Strategies and influencing factors for the treatment of advanced non-small cell lung cancer based on epidermal growth factor receptor tyrosine kinase inhibitors: a narrative review
Review Article

Strategies and influencing factors for the treatment of advanced non-small cell lung cancer based on epidermal growth factor receptor tyrosine kinase inhibitors: a narrative review

Linwu Kuang, Peng Wang, Lin Zhou, Yangkai Li ORCID logo

Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China

Contributions: (I) Conception and design: L Kuang; (II) Administrative support: P Wang, L Zhou, Y Li; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Yangkai Li, MD, PhD. Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan 430030, China. Email: doclyk@163.com.

Background and Objective: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are the primary treatment for advanced non-small cell lung cancer (NSCLC) patients with EGFR mutations, significantly enhancing patient prognosis. Despite the efficacy of EGFR-TKIs, monotherapy faces challenges such as variability among individuals and early drug resistance. This article aims to explore the treatment strategies and influencing factors for advanced NSCLC patients treated with EGFR-TKIs, optimize treatment plans, and improve the prognosis of patients with advanced NSCLC.

Methods: We undertook a comprehensive, narrative review of the latest literature to define the current application and progress of EGFR-TKIs in treating patients with advanced NSCLC.

Key Content and Findings: The efficacy and promise of EGFR-TKIs, both as monotherapy and combined with other agents, for treating patients with advanced NSCLC are outlined. The study delves into the mechanisms of resistance and the ongoing development of EGFR-TKIs. Various factors influencing the treatment of advanced NSCLC patients with EGFR-TKIs are also examined.

Conclusions: EGFR-TKIs alone improve survival in patients with advanced NSCLC. Combined with other agents, some regimens have shown improved benefits in overcoming drug resistance and prolonging patient survival. It is imperative to focus on developing novel EGFR-TKIs and investigate innovative combination therapies to maximize patient benefit.

Keywords: Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs); advanced non-small cell lung cancer (advanced NSCLC); EGFR mutations; prognostic analysis; combination therapy


Submitted Apr 18, 2024. Accepted for publication Jul 19, 2024. Published online Sep 05, 2024.

doi: 10.21037/tcr-24-637


Introduction

Lung cancer, the most deadly malignant tumor globally, often goes undetected in its early stages, leading to diagnosis in advanced stages (1). Non-small cell lung cancer (NSCLC) is the most prevalent type, making up 85% of cases, with a grim prognosis (2). A significant portion of NSCLC patients (46.8–61.2%) receive a stage IV diagnosis initially (3). Advanced NSCLC is typically deemed incurable, palliative care is the primary approach. In the past, systemic chemotherapy was the primary treatment for advanced NSCLC patients. However, the advent of targeted therapy has significantly enhanced the survival rates of stage IV NSCLC patients, although the 5-year survival rate remains less than 10% (3,4).

Epidermal growth factor receptor (EGFR), a protein molecule found on cell surfaces, regulates cell growth and division. Altered EGFR function in EGFR-mutant NSCLC leads to accelerated growth (5). These mutations are the main driving force behind lung cancer, with approximately 50% of Asian NSCLC patients (6,7) and 10–20% of patients in other regions carrying EGFR mutations (5). The most common EGFR mutations are 19del (deletion of five amino acids in exon 19, in which delE746_A750 is the most common subtype) and L858R, each accounting for 45% of all EGFR mutations (5). High levels of EGFR expression are often linked to a poor prognosis in lung cancer (8). Targeted therapy with EGFR tyrosine kinase inhibitors (TKIs) is the standard first-line treatment for advanced NSCLC patients with EGFR mutations, improving survival in those with sensitive mutations (9). Researchers are now investigating treatment strategies combining EGFR-TKIs with anti-vascular endothelial growth factor (VEGF) therapies, Janus kinase (JAK) inhibitors, mesenchymal epithelial transition factor (MET) TKIs, immune checkpoint inhibitors (ICIs), chemotherapy, radiotherapy, and surgery to delay TKI resistance, extend progression-free survival (PFS), and enhance overall survival (OS) in patients (10-16). In this review, we aimed to explore treatment strategies and factors affecting outcomes in patients with advanced NSCLC treated with EGFR-TKIs, optimize treatment regimens, and improve the prognosis of patients with advanced NSCLC. We present this article in accordance with the Narrative Review reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-637/rc).


Methods

A comprehensive, narrative review of literature was conducted to determine the progress of EGFR-TKIs in treating patients with advanced NSCLC. Studies from 2017 to 2024 were reviewed from PubMed using the keywords “EGFR-TKIs”, “epidermal growth factor receptor tyrosine kinase inhibitors”, “advanced NSCLC”, and “stage IV”. Some important studies published before 2017 cited in the literature we searched were also added to our review.

Articles related to the topic of this study were thoroughly reviewed. Table 1 summarizes the search strategy.

Table 1

The search strategy summary

Items Specification
Date of search November 3, 2023 to April 3, 2024; June 3, 2024 to June 6, 2024
Databases and other sources searched PubMed
Search terms used (“EGFR-TKIs” OR “epidermal growth factor receptor tyrosine kinase inhibitors”) and (“advanced NSCLC” OR “stage IV”)
Timeframe 2017–2024
Inclusion and exclusion criteria Inclusion criteria: clinical trial; meta-analysis; randomized controlled trial; review; systematic review; exclusion criteria: written in non-English language
Selection process All authors selected studies together

EGFR-TKI, epidermal growth factor receptor tyrosine kinase inhibitor; NSCLC, non-small cell lung cancer.


