be surrounded by cells that can suppress the immune response, making it difficult for T cells to attack the tumor cells. Therefore, these TMEs may be responsible for the poor efficacy of ICI monotherapy in EGFR-mutated NSCLC. These trials are summarized in Table 1.

    Immune checkpoint inhibitors monotherapy for EGFR-mutated NSCLC

    TreatmentStudy nameSettingDrugsPhaseEfficacyAEsReference
    IO monotherapyKEYNOTE-001PretreatedPembrolizumabIORR 50%, mPFS of 157.5 days in four EGFR-TKI-naive patients; ORR 4%, mPFS 56 days in EGFR-TKI treated patients.No report for EGFR patients.[34]
    NCT028799941stPembrolizumabIIORR 0%.TRAE: 46%, no grade 4–5 (38%). 6/7 patients had a TRAE on second-line EGFR-TKI.[35]
    Checkmate0122ndNivolumabIORR: 14% vs. 30%; mPFS: 1.8 months vs. 8.8 months.Grade 3–4 in 14 (37%), no G5, in the ITT population.[36]
    WJOG8515L2ndNivolumab vs. Cb + pemetrexedIINivolumab/Cb + pemetrexed, ORR 9.6% vs. 36.0%, mPFS 1.7 months vs. 5.6 months.Serious AEs: 25.5% in nivolumab and 16.0% in chemotherapy.[39]
    BIRCH1st to 3rdAtezolizumabIIORRs for mutant/wild-type in cohorts 1, 2, and 3 were 23%/19%, 0%/21%, and 7%/18%, respectively.Grade 3 to 4 AEs occurred in 42% of patients (12% treatment-related).[40]
    ATRANTICLess than 3rdDurvalumabIIORR was 12%.Treatment-related serious adverse events occurred in 5%.[41]
    Dual-IONCT030914912ndNivolumab vs. nivolumab + ipillimumabIORR 3.2%, PFS 1.22 months in overall cohort.2/31 of grade 3 TRAE in the overall cohort[43]
    KEYNOTE 021Less than 2ndPembrolizumab plus ipilimumabI/IIOne of the 12 patients showed an objective response.-[44]
    Display full size

    IO: immune-oncology; ORR: objective response rate; EGFR: epidermal growth factor receptor; TKIs: tyrosine kinase inhibitors; TRAE: treatment related adverse event; mPFS: median progression free survival; ITT: intent-to-treat; AEs: adverse events; ALK: anaplastic lymphoma kinase. -: no data

    ICIs + EGFR-TKI therapy

    The CheckMate012 trial evaluated the combination of nivolumab and erlotinib in 21 patients with EGFR-mutant NSCLC. The PFS of patients previously treated with EGFR-TKIs (n = 20) was 5.1 months. Responders were PD-L1 positive or PD-L1 status unknown. No grade 4 or 5 treatment related adverse events (TRAEs) were reported, and 2/21 patients discontinued the study due to toxicity [45]. In contrast, the TATTON trial, which evaluated a combination of osimertinib and durvalumab, raised serious safety concerns. In this trial, 48% of the patients developed at least one grade 3 TRAE, 5/23 developed interstitial lung disease, and all patients discontinued the trial [14]. Furthermore, the CAURAL trial comparing osimertinib with durvalumab as second-line treatment was stopped early due to the early discontinuation of the TATTON trial reported at the same time, and one patient developed pneumonitis [46]. In addition, the combination of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and EGFR-TKI for previously treated EGFR-mutant NSCLC was investigated, but most studies were discontinued in the early phase because of low efficacy and toxicity (e.g., tremelimumab and gefitinib [47], ipilimumab and EGFR-TKI or ALK-TKI [44], erlotinib and atezolizumab [48], and pembrolizumab plus gefitinib or erlotinib [49]). These trials are summarized in Table 2.

    Immune checkpoint inhibitors + EGFR-TKI therapy

    Study nameSettingDrugsPhaseEfficacyAEsReferences
    CheckMate012≥ 2ndNivolumab and erlotinibIbORR 15%, DCR 65%, PFS: 5.1 monthsG3: 24%, no G4 or G5 TRAEs[49]
    TATTON≥ 2ndDurvalumab + osimertinibIbORR 43%≥ G3: 48%; ILD occurred in 22% (≥ G3, 8.7%)[14]
    CAURAL≥ 2ndDurvalumab + osimertinibIIIORR 64% in the combination armNot sufficient data, grade 2 interstitial lung disease occurred in 1 patient.[46]
    NCT020400642ndTremelimumab and gefitinibIPFS of 2.2 monthsG3 TRAE 81%[50]
    NCT019981261stIpilimumab and erlotinibIPFS 27.8 monthsFour of the 11 patients had G3 colitis.[51]
    NCT020132191st and anyAtezolizumab + erlotinibIbPFS 15.4 monthsG3 46%, no G4 or G5 TRAE.[52]
    KEYNOTE 0211stPembrrolizumab + gefitinib (cohort F), Pembrrolizumab + erlotinib (cohort E)Phase I/IIORR 41.7% in cohort F and 14.3% in cohort EG3: 33.3% in cohort F, G3–4: 71.4% in cohort E[44]
    Display full size

    ORR: objective response rate; DCR: dacryocystorhinostomy; PFS: progression free survival; TRAEs: treatment related adverse event; ILD: interstitial lung disease; EGFR: epidermal growth factor receptor; TKI: tyrosine kinase inhibitor

    Dual ICI therapy for EGFR-mutated NSCLC

    The KEYNOTE 021 phase I/II study evaluated pembrolizumab plus ipilimumab as second-line or later therapy. In this study, 27% (12/44) of the patients harbored EGFR or ALK, of whom only one showed an objective response [44]. Although several trials have been conducted [43], severe toxicity concerns and limited efficacy were shown.

