Preprint
Review

It is too Early to Say NO IMMUNOTHERAPY for EGFR‐Mutant NSCLC Patients

Altmetrics

Downloads

154

Views

28

Comments

0

A peer-reviewed article of this preprint also exists.

DAN YAN  *

This version is not peer-reviewed

Submitted:

06 September 2023

Posted:

08 September 2023

You are already at the latest version

Alerts
Abstract
EGFR tyrosine kinase inhibitors (TKIs) are the preferred initial treatment for non-small cell lung cancer (NSCLC) patients harboring EGFR mutations. However, remission is transient, and no further effective treatment options are available for EGFR-TKI-advanced EGFR-mutant NSCLCs. Immunotherapy with immune checkpoint inhibitors (ICIs) induces sustained cancer remission in a subset of NSCLCs. However, ICI therapy exhibits limited activity in most EGFR-mutant NSCLCs. Mechanistically, the strong oncogenic EGFR signaling in EGFR-mutant NSCLCs contributes to the non-inflamed tumor immune microenvironment (TIME), characterized by a limited number of CD8+ T cell infiltration, a high number of regulatory CD4+ T cells and a high number of inactivated infiltrated T cells. Besides, EGFR-mutant NSCLC patients are generally non-smokers with low levels of PD-L1 expression and tumor mutation burden. However, current understanding only partially explains why a small population of EGFR-mutant NSCLCs durably respond to ICI therapy, resulting in many researchers actively working in this field. This review reviews the hope seen from pre-clinical studies and clinical trials which may be adopted to improve the outcome of ICI therapy in EGFR-mutant NSCLCs. Besides, the underlying mechanisms leading to the inferior clinical outcome of ICI therapy in EGFR-mutant NSCLCs are discussed.
Keywords: 
Subject: Biology and Life Sciences  -   Immunology and Microbiology

Introduction

Lung cancer is still the leading cause of cancer-related death worldwide [1]. Non-small cell lung cancer (NSCLC) accounts for ~85% of all lung cancers. Up to 50% of NSCLCs in Eastern Asia and 8-16% of NSCLCs in the West have epidermal growth factor receptor (EGFR) mutations [2,3,4]. EGFR L858R missense mutation and in-frame exon 19 deletions (19del) are the most common mutations found in EGFR-mutant NSCLCs, accounting for ~85% of all EGFR-activating mutations [3,4,5,6]. NSCLCs with EGFR L858R or 19del are sensitive to initial EGFR tyrosine kinase inhibitor (TKI) therapy [7,8]. Relative to traditional chemotherapy, EGFR-TKI therapy significantly extends progression-free survival (PFS), overall survival (OS), and improved quality of life due to relatively low toxicity for NSCLC patients harboring sensitive EGFR mutations [9,10,11,12,13,14,15,16,17,18]. EGFR-TKI therapy is the first-line treatment for NSCLC patients with sensitive EGFR mutations in the National Comprehensive Cancer Network (NCCN) guidelines [19]. However, the remission is still transient for only nine months to around two years [9,10,11,12,13,14,15,16,20]. With the universal application of EGFR TKIs to treat the EGFR-mutant NSCLCs, EGFR-TKI resistance represents an unmet clinical problem. So far, no further effective treatment options have been approved after progression to EGFR TKIs in EGFR-mutant NSCLCs, most of whom need chemotherapy-consisting therapy. However, the clinical benefit of the chemotherapy-consisting therapy is modest, with a high potential for toxicity.
Other than targeted therapies, immunotherapy with immune checkpoint inhibitors (ICIs), represented by anti-programmed cell death-(ligand) 1 [anti-PD-(L)1] therapy, has demonstrated improved survival rates, long-term survival benefits, and favorable safety profile over chemotherapy in a subset of advanced NSCLCs [21,22,23,24,25,26,27], leading 6 ICIs (ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, and durvalumab) approved by FDA as first- or second-line treatments for advanced NSCLCs but not suggested for patients with sensitive EGFR mutations [28]. Relative to NSCLCs with EGFR wild-type, ICI therapy fails to improve survival benefits in most EGFR-mutant NSCLCs [29,30,31,32,33,34]. Clinical benefit from ICI therapy is associated with inflamed tumors with infiltrated immune cells, high tumor mutation burden (TMBs), and high expression of PD-L1 on tumor cells [35]. However, the oncogenic EGFR signaling in lung cancer with EGFR mutations contributes to a non-inflamed tumor immune microenvironment (TIME) characterized by limited infiltration of CD8+ T cells [36,37,38], a high number of infiltrations of regulatory CD4+ T cells [38], and a high number of inactivated infiltrated T cells [39]. Besides, EGFR-mutant NSCLCs are generally non-smokers [34] with low PD-L1 expression [36,37,39] and low tumor mutation burden [36,37]. The mechanisms mediating durable effects in a small proportion of EGFR-mutant NSCLCs are worth further investigation [40].

Hope:

  • The Efficacy of Osimertinib for EGFR-Mutant NSCLCs is Independent of PD-L1 Levels
Although osimertinib treatment decreased PD-L1 expression on EGFR-mutant NSCLC cell lines by reducing PD-L1 at mRNA level and promoting PD-L1 degradation by proteasomes [41], the median PFS (mPFS) of osimertinib was similar in PD-L1 negative patients (TC<1%) (18.9 months) and in PD-L1 positive patients (TC≥1%) (18.4 months) [42], suggesting the osimertinib efficacy was independent of PD-L1 status in EGFR-mutant NSCLCs.
2.
Subtypes of EGFR Alterations May be Sensitive to ICI Therapy
ICI efficacy varies in subtypes of EGFR alterations in EGFR-mutant NSCLCs. EGFR wild-type and L858R NSCLCs had a similar outcome, while EGFR 19del NSCLCs had a worse outcome when treated with ICI therapy [43]. One of the possible explanations was that EGFR L858R-mutant tumors were more likely to have significantly increased CD8+ T cell infiltration than EGFR 19del tumors [39]. The findings were also supported by other groups that ICI therapy alone or in combination exhibited improved survival outcomes in the EGFR L858R group compared with that in the EGFR 19del group [44,45,46,47]. Besides, EGFR T790M-negative patients were more likely to benefit from ICI therapy than EGFR T790M-positive patients [48,49], as the EGFR T790M-negative tumors had higher PD-L1 expression than the EGFR T790M-positive tumors [48,50]. Other than the common mutations, 10-20 % of EGFR-mutant NSCLCs harbor non-EGFR L858R or 19del mutations, known as uncommon mutations, such as S768I and G719X [5,37,51,52]. Relative to common EGFR mutations, uncommon EGFR-mutant tumors were likely to express high PD-L1 (≥50%), high TMB, and high infiltration of CD8+ T cells [37,49,52,53]. Further, 36.7% uncommon EGFR-mutant NSCLCs with dual high PD-L1 expression and abundant CD8+ T cell infiltration showed a favorable response to ICI therapy [49,53].
3.
Heavily Pre-Treated EGFR-Mutant NSCLCs are Likely to Respond to ICI Therapy Relative to Treatment-Naïve EGFR-Mutant NSCLCs
EGFR-TKI treatment results in dynamic changes in host immunity. EGFR-TKI-advanced EGFR-mutant NSCLC patients trended to have concurrent high levels of PD-L1 expression and high number of CD8+ T cell infiltration [34]. It was further found that the proportion of EGFR-mutant NSCLCs with high PD-L1 (≥50%) was increased from 14% to 28% after EGFR-TKI treatment [54]. Further, heavily pre-treated EGFR-TKI-advanced EGFR-mutant NSCLCs with high expression of PD-L1 (≥25% of tumor cells) had a better median OS (mOS) (13.3 vs. 9.9 months) than that with low expression of PD-L1 (<25% of tumor cells) after ICI therapy in a Phase II ATLANTIC study [29,55]. It was also found that 2 of 5 EGFR-TKI-advanced EGFR-mutant NSCLCs with increased PD-L1 after EGFR blockade showed a durable response to subsequent ICI therapy [54]. Overall, EGFR-TKI-advanced EGFR-mutant NSCLCs may be the groups responding to ICI therapy. However, ICI therapy immediately followed by EGFR TKI therapy may cause immune-related adverse effects (irAEs), as discussed below.
4.
IrAEs Observed During ICI Treatment Are Associated with the Efficacy
Even though irAEs are not necessary for treatment benefit, a growing body of literature supports that the occurrence of irAEs during ICI therapy is associated with improved treatment efficacy. A retrospective analysis proved that the 270 metastatic NSCLCs who experienced irAEs after ICI therapy had improved PFS, overall response rate (ORR), disease control rate (DCR), and OS relative to those who did not experience irAEs (PFS: 5.2 vs. 1.97 months; ORR: 22.9% vs. 5.7%; DCR: 76% vs. 58%; OS: not reached vs. 8.21 months) [56]. Similarly, secondary analysis of the Phase I CA209-003 clinical trial conducted at 13 US medical centers led to findings that the mOS was significantly longer among patients with irAEs of any grade (19.8 months; 95% CI, 13.8-26.9 months) or grade 3 or more (20.3 months; 95% CI, 12.5-44.9 months) compared with those without treatment-related AEs (5.8 months; 95% CI, 4.6-7.8 months) (p<0.001 for both comparisons based on hazard ratios) [21]. Another retrospective study found that around 43.6% of 195 advanced NSCLCs treated with ICI therapy experienced irAEs. Significantly longer mPFS (5.7 s. 2.0 months), PFS (8.5 vs. 4.6 months), mOS (17.8 vs. 4.0 months), and OS (26.8 vs. 11.9 months) relative to those who did not experience irAEs was observed [57]. Besides the retrospective studies, forty-three advanced NSCLCs were enrolled in a prospective study to evaluate the association between clinical outcomes and irAEs after ICI therapy. Consistently, earlier irAEs were associated with better objective response and disease control rates than those without irAEs after ICI therapy (37% vs. 17% and 74% vs. 29%, respectively) [58]. Another prospective study with 76 advanced NSCLCs also supported that the mPFS was significantly longer when the NSCLCs experienced irAEs within two weeks of beginning ICI than those who did not (5.0 vs. 2.0 months) [59]. However, it is difficult to predict the treatment-related toxicity.
5.
Positive Clinical Studies Support the Use of ICI Therapy in EGFR-Mutant NSCLCs
Even though the ORR for KRAS-mutant NSCLCs to ICI therapy is around 20%, 7% of EGFR-mutant NSCLCs respond to single-agent ICI therapy [40]. A female NSCLC patient harboring EGFR L858R mutation treated with ICI therapy resulted in a prolonged PFS (~23 months) [60]. To achieve further improved outcomes, anti-PD-1/PD-L1 antibody-based ICI therapy, combined with other checkpoint inhibitors or platinum-based chemotherapy, has been widely applied in advanced lung cancers [28,61]. Yang et al. observed that chemo-immunotherapy was better than immunotherapy alone for EGFR-TKI advanced EGFR-mutant NSCLCs (mPFS: 3.42 vs. 1.58 months, p=0.027) [62]. A retrospective study of 122 EGFR-TKI-advanced EGFR-mutant NSCLC patients, especially with EGFR L858R mutation, revealed better mPFS (5.0 vs. 2.2 months) and mOS (14.4 vs. 7 months) in ICI-based combination therapy than that in ICI alone [45]. Further, front-line ICI therapy reached better survival benefits than later-line ICI therapy [45]. In another retrospective study, chemo-immunotherapy also achieved better outcomes than ICI therapy alone of mPFS (4.3 vs. 1.5 months), mOS (14.92 vs. 7.41 months) and ORR (23.1% vs. 3.1%) in the EGFR-TKI-advanced EGFR-mutant NSCLCs especially with EGFR L858R mutation, respectively [63]. Consistent with the findings, patients with EGFR L858R exhibited a trend of longer mPFS (7.6 vs. 5.4 months) and mOS (23.5 vs. 18.0 months) versus patients with EGFR 19del receiving chemo-immunotherapy [46].
VEGF is not only associated with NSCLC progression, recurrence, and metastasis; VEGF, complimentarily with EGFR pathways, promotes an immunosuppressive TIME [64], and anti-angiogenic therapy can reprogram the TIME from immunosuppressive to immune-supportive [65,66]. Currently, anti-angiogenic drugs plus chemotherapy are the most common regimen for EGFR-TKI-advanced NSCLCs [67]. Chemo-immunotherapy combinations achieved a higher objective response rate (ORR) relative to the chemo-anti-angiogenesis combination (29.5% vs. 13%) [67]. Longer PFS was also associated with previous anti-angiogenic drug applications in patients receiving chemo-immunotherapy [62]. Chemo-immuno-anti-angiogenesis combination was superior to either chemo-immunotherapy or chemo-anti-angiogenesis combination on mPFS (10.2, 6.9, 6.9 months), and the mOS (29.4, 19.0, 18.1 months) in the Phase III IMpower150 study in a subgroup of EGFR-TKI-advanced EGFR-mutant NSCLCs [68], suggesting that the combination of ICI, anti-angiogenesis, and chemotherapy may be a novel therapy for EGFR-TKI-advanced EGFR-mutant NSCLCs. Chemo-immuno-anti-angiogenesis combination therapy correlated with the highest rate of grade 3/4 treatment-related AEs compared to chemo-immunotherapy and chemo-anti-angiogenesis (66.7%, 42.4%, and 13.6%) [68]. Like the IMpower150 clinical trial, 40 EGFR-TKI-advanced EGFR-mutant NSCLC patients received chemo-immuno-anti-angiogenesis therapy enrolled in a Phase II clinical trial, and the patients achieved impressive mPFS (9.4 months), one-year OS (72.5%), and ORR (62.5%) with only 37.5% reported irAEs [69]. These positive clinical trials may identify novel biomarkers that predict responders to ICI therapy alone or in combination for EGFR-TKI-advanced EGFR-mutant NSCLCs.

Challenges:

  • PD-L1 is a Debating exclusive Predictive and Prognostic Biomarker for ICI Therapy in EGFR-Mutant NSCLCs
Expression of PD-1 on activated T cells, B cells, and natural killer (NK) cells blunts the immune response through interaction with its major ligand PD-L1, expressed on tumor cells and infiltrating immune cells [70,71,72]. Disruption of the PD-1/PD-L1 interaction reactivates the anti-tumor T cell-mediated cell cytotoxicity. However, researchers do not consistently agree on PD-L1 expression levels in EGFR-mutant NSCLCs. In earlier studies, activation of the PI3K-AKT, MAPK-ERK, or JAK-STAT3 pathway by aberrant oncogenic EGFR signaling upregulated PD-L1 expression in EGFR-mutant NSCLC cell lines [73,74,75,76,77], and EGFR TKI treatment decreased PD-L1 expression [73,74]. Some studies reported no correlation between PD-L1 expression and EGFR mutation status [78,79]. In more recent studies with clinical samples, the EGFR-mutant NSCLC group expressed significantly lower PD-L1 than the EGFR wild-type NSCLC group [80,81,82,83,84]{Ji, 2016 #18530}{Dong, 2017 #17138}{Li, 2017 #18540}{Takada, 2018 #18462}{Lee, 2019 #18499}. A pooled analysis of 15 public studies further suggested that EGFR-mutant NSCLCs have a decreased PD-L1 expression [36]. PD-L1 expression was more accentuated at portions with higher PD-L1 expression in EGFR-mutant versus EGFR wild-type group (51% vs. 68% at TC≥1%, 8% vs. 35% at TC≥25% and 5% vs. 28% at TC≥50%) [42]. Furthermore, EGFR blockade upregulated PD-L1 expression in EGFR-mutant NSCLCs [34,54,85], supporting that EGFR-TKI-advanced EGFR-mutant NSCLC patients probably benefit from ICI therapy as discussed above.
PD-L1 was a predictive and prognostic biomarker for response to ICI therapy in NSCLCs [25]. Several clinical trials reported the association between PD-L1 expression and clinical outcomes in NSCLC patients [25,33,86]. However, PD-L1 levels do not consistently indicate ICI response and correlate with prognosis, especially for EGFR-mutant NSCLCs [39]. ICI therapy did not show clinical benefit in TKI naïve EGFR-mutant NSCLCs, even with high PD-L1 expression. A Phase II trial, despite 73% of enrolled NSCLC patients with more than 50% PD-L1 expression, mostly treatment-naïve with sensitive EGFR mutations, was discontinued after 11 patients due to lack of efficacy PD-L1 [33,86]. Patients with low or even undetectable PD-L1 expression also had improved survival with ICI therapy versus chemotherapy [25]. Cross-comparison is sometimes challenging due to various immunohistochemical (IHC) assays with different scoring systems and cutoff values [87,88], making a universal assay for assessing PD-L1 expression with appropriate cutoff points important.
Besides, the disconnection might be because PD-L1 expression could not reflect the underlying T cell activity in EGFR-mutant NSCLCs in earlier studies [39]. PD-L1 is also highly expressed in circulating immune cells, such as dendritic cells and myeloid-derived immune suppressor cells [89,90]. In contrast, most clinical trials only assess tumoral PD-L1 expression when investigating ICI efficacy. Assessing PD-L1 expression in tumor cells and tumor-infiltrating immune cells in both POPLAR and OAK trials led to the finding that higher PD-L1 levels in both tumor cells and tumor-infiltrating immune cells were associated with improved patient survival after ICI therapy [25,91]. Patients with more than 30% of PD-L1+CD11b+ myeloid cells before ICI therapy exhibited a 50% superior response rate [92]. Furthermore, PD-L2, another ligand identified for PD-1 T cell receptor (TCR) [93], could compete with PD-L1 with 2-6 fold higher affinity to PD-1 [94]. EGFR signaling could also regulate PD-L2 expression in NSCLCs [95].
2.
TMB is Low in EGFR-Mutant NSCLCs
TMB is the total number of gene alterations, including substitutions, insertions, and deletions, correlated with higher levels of neoantigens. NSCLC patients with high TMB respond better to ICI therapy [96]. However, EGFR mutations are associated with decreased tumor mutation burden compared with tumors with EGFR wild-type [36,37,97]. Lung tumors with EGFR 19del harbored an even lower tumor mutation burden than EGFR L858R lung tumors [43,97]. Older people have increased TMB [98]. EGFR 19del was commonly found in the young, while EGFR L858R predominated in the elderly [5]. In contrast to NSCLCs harboring common EGFR mutations, patients with uncommon EGFR mutations, especially the G719X mutation, showed the highest TMB (7.5 mutations/Mb), followed by EGFR exon 20 ins (4.6 mutations/Mb), EGFR T790M (4.05 mutations/Mb), EGFR L858R (3.4 mutations/Mb), and EGFR 19del (3.1 mutations/Mb) (p<0.05) [37]. Similar to PD-L1, high TMB has been detected in responders and non-responders receiving ICI therapy [99], indicating TMB is not the only determinant for ICI response.
3.
EGFR-Mutant NSCLCs are Non-Smokers, Generally
Tobacco is a known risk factor for lung cancer. However, Smokers are more likely to benefit from ICI therapy [100,101,102]. Although EGFR mutations are more enriched in never-smokers, EGFR mutations in NSCLCs are also found in ever-smokers [6,103,104,105]. Compared with common EGFR mutations, uncommon EGFR mutations were significantly associated with smoking [106]. PD-L1 positivity was also associated with smoking history [80,81,82]. These findings further support the above discussed findings that the NSCLCs with uncommon EGFR mutations are more likely to have a favorable response to ICI therapy than NSCLCs with common EGFR mutations [49,53].
4.
EGFR-Mutant NSCLCs Have a Lymphocyte-Depleted TIME
The crosstalk between cancer cells and TIME has become a hot research topic with the rapid development of immunotherapy in cancer, including lung cancer. Based on the presence or absence of tumor-infiltrating lymphocytes (TILs), there are four different types of TIME in tumors: type I: TIL+PD-L1+; type II: TIL-PD-L1-; type III: TIL-PD-L1+; type IV: TIL+PD-L1-, and only type I tumors with lymphocyte infiltration and PD-L1 expression can respond to ICI therapy [107]. To better reflect the complex relationship of the tumor, host, and environmental factors, tumors have also been classified into the following types: the immune-dessert tumor, the immune-excluded tumor, and the inflamed tumor [108]. The immune-dessert and immune-excluded tumors are naturally resistant to ICI therapy. EGFR-mutant tumors have a “lymphocyte depletion” phenotype [34,36,38,109,110], characterized by a pronounced lack of the infiltration of CD8+ T cells [34,36,38,111], suggesting an immunosuppressive TIME. Single-cell RNA sequencing supported the findings that fewer CD8+ T cells and more macrophages were in the TIME when tumors were resistant to EGFR-TKI treatment than when EGFR TKIs were effective [112]. Additionally, EGFR-mutant NSCLC tumors had markedly less crosstalk between T cells and other cell types via the PD-1/PD-L1 pathway than EGFR-negative NSCLCs [113].
Regulatory T cells (Tregs), especially the Forkhead box P3 (Foxp3)+, play essential roles in immune suppression [114]. EGFR-mutant NSCLC tumors showed high infiltration of Foxp3+CD4+ regulatory T cells [38,115,116]. Retrospective immunohistochemistry analysis of 164 EGFR-mutant and 159 EGFR wild-type tumors revealed that the expression of CD3, CD4, and Foxp3 was significantly higher in EGFR-mutant NSCLC tumors than that in the EGFR wild-type tumors [117]. EGFR blockade increased intratumor CD8+ T cells and decreased Tregs infiltration in the TIME [38,118,119,120,121]. Similar as Tregs, myeloid-derived suppressor cells (MDSCs), known to suppress immune response [122,123,124], were also found to be elevated in EGFR-mutant NSCLC tumors [121].
Tumors without detectable EGFR expression responded to EGFR inhibition [125], implicating that EGFR blocking blockade potentially influenced the tumor-specific immune responses. Deficient Egfr in murine myeloid cells decreased carcinogenesis, suggesting a tumor-promoting function by myeloid cell-intrinsic EGFR signaling [126]. Macrophages in the TIME express EGFR [126,127]. EGF secreted by tumor cells promoted M2 polarization of tumor-associated macrophages (TAMs), associated with suppression of cytotoxic T cell function [128]. In contrast to M2-type TAMs, higher infiltrated M1-type TAMs found in NSCLC with uncommon EGFR mutations (G719X and exon 20s) correlated with longer PFS than common EGFR mutations [37].
EGFR-mutant NSCLC cell lines significantly downregulated MHC class I molecule expression compared with the EGFR wild-type NSCLC cell lines in response to IFNγ [129]. PI3K-AKT and MAPK pathways, the primary downstream signaling pathways of EGFR {Yan, 2018 #7245}{Yan, 2022 #15218}, also suppressed MHC class I molecule expression [129,130,131,132,133], suggesting the downregulation of MHC class I molecules in EGFR-mutant NSCLCs is through abnormal EGFR signaling. Indeed, EGFR inhibition augmented the expression of MHC class I molecules [134,135,136].
5.
EGFR-Mutant NSCLCs Respond Poorly to ICI Therapy Alone or in Combination
Unlike those without driver mutations, EGFR-mutant NSCLCs generally respond poorly to ICI therapy [30,34]. Subgroup analysis of the patients with activating EGFR mutations in the first Phase III clinical trial of CheckMate-057 revealed no PFS and OS benefit from ICI treatment [23]. A retrospective analysis revealed that EGFR mutations were associated with low clinical response to ICI blockade in NSCLCs [34]. Subgroup analysis of the data from the Phase III clinical trial of KEYNOTE-010 also indicated no improved OS benefit from ICI treatment in EGFR-mutant NSCLCs [27]. Similarly, EGFR-mutant NSCLCs failed to achieve prolonged OS from ICI vs. chemotherapy in Phase III clinical trial OAK [25]. It was further confirmed in a pooled analysis from 3 clinical trials (CheckMate-057, KEYNOTE-010, and POPLAR) that the EGFR wild-type but not the EGFR-mutant NSCLCs had prolonged OS [31]. Combining data from 5 trials (CheckMate-017, CheckMate-057, KEYNOTE-010, OAK, and POPLAR), Lee et al. additionally confirmed no prolonged OS in EGFR-mutant NSCLCs receiving ICIs relative to chemotherapy [32]. In a Phase II trial, treatment of EGFR-TKI naïve EGFR-mutant patients with ICI was halted due to lack of efficacy and two deaths within six months of enrollment, 1 of which was from pneumonitis [33]. In the study of WJOG8515L, nivolumab was inferior to the chemotherapy treatment in EGFR-TKI-advanced EGFR-mutant NSCLCs without T790M mutation with worse mPFS (1.7 vs. 5.6 months) and ORR (9.6% vs. 36%) [137]. In a retrospective study, 58 EGFR-mutant NSCLC patients who responded to prior EGFR TKIs for more than ten months displayed significantly shorter PFS of ICIs compared to those responding to prior EGFR TKIs for less than ten months (1.6 vs. 1.9 months, p=0.009) [138]. More recently, ICI was unfavorable versus chemotherapy in EGFR-TKI-advanced NSCLCs harboring a secondary T790M mutation with worse mPFS (1.7 vs. 5.6 months) and response rate (9.6% vs. 36.0%) and similar OS (20.7 vs. 19.9 months) in Phase II WJOG8515L clinical trial [137]. Adding ICIs to chemotherapy was associated with worse survival than platinum doublet chemotherapy alone in osimertinib-advanced EGFR-mutant NSCLCs [139]. Phase III clinical trial IMpower130 also found no benefit in the EGFR-mutant subgroup treated with ICIs and chemotherapy combined versus chemotherapy alone [140]. Based on all these negative findings, the National Comprehensive Cancer Network (NCCN) clinical practice guidelines of NSCLC (version 4, 2021) did not recommend immunotherapy for treating EGFR-mutant NSCLCs.
6.
Safety Concerns and Lower Clinical Outcomes Regarding EGFR TKI and ICI Combined for the Treatment of EGFR-Mutant NSCLCs
Compared to chemotherapy, ICI therapy correlates fewer adverse reactions. However, ICI therapy was associated with immune-related adverse events (irAEs) caused probably due to the disruption of immunologic homeostasis [141]. There is growing concern that a combination of EGFR TKIs, especially osimertinib and gefitinib, and ICI therapy may be associated with an increased risk of toxicity. Five of seven untreated stage IIIB/IV EGFR-mutant NSCLC patients (71.4%) treated with ICI plus gefitinib had grade 3/4 liver toxicity, leading to permanent treatment discontinuation in four patients in the Phase 1/2 KEYNOTE-021 clinical trial [142]. A lung adenocarcinoma patient bearing EGFR 19del treated with osimertinib following ICI therapy had Stevens–Johnson syndrome and hepatotoxicity [143]. In a Phase Ib clinical trial of TATTON, 38.5-60% of EGFR-TKI-advanced EGFR-mutant NSCLCs experienced grade 3/4 irAEs, and 30-40% discontinued to osimertinib and ICI combined therapy due to irAEs [144]. Concurrent osimertinib and ICI therapy were associated with an increased incidence of irAEs, leading to early termination of a Phase III CAURAL recruitment [145]. Interstitial lung disease (ILD) is a severe adverse response to EGFR TKIs [146]. Osimertinib treatment immediately followed prior ICI therapy, which also caused a high incidence of ILD [147,148]. A meta-analysis of eight studies further concluded that a higher chance of irAEs was observed in EGFR-TKI plus ICI therapy in EGFR-TKI-advanced EGFR-mutant NSCLCs [149].
Other than toxicity concerns, lower clinical outcomes have also been observed in EGFR-mutant NSCLCs treated with EGFR TKI and ICI combined than those treated with EGFR TKI alone. The Phase Ib clinical trial of TATTON showed that osimertinib and ICI combination therapy only achieved 43% of ORR in the EGFR-TKI-advanced EGFR-mutant NSCLCs [144]. Besides, the ORR in gefitinib plus ICI therapy (14.3%) was much worse than the 55% of RR in gefitinib alone for EGFR-mutant NSCLC patients treated with gefitinib alone [150]. In contrast to gefitinib plus ICI therapy, the untreated stage IIIB/IV EGFR-mutant NSCLCs tolerated erlotinib plus ICI therapy and better ORR (41.7%) in the KEYNOTE-021 [142]. The RR was much lower than ~70% when using erlotinib alone as a first-line treatment in clinical trials [151,152]. Decreased ORR was also observed in the osimertinib plus ICI group versus osimertinib alone in the Phase III CAURAL recruitment [145].
7.
Hyper-Progressive Disease (HPD)
Hyper-progressive disease (HPD), characterized by unexpected fast tumor enlargement both at rate and volume and early fatality of patients [153,154], was observed in 13.8% of NSCLC patients during treatment with ICIs in a retrospective study [155]. In contrast, around 20% of EGFR-mutant NSCLCs showed risk of HPD after ICI therapy [153].