EGFR-TKIs monotherapy

First-generation EGFR-TKIs, such as gefitinib, erlotinib, and icotinib, are competitive inhibitors that bind reversibly to the adenosine triphosphate (ATP)-binding site of the EGFR (7,17-19). Studies have indicated that both first- and second-generation EGFR-TKIs are more effective and tolerable than platinum-based chemotherapy (20-22), with the erlotinib group demonstrating a median PFS of 9.7 versus 5.2 months in the standard chemotherapy group. Furthermore, the incidence of treatment-related severe adverse events is lower in the erlotinib group compared to the chemotherapy group (20). Despite the initial efficacy, many EGFR-mutant NSCLC patients eventually develop resistance to first-generation EGFR-TKIs, with a significant proportion developing the EGFR T790M mutation (15).

To address acquired resistance following treatment with first-generation EGFR-TKIs, researchers have developed second-generation EGFR-TKIs like afatinib and dacomitinib, which irreversibly inhibit the tyrosine kinase structural domain of EGFR (22,23). In a study by Wu et al., 364 patients were randomized into targeted therapy and chemotherapy groups (242 received afatinib; 122 received gemcitabine and cisplatin), with the afatinib group showing significantly longer median PFS compared to the gemcitabine and cisplatin group (11.0 vs. 5.6 months, P<0.0001) (22). Data from phase III clinical trials (LUX-Lung 3 and LUX-Lung 6) demonstrate that afatinib significantly enhances OS in patients with 19del mutation compared to platinum-containing chemotherapy (24). However, second-generation EGFR-TKIs do not effectively address the T790M mutation nor improve survival in NSCLC patients who have progressed after initial treatment with first-generation EGFR-TKIs (15).

The third-generation EGFR-TKIs have been developed to target the T790M mutation, inhibiting EGFR activity from preventing, slowing, or halting the growth of NSCLC. These personalized treatment options include osimertinib, aumolertinib, furmonertinib, and lazertinib (5,25-29). Currently, osimertinib is the standard first-line treatment for metastatic EGFR-mutant NSCLC (5). For advanced EGFR-mutant NSCLC, receiving EGFR-TKIs is superior to chemotherapy, which is the consensus of many investigators (20). However, EGFR-TKIs are cytostatic rather than cytotoxic agents and cannot eradicate micrometastatic tumor cells, even with highly significant clinical efficacy (30). Despite durable remissions, the median PFS of first-generation EGFR-TKIs (gefitinib and erlotinib) is approximately 1 year due to acquired resistance (9). The FLAURA study shows a median PFS of 18.9 months in locally advanced or metastatic NSCLC patients treated with third-generation EGFR-TKI (osimertinib) (31). LUX-Lung 7 (afatinib vs. gefitinib, phase IIB) (32), ARCHER-1050 (dacomitinib vs. gefitinib, phase III) (33) and FLAURA trial (osimertinib vs. gefitinib or erlotinib, phase III) (34) demonstrate that second- and third-generation EGFR-TKIs have better outcome superior to first-generation EGFR-TKIs (Table 2).

Table 2

The efficacy of EGFR-TKIs alone in treating NSCLC is as follows

Representative drugs Types of inhibitors Targets of EGFR PFS (months) OS (months) Ref
First-generation EGFR-TKIs
   Gefitinib Reversible 19del, L858R 8.0 17.0 (11)
   Erlotinib Reversible 19del, L858R 13.1 22.8 (17,18)
   Icotinib Reversible 19del, L858R 11.2 30.5 (19)
Second-generation EGFR-TKIs
   Afatinib Irreversible 19del, L858R 11.0 23.1 (22,24)
   Dacomitinib Irreversible 19del, L858R 14.7 34.1 (23,33)
Third-generation EGFR-TKIs
   Osimertinib Irreversible 19del, L858R, T790M 18.9 38.6 (28,34)
   Aumolertinib Irreversible 19del, L858R, T790M 19.3 NR (25)
   Furmonertinib Irreversible 19del, L858R, T790M 20.8 NR (26)
   Lazertinib Irreversible 19del, L858R, T790M 11.1 NR (29)

EGFR-TKI, epidermal growth factor receptor tyrosine kinase inhibitor; NSCLC, non-small cell lung cancer; PFS, progression-free survival; OS, overall survival; NR, not reached.

Osimertinib, the most popular third-generation EGFR-TKI, demonstrates efficacy in EGFR T790M-positive NSCLC patients who experience disease progression following treatment with first or second-generation EGFR-TKIs (7). The median PFS for patients with sensitive mutations treated with osimertinib is 19.17 months, while for EGFR T790M-positive patients, it is 10.58 months (35). Osimertinib’s high selectivity in inhibiting EGFR-sensitive mutations and T790M mutations prevents the development of T790M mutations in patients when used as a first-line therapy (34,36). Some advanced EGFR T790M NSCLC patients may lose the mutation after initial treatment with osimertinib (37). However, not all EGFR-mutant NSCLC patients benefit from EGFR-TKIs and some still experience disease progression (2,38). Following resistance to third-generation EGFR-TKIs, patients have limited standard targeted agent options, with little benefit from cytotoxic chemotherapy or immunotherapy. Therefore, personalized treatment and combination therapy involving EGFR-TKIs are crucial to enhance patient survival outcomes (9).