    ICIs + chemotherapy for EGFR-mutated NSCLC

    As a subgroup analysis of patients with EGFR mutations, in the CheckMate012 study evaluating the efficacy of nivolumab in combination with chemotherapy, mPFS and OS were shorter in the EGFR mutation arm than those in the wild-type arm (mPFS: 4.8 months vs. 7.5 months; mOS: 20.5 months vs. 24.5 months) [50]. Additionally, in the IMpower130 trial, CBDCA + nanoparticle albumin-bound paclitaxel (nab-PTX) + atezolizumab did not show superiority compared to chemotherapy in EGFR HR (mPFS: 7.0 months vs. 6.0 months, HR 0.75; 95% CI: 0.36–1.54; mOS: 14.4 months vs. 10.0 months, HR 0.98, 95% CI: 0.41–2.31) [51].

    In addition, the CheckMate722 and KEYNOTE789 trials validated chemotherapy + ICI treatment in patients with EGFR mutations. The CheckMate722 trial included 294 patients with EGFR-mutated NSCLC who had progressed with first- or second-generation EGFR-TKIs and did not have the T790M mutation and patients who had progressed with osimertinib with or without the T790M mutation. Nivolumab and chemotherapy could not show superiority in PFS and OS [52].

    The KEYNOTE789 study evaluated CBDCA + pemetrexed + pembrolizumab as a treatment after EGFR-TKI failure in NSCLC harboring EGFR-sensitive mutations (19del or L858R). Similar to the Checkmate722 study, patients who progressed to osimertinib and those who progressed to first- or second-generation EGFR-TKIs without T790M mutations were included in this study. The PFS was set to be achieved if HR was 0.70 or less, but resulting in HR 0.80 (95% CI: 0.65–0.97), and OS was set to be achieved if HR was 0.72 or less, but resulting in HR 0.84 (95% CI: 0.69–1.02), exceeding both primary endpoints could not be achieved. Subgroup analysis showed a slightly better OS in the PD-L1-positive group; however, none of the subgroups appeared particularly effective [53].

    The ILLUMINATE phase II study evaluated the combination of durvalumab and tremelimumab plus platinum-pemetrexed in EGFR-mutated NSCLC who had experienced disease progression with EGFR-TKIs. The study included T790M-negative cohort 1 and T790M-positive cohort 2. The ORR was 42% in cohort 1 and 35% in cohort 2, with mPFS of 6.5 months and 4.9 months, respectively, demonstrating modest efficacy for this population. In T790M-negative patients, high PD-L1 expression (PD-L1 ≥ 50%) was associated with greater efficacy compared with low expression [54]. These studies are summarized in Table 3.

    Immune checkpoint inhibitors + chemotherapy ± anti-VEGF antibodies for EGFR mutant NSCLC

    TreatmentStudy nameSettingDrugsPhaseEfficacyAEsReferences
    Chemotherapy + IOCheckMate0122ndNivolumab + platinum doubletIEGFR mutated vs. wild type, ORR: 17% vs. 47%, PFS: 4.8 months vs. 7.5 months, OS: 20.5 months vs. 24.5 months.G3–4: 50%, G5: 0%. (All patients, not only EGFR).[50]
    IMpower1302ndComparing CBDCA + nab-PTX + atezolizumab with CBDCA + nab-PTXIIIIn the subgroup of EGFR/ALK, the mPFS was 7.0 months vs. 6.0 months (HR 0.75, 95% CI: 0.36–1.54).G3–4: 81% in the combination arm. (All patients, not only EGFR).[51]
    CheckMate7222ndNivolumab plus chemotherapy vs. chemotherapyIIIPFS: 5.6 months in the nivolumab plus chemotherapy group and 5.4 months in the chemotherapy group.G3–4: 45% in nivolumab-based therapy and 29% in chemotherapy.[55]
    KEYNOTE7892ndPembrolizumab plus chemotherapy vs. chemotherapyIIIPFS: 5.6 months in the pembrolizumab plus chemotherapy group and 5.5 months in the chemotherapy group.G3 ≤ TRAE; 43.7%, irAE 4.5% in combination arm.[56]
    Chemotherapy + dual-IOILLUMINATE2ndDurvalumab and tremelimumab plus platinum-pemetrexedIIThe ORR was 42% in cohort 1 and 35% in cohort 2, with mPFS of 6.5 months and 4.9 months.G3–4 colitis 8%, hepatitis 4%, ILD 1%.[54]
    Chemotherapy + IO + anti-VEGFIMpower1502ndAtezolizumab, bevacizumab, carboplatin-paclitaxel (CP). Control arm: BCP, study arm: ACP, ABCPIIIORR 70.6% for ABCP, 35.6% for ACP, 41.9% for BCP.G3–4: 64% of ABCP, 68% of ACP, and 64% of BCP.[57, 58]
    Chemotherapy + IO + anti-VEGFORIENT2ndScintilimab, IBI305 (bevacizumab biosimilar), pemetrexed + cisplatin (PC). Arm A: SIPC, arm B: SPC, arm C: PC→SIIIConfirmed ORR were 43.9%, 33.1%, and 25.2% in arm A, B, and C, PFS 6.9 months for arm A, 5.5 months for arm B, 4.3 months for arm C.Grade ≥ 3 treatment-emergent AEs were 54.7% (arm A), 39.3% (arm B), and 51.0% (arm C).[55]
    Chemotherapy + IO + anti-VEGFIMpower1512ndAtezolizumab, bevacizumab, carboplatin-pemetrexed. Control arm: bevacizumab + carboplatin-pemetrexed, study arm: atezolizumab + carboplatin-pemetrexed, atezolizumab + bevacizumab + carboplatin-pemetrexed. Over half of the patients had EGFR/ALKIIIIn the subgroup of EGFR/ALK, the mPFS was 8.5 months (95% CI: 6.9–10.3) for atezolizumab + bevacizumab + carboplatin-pemetrexed and 8.3 months (95% CI: 6.9–10.1) for bevacizumab + carboplatin-pemetrexed (HR 0.86, 95% CI: 0.55–1.19).G3–4: 67.1% of ABCP, G5 5.9% in ABCP.[59]
    Display full size