Conclusions

So far, it has been agreed that ICI-based immunotherapy is ineffective for treatment-naïve EGFR-mutant NSCLC patients. Whether EGFR-TKI-advanced EGFR-mutant NSCLC patients may benefit from the ICI-based therapy is worth investigating, considering no further approved effective treatment for this population. irAEs, symptoms that oncologists try to avoid during the immunotherapy application, will be an unmet roadblock during the evaluation. However, irAEs may not always be bad for the patients, as it may indicate that the immune system in the body is awakened. However, it is hard to predict irAEs, and some strong irAEs, if not controlled, may be deadly. A better understanding of what types of irAEs may correlate with efficacy and when the correlated irAEs happen may be helpful for future clinical monitoring during immunotherapy application. Other than irAEs, many other questions remain unsolved. Who may benefit: PD-L1 TPS>50%, smoking history, high tumor mutation burden, high CD8+ T cells, and specific subtypes of EGFR mutations? Besides, the combination of ICI-based immunotherapy and several other treatment modalities is still under active investigation. Furthermore, immunotherapy is not limited to anti-PD-1/PD-L1 monoclonal antibodies and could go beyond them.

References

  1. Siegel, R.L., et al., Cancer statistics, 2022. CA Cancer J Clin, 2022. 72(1): p. 7-33. [CrossRef]
  2. Barlesi, F.; Mazieres, J.; Merlio, J.-P.; Debieuvre, D.; Mosser, J.; Lena, H.; Ouafik, L.H.; Besse, B.; Rouquette, I.; Westeel, V.; et al. Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT). Lancet 2016, 387, 1415–1426. [CrossRef]
  3. Graham, R.P.; Treece, A.L.; Lindeman, N.I.; Vasalos, P.; Shan, M.; Jennings, L.J.; Rimm, D.L. Worldwide Frequency of Commonly Detected EGFR Mutations. Arch. Pathol. Lab. Med. 2018, 142, 163–167. [CrossRef]
  4. Zhang, Y.-L.; Yuan, J.-Q.; Wang, K.-F.; Fu, X.-H.; Han, X.-R.; Threapleton, D.; Yang, Z.-Y.; Mao, C.; Tang, J.-L. The prevalence of EGFR mutation in patients with non-small cell lung cancer: a systematic review and meta-analysis. Oncotarget 2016, 7, 78985–78993. [CrossRef]
  5. Evans, M.; O’sullivan, B.; Smith, M.; Hughes, F.; Mullis, T.; Trim, N.; Taniere, P. Large-Scale EGFR Mutation Testing in Clinical Practice: Analysis of a Series of 18,920 Non-Small Cell Lung Cancer Cases. Pathol. Oncol. Res. 2018, 25, 1401–1409. [CrossRef]
  6. Shigematsu, H.; Lin, L.; Takahashi, T.; Nomura, M.; Suzuki, M.; Wistuba, I.I.; Fong, K.M.; Lee, H.; Toyooka, S.; Shimizu, N.; et al. Clinical and Biological Features Associated With Epidermal Growth Factor Receptor Gene Mutations in Lung Cancers. JNCI J. Natl. Cancer Inst. 2005, 97, 339–346. [CrossRef]
  7. Paez, J.G., et al., EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science, 2004. 304(5676): p. 1497-500. [CrossRef]
  8. Lynch, T.J., et al., Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med, 2004. 350(21): p. 2129-39. [CrossRef]
  9. Mok, T.S.; Wu, Y.-L.; Thongprasert, S.; Yang, C.-H.; Chu, D.-T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or Carboplatin–Paclitaxel in Pulmonary Adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957. [CrossRef]
  10. Maemondo, M., et al., Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med, 2010. 362(25): p. 2380-8. [CrossRef]
  11. Mitsudomi, T.; Morita, S.; Yatabe, Y.; Negoro, S.; Okamoto, I.; Tsurutani, J.; Seto, T.; Satouchi, M.; Tada, H.; Hirashima, T.; 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. 2009, 11, 121–128. [CrossRef]
  12. Zhou, C.; Wu, Y.-L.; Chen, G.; Feng, J.; Liu, X.-Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011, 12, 735–742. [CrossRef]
  13. Wu, Y.-L.; Zhou, C.; Liam, C.-K.; Wu, G.; Liu, X.; Zhong, Z.; Lu, S.; Cheng, Y.; Han, B.; Chen, L.; et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: analyses from the phase III, randomized, open-label, ENSURE study. Ann. Oncol. 2015, 26, 1883–1889. [CrossRef]
  14. Sequist, L.V.; Yang, J.C.-H.; Yamamoto, N.; Obyrne, K.; Hirsh, V.; Mok, T.; Geater, S.L.; Orlov, S.; Tsai, C.-M.; Boyer, M.; 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, 3327–3334. [CrossRef]
  15. Soria, J.C., et al., Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med, 2018. 378(2): p. 113-125. [CrossRef]
  16. Mok, T.S., et al., Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N Engl J Med, 2017. 376(7): p. 629-640. [CrossRef]
  17. Cohen, M.H.; Johnson, J.R.; Chen, Y.-F.; Sridhara, R.; Pazdur, R. FDA Drug Approval Summary: Erlotinib (Tarceva) Tablets. Oncologist 2005, 10, 461–466. [CrossRef]
  18. Janne, P.A., et al., AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med, 2015. 372(18): p. 1689-99. [CrossRef]
  19. Ettinger, D.S., et al., NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 2.2021. J Natl Compr Canc Netw, 2021. 19(3): p. 254-266. [CrossRef]
  20. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. 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, 239–246. [CrossRef]
  21. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Sosman, J.A.; Atkins, M.B.; Leming, P.D.; et al. Five-Year Survival and Correlates Among Patients With Advanced Melanoma, Renal Cell Carcinoma, or Non–Small Cell Lung Cancer Treated With Nivolumab. JAMA Oncol. 2019, 5, 1411–1420. [CrossRef]
  22. Keith, R.; New, M.; J, B.; Kl, R.; P, B.; L, C.; We, E.; E, P.; S, A.; A, P.; et al. Faculty Opinions recommendation of Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer.. 2018, 373. [CrossRef]
  23. Borghaei, H., et al., Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N Engl J Med, 2015. 373(17): p. 1627-39. [CrossRef]
  24. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [CrossRef]
  25. Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; 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, 255–265; Erratum in Lancet 2017, 389, e5. [CrossRef]
  26. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [CrossRef]
  27. Herbst, R.S.; Baas, P.; Kim, D.-W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.-Y.; Molina, J.; Kim, J.-H.; Arvis, C.D.; Ahn, M.-J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2015, 387, 1540–1550. [CrossRef]
  28. Hargadon, K.M.; Johnson, C.E.; Williams, C.J. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018, 62, 29–39. [CrossRef]
  29. Garassino, M.C.; Cho, B.-C.; Kim, J.-H.; Mazières, J.; Vansteenkiste, J.; Lena, H.; Jaime, J.C.; Gray, J.E.; Powderly, J.; Chouaid, C.; et al. 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, 521–536. [CrossRef]
  30. Mazieres, J.; Drilon, A.; Lusque, A.B.; Mhanna, L.; Cortot, A.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [CrossRef]
  31. Lee, C.K.; Man, J.; Lord, S.; Links, M.; Gebski, V.; Mok, T.; Yang, J.C.-H. Checkpoint Inhibitors in Metastatic EGFR- Mutated Non–Small Cell Lung Cancer—A Meta-Analysis. J. Thorac. Oncol. 2017, 12, 403–407. [CrossRef]
  32. Lee, C.K., 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 Oncol, 2018. 4(2): p. 210-216. [CrossRef]
  33. Lisberg, A., 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(8): p. 1138-1145. [CrossRef]
  34. Gainor, J.F., et al., EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin Cancer Res, 2016. 22(18): p. 4585-93. [CrossRef]