Substituting the cysteine residue at position 797 in the ATP-binding pocket with a serine prevents the covalent binding of osimertinib to EGFR, resulting in acquired resistance to osimertinib in NSCLC patients. The incidence of the C797S mutation is 7% in first-line treatment with osimertinib and 26% in second-line treatment (36). Rare EGFR mutations that lead to osimertinib resistance include G796X, which disrupts osimertinib binding; L792X, affecting the kinase’s ‘hinge’ region; L718X in the P-loop causing spatial blockage; and G724S inducing conformational changes incompatible with drug binding (39). Studies also suggest that the G796 R/S mutation, L792 residue mutation, transmutation of C797, and L718V mutation can interfere with osimertinib binding to EGFR (40). Understanding resistance mechanisms to EGFR-TKIs is crucial for progress in this field and the development of new therapies (39). Despite advancements, a large percentage of resistance mechanisms to EGFR-TKIs, particularly after first-line treatment with osimertinib, remain unknown. Resistance to EGFR-TKIs is complex, involving multiple mechanisms and potential tumor heterogeneity, posing a significant challenge to overcome (39).

ASP8273 has shown antitumor activity by inhibiting 19del, L858R, and T790M. In phase III clinical trials comparing ASP8273 with erlotinib or gefitinib, it effectively targeted inhibition of T790M but resulted in higher toxic effects like hyponatremia and peripheral sensory neuropathy. For stage IIIB/IV NSCLC patients, ASP8273 did not demonstrate improved PFS compared to erlotinib or gefitinib (9.3 vs. 9.6 months) (41). Ongoing development of new EGFR-TKIs includes third-generation EGFR-TKIs (rociletinib and PF 06747775) and fourth-generation EGFR-TKIs (EAI 045, CH 7233163, JBJ-04-125-02, BLU-945, and BLU-701). EAI 045 targets the C797S mutation found in third-generation drugs. In vitro, EAI 045 inhibits the L858R-T790M mutant form of EGFR alone but not in vivo; however, when combined with cetuximab, it successfully inhibits L858R-T790M and L858R-T790M-C797S-driven lung cancer in a mouse model. This combination significantly enhances efficacy, though the associated toxicity of cetuximab may restrict its clinical use (42,43). CH 7233163 has shown inhibition of NSCLC cells with the 19del-T790M-C797S triple EGFR mutation both in vitro and in vivo (44). JBJ-04-125-02 has demonstrated inhibition of the L858R-T790M-C797S form of EGFR in both in vitro and in vivo studies, potentially showing a more robust response when combined with osimertinib (45). BLU-945 and BLU-701 are new potent fourth-generation EGFR-TKIs that can cross the blood-brain barrier and retain wild-type EGFR. In preclinical studies, BLU-945 has demonstrated effectiveness against triple-mutant EGFR isoforms (L858R or 19del, T790M, and C797S), while BLU-701 has shown activity against double mutations (the sensitive mutation and C797S) (39,46). Given that T790M did not develop with first-line osimertinib treatment, BLU-701 could be a crucial treatment option. Both BLU-945 (NCT 04862780) and BLU-701 (NCT 05153408) are currently undergoing early clinical trials (39). Despite the recent success in EGFR-TKI research, there is still significant potential in exploring combination therapy regimens involving EGFR-TKIs (9).

Analysis of the EGFR-TKIs’ molecular structure allows us to gain a more comprehensive understanding. Like the first-generation TKIs, second-generation TKIs feature an anilinoquinazoline core, but they can form covalent bonds with the sulfur atoms of Cys797, enabled by the inclusion of an acrylamide slug moiety. However, second-generation TKIs exhibit reduced selectivity for mutant EGFR, potentially resulting in epithelial toxicity. Additionally, the quinazoline portion of second-generation TKIs interacts adversely with the side chain of Met790, necessitating the administration of relatively high doses. Third-generation TKIs offer irreversible, selective inhibition of the T790M mutation and use an anilinopyrimidine scaffold that maximizes and stabilizes their interactions within the ATP-binding pocket. To address the C797S mutation, researchers have developed fourth-generation TKIs that target triple mutants, including both variant and ATP-competitive inhibitors. Variant inhibitors are derivatives of oxoisoindoline phenylacetamide. Currently, at least five fourth-generation ATP-competitive reversible EGFR-TKIs are undergoing clinical trials. These compounds share an anilinopyrimidine scaffold, highlighting the heterocyclic portion’s critical role in interacting with the ATP-binding pocket (47).


Combination therapies based on EGFR-TKIs

Combining EGFR-TKIs with anti-VEGF therapy

VEGF is a group of genetically encoded polypeptide molecules, such as VEGF-A, -B, -C, and -D (48), that promote angiogenesis to support tumor growth (49). The VEGF and EGFR pathways share common downstream signaling pathways and interact to affect tumor growth (48). In EGFR-mutant NSCLC, overexpressed EGFR can upregulate hypoxia-inducible factors like HIF-1 through a mechanism independent of hypoxia, leading to increased VEGF expression (50). This elevated VEGF level contributes to the development of resistance to EGFR-TKIs (51). VEGF signaling also aids tumor growth by activating the pro-survival effects of the PI3K/AKT pathway and the MAPK pathway for proliferation (51). Anti-angiogenic monoclonal antibodies that block the VEGF pathway, bevacizumab, and ramucirumab, were approved by the United States Food and Drug Administration as first-line treatments for advanced or metastatic non-squamous NSCLC in 2006 and 2014, respectively (48,52). Bevacizumab inhibits tumor angiogenesis and improves EGFR-TKIs delivery, making it a promising antitumor agent (9). Ramucirumab selectively targets VEGFR2 and blocks VEGFA, VEGFC, and VEGFD-mediated signaling in NSCLC. Thus, ramucirumab has the potential for broader antitumor activity than VEGF inhibitors (51). Vandetanib (53), nintedanib (54) and cediranib (55) are VEGFR-TKIs used for NSCLC. The mechanisms involved in combination therapy can be seen in Figure 1.