    irAE: immune-related adverse events; CP: carboplatin-paclitaxel; BCP: bevacizumab carboplatin-paclitaxel; ACP: atezolizumab carboplatin-paclitaxel; PC: pemetrexed + cisplatin; SIPC: scintilimab + IBI305 + pemetrexed + cisplatin; SPC: scintilimab + pemetrexed + cisplatin; EGFR: epidermal growth factor receptor; TRAE: treatment related adverse event; mPFS: median progression free survival; ORR: objective response rate; IO: immune-oncology; OS: overall survival; CBDCA: carboplatin; nab-PTX: nanoparticle albumin-bound paclitaxel; VEGF: vascular endothelial growth factor; NSCLC: non-small cell lung cancer; HR: hazard ratio; CI: confidence intervals; TRAE: treatment related adverse event; ILD: interstitial lung disease; ALK: anaplastic lymphoma kinase. PC→S: pemetrexed + cisplatin → sincilimab

    ICIs + chemotherapy + anti-VEGF antibodies for EGFR-mutated NSCLC

    In a subgroup analysis of patients with EGFR mutation in a phase III trial (IMpower150) comparing CBDCA + PTX + bevacizumab + atezolizumab (ABCP) with CBDCA + PTX + bevacizumab (BCP) in the first-line treatment of non-squamous NSCLC, mOS was not reached vs. 18.7 months (HR 0.61, 95% CI: 0.29–1.28) and mPFS was 10.2 months vs. 6.9 months (HR 0.61, 95% CI: 0.36–1.28), showing a trend towards better treatment response in the atezolizumab combination group [60]. Furthermore, OS improvements were sustained with ABCP vs. BCP in sensitizing EGFR mutations in updated analysis (mOS 29.4 months vs. 18.1 months, HR 0.74, 95% CI: 0.38–1.46) [61]. However, this subgroup analysis was not planned in the protocol, and the presence of EGFR mutations was not set as a pre-planned stratification factor, which should be cautioned.

    The ORIENT-31 trial is the first prospective phase III trial to show the benefit of anti-PD-1 antibody plus chemotherapy in patients with EGFR-mutated NSCLC who have progressed after treatment with tyrosine kinase inhibitors. In this study, sintilimumab (PD-1 inhibitor) + IBI305 (anti-VEGF) + cisplatin + pemetrexed showed superiority in terms of PFS over chemotherapy and was well tolerated [55]. VEGF is involved in angiogenesis and the formation of a broad immunosuppressive environment. VEGF promotes Treg differentiation and proliferation and inhibits dendritic cell maturation [56]. The combination of PD-1/L1 inhibition and VEGF blockade enhances antigen-specific T-cell migration and modulates the expression of the CD8+ T-cell inhibitory checkpoint in tumors [57, 58, 62]. Therefore, the role of VEGF as an immunomodulator is expected, and elevated VEGF levels have been reported in EGFR-mutant NSCLC [63]. These associations between VEGF and the TME in EGFR-mutated NSCLC support the combined strategy of PD-1 and VEGF inhibition in EGFR cases. However, contradictory results have recently been reported. The IMpower151 trial was presented at the 2023 IASLC World Lung Cancer for the Study of ABCP, or BCP. In the subgroup of patients with EGFR/ALK-positive (n = 163), the mPFS was 8.5 months (95% CI: 6.9–10.3) in the ABCP group vs. 8.3 months (95% CI: 6.9–10.1) in the BCP group (non-statistical HR, 0.86; 95% CI: 0.55–1.19) [59]. These results are inconsistent with the improvements in PFS and OS with ABCP observed in the IMpower150 trial. Therefore, the strategy for combination with VEGF blockade remains unknown. However, this is currently the only regimen with promising efficacy.

    ICI therapy for locally advanced NSCLC harboring an EGFR mutation

    As mentioned above, the efficacy of ICI treatment in EGFR-mutated cases is limited to advanced stages. However, this does not appear to be the case in early-stage EGFR-mutated NSCLC. The IMpower010 study was a phase III open-label study comparing atezolizumab with placebo after postoperative adjuvant platinum-based chemotherapy for completely resected NSCLC. Disease-free survival (DFS) in stages II–III was significantly longer in the atezolizumab group than in the best supportive care group. In this study, the DFS in the EGFR mutation subgroup in the overall population was HR 0.99 (0.60–1.62), while PD-L1 positive cases showed that the DFS in the EGFR mutation subgroup was HR 0.57 (0.26–1.24), similar to cases without EGFR mutations [17].