  35. Kim, J.M. and D.S. Chen, Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol, 2016. 27(8): p. 1492-504. [CrossRef]
  36. Dong, Z.-Y.; Zhang, J.-T.; Liu, S.-Y.; Su, J.; Zhang, C.; Xie, Z.; Zhou, Q.; Tu, H.-Y.; Xu, C.-R.; Yan, L.-X.; et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. OncoImmunology 2017, 6, e1356145. [CrossRef]
  37. Ma, T.; Jiao, J.; Huo, R.; Li, X.; Fang, G.; Zhao, Q.; Liu, W.; Han, X.; Xi, C.; Wang, Y.; et al. PD-L1 expression, tumor mutational burden, and immune cell infiltration in non-small cell lung cancer patients with epithelial growth factor receptor mutations. Front. Oncol. 2022, 12, 922899. [CrossRef]
  38. Sugiyama, E.; Togashi, Y.; Takeuchi, Y.; Shinya, S.; Tada, Y.; Kataoka, K.; Tane, K.; Sato, E.; Ishii, G.; Goto, K.; et al. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR -mutated non–small cell lung cancer. Sci. Immunol. 2020, 5. [CrossRef]
  39. Toki, M.I.; Mani, N.; Smithy, J.W.; Liu, Y.; Altan, M.; Wasserman, B.; Tuktamyshov, R.; Schalper, K.; Syrigos, K.N.; Rimm, D.L. Immune Marker Profiling and Programmed Death Ligand 1 Expression Across NSCLC Mutations. J. Thorac. Oncol. 2018, 13, 1884–1896. [CrossRef]
  40. Guaitoli, G.; Tiseo, M.; Di Maio, M.; Friboulet, L.; Facchinetti, F. Immune checkpoint inhibitors in oncogene-addicted non-small cell lung cancer: a systematic review and meta-analysis. Transl. Lung Cancer Res. 2021, 10, 2890–2916. [CrossRef]
  41. Jiang, X.-M.; Xu, Y.-L.; Huang, M.-Y.; Zhang, L.-L.; Su, M.-X.; Chen, X.; Lu, J.-J. Osimertinib (AZD9291) decreases programmed death ligand-1 in EGFR-mutated non-small cell lung cancer cells. Acta Pharmacol. Sin. 2017, 38, 1512–1520. [CrossRef]
  42. 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, 138–143. [CrossRef]
  43. Hastings, K.; Yu, H.; Wei, W.; Sanchez-Vega, F.; DeVeaux, M.; Choi, J.; Rizvi, H.; Lisberg, A.; Truini, A.; Lydon, C.; et al. EGFR mutation subtypes and response to immune checkpoint blockade treatment in non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1311–1320. [CrossRef]
  44. Zhou, C.; Wang, Z.; Fu, C.; Tao, H.; Liu, C. The efficacy and safety of PD-1 inhibitors for EGFR-mutant non-small cell lung cancer after tyrosine kinase inhibitor failure: a retrospective real-world cohort study. Ann. Transl. Med. 2023, 11, 157–157. [CrossRef]
  45. Tian, T.; Yu, M.; Li, J.; Jiang, M.; Ma, D.; Tang, S.; Lin, Z.; Chen, L.; Gong, Y.; Zhu, J.; et al. Front-Line ICI-Based Combination Therapy Post-TKI Resistance May Improve Survival in NSCLC Patients With EGFR Mutation. Front. Oncol. 2021, 11. [CrossRef]
  46. Jiang, T.; Wang, P.; Zhang, J.; Zhao, Y.; Zhou, J.; Fan, Y.; Shu, Y.; Liu, X.; Zhang, H.; He, J.; et al. Toripalimab plus chemotherapy as second-line treatment in previously EGFR-TKI treated patients with EGFR-mutant-advanced NSCLC: a multicenter phase-II trial. Signal Transduct. Target. Ther. 2021, 6, 1–9. [CrossRef]
  47. Chen, C.-J.; Liu, Y.-P. MERTK Inhibition: Potential as a Treatment Strategy in EGFR Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer. Pharmaceuticals 2021, 14, 130. [CrossRef]
  48. Haratani, K.; Hayashi, H.; Tanaka, T.; Kaneda, H.; Togashi, Y.; Sakai, K.; Hayashi, K.; Tomida, S.; Chiba, Y.; Yonesaka, K.; et al. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann. Oncol. 2017, 28, 1532–1539. [CrossRef]
  49. Yamada, T.; Hirai, S.; Katayama, Y.; Yoshimura, A.; Shiotsu, S.; Watanabe, S.; Kikuchi, T.; Hirose, K.; Kubota, Y.; Chihara, Y.; et al. Retrospective efficacy analysis of immune checkpoint inhibitors in patients with EGFR-mutated non-small cell lung cancer. Cancer Med. 2019, 8, 1521–1529. [CrossRef]
  50. Hata, A.; Katakami, N.; Nanjo, S.; Okuda, C.; Kaji, R.; Masago, K.; Fujita, S.; Yoshida, H.; Zama, K.; Imai, Y.; et al. Programmed death-ligand 1 expression and T790M status in EGFR -mutant non-small cell lung cancer. Lung Cancer 2017, 111, 182–189. [CrossRef]
  51. Brindel, A.; Althakfi, W.; Barritault, M.; Watkin, E.; Maury, J.-M.; Bringuier, P.-P.; Girard, N.; Brevet, M. Uncommon EGFR mutations in lung adenocarcinoma: features and response to tyrosine kinase inhibitors. J. Thorac. Dis. 2020, 12, 4643–4650. [CrossRef]
  52. Miyawaki, E.; Murakami, H.; Mori, K.; Mamesaya, N.; Kawamura, T.; Kobayashi, H.; Omori, S.; Wakuda, K.; Ono, A.; Kenmotsu, H.; et al. PD-L1 expression and response to pembrolizumab in patients with EGFR-mutant non-small cell lung cancer. Ultrasound Med. Biol. 2020, 50, 617–622. [CrossRef]
  53. Chen, K.; Cheng, G.; Zhang, F.; Zhu, G.; Xu, Y.; Yu, X.; Huang, Z.; Fan, Y. PD-L1 expression and T cells infiltration in patients with uncommon EGFR-mutant non-small cell lung cancer and the response to immunotherapy. Lung Cancer 2020, 142, 98–105. [CrossRef]
  54. 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, 2037–2046. [CrossRef]
  55. Garassino, M.C.; Cho, B.-C.; Kim, J.-H.; Mazières, J.; Vansteenkiste, J.; Lena, H.; Jaime, J.C.; Gray, J.E.; Powderly, J.; Chouaid, C.; et al. Final overall survival and safety update for durvalumab in third- or later-line advanced NSCLC: The phase II ATLANTIC study. Lung Cancer 2020, 147, 137–142. [CrossRef]
  56. Grangeon, M.; Tomasini, P.; Chaleat, S.; Jeanson, A.; Souquet-Bressand, M.; Khobta, N.; Bermudez, J.; Trigui, Y.; Greillier, L.; Blanchon, M.; et al. Association Between Immune-related Adverse Events and Efficacy of Immune Checkpoint Inhibitors in Non–small-cell Lung Cancer. Clin. Lung Cancer 2018, 20, 201–207. [CrossRef]
  57. Ricciuti, B.; Genova, C.; De Giglio, A.; Bassanelli, M.; Bello, M.G.D.; Metro, G.; Brambilla, M.; Baglivo, S.; Grossi, F.; Chiari, R. Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab: long-term outcomes from a multi-institutional analysis. J. Cancer Res. Clin. Oncol. 2018, 145, 479–485. [CrossRef]
  58. Teraoka, S.; Fujimoto, D.; Morimoto, T.; Kawachi, H.; Ito, M.; Sato, Y.; Nagata, K.; Nakagawa, A.; Otsuka, K.; Uehara, K.; et al. Early Immune-Related Adverse Events and Association with Outcome in Advanced Non–Small Cell Lung Cancer Patients Treated with Nivolumab: A Prospective Cohort Study. J. Thorac. Oncol. 2017, 12, 1798–1805. [CrossRef]
  59. Hosoya, K.; Fujimoto, D.; Morimoto, T.; Kumagai, T.; Tamiya, A.; Taniguchi, Y.; Yokoyama, T.; Ishida, T.; Hirano, K.; Matsumoto, H.; et al. Association Between Early Immune-related Adverse Events and Clinical Outcomes in Patients With Non–Small Cell Lung Cancer Treated With Immune Checkpoint Inhibitors. Clin. Lung Cancer 2020, 21, e315–e328. [CrossRef]
  60. Peng, J.; Zhao, X.; Zhao, K.; Meng, X. Case Report: Long Progression-Free Survival of Immunotherapy for Lung Adenocarcinoma With Epidermal Growth Factor Receptor Mutation. Front. Oncol. 2021, 11. [CrossRef]
  61. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [CrossRef]
  62. Yang, L., et al., Superior efficacy of immunotherapy-based combinations over monotherapy for EGFR-mutant non-small cell lung cancer acquired resistance to EGFR-TKIs. Thorac Cancer, 2020. 11(12): p. 3501-3509. [CrossRef]
  63. Chen, Y.; Yang, Z.; Wang, Y.; Hu, M.; Zhang, B.; Zhang, Y.; Qian, F.; Zhang, W.; Han, B. Pembrolizumab Plus Chemotherapy or Anlotinib vs. Pembrolizumab Alone in Patients With Previously Treated EGFR-Mutant NSCLC. Front. Oncol. 2021, 11. [CrossRef]