Figure 1 Molecular mechanisms of drug combination therapy for advanced NSCLC. Both EGFR and VEGFR-2 signaling activate the PI3K/AKT and Ras/Raf/Mek pathways. EGFR activation induces HIF-1 upregulation, leading to VEGF gene expression and a subsequent positive feedback loop. Small molecule inhibitors of the EGFR (erlotinib, gefitinib, icotinib, afatinib, dacomitinib, osimertinib, and vandetanib), small molecule inhibitors of VEGFR-2 (vandetanib, nintedanib and cediranib), as well as monoclonal antibodies targeting the extracellular structural domain of VEGFR-2 (ramucirumab) or inhibiting the VEGF-A protein (bevacizumab) have demonstrated efficacy. Pemetrexed inhibits thymidylate synthase, dihydrofolate reductase, and glycinamide ribonucleotide formyltransferase activities to impede DNA synthesis and cell division. Pemetrexed also suppresses the Akt pathway. MET amplification activates the PI3K/Akt pathway; capmatinib is an inhibitor of MET. Activation of the EGFR pathway up-regulates PD-L1 expression; durvalumab inhibits PD-L1 while nivolumab, sintilimab, and tislelizumab inhibit PD-1. IL-6 triggers JAK1/STAT3 signaling pathway activation leading to TKI resistance; AZD 4205, itacitinib and ruxolitinib are inhibitors of JAK1. This picture was drawn using Figdraw (www.figdraw.com). AKT, protein kinase B; c-Jun, jun proto-oncogene; DHFR, dihydrofolate reductase; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; Erk, extracellular signal-regulated kinases; FAK, focal adhesion kinase; GARFT, glycinamide ribonucleotide formyltransferase; HIF-1, hypoxia-inducible factor 1; IL-6, interleukin-6; JAK1, Janus kinase 1; MAPK, mitogen-activated protein kinase; Mek, mitogen-activated extracellular signal-regulated kinase; MET, mesenchymal epithelial transition factor; MHC, major histocompatibility complex; MTORC1, mechanistic target of rapamycin complex 1; PD-1, programed death receptor-1; PD-L1, programed death ligand-1; PI3K, phosphatidylinositol 3-kinase; Raf, rapidly accelerated fibrosarcoma; Ras, rat sarcoma; SFK, Src family kinase; STAT3, signal transducer and activator of transcription 3; TCR, T-cell receptor; TKIs, tyrosine kinase inhibitors; TS, thymidylate synthase; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; NSCLC, non-small cell lung cancer.

In clinical trials, the addition of anti-VEGF agents to EGFR-TKI therapy has significantly improved clinical outcomes (51). CTONG1706, the first phase III clinical trial conducted in China, evaluated gefitinib in combination with apatinib for first-line treatment of EGFR-mutant NSCLC. The study randomized 313 patients with primary advanced NSCLC harboring EGFR 19del or L858R mutations equally into the experimental group (gefitinib, 250 mg/d; apatinib, 500 mg/d) and the placebo group (gefitinib, 250 mg/d; placebo, 500 mg/d). The Independent Radiologic Review Board reported a median PFS of 13.7 months in the experimental group and 10.2 months in the placebo group. Although combination therapy led to more complications like hypertension (46.5%) and proteinuria (17.8%), it did not impact the quality of life (16). Stratified analysis based on mutation types showed that the combination therapy prolonged PFS in the 19del subgroup but not significantly in the L858R subgroup. Patients with TP53 exon 8 mutation tended to benefit from the experimental group, suggesting that TP53 mutation status could potentially serve as a biomarker to predict the efficacy of EGFR-TKIs combined with anti-angiogenic drugs (16).

Two-phase III clinical trials (CTONG1509, NEJ026) have shown that the median PFS with bevacizumab in combination with erlotinib was significantly longer than with erlotinib alone (9,56). Although Kawashima et al.’s study did not find a survival benefit with the combination, hypothesizing that the treatment regimen in advanced patients with disease progression may impact the efficacy of this combination (9). It is worth noting that the evidence category for erlotinib in combination with bevacizumab has been upgraded from 2B to 2A in the National Comprehensive Cancer Network guidelines (16).

Several studies have investigated the combination of EGFR-TKIs and anti-VEGF agents, including JO 25567 (bevacizumab + erlotinib vs. erlotinib; phase II), ACCRURC 1126 (bevacizumab + erlotinib vs. erlotinib; phase II), RELAY (ramucirumab + erlotinib vs. erlotinib; phase III), and BEVERLY (bevacizumab + erlotinib vs. erlotinib; phase III) (17). These studies demonstrated improved PFS with the combination therapy (57). The RELAY regimen has shown promise in treating advanced EGFR-mutated NSCLC patients (58). Clinical trials, such as the phase II trial NCT 02803203, have indicated that osimertinib, in combination with bevacizumab, can improve PFS (59). However, other trials have not shown the same benefit (60). Ongoing clinical trials also investigate the combination of osimertinib and ramucirumab, as seen in NCT 02789345 (58).

The selection of combination therapy regimens for patients with different EGFR-mutant NSCLCs is still controversial, particularly concerning the use of higher-generation EGFR-TKIs (16). Although this combination therapy may lead to more grade 3 or worse adverse events, it has not resulted in patient mortality (31). Further research is necessary to determine the optimal combination of EGFR TKIs and anti-VEGF therapies (9).