    In the EGFR mutation subgroup of the KEYNOTE091 trial evaluating postoperative adjuvant pembrolizumab for similar populations, the HR was 0.44 (0.23–0.84), suggesting that it may be better than in non-EGFR mutation cases [64].

    KEYNOTE671 is a randomized, double-blind, phase III study that compared pembrolizumab with a placebo after postoperative adjuvant platinum-based chemotherapy for completely resected NSCLC. The primary endpoint, event-free survival, was better in the subgroup of patients with EGFR mutations [HR 0.09 (0.01–0.74)] than in those without EGFR mutations [65].

    The AEGEAN study included EGFR mutation cases, but only the results from subjects excluding EGFR mutation cases are available [66].

    Thus, unlike patients with advanced disease, patients with EGFR mutations do not seem to be particularly less affected by perioperative treatment than wild-type patients. However, the number of patients with EGFR mutations was small in both studies, which were subgroup analyses. In resectable NSCLC, the levels of CD8+ cytotoxic T cells and CD20+ B cells are associated with OS and DFS, and it has been reported that the higher the number of Tregs, the shorter the OS [67]. In contrast, although PD-L1 expression reflects the presence of tumor-infiltrating lymphocytes (TILs), there are reports that PD-L1 expression is a prognostic marker for resectable NSCLC harboring an EGFR mutation [68]. Therefore, there is no consensus regarding the TME of resectable EGFR-mutated NSCLC. However, osimertinib as a postoperative adjuvant showed a remarkable increase in DFS. Although it is necessary to compare the long-term prognosis, EGFR-TKIs currently have greater benefits as adjuvant treatments. Although immunotherapy for resectable EGFR-mutated NSCLC has DFS benefits, it is less effective than EGFR-TKIs, and there is little need for immunotherapy in clinical practice.

    For unresectable stage III NSCLC, durvalumab is the standard treatment after concurrent chemoradiotherapy (CCRT); however, it is reported to be less effective in patients with EGFR [69]. Osimertinib consolidation therapy after CCRT is currently being investigated in the LAURA study.

    Biomarkers and TME of EGFR-mutated NSCLC

    Several reports have indicated that the TME has a significant impact on the therapeutic effects of ICIs [7072].

    The most representative biomarker is PD-L1 expression, and it has been reported that PD-L1 expression may be upregulated by multiple pathways in EGFR-mutated NSCLC [9]. However, there are no certain opinions on whether PD-L1 expression is high in EGFR-mutated cases, as a few indicate that PD-L1 expression is high, whereas others indicate that it is low [10, 73]. Real-world studies have reported that PD-L1 expression correlates with the response to first-line osimertinib therapy, and PD-L1 expression is associated with prognosis [74].

    No consensus has been observed on the importance of PD-L1 expression in EGFR-mutated NSCLC. However, PD-L1 expression does not appear to be a biomarker for ICI treatment in advanced EGFR cases, and there have been few clinically significant results.

    TMB was defined as the number of somatic mutations per megabase in the coding region of a tumor. In advanced NSCLC, there is a significant association between smoking history and genetic alterations and TMB [75], and patients with EGFR-mutated NSCLC are less affected by smoking and therefore have fewer somatic mutations and neoantigens [76]. However, it is reported that TMB is not associated with the therapeutic efficacy of PD-1/L1 blockade in patients with driver mutations [77].

    In addition, lymphocyte infiltration into the tumor and surrounding stroma is associated with ICI efficacy, and a higher density of CD8+ TILs is associated with a better ICI response. In contrast, Treg infiltration was associated with poor ICI efficacy. Fewer CD8+ TILs and more Tregs were observed in EGFR-mutated mutations. EGFR-mutated NSCLC is a cold tumor with a non-inflammatory microenvironment. However, the high prevalence of Treg infiltration, which is usually observed in inflammatory microenvironments, is unique. Tregs are induced by EGFR [78].

    Various soluble molecules have been reported to interact with EGFR. For example, interleukin-6 (IL6) is reported to be overexpressed in EGFR-mutated mouse models [79], and transforming growth factor-beta (TGF-β) and tumor necrosis factor (TNF) are reported to be increased by EGFR expression [80, 81].

    Another potential influence on the TME is the effect of previous EGFR-TKI treatment. Several studies have indicated that EGFR inhibition affects TMEs [73, 78, 79, 82]. Reports have shown that EGFR inhibition improves the TME [78, 82], but the combination of EGFR-TKIs and ICI has shown less clinical benefit in clinical trials.

    Conclusions

    Current evidence suggests that ICI therapy for EGFR-positive lung cancer remains inadequate, probably due to the EGFR-specific TME. Various attempts have been made to increase the efficacy of ICI in this population. Combination therapy with CTLA-4 has not shown good results in EGFR cases, whereas the combination of VEGF, chemotherapy, and ICI has shown good results. A better understanding of EGFR-specific TME and consideration of suitable combinations is required to establish treatment strategies, including optimal ICI for this population. However, the evidence currently described is still insufficient for ICI to prolong the prognosis of EGFR-mutated NSCLC, and there is hope for the development of new agents such as ADC drugs and dual antibodies. In addition, adjuvant therapy with PD-L1 inhibitors has been introduced for resectable NSCLC and unresectable stage III NSCLC; however, EGFR-positive NSCLC is unlikely to benefit from ICI, even in these patients, and targeted therapy seems to be the most promising.