  64. Zhao, Y.; Guo, S.; Deng, J.; Shen, J.; Du, F.; Wu, X.; Chen, Y.; Li, M.; Chen, M.; Li, X.; et al. VEGF/VEGFR-Targeted Therapy and Immunotherapy in Non-small Cell Lung Cancer: Targeting the Tumor Microenvironment. Int. J. Biol. Sci. 2022, 18, 3845–3858. [CrossRef]
  65. Choi, S.H.; Yoo, S.S.; Lee, S.Y.; Park, J.Y. Anti-angiogenesis revisited: reshaping the treatment landscape of advanced non-small cell lung cancer. Arch. Pharmacal Res. 2022, 45, 263–279. [CrossRef]
  66. Ren, S.; Xiong, X.; You, H.; Shen, J.; Zhou, P. The Combination of Immune Checkpoint Blockade and Angiogenesis Inhibitors in the Treatment of Advanced Non-Small Cell Lung Cancer. Front. Immunol. 2021, 12. [CrossRef]
  67. Yu, X.; Li, J.; Ye, L.; Zhao, J.; Xie, M.; Zhou, J.; Shen, Y.; Zhou, F.; Wu, Y.; Han, C.; et al. Real-world outcomes of chemo-antiangiogenesis versus chemo-immunotherapy combinations in EGFR-mutant advanced non-small cell lung cancer patients after failure of EGFR-TKI therapy. Transl. Lung Cancer Res. 2021, 10, 3782–3792. [CrossRef]
  68. Nogami, N.; Barlesi, F.; Socinski, M.A.; Reck, M.; Thomas, C.A.; Cappuzzo, F.; Mok, T.S.; Finley, G.; Aerts, J.G.; Orlandi, 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. 2021, 17, 309–323. [CrossRef]
  69. Lam, T.; Tsang, K.; Choi, H.; Lee, V.; Lam, K.; Chiang, C.; So, T.; Chan, W.; Nyaw, S.; Lim, F.; et al. Combination atezolizumab, bevacizumab, pemetrexed and carboplatin for metastatic EGFR mutated NSCLC after TKI failure. Lung Cancer 2021, 159, 18–26. [CrossRef]
  70. Tarleton, R.; Ye, L.; Sc, L.; Y, W.; T, C.; Ra, S.; M, K.; Vk, K.; Gj, F.; Ah, S. Faculty Opinions recommendation of PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells.. 2004, 101. [CrossRef]
  71. Keir, M.E.; Liang, S.C.; Guleria, I.; Latchman, Y.E.; Qipo, A.; Albacker, L.A.; Koulmanda, M.; Freeman, G.J.; Sayegh, M.H.; Sharpe, A.H. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J. Exp. Med. 2006, 203, 883–895. [CrossRef]
  72. Fife, B.T.; Pauken, K.E.; Eagar, T.N.; Obu, T.; Wu, J.; Tang, Q.; Azuma, M.; Krummel, M.F.; Bluestone, J.A. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR–induced stop signal. Nat. Immunol. 2009, 10, 1185–1192. [CrossRef]
  73. Akbay, E.A., et al., Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov, 2013. 3(12): p. 1355-63. [CrossRef]
  74. 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, 910–923. [CrossRef]
  75. Azuma, K.; Ota, K.; Kawahara, A.; Hattori, S.; Iwama, E.; Harada, T.; Matsumoto, K.; Takayama, K.; Takamori, S.; Kage, M.; et al. Association of PD-L1 overexpression with activating EGFR mutations in surgically resected nonsmall-cell lung cancer. Ann. Oncol. 2014, 25, 1935–1940. [CrossRef]
  76. Lastwika, K.J., et al., Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. Cancer Res, 2016. 76(2): p. 227-38. [CrossRef]
  77. Zhang, N.; Zeng, Y.; Du, W.; Zhu, J.; Shen, D.; Liu, Z.; Huang, J.-A. The EGFR pathway is involved in the regulation of PD-L1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer. Int. J. Oncol. 2016, 49, 1360–1368. [CrossRef]
  78. Cooper, W.A.; Tran, T.; Vilain, R.E.; Madore, J.; Selinger, C.I.; Kohonen-Corish, M.; Yip, P.; Yu, B.; O’toole, S.A.; McCaughan, B.C.; et al. PD-L1 expression is a favorable prognostic factor in early stage non-small cell carcinoma. Lung Cancer 2015, 89, 181–188. [CrossRef]
  79. Schmidt, L.H.; Kümmel, A.; Görlich, D.; Mohr, M.; Bröckling, S.; Mikesch, J.H.; Grünewald, I.; Marra, A.; Schultheis, A.M.; Wardelmann, E.; et al. PD-1 and PD-L1 Expression in NSCLC Indicate a Favorable Prognosis in Defined Subgroups. PLOS ONE 2015, 10, e0136023. [CrossRef]
  80. Inoue, Y.; Yoshimura, K.; Mori, K.; Kurabe, N.; Kahyo, T.; Mori, H.; Kawase, A.; Tanahashi, M.; Ogawa, H.; Inui, N.; et al. Clinical significance of PD-L1 and PD-L2 copy number gains in non-small-cell lung cancer. Oncotarget 2016, 7, 32113–32128. [CrossRef]
  81. Cha, Y.J.; Kim, H.R.; Lee, C.Y.; Cho, B.C.; Shim, H.S. Clinicopathological and prognostic significance of programmed cell death ligand-1 expression in lung adenocarcinoma and its relationship with p53 status. Lung Cancer 2016, 97, 73–80. [CrossRef]
  82. Takada, K.; Okamoto, T.; Shoji, F.; Shimokawa, M.; Akamine, T.; Takamori, S.; Katsura, M.; Suzuki, Y.; Fujishita, T.; Toyokawa, G.; et al. Clinical Significance of PD-L1 Protein Expression in Surgically Resected Primary Lung Adenocarcinoma. J. Thorac. Oncol. 2016, 11, 1879–1890. [CrossRef]
  83. Tsao, M.-S.; Le Teuff, G.; Shepherd, F.A.; Landais, C.; Hainaut, P.; Filipits, M.; Pirker, R.; Le Chevalier, T.; Graziano, S.; Kratze, R.; et al. PD-L1 protein expression assessed by immunohistochemistry is neither prognostic nor predictive of benefit from adjuvant chemotherapy in resected non-small cell lung cancer. Ann. Oncol. 2017, 28, 882–889. [CrossRef]
  84. Yang, H., et al., EGFR mutation status in non-small cell lung cancer receiving PD-1/PD-L1 inhibitors and its correlation with PD-L1 expression: a meta-analysis. Cancer Immunol Immunother, 2022. 71(5): p. 1001-1016. [CrossRef]
  85. Han, J.J.; Kim, D.-W.; Koh, J.; Keam, B.; Kim, T.M.; Jeon, Y.K.; Lee, S.-H.; Chung, D.H.; Heo, D.S. Change in PD-L1 Expression After Acquiring Resistance to Gefitinib in EGFR-Mutant Non–Small-Cell Lung Cancer. Clin. Lung Cancer 2015, 17, 263–270.e2. [CrossRef]
  86. Zhao, Q.; Zhang, X.; Ma, Q.; Luo, N.; Liu, Z.; Wang, R.; He, Y.; Li, L. Case Report: An “Immune-Cold” EGFR Mutant NSCLC With Strong PD-L1 Expression Shows Resistance to Chemo-Immunotherapy. Front. Oncol. 2022, 12, 765997. [CrossRef]
  87. Kerr, K.M. and F.R. Hirsch, Programmed Death Ligand-1 Immunohistochemistry: Friend or Foe? Arch Pathol Lab Med, 2016. 140(4): p. 326-31. [CrossRef]
  88. Ratcliffe, M.J.; Sharpe, A.; Midha, A.; Barker, C.; Scott, M.; Scorer, P.; Al-Masri, H.; Rebelatto, M.C.; Walker, J. Agreement between Programmed Cell Death Ligand-1 Diagnostic Assays across Multiple Protein Expression Cutoffs in Non–Small Cell Lung Cancer. Clin. Cancer Res. 2017, 23, 3585–3591. [CrossRef]
  89. Karwacz, K., et al., PD-L1 co-stimulation contributes to ligand-induced T cell receptor down-modulation on CD8+ T cells. EMBO Mol Med, 2011. 3(10): p. 581-92. [CrossRef]
  90. Gato-Cañas, M.; de Morentin, X.M.; Blanco-Luquin, I.; Fernandez-Irigoyen, J.; Zudaire, I.; Liechtenstein, T.; Arasanz, H.; Lozano, T.; Casares, N.; Chaikuad, A.; et al. A core of kinase-regulated interactomes defines the neoplastic MDSC lineage. Oncotarget 2015, 6, 27160–27175. [CrossRef]
  91. Nana-Sinkam, P.; L, F.; A, S.; M, B.; M, K.; J, V.; J, M.; K, P.; D, S.; A, A.-C.; et al. Faculty Opinions recommendation of Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial.. 2017, 387. [CrossRef]
  92. Bocanegra, A., et al., PD-L1 Expression in Systemic Immune Cell Populations as a Potential Predictive Biomarker of Responses to PD-L1/PD-1 Blockade Therapy in Lung Cancer. Int J Mol Sci, 2019. 20(7). [CrossRef]
  93. Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [CrossRef]