Combining EGFR-TKIs with JAK inhibitors

The signal transducer and activator of transcription (STAT) is phosphorylated by JAK, dimerizes, and then translocates through the nuclear membrane into the nucleus to regulate the expression of relevant genes. This pathway is known as the JAK/STAT signaling pathway (61). In the presence of EGFR-TKIs, NSCLC cells with the EGFR T790M mutation develop drug resistance by producing interleukin-6 and activating the JAK1/STAT3 signaling pathway (15).

Knockdown of STAT3 through siRNA or JAK inhibitors enhances the antitumor effects of afatinib in the EGFR T790M NSCLC xenograft model. In a study by Park et al., 30 advanced NSCLC patients who had progressed after EGFR-TKI treatment were treated with afatinib monotherapy for 8 days, followed by concurrent administration of ruxolitinib and afatinib until disease progression. The median PFS was 4.9 months [95% confidence interval (CI): 2.5–7.3] in T790M+ patients and 3.1 months (95% CI: 0–8.8) in T790M− patients (P=0.21). While this was lower than results from other studies like the AURA study, the AURA study phase II extension, and the AURA phase III study with osimertinib, the combination of afatinib and ruxolitinib showed promising clinical efficacy in T790M+ patients. Targeting the JAK1/STAT3 pathway could be a potential therapeutic strategy for EGFR-mutant NSCLC patients, although it may not be sufficient to overcome drug resistance in T790M NSCLC (15). A novel JAK1 inhibitor, AZD 4205, has shown enhanced antitumor activity when combined with osimertinib in mouse transplantation models of NSCLC (62). Clinical trials also investigate the combination of osimertinib with itacitinib (JAK1 inhibitor) for NSCLC (NCT 02917993) (7).

Combining EGFR-TKIs with MET-TKIs

MET plays a crucial role in promoting tumor cell migration, invasion, and proliferation through various transduction pathways, including PI3K-AKT-mTOR, RAS-MAPK, RASCDC42-PAK-Rho kinase, and β-collagen signaling pathways. Driver mutations in MET, such as exon 14 jump alterations and MET amplification, have been linked to cancer progression and a poorer prognosis for patients (63). Specifically, patients with MET amplification have shown limited response to EGFR-TKIs in targeted therapy for NSCLC (64). Approximately 3–4% of stage IV non-squamous NSCLC patients exhibit tumor progression due to MET pathway activation through MET mutations or amplification. MET amplification is a known mechanism of acquired resistance to EGFR-TKIs, particularly after treatment with third-generation EGFR-TKIs like osimertinib, with around 15% of resistant patients showing MET amplification. However, the actual prevalence may be higher, as this data was derived from liquid biopsy analysis, which is less sensitive than tissue testing (14).

In EGFR-mutant NSCLC patients, combining EGFR-TKIs with MET-TKIs like capmatinib shows promise in improving outcomes for patients resistant to various generations of EGFR-TKIs. Capmatinib, a potent MET pathway inhibitor, demonstrates antitumor activity in NSCLC both in vitro and in vivo. The combination therapy may help overcome resistance to EGFR-TKIs through the MET pathway (14). Studies indicate that capmatinib treatment leads to tumor regression in MET-dependent models (65,66) and reverses the effects of MET activation on EGFR and HER-3 pathways (65), potentially restoring sensitivity to EGFR-TKIs in resistant NSCLC cell lines (67). These findings highlight the potential of capmatinib in inhibiting the MET pathway and enhancing the effectiveness of EGFR-TKIs in treating resistant tumors (14).

A study evaluating the use of third-generation EGFR-TKI (nazartinib) in combination with capmatinib for NSCLC treatment reported an overall response rate (ORR) of 42% across all dose levels, increasing to 50% at the 400 mg capmatinib/100 mg nazartinib dose level. Initial antitumor activity was observed at various dose levels (14). Another phase Ib/II study combining capmatinib with gefitinib in NSCLC patients with acquired resistance to EGFR-mutated disease (without T790M mutation but altered MET) showed an ORR of 23% and disease control rate (DCR) of 57% across all dosage groups, irrespective of MET status. In a subgroup analysis by MET gene copy number category, the best ORR observed was 47% in patients with MET gene copy number ≥6 (n=36), compared with 32% in patients with MET immunohistochemistry 3+ (n=78) (68). In EGFR-mutant NSCLC patients with acquired resistance due to high MET amplification, the combination of EGFR-TKIs and MET-TKIs demonstrated promising outcomes in advanced cases (14).

Combining EGFR-TKIs with ICIs

Cell adhesion molecules play a crucial role in tumor progression and immune evasion. ICIs combat tumor development by targeting irregularly expressed cell adhesion molecules. Among these molecules, programmed cell death-1 protein/ligand (PD-1/L1) and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) have been extensively studied for their role in inducing tumor immunosuppression (69). Anti-PD-1/L1 and anti-CTLA-4 therapies have been utilized as primary or secondary treatments for advanced NSCLC, often in combination with radiotherapy, chemotherapy, yielding positive outcomes (69). ICIs have been advocated as standard treatment for advanced NSCLC without EGFR or ALK mutations. This is because few previous studies have used ICIs for advanced EGFR-mutant NSCLC, and ICIs should not be used in EGFR-mutant NSCLC based on several retrospective single-agent studies of ICIs (70). The preclinical study indicates that activation of the EGFR pathway may upregulate PD-L1 expression on tumor cells, supporting the potential benefit of combining EGFR-TKIs with ICIs to enhance antitumor effects (71).