    Abbreviations

    ABCP:

    carboplatin + paclitaxel +bevacizumab + atezolizumab

    ALK:

    anaplastic lymphoma kinase

    BCP:

    carboplatin + paclitaxel + bevacizumab

    CBDCA:

    carboplatin

    CTLA-4:

    cytotoxic T-lymphocyte-associated protein 4

    DFS:

    disease-free survival

    EGFR:

    epidermal growth factor receptor

    HR:

    hazard ratio

    ICIs:

    immune checkpoint inhibitors

    mPFS:

    median progression free survival

    NSCLC:

    non-small cell lung cancer

    ORR:

    objective response rate

    OS:

    overall survival

    PD-L1:

    programmed cell death ligand 1

    PTX:

    paclitaxel

    TILs:

    tumor-infiltrating lymphocytes

    TKIs:

    tyrosine kinase inhibitors

    TMB:

    tumor mutation burden

    TME:

    tumor microenvironment

    VEGF:

    vascular endothelial growth factor

    Declarations

    Author contributions

    KA, TS, AI, TH, YG, NH, and MK: Writing—review & editing. KI: Writing—review & editing, Supervision. YO: Conceptualization, Data curation, Writing—review & editing. All authors read and approved the submitted version.

    Conflicts of interest

    YO reports personal fees from Bristol Myers Squibb, Eli Lilly, Takeda, Daiichi Sankyo, AstraZeneca, Chugai Pharma, Amgen, and Novartis, and personal fees from Taiho outside the submitted work. YG reports personal fees from Bristol Myers Squibb, Eli Lilly, Takeda, AstraZeneca, Chugai Pharma, Taiho, and Boehringer Ingelheim outside the submitted work. NH received a research grant from Boehringer Ingelheim and lecture fees from GlaxoSmithKline, AstraZeneca, Novartis, and Boehringer Ingelheim, outside the submitted work. MK received personal fees from Bristol Myers Squibb, Eli Lilly, Takeda, Daiichi Sankyo, AstraZeneca, Chugai Pharma, MSD, and Taiho outside the submitted work. KI reports personal fees from GSK, MSD, Sanofi, AstraZeneca, and Chugai Pharmaceutical Co., and he received research grants from Chugai Pharmaceutical Co. and Taiho Pharmaceutical Co. The other authors declare that they have no conflict of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Kohno T, Nakaoku T, Tsuta K, Tsuchihara K, Matsumoto S, Yoh K, et al. Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer. Transl Lung Cancer Res. 2015;4:15664. [DOI] [PubMed] [PMC]
    Gahr S, Stoehr R, Geissinger E, Ficker JH, Brueckl WM, Gschwendtner A, et al. EGFR mutational status in a large series of Caucasian European NSCLC patients: data from daily practice. Br J Cancer. 2013;109:18218. [DOI] [PubMed] [PMC]
    Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al.; West Japan Oncology Group. 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;11:1218. [DOI] [PubMed]
    Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al.; Spanish Lung Cancer Group in collaboration with Groupe Français de Pneumo-Cancérologie and Associazione Italiana Oncologia Toracica. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:23946. [DOI] [PubMed]
    Sequist LV, Yang JC, Yamamoto N, O’Byrne K, Hirsh V, Mok T, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31:332734. [DOI] [PubMed]
    Wu YL, Zhou C, Hu CP, Feng J, Lu S, Huang Y, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:21322. [DOI] [PubMed]
    Ohashi K, Maruvka YE, Michor F, Pao W. Epidermal growth factor receptor tyrosine kinase inhibitor-resistant disease. J Clin Oncol. 2013;31:107080. [DOI] [PubMed] [PMC]
    Hanna NH, Robinson AG, Temin S, Baker Jr S, Brahmer JR, Ellis PM, et al. Therapy for stage IV non-small-cell lung cancer with driver alterations: ASCO and OH (CCO) joint guideline update. J Clin Oncol. 2021;39:104091. Erratum in: J Clin Oncol. 2021;39:2520. [DOI] [PubMed]
    Chen N, Fang W, Zhan J, Hong S, Tang Y, Kang S, 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;10:91023. [DOI] [PubMed]
    Azuma K, Ota K, Kawahara A, Hattori S, Iwama E, Harada T, et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann Oncol. 2014;25:193540. [DOI] [PubMed]
    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;20:3821. [DOI] [PubMed] [PMC]
    Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 2013;3:135563. [DOI] [PubMed] [PMC]
    Gemma A, Kusumoto M, Sakai F, Endo M, Kato T, Saito Y, et al. Real-world evaluation of factors for interstitial lung disease incidence and radiologic characteristics in patients with EGFR T790M-positive NSCLC treated with osimertinib in Japan. J Thorac Oncol. 2020;15:1893906. [DOI] [PubMed]
    Oxnard GR, Yang JC, Yu H, Kim SW, Saka H, Horn L, et al. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann Oncol. 2020;31:50716. [DOI] [PubMed]
    Schoenfeld AJ, Arbour KC, Rizvi H, Iqbal AN, Gadgeel SM, Girshman J, et al. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann Oncol. 2019;30:83944. [DOI] [PubMed] [PMC]
    Antonia SJ. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med. 2019;380:989990. [DOI] [PubMed]
    Felip E, Altorki N, Zhou C, Csoszi T, Vynnychenko I, Goloborodko O, et al.; IMpower010 Investigators. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet. 2021;398:134457. [DOI] [PubMed]
    Felip E, Altorki N, Zhou C, Vallieres E, Martinez-Marti A, Rittmeyer A, et al. Overall survival with adjuvant atezolizumab after chemotherapy in resected stage II-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase III trial. Ann Oncol. 2023;34:90719. [DOI] [PubMed]
    Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR TKI therapy in 155 patients with EGFR mutant lung cancers. Clin Cancer Res. 2013:19:22407. [DOI] [PubMed] [PMC]
    Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014;4:104661. [DOI] [PubMed] [PMC]
    Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, et al.; AURA3 Investigators. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med. 2017;376:62940. [DOI] [PubMed] [PMC]
    Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al.; FLAURA Investigators. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N Engl J Med. 2018;378:11325. [DOI] [PubMed]
    Chmielecki J, Gray JE, Cheng Y, Ohe Y, Imamura F, Cho BC, et al. Candidate mechanisms of acquired resistance to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer. Nat Commun. 2023;14:1070. Erratum in: Nat Commun. 2023;14:3179. [DOI] [PubMed] [PMC]
    Schmid S, Li JJN, Leighl NB. Mechanisms of osimertinib resistance and emerging treatment options. Lung Cancer. 2020;147:1239. [DOI] [PubMed]
    Leonetti A, Verze M, Minari R, Perrone F, Gnetti L, Bordi P, et al. Resistance to osimertinib in advanced EGFR-mutated NSCLC: a prospective study of molecular genotyping on tissue and liquid biopsies. Br J Cancer. 2024;130:13542. [DOI] [PubMed]
    Piotrowska Z, Isozaki H, Lennerz JK, Gainor JF, Lennes IT, Zhu VW, et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov. 2018;8:152939. [DOI] [PubMed] [PMC]
    Yu HA, Goldberg SB, Le X, Piotrowska Z, Goldman JW, De Langen AJ, et al. Biomarker-directed phase II platform study in patients with EGFR sensitizing mutation-positive advanced/metastatic non-small cell lung cancer whose disease has progressed on first-line osimertinib therapy (ORCHARD). Clin Lung Cancer. 2021;22:6016. [DOI] [PubMed] [PMC]
    Passaro A, Wang J, Wang Y, Lee SH, Melosky B, Shih JY, et al.; MARIPOSA-2 Investigators. Amivantamab plus chemotherapy with and without lazertinib in EGFR-mutant advanced NSCLC after disease progression on osimertinib: primary results from the phase III MARIPOSA-2 study. Ann Oncol. 2024;35:7790. [DOI] [PubMed]
    Yu HA, Goto Y, Hayashi H, Felip E, Chih-Hsin Yang J, Reck M, et al. HERTHENA-Lung01, a phase II trial of patritumab deruxtecan (HER3-DXd) in epidermal growth factor receptor-mutated non-small-cell Lung cancer after epidermal growth factor receptor tyrosine kinase inhibitor therapy and platinum-based chemotherapy. J Clin Oncol. 2023;41:536375. [DOI] [PubMed] [PMC]
    Inoue A, Kobayashi K, Maemondo M, Sugawara S, Oizumi S, Isobe H, et al.; North-East Japan Study Group. Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin-paclitaxel for chemo-naive non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002). Ann Oncol. 2013;24:549. [DOI] [PubMed]
    Yoshioka H, Shimokawa M, Seto T, Morita S, Yatabe Y, Okamoto I, et al. Final overall survival results of WJTOG3405, a randomized phase III trial comparing gefitinib versus cisplatin with docetaxel as the first-line treatment for patients with stage IIIB/IV or postoperative recurrent EGFR mutation-positive non-small-cell lung cancer. Ann Oncol. 2019;30:197884. [DOI] [PubMed]
    McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:61328. [DOI] [PubMed]
    Burrell RA, Swanton C. Tumour heterogeneity and the evolution of polyclonal drug resistance. Mol Oncol. 2014;8:1095111. [DOI] [PubMed] [PMC]
    Garon EB, Wolf B, Lisberg A, Kim KY, Horton JM, Kamranpour N, et al. Prior TKI therapy in NSCLC EGFR mutant patients associates with lack of response to anti-PD-1 treatment. J Thorac Oncol. 2015;10:S269. [DOI] [PubMed]
    Lisberg A, Cummings A, Goldman JW, Bornazyan K, Reese N, Wang T, et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naive patients with advanced NSCLC. J Thorac Oncol. 2018;13:113845. [DOI] [PubMed] [PMC]
    Gettinger S, Rizvi NA, Chow LQ, Borghaei H, Brahmer J, Ready N, et al. Nivolumab monotherapy for first-line treatment of advanced non-small-cell lung cancer. J Clin Oncol. 2016;34:29807. [DOI] [PubMed] [PMC]
    Lee CK, Man J, Lord S, Cooper W, Links M, Gebski V, et al. 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 oncology. 2018;4:2106. [DOI] [PubMed] [PMC]