  94. Tang, S. and P.S. Kim, A high-affinity human PD-1/PD-L2 complex informs avenues for small-molecule immune checkpoint drug discovery. Proc Natl Acad Sci U S A, 2019. 116(49): p. 24500-24506. [CrossRef]
  95. Shibahara, D.; Tanaka, K.; Iwama, E.; Kubo, N.; Ota, K.; Azuma, K.; Harada, T.; Fujita, J.; Nakanishi, Y.; Okamoto, I. Intrinsic and Extrinsic Regulation of PD-L2 Expression in Oncogene-Driven Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2018, 13, 926–937. [CrossRef]
  96. Rizvi, N.A., et al., Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science, 2015. 348(6230): p. 124-8. [CrossRef]
  97. Offin, M.; Rizvi, H.; Tenet, M.; Ni, A.; Sanchez-Vega, F.; Li, B.T.; Drilon, A.; Kris, M.G.; Rudin, C.M.; Schultz, N.; et al. Tumor Mutation Burden and Efficacy of EGFR-Tyrosine Kinase Inhibitors in Patients with EGFR-Mutant Lung Cancers. Clin. Cancer Res. 2019, 25, 1063–1069. [CrossRef]
  98. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [CrossRef]
  99. Yang, R.K.; Qing, Y.; Jelloul, F.Z.; Routbort, M.J.; Wang, P.; Shaw, K.; Zhang, J.; Lee, J.; Medeiros, L.J.; Kopetz, S.; et al. Identification of biomarkers of immune checkpoint blockade efficacy in recurrent or refractory solid tumor malignancies. Oncotarget 2020, 11, 600–618. [CrossRef]
  100. Mo, J.; Hu, X.; Gu, L.; Chen, B.; Khadaroo, P.A.; Shen, Z.; Dong, L.; Lv, Y.; Chitumba, M.N.; Liu, J. Smokers or non-smokers: who benefits more from immune checkpoint inhibitors in treatment of malignancies? An up-to-date meta-analysis. World J. Surg. Oncol. 2020, 18, 1–12. [CrossRef]
  101. Corke, L.K.; Li, J.J.N.; Leighl, N.B.; Eng, L. Tobacco Use and Response to Immune Checkpoint Inhibitor Therapy in Non-Small Cell Lung Cancer. Curr. Oncol. 2022, 29, 6260–6276. [CrossRef]
  102. Ng, T.L., et al., Predictive value of oncogenic driver subtype, programmed death-1 ligand (PD-L1) score, and smoking status on the efficacy of PD-1/PD-L1 inhibitors in patients with oncogene-driven non-small cell lung cancer. Cancer, 2019. 125(7): p. 1038-1049. [CrossRef]
  103. Saito, M.; Shiraishi, K.; Kunitoh, H.; Takenoshita, S.; Yokota, J.; Kohno, T. Gene aberrations for precision medicine against lung adenocarcinoma. Cancer Sci. 2016, 107, 713–720. [CrossRef]
  104. Dogan, S., et al., Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRAS-mutant cancers. Clin Cancer Res, 2012. 18(22): p. 6169-77. [CrossRef]
  105. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [CrossRef]
  106. Lohinai, Z.; Hoda, M.A.; Fabian, K.; Ostoros, G.; Raso, E.; Barbai, T.; Timar, J.; Kovalszky, I.; Cserepes, M.; Rozsas, A.; et al. Distinct Epidemiology and Clinical Consequence of Classic Versus Rare EGFR Mutations in Lung Adenocarcinoma. J. Thorac. Oncol. 2015, 10, 738–746. [CrossRef]
  107. Teng, M.W.; Ngiow, S.F.; Ribas, A.; Smyth, M.J. Classifying Cancers Based on T-cell Infiltration and PD-L1. Cancer Res 2015, 75, 2139–2145. [CrossRef]
  108. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [CrossRef]
  109. Thorsson, V., et al., The Immune Landscape of Cancer. Immunity, 2018. 48(4): p. 812-830 e14. [CrossRef]
  110. MacDonald, F. and D.M.W. Zaiss, The Immune System's Contribution to the Clinical Efficacy of EGFR Antagonist Treatment. Front Pharmacol, 2017. 8: p. 575. [CrossRef]
  111. Taube, J.M.; Galon, J.; Sholl, L.M.; Rodig, S.J.; Cottrell, T.R.; A Giraldo, N.; Baras, A.S.; Patel, S.S.; A Anders, R.; Rimm, D.L.; et al. Implications of the tumor immune microenvironment for staging and therapeutics. Mod. Pathol. 2018, 31, 214–234. [CrossRef]
  112. Maynard, A.; McCoach, C.E.; Rotow, J.K.; Harris, L.; Haderk, F.; Kerr, D.L.; Yu, E.A.; Schenk, E.L.; Tan, W.; Zee, A.; et al. Therapy-Induced Evolution of Human Lung Cancer Revealed by Single-Cell RNA Sequencing. Cell 2020, 182, 1232–1251.e22. [CrossRef]
  113. Yang, L.; He, Y.-T.; Dong, S.; Wei, X.-W.; Chen, Z.-H.; Zhang, B.; Chen, W.-D.; Yang, X.-R.; Wang, F.; Shang, X.-M.; et al. Single-cell transcriptome analysis revealed a suppressive tumor immune microenvironment in EGFR mutant lung adenocarcinoma. J. Immunother. Cancer 2022, 10, e003534. [CrossRef]
  114. Hori, S., T. Nomura, and S. Sakaguchi, Control of regulatory T cell development by the transcription factor Foxp3. Science, 2003. 299(5609): p. 1057-61. [CrossRef]
  115. Zaiss, D.M.W.; van Loosdregt, J.; Gorlani, A.; Bekker, C.P.J.; Gröne, A.; Sibilia, M.; van Bergen en Henegouwen, P.M.P.; Roovers, R.C.; Coffer, P.J.; Sijts, A.J.A.M. Amphiregulin Enhances Regulatory T Cell-Suppressive Function via the Epidermal Growth Factor Receptor. Immunity 2013, 38, 275–284. [CrossRef]
  116. Mascia, F.; Schloemann, D.T.; Cataisson, C.; McKinnon, K.M.; Krymskaya, L.; Wolcott, K.M.; Yuspa, S.H. Cell autonomous or systemic EGFR blockade alters the immune-environment in squamous cell carcinomas. Int. J. Cancer 2016, 139, 2593–2597. [CrossRef]
  117. Luo, J.-W.; Guo, Y.-H.; Wu, F.-Y.; Li, X.-F.; Sun, X.-C.; Wang, J.-L.; Zhou, C.-C. Differences in Immunological Landscape between EGFR-Mutated and Wild-Type Lung Adenocarcinoma. Dis. Markers 2021, 2021, 1–8. [CrossRef]
  118. Selenz, C.; Compes, A.; Nill, M.; Borchmann, S.; Odenthal, M.; Florin, A.; Brägelmann, J.; Büttner, R.; Meder, L.; Ullrich, R.T. EGFR Inhibition Strongly Modulates the Tumour Immune Microenvironment in EGFR-Driven Non-Small-Cell Lung Cancer. Cancers 2022, 14, 3943. [CrossRef]
  119. Ayeni, D.; Miller, B.; Kuhlmann, A.; Ho, P.-C.; Robles-Oteiza, C.; Gaefele, M.; Levy, S.; de Miguel, F.J.; Perry, C.; Guan, T.; et al. Tumor regression mediated by oncogene withdrawal or erlotinib stimulates infiltration of inflammatory immune cells in EGFR mutant lung tumors. J. Immunother. Cancer 2019, 7, 172. [CrossRef]
  120. Venugopalan, A.; Lee, M.-J.; Niu, G.; Medina-Echeverz, J.; Tomita, Y.; Lizak, M.J.; Cultraro, C.M.; Simpson, R.M.; Chen, X.; Trepel, J.B.; et al. EGFR-targeted therapy results in dramatic early lung tumor regression accompanied by imaging response and immune infiltration in EGFR mutant transgenic mouse models. Oncotarget 2016, 7, 54137–54156. [CrossRef]
  121. Jia, Y.; Li, X.; Jiang, T.; Zhao, S.; Zhao, C.; Zhang, L.; Liu, X.; Shi, J.; Qiao, M.; Luo, J.; et al. EGFR-targeted therapy alters the tumor microenvironment in EGFR-driven lung tumors: Implications for combination therapies. Int. J. Cancer 2019, 145, 1432–1444. [CrossRef]
  122. Yu, J., et al., Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol, 2013. 190(7): p. 3783-97. [CrossRef]
  123. Almand, B.; Clark, J.I.; Nikitina, E.; van Beynen, J.; English, N.R.; Knight, S.C.; Carbone, D.P.; Gabrilovich, D.I. Increased Production of Immature Myeloid Cells in Cancer Patients: A Mechanism of Immunosuppression in Cancer. J. Immunol. 2001, 166, 678–689. [CrossRef]
  124. Suzuki, E., et al., Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res, 2005. 11(18): p. 6713-21. [CrossRef]
  125. Chung, K.Y.; Shia, J.; Kemeny, N.E.; Shah, M.; Schwartz, G.K.; Tse, A.; Hamilton, A.; Pan, D.; Schrag, D.; Schwartz, L.; et al. Cetuximab Shows Activity in Colorectal Cancer Patients With Tumors That Do Not Express the Epidermal Growth Factor Receptor by Immunohistochemistry. J. Clin. Oncol. 2005, 23, 1803–1810. [CrossRef]