Yang et al. compared the efficacy and safety of osimertinib in combination with durvalumab (n=12) with osimertinib monotherapy (n=17) in 29 advanced EGFR-mutant patients who had acquired the T790M mutation after treatment with an EGFR-TKI in the phase III clinical trial (CAURAL). CAURAL enrollment was terminated early due to the high incidence (38%, 13/34) of interstitial lung disease in a phase Ib clinical trial of osimertinib combined with durvalumab for EGFR-mutant NSCLC (NCT 02143466). Unfortunately, the small sample size of the CAURAL trial did not allow for formal safety and efficacy comparisons (12). However, trials in which erlotinib and nivolumab were treated proved that the combination was tolerable (72). There is no clear evidence that PD-1/L1 inhibitors combined with EGFR-TKIs can enhance anti-NSCLC efficacy and benefit advanced EGFR-mutant NSCLC patients (12,73). The other study showed that the use of osimertinib after treatment with ICIs was associated with a high incidence of severe immune-related adverse reactions (irAEs) (14.6%, 6/41), which did not occur in patients receiving osimertinib or other EGFR-TKIs followed by ICIs. These cases guide the clinical use of ICIs in advanced NSCLC patients resistant to EGFR-TKIs (70). In advanced NSCLC patients with the T790M mutation, the sequential administration of nivolumab followed by osimertinib can lead to grade 3 or higher hepatotoxicity (69). While ICIs have shown significant advancements in NSCLC treatment, drug resistance, and irAEs have hindered their full potential (69). Interestingly, one study found that responders had a higher incidence of irAEs compared to non-responders (65.2% vs. 19.3%, P<0.01), suggesting a potential link between irAEs and treatment efficacy (74). Common irAEs include pruritus, colitis, cardiovascular toxicity, peritonitis, and interstitial lung disease (69). Sintilimab and tislelizumab are two cost-effective domestic anti-PD-1 monoclonal antibodies. The future of NSCLC treatment may involve dual ICI therapy (combination of anti-PD-1 and anti-CTLA-4), as well as exploration of tumor vaccines and CAR-T cell therapies (69). Further research on the combination of EGFR-TKIs with ICIs holds promise for achieving more excellent treatment outcomes.

Combining EGFR-TKIs with chemotherapy

Chemotherapy and EGFR-TKIs synergistically induce apoptosis and inhibit Akt and extracellular signal-regulated kinase phosphorylation, potentially delaying the development of resistance (75). Pemetrexed reduces thymidine synthase expression and inhibits T790M production, exerting its anti-NSCLC effect (76,77). Gefitinib selectively inhibits EGFR into the nucleus, reduces DNA-dependent protein kinase activity, and enhances the cytotoxicity of cisplatin in NSCLC. This is how targeted agents are combined with chemotherapy to enhance efficacy (78).

Multiple studies have shown that combining EGFR-TKIs and chemotherapy is more effective in advanced EGFR-mutant NSCLC patients (11,79-81). Combination chemotherapy with gefitinib plus pemetrexed and platinum significantly improved PFS and OS, with more significant benefit achieved in the combination therapy group (estimated median PFS 16 months; OS not reached) compared with the gefitinib group (estimated median PFS =8 months; OS =17 months). However, this benefit came at the cost of a higher incidence of grade 3 or higher toxicity in the combination therapy group (75%) compared to the gefitinib group (49.4%) (11). Previous studies on gefitinib combined with chemotherapy for the initial treatment of advanced EGFR-mutant NSCLC have reported improvements in PFS and OS in the combination therapy group (11,82-84). A meta-analysis involving 1,349 individuals concluded that first-generation TKIs combined with platinum-based two-agent chemotherapy provided significant benefits in terms of ORR, PFS, and OS (85). Patients with low-abundance EGFR-mutated NSCLC had inferior outcomes compared to those with high-abundance EGFR-mutated phenotypes when treated with EGFR-TKIs (86). However, combining EGFR-TKIs with chemotherapy improved both PFS and OS in patients with low-abundance EGFR-mutated phenotypes (87). A study demonstrated that concurrent treatment with gefitinib in combination with pemetrexed and carboplatin leads to longer OS compared to sequential therapy (88). Therefore, concurrent administration of TKI and chemotherapy is recommended for patients with T790M-negative advanced lung cancer who have experienced progression after initial EGFR-TKI therapy (89). In a study comparing gefitinib or icotinib combined with pemetrexed for EGFR-mutant advanced NSCLC, stratified analysis revealed that patients with concurrent mutations in TP53, KRAS, PIK3CA, and MLH1 had a poorer PFS with EGFR-TKIs monotherapy (P=0.002) (79). In a retrospective study, patients with advanced NSCLC who progressed during treatment with osimertinib had a higher survival rate when receiving chemotherapy regimens compared to non-chemotherapy regimens. The median OS was 25.0 months for those receiving chemotherapy regimens, compared to 11.8 months for those receiving non-chemotherapy regimens (90). Shang et al. evaluated the safety and efficacy of EGFR-TKIs monotherapy (Group T) or combined with pemetrexed-based chemotherapy (Group TC) in 95 advanced NSCLC patients with EGFR/TP53 co-mutations and compared with Group T, Group TC showed significantly improved ORR (55.9% vs. 34.4%, P=0.042), median time to progression (16.1 vs. 11.1 months, P=0.002) were improved, but OS was not significantly prolonged. The median OS of patients without brain metastases in Group TC was longer than in Group T (48.4 vs. 28.8 months, P=0.003) (91). Although grade 3 treatment-related AEs occurred higher in Group TC than in Group T (32.4% vs. 13.1%), combination therapy remains a promising treatment approach for advanced NSCLC patients harboring EGFR/TP 53 co-mutations without brain metastases, especially considering that there are no drugs on the market that specifically target TP53 for the treatment of advanced NSCLC (91).

Brain metastases are a significant challenge in EGFR-mutant NSCLC patients undergoing first-generation EGFR-TKI treatment (92). The combination of gefitinib, carboplatin, and pemetrexed can delay drug resistance but does not prevent brain metastases (84). Approximately 30% of NSCLC patients develop brain metastases, with around half presenting with brain metastases at the time of diagnosis and the remainder developing them during treatment (93). Systemic therapy and whole-brain radiotherapy show promise in treating intracranial progression of baseline brain tumors (40). Osimertinib, particularly when combined with platinum-based chemotherapy, demonstrates superior efficacy in patients with brain metastases, leading to fewer central nervous system progressions, improved remission rates, and enhanced quality of life (80). Despite limited central nervous system penetration by certain drugs, such as cisplatin, carboplatin, and pemetrexed (80), there is speculation that their efficacy in treating brain metastases may be linked to blood-brain barrier disruption (94).

Combining EGFR-TKIs with radiotherapy

Previous randomized trials have demonstrated the efficacy of radiation therapy in treating oligometastatic NSCLC. However, the effectiveness of this treatment in advanced EGFR-mutant NSCLC patients remains uncertain (95,96).

The SINDAS phase III clinical trial revealed that local radiotherapy is safe and improves PFS and OS in patients with oligometastatic disease and EGFR mutations. Median PFS was 12.5 months in the EGFR-TKI-treated group and 20.2 months in the group that received TKI + radiotherapy. Median survival was 17.4 and 25.5 months, respectively, indicating a statistically significant difference (97). A prospective study involving advanced NSCLC patients with EGFR-sensitive mutations compared EGFR-TKI treatment alone and stereotactic body radiation therapy (SBRT) + EGFR-TKI treatment. After 3 months of EGFR-TKI treatment, patients in the combination group received SBRT. As of February 14, 2022, the SBRT + EGFR-TKI group had a median PFS of 17.6 months and a median OS of 33.6 months, while the EGFR-TKI group had a median PFS of 9 months and a median OS of 23.2 months. The addition of SBRT delayed the onset of acquired resistance to EGFR-TKIs, leading to prolonged PFS and OS. This study highlights the potential of SBRT in improving outcomes for patients with oligometastatic EGFR-mutant advanced NSCLC receiving first-generation EGFR-TKIs. Notably, patients who received primary site radiotherapy alone had the most favorable prognosis, with PFS and OS of 27.3 and 49.1 months, respectively (10).

The SINDAS trial illustrated the potential survival benefits of radiotherapy in patients with oligometastatic EGFR-mutant NSCLC. However, this study had limitations, such as excluding patients with brain metastases at enrollment and focusing on first-generation EGFR-TKIs (gefitinib, erlotinib, and icotinib) without considering other TKIs (97). The NORTHSTAR trial, utilizing osimertinib and allowing for some brain metastases, will address the gaps identified in the SINDAS trial (98). Combining EGFR-TKIs with local radiotherapy may still provide additional benefits to patients. Therefore, radiation therapy should be considered even when targeted therapy is being administered (97).

Combining EGFR-TKIs with surgery

Resistance to targeted therapies leads to tumor recurrence in a majority of patients with oncogene-driven advanced NSCLC. Around 60% of NSCLC patients experience their initial disease progression at the primary site (99,100). Following a positive response to EGFR-TKIs, local consolidation therapy directed at residual cancer cells may help overcome resistance and enhance survival (30). Traditionally, treating advanced NSCLC has focused on disease control, symptom management, and enhancing quality of life rather than curative surgery (101). Nevertheless, several studies have highlighted the survival advantages of surgical resection in carefully selected advanced patients, some of whom have extended their survival through surgery (102,103). Surgery can play a critical role in a comprehensive treatment strategy (13).

Kuo et al. conducted a retrospective analysis comparing stage IV patients who underwent primary tumor resection and targeted therapy (n=56) to those who only received targeted therapy (n=224). The long-term follow-up revealed a median PFS of 29.6 months (95% CI: 18.9–40.3) for the surgical group and 13.0 months (95% CI: 11.8–14.2) for the control group (P<0.001). Progression rates were 51.8% and 92.4% for the surgical and control groups, respectively, indicating the potential benefits of primary tumor resection alongside EGFR-TKIs for EGFR-mutant NSCLC patients (13). Park et al. retrospectively analyzed 44 patients who underwent lung surgery post-targeted therapy, reporting 2-year PFS and OS rates of 70.8% and 95.0%, respectively, with minimal complications. However, the long-term impact of pneumonectomy on survival post-targeted therapy requires further investigation, particularly considering the inclusion of patients with non-EGFR mutations in the study (30). The survival benefits observed after resection of primary lung tumors may be attributed to reduced tumor load and increased heterogeneity at the primary site (13).

Lung surgery is a viable option for advanced NSCLC patients who have previously undergone targeted therapies. It can provide adequate specimens for detailed genetic analysis, aiding in developing newer treatment plans. Moreover, surgery can uncover pleural metastases that may be missed by imaging and assess the thoracic cavity for metastases (30). As targeted therapies continue to advance, surgery may have a role in local consolidation therapy for these patients (101). The combination of EGFR-TKIs with surgery shows promise as a therapeutic approach. Several important aspects need to be carefully considered and further investigated in this study. These include the optimal timing of surgery, the duration of preoperative targeted therapy, specific patient selection criteria for surgery, and the extent of surgical resection (30). Some cited articles did not provide trial numbers. Table 3 only summarizes a part of clinical trials.

Table 3

Some clinical trials of the combination therapy are as follows

Experiment registration number Phase Pts Stage Treatment arm(s) Primary endpoint(s) Ref
JPRN-UMIN 000017069 3 226 IIIB, IV Erlotinib + bevacizumab; erlotinib PFS (9)
NCT02824458 3 313 IIIB, IV Gefitinib + apatinib; gefitinib PFS (16)
NCT02759614 3 311 IIIB, IV Bevacizumab + erlotinib; erlotinib PFS (56)
NCT02633189 3 160 IIIB, IV Erlotinib + bevacizumab; erlotinib PFS (17)
NCT02411448 3 211 IV Ramucirumab + erlotinib; erlotinib PFS (57)
NCT02803203 1/2 49 IV Osimertinib + bevacizumab Number of patients who were progression-free at 12 months (59)
JPRN-UMIN 000023761 1/2 80 IV Osimertinib + bevacizumab; osimertinib PFS (60)
NCT02145637 1 30 IV Afatinib + ruxolitinib RP2D, DLT, MTD (15)
NCT02454933 3 29 IIIA, IIIB, IV Osimertinib + durvalumab; osimertinib AEs (12)
NCT01454102 1 21 IIIB, IV Nivolumab + erlotinib; AEs (72)
CTRI/2016/08/007149 3 350 IIIB, IV Gefitinib + pemetrexed + carboplatin; gefitinib PFS (11)
NCT02148380. 3 81 IIIB, IV Pemetrexed + carboplatin + gefitinib; gefitinib PFS (82)
JPRN-UMIN 000006340 3 345 IIIA, IIIB, IV Pemetrexed + carboplatin + gefitinib; gefitinib PFS, PFS2, OS (83)
NCT02886195 2 21 IV Gefitinib + carboplatin + pemetrexed PFS (84)
JPRN-UMIN 000002789 2 80 IIIB, IV Concurrent gefitinib + carboplatin + pemetrexed; sequential alternating gefitinib + carboplatin + pemetrexed PFS (88)
NCT03544814 2 99 IIIB, IV First icotinib and then pemetrexed + cisplatin; icotinib combined with pemetrexed + cisplatin PFS (89)
NCT02893332 3 133 IV First-generation TKI (gefitinib/erlotinib/icotinib); first-generation TKI (gefitinib/erlotinib/icotinib) + RT PFS (97)
NCT03595644 2 62 IV First-generation TKI (gefitinib/erlotinib/icotinib); first-generation TKI (gefitinib/erlotinib/icotinib) + SBRT PFS (10)
NCT03410043 2 143 IIIB, IV Osimertinib + surgery or RT; osimertinib PFS (98)

Pts, patients; PFS, progression-free survival; RP2D, recommended phase 2 dose; DLT, dose-limiting toxicities; MTD, maximum tolerated dose; AEs, adverse events; PFS2, progression-free survival 2; OS, overall survival; TKI, tyrosine kinase inhibitor; RT, radiotherapy; SBRT, stereotactic body radiation therapy.


Factors affecting EGFR-TKIs in treating patients with advanced NSCLC

Various factors influence the clinical prognosis and effectiveness of EGFR-TKIs in treating advanced NSCLC patients, with genetic mutations playing a significant role. These include EGFR mutations (19del, L858R, T790M, exon20ins), and other conditions (20,104-106). Research indicates that patients with 19del mutations may have a more favorable prognosis than those with L858R mutations (20,104,107). Moreover, a retrospective study has shown that osimertinib demonstrates superior efficacy in patients who develop T790M mutations following first-line EGFR-TKI treatment (104). Additionally, analysis by Bazhenova et al. involving 2,825 patients treated with EGFR-TKIs revealed that patients with exon20ins mutations had a poor prognosis and minimal benefit, with a median real-world OS of 2.9 months compared to 10.5 months in patients with common EGFR mutations (107). Genetic factors such as BIM deletion polymorphisms, MET amplification, HER2 amplification, and AXL activation and amplification have been observed to impact the clinical outcomes and effectiveness of EGFR-TKIs in treating advanced NSCLC patients (7). A study examined the impact of PD-L1 expression on the prognosis of advanced NSCLC patients treated with first/second-generation EGFR-TKIs. Patients with high PD-L1 expression had a PFS of 6.6 months and an OS of 11.5 months, significantly lower than those with low PD-L1 expression (PFS: 13.0 months, OS: 32.9 months). This suggests that high PD-L1 expression is linked to early EGFR-TKIs resistance and lower survival rates, making it a more effective predictive biomarker than physical status, mutation type, and disease site (105). Additionally, the presence of seven driver genes (ALK, KRAS, BIM, PIK3CA, MET, IGF1R, and PTEN) as resistance genes was identified through multigene coanalysis of EGFR-sensitive mutations in advanced patients. It was observed that carriers of resistance genes with aberrant alterations in the EGFR signaling pathway had significantly lower PFS compared to those with normal EGFR signaling. Furthermore, phosphatase and tensin homolog (PTEN) deletion, low expression, and MET fluorescence in situ hybridization (FISH)+ were identified as independent predictors of PFS in patients with EGFR-TKI prolongation after adjusting for multiple factors (108).

In addition to genetic background, clinical factors can impact the effectiveness of EGFR-TKIs in treating advanced EGFR-