    Hayashi H, Sugawara S, Fukuda Y, Fujimoto D, Miura S, Ota K, et al. A randomized phase II study comparing nivolumab with carboplatin-pemetrexed for EGFR-mutated NSCLC with resistance to EGFR tyrosine kinase inhibitors (WJOG8515L). Clin Cancer Res. 2022;28:893902. [DOI] [PubMed] [PMC]
    Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol. 2019;30:13218. [DOI] [PubMed] [PMC]
    Peters S, Gettinger S, Johnson ML, Janne PA, Garassino MC, Christoph D, et al. Phase II trial of atezolizumab as first-line or subsequent therapy for patients with programmed death-ligand 1-selected advanced non-small-cell lung cancer (BIRCH). J Clin Oncol. 2017;35:27819. Erratum in: J Clin Oncol. 2018;36:931. [DOI] [PubMed] [PMC]
    Garassino MC, Cho BC, Kim JH, Mazieres J, Vansteenkiste J, Lena H, et al.; ATLANTIC Investigators. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): an open-label, single-arm, phase 2 study. Lancet Oncol. 2018;19:52136. [DOI] [PubMed] [PMC]
    Oya Y, Kuroda H, Nakada T, Takahashi Y, Sakakura N, Hida T. Efficacy of immune checkpoint inhibitor monotherapy for advanced non-small-cell lung cancer with ALK rearrangement. Int J Mol Sci. 2020;21:2623. [DOI] [PubMed] [PMC]
    Lai GGY, Yeo JC, Jain A, Zhou S, Pang M, Alvarez JJS, et al. A randomized phase 2 trial of nivolumab versus nivolumab-ipilimumab combination in EGFR-mutant NSCLC. JTO Clin Res Rep. 2022;3:100416. [DOI] [PubMed] [PMC]
    Chalmers AW, Patel S, Boucher K, Cannon L, Esplin M, Luckart J, et al. Phase I trial of targeted EGFR or ALK therapy with ipilimumab in metastatic NSCLC with long-term follow-up. Target Oncol. 2019;14:41721. [DOI] [PubMed]
    Gettinger S, Hellmann MD, Chow LQM, Borghaei H, Antonia S, Brahmer JR, et al. Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J Thorac Oncol. 2018;13:136372. [DOI] [PubMed]
    Yang JC, Shepherd FA, Kim DW, Lee GW, Lee JS, Chang GC, et al. Osimertinib plus durvalumab versus osimertinib monotherapy in EGFR T790M-positive NSCLC following previous EGFR TKI therapy: CAURAL brief report. J Thorac Oncol. 2019;14:9339. [DOI] [PubMed]
    Riudavets M, Naigeon M, Texier M, Dorta M, Barlesi F, Mazieres J, et al. Gefitinib plus tremelimumab combination in refractory non-small cell lung cancer patients harbouring EGFR mutations: the GEFTREM phase I trial. Lung Cancer. 2022;166:25564. [DOI] [PubMed]
    Rudin CM, Cervantes A, Dowlati A, Besse B, Ma B, Costa DB, et al. Safety and clinical activity of atezolizumab plus erlotinib in patients with non-small-cell lung cancer. ESMO Open. 2023;8:101160. [DOI] [PubMed] [PMC]
    Yang JC, Gadgeel SM, Sequist LV, Wu CL, Papadimitrakopoulou VA, Su WC, et al. Pembrolizumab in combination with erlotinib or gefitinib as first-line therapy for advanced NSCLC with sensitizing EGFR mutation. J Thorac Oncol. 2019;14:5539. [DOI] [PubMed]
    Rizvi NA, Hellmann MD, Brahmer JR, Juergens RA, Borghaei H, Gettinger S, et al. Nivolumab in combination with platinum-based doublet chemotherapy for first-line treatment of advanced non-small-cell lung cancer. J Clin Oncol. 2016;34:296979. [DOI] [PubMed] [PMC]
    West H, McCleod M, Hussein M, Morabito A, Rittmeyer A, Conter HJ, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20:92437. [DOI] [PubMed]
    Mok T, Nakagawa K, Park K, Ohe Y, Girard N, Kim HR, et al. Nivolumab plus chemotherapy in epidermal growth factor receptor-mutated metastatic non-small-cell lung cancer after disease progression on epidermal growth factor receptor tyrosine kinase inhibitors: final results of CheckMate 722. J Clin Oncol. 2024;42:125264. [DOI] [PubMed] [PMC]
    Yang JCH, Lee DH, Lee JS, Fan Y, de Marinis F, Okamoto I, et al. Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: Phase 3 KEYNOTE-789 study. In: 2023 ASCO Annual Meeting; 2023 May 30–June 3; Chicago, USA. J Clin Oncol; 2023. p. LBA9000.
    Lee CK, Subramaniam S, Mersiades A, Mitchell J, Jurkovic H, Walker M, et al. A phase II trial of durvalumab (MEDI4736) and tremelimumab with chemotherapy in metastatic EGFR mutant non-squamous non-small cell lung cancer (NSCLC) following progression on EGFR tyrosine kinase inhibitors (TKIs)(ILLUMINATE). J Clin Oncol. 2020;38:TPS9631. [DOI]
    Lu S, Wu L, Jian H, Chen Y, Wang Q, Fang J, et al. Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): first interim results from a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2022;23:116779. Erratum in: Lancet Oncol. 2022;23:e404. [DOI] [PubMed]
    Bourhis M, Palle J, Galy-Fauroux I, Terme M. Direct and indirect modulation of T cells by VEGF-A counteracted by anti-angiogenic treatment. Front Immunol. 2021;12:616837. [DOI] [PubMed] [PMC]
    Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015;212:13948. [DOI] [PubMed] [PMC]
    Heinemann AS, Pirr S, Fehlhaber B, Mellinger L, Burgmann J, Busse M, et al. In neonates S100A8/S100A9 alarmins prevent the expansion of a specific inflammatory monocyte population promoting septic shock. FASEB J. 2017;31:115364. [DOI] [PubMed]
    Zhou C, Dong X, Chen G, Wang Z, Wu X, Yao Y, et al. OA09. 06 IMpower151: phase III study of atezolizumab + bevacizumab + chemotherapy in 1L metastatic nonsquamous NSCLC. J Thorac Oncol. 2023;18:S645. [DOI]
    Reck M, Mok TSK, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, et al.; IMpower150 Study Group. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir Med. 2019;7:387401. [DOI] [PubMed]
    Nogami N, Barlesi F, Socinski MA, Reck M, Thomas CA, Cappuzzo F, et al. IMpower150 final exploratory analyses for atezolizumab plus bevacizumab and chemotherapy in key NSCLC patient subgroups with EGFR mutations or metastases in the liver or brain. J Thorac Oncol. 2022;17:30923. [DOI] [PubMed]
    Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624. [DOI] [PubMed] [PMC]
    Hung MS, Chen IC, Lin PY, Lung JH, Li YC, Lin YC, et al. Epidermal growth factor receptor mutation enhances expression of vascular endothelial growth factor in lung cancer. Oncol Lett. 2016;12:4598604. [DOI] [PubMed] [PMC]
    O’Brien M, Paz-Ares L, Marreaud S, Dafni U, Oselin K, Havel L, et al.; EORTC-1416-LCG/ETOP 8-15 – PEARLS/KEYNOTE-091 Investigators. Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer (PEARLS/KEYNOTE-091): an interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol. 2022;23:127486. [DOI] [PubMed]
    Wakelee H, Liberman M, Kato T, Tsuboi M, Lee SH, Gao S, et al.; KEYNOTE-67. Investigators. Perioperative pembrolizumab for early-stage non-small-cell lung cancer. N Engl J Med. 2023;389:491503. [DOI] [PubMed] [PMC]
    Heymach JV, Harpole D, Reck M. Perioperative durvalumab for resectable non-small-cell lung cancer. N Engl J Med. 2024;390:2878. [DOI] [PubMed]
    Tuminello S, Veluswamy R, Lieberman-Cribbin W, Gnjatic S, Petralia F, Wang P, et al. Prognostic value of immune cells in the tumor microenvironment of early-stage lung cancer: a meta-analysis. Oncotarget. 2019;10:714255. [DOI] [PubMed] [PMC]
    Saw SP, Ng WP, Zhou S, Lai GG, Tan AC, Ang MK, et al. PD-L1 score as a prognostic biomarker in asian early-stage epidermal growth factor receptor-mutated lung cancer. Eur J Cancer. 2023;178:13949. [DOI] [PubMed]
    Hellyer JA, Aredo JV, Das M, Ramchandran K, Padda SK, Neal JW, et al. Role of consolidation durvalumab in patients with EGFR- and HER2-mutant unresectable stage III NSCLC. J Thorac Oncol. 2021;16:86872. [DOI] [PubMed]
    Brambilla E, Le Teuff G, Marguet S, Lantuejoul S, Dunant A, Graziano S, et al. Prognostic effect of tumor lymphocytic infiltration in resectable non-small-cell lung cancer. J Clin Oncol. 2016;34:122330. [DOI] [PubMed] [PMC]
    Gettinger SN, Choi J, Mani N, Sanmamed MF, Datar I, Sowell R, et al. A dormant TIL phenotype defines non-small cell lung carcinomas sensitive to immune checkpoint blockers. Nat Commun. 2018;9:3196. [DOI] [PubMed] [PMC]
    Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:1248. [DOI] [PubMed] [PMC]
    Toki MI, Mani N, Smithy JW, Liu Y, Altan M, Wasserman B, et al. Immune marker profiling and programmed death ligand 1 expression across NSCLC mutations. J Thorac Oncol. 2018;13:188496. [DOI] [PubMed] [PMC]
    Sakata Y, Sakata S, Oya Y, Tamiya M, Suzuki H, Shibaki R, 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;159:14453. [DOI] [PubMed]
    Wang X, Ricciuti B, Nguyen T, Li X, Rabin MS, Awad MM, et al. Association between smoking history and tumor mutation burden in advanced non-small cell lung cancer. Cancer Res. 2021;81:256673. [DOI] [PubMed] [PMC]
    Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:6974. [DOI] [PubMed]
    Vokes N, Alguilar EJ, Adeni A, Umeton R, Sholl L, Rizvi H, et al. MA19. 01 efficacy and genomic correlates of response to anti-PD1/PD-L1 blockade in non-small cell lung cancers harboring targetable oncogenes. J Thorac Oncol. 2018;13:S422. [DOI]
    Sugiyama E, Togashi Y, Takeuchi Y, Shinya S, Tada Y, Kataoka K, et al. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR-mutated non-small cell lung cancer. Sci Immunol. 2020;5:eaav3937. [DOI] [PubMed]
    Busch SE, Hanke ML, Kim KH, Kargl J, Houghton AM. Abstract 3181: EGFR and KRAS activation generate discrete inflammatory responses within the lung tumor microenvironment. Cancer Res. 2015;75:3181. [DOI]
    Zhao Y, Ma J, Fan Y, Wang Z, Tian R, Ji W, et al. TGF-β transactivates EGFR and facilitates breast cancer migration and invasion through canonical Smad3 and ERK/Sp1 signaling pathways. Mol Oncol. 2018;12:30521. [DOI] [PubMed] [PMC]
    Gong K, Guo G, Gerber DE, Gao B, Peyton M, Huang C, et al. TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer. J Clin Invest. 2018;128:250018. [DOI] [PubMed] [PMC]
    Madeddu C, Donisi C, Liscia N, Lai E, Scartozzi M, Maccio A. EGFR-mutated non-small cell lung cancer and resistance to immunotherapy: role of the tumor microenvironment. Int J Mol Sci. 2022;23:6489. [DOI] [PubMed] [PMC]