  126. Srivatsa, S.; Paul, M.C.; Cardone, C.; Holcmann, M.; Amberg, N.; Pathria, P.; Diamanti, M.A.; Linder, M.; Timelthaler, G.; Dienes, H.P.; et al. EGFR in Tumor-Associated Myeloid Cells Promotes Development of Colorectal Cancer in Mice and Associates With Outcomes of Patients. Gastroenterology 2017, 153, 178–190.e10. [CrossRef]
  127. Rose-John, S.; H, L.; A, N.; K, K.; L, L.; N, A.; L, C.; Sk, W.; M, H.; R, Z.; et al. Faculty Opinions recommendation of EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation.. 2020, 16. [CrossRef]
  128. Lian, G.; Chen, S.; Ouyang, M.; Li, F.; Chen, L.; Yang, J. Colon Cancer Cell Secretes EGF to Promote M2 Polarization of TAM Through EGFR/PI3K/AKT/mTOR Pathway. Technol. Cancer Res. Treat. 2019, 18. [CrossRef]
  129. Watanabe, S.; Hayashi, H.; Haratani, K.; Shimizu, S.; Tanizaki, J.; Sakai, K.; Kawakami, H.; Yonesaka, K.; Tsurutani, J.; Togashi, Y.; et al. Mutational activation of the epidermal growth factor receptor down-regulates major histocompatibility complex class I expression via the extracellular signal-regulated kinase in non-small cell lung cancer. Cancer Sci. 2018, 110, 52–60. [CrossRef]
  130. Chandrasekaran, S.; Sasaki, M.; Scharer, C.D.; Kissick, H.T.; Patterson, D.G.; Magliocca, K.R.; Seykora, J.T.; Sapkota, B.; Gutman, D.A.; Cooper, L.A.; et al. Phosphoinositide 3-Kinase Signaling Can Modulate MHC Class I and II Expression. Mol. Cancer Res. 2019, 17, 2395–2409. [CrossRef]
  131. Marijt, K.A., et al., Metabolic stress in cancer cells induces immune escape through a PI3K-dependent blockade of IFNgamma receptor signaling. J Immunother Cancer, 2019. 7(1): p. 152. [CrossRef]
  132. Sivaram, N., et al., Tumor-intrinsic PIK3CA represses tumor immunogenecity in a model of pancreatic cancer. J Clin Invest, 2019. 129(8): p. 3264-3276. [CrossRef]
  133. Mimura, K.; Shiraishi, K.; Mueller, A.; Izawa, S.; Kua, L.-F.; So, J.; Yong, W.-P.; Fujii, H.; Seliger, B.; Kiessling, R.; et al. The MAPK Pathway Is a Predominant Regulator of HLA-A Expression in Esophageal and Gastric Cancer. J. Immunol. 2013, 191, 6261–6272. [CrossRef]
  134. Pollack, B.P., EGFR inhibitors, MHC expression and immune responses : Can EGFR inhibitors be used as immune response modifiers? Oncoimmunology, 2012. 1(1): p. 71-74. [CrossRef]
  135. Brea, E.J.; Oh, C.Y.; Manchado, E.; Budhu, S.; Gejman, R.S.; Mo, G.; Mondello, P.; Han, J.E.; Jarvis, C.A.; Ulmert, D.; et al. Kinase Regulation of Human MHC Class I Molecule Expression on Cancer Cells. Cancer Immunol. Res. 2016, 4, 936–947. [CrossRef]
  136. Pollack, B.P., B. Sapkota, and T.V. Cartee, Epidermal growth factor receptor inhibition augments the expression of MHC class I and II genes. Clin Cancer Res, 2011. 17(13): p. 4400-13. [CrossRef]
  137. Hayashi, H.; Sugawara, S.; Fukuda, Y.; Fujimoto, D.; Miura, S.; Ota, K.; Ozawa, Y.; Hara, S.; Tanizaki, J.; Azuma, 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. 2021, 28, 893–902. [CrossRef]
  138. Ichihara, E.; Harada, D.; Inoue, K.; Shibayama, T.; Hosokawa, S.; Kishino, D.; Harita, S.; Ochi, N.; Oda, N.; Hara, N.; et al. Characteristics of patients with EGFR-mutant non-small-cell lung cancer who benefited from immune checkpoint inhibitors. Cancer Immunol. Immunother. 2020, 70, 101–106. [CrossRef]
  139. White, M.N.; Piper-Vallillo, A.J.; Gardner, R.M.; Cunanan, K.; Neal, J.W.; Das, M.; Padda, S.K.; Ramchandran, K.; Chen, T.T.; Sequist, L.V.; et al. Chemotherapy Plus Immunotherapy Versus Chemotherapy Plus Bevacizumab Versus Chemotherapy Alone in EGFR-Mutant NSCLC After Progression on Osimertinib. Clin. Lung Cancer 2021, 23, e210–e221. [CrossRef]
  140. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.-G.; Daniel, D.; McCune, S.; Mekhail, T.; 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, 924–937. [CrossRef]
  141. Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168. [CrossRef]
  142. Yang, J.C.-H.; Gadgeel, S.M.; Sequist, L.V.; Wu, C.-L.; Papadimitrakopoulou, V.A.; Su, W.-C.; Fiore, J.; Saraf, S.; Raftopoulos, H.; Patnaik, A. Pembrolizumab in Combination With Erlotinib or Gefitinib as First-Line Therapy for Advanced NSCLC With Sensitizing EGFR Mutation. J. Thorac. Oncol. 2019, 14, 553–559. [CrossRef]
  143. Gianni, C.; Bronte, G.; Delmonte, A.; Burgio, M.A.; Andrikou, K.; Monti, M.; Menna, C.; Frassineti, G.L.; Crinò, L. Case Report: Stevens-Johnson Syndrome and Hepatotoxicity Induced by Osimertinib Sequential to Pembrolizumab in a Patient With EGFR-Mutated Lung Adenocarcinoma. Front. Pharmacol. 2021, 12. [CrossRef]
  144. Oxnard, G.R.; Yang, J.C.-H.; Yu, H.; Kim, S.-W.; Saka, H.; Horn, L.; Goto, K.; Ohe, Y.; Mann, H.; Thress, K.S.; 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, 507–516. [CrossRef]
  145. Yang, J.C.-H.; Shepherd, F.A.; Kim, D.-W.; Lee, G.-W.; Lee, J.S.; Chang, G.-C.; Lee, S.S.; Wei, Y.-F.; Lee, Y.G.; Laus, G.; 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, 933–939. [CrossRef]
  146. Inoue, A.; Saijo, Y.; Maemondo, M.; Gomi, K.; Tokue, Y.; Kimura, Y.; Ebina, M.; Kikuchi, T.; Moriya, T.; Nukiwa, T. Severe acute interstitial pneumonia and gefitinib. Lancet 2003, 361, 137–139. [CrossRef]
  147. Kotake, M.; Murakami, H.; Kenmotsu, H.; Naito, T.; Takahashi, T. High incidence of interstitial lung disease following practical use of osimertinib in patients who had undergone immediate prior nivolumab therapy. Ann. Oncol. 2016, 28, 669–670. [CrossRef]
  148. Schoenfeld, A.; Arbour, K.; Rizvi, H.; Iqbal, A.; Gadgeel, S.; Girshman, J.; Kris, M.; Riely, G.; Yu, H.; Hellmann Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann. Oncol. 2019, 30, 839–844. [CrossRef]
  149. Chan, D.W.-K.; Choi, H.C.-W.; Lee, V.H.-F. Treatment-Related Adverse Events of Combination EGFR Tyrosine Kinase Inhibitor and Immune Checkpoint Inhibitor in EGFR-Mutant Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 2157. [CrossRef]
  150. Sequist, L.V., et al., First-line gefitinib in patients with advanced non-small-cell lung cancer harboring somatic EGFR mutations. J Clin Oncol, 2008. 26(15): p. 2442-9. [CrossRef]
  151. Park, K., et al., First-Line Erlotinib Therapy Until and Beyond Response Evaluation Criteria in Solid Tumors Progression in Asian Patients With Epidermal Growth Factor Receptor Mutation-Positive Non-Small-Cell Lung Cancer: The ASPIRATION Study. JAMA Oncol, 2016. 2(3): p. 305-12. [CrossRef]
  152. Lim, S.H.; Lee, J.Y.; Sun, J.-M.; Ahn, J.S.; Park, K.; Ahn, M.-J. Comparison of Clinical Outcomes Following Gefitinib and Erlotinib Treatment in Non–Small-Cell Lung Cancer Patients Harboring an Epidermal Growth Factor Receptor Mutation in Either Exon 19 or 21. J. Thorac. Oncol. 2014, 9, 506–511. [CrossRef]
  153. Wang, X., et al., The biomarkers of hyperprogressive disease in PD-1/PD-L1 blockage therapy. Mol Cancer, 2020. 19(1): p. 81. [CrossRef]
  154. Fuentes-Antrás, J.; Provencio, M.; Díaz-Rubio, E. Hyperprogression as a distinct outcome after immunotherapy. Cancer Treat. Rev. 2018, 70, 16–21. [CrossRef]
  155. Ferrara, R., et al., Hyperprogressive Disease in Patients With Advanced Non-Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy. JAMA Oncol, 2018. 4(11): p. 1543-1552. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2024 MDPI (Basel, Switzerland) unless otherwise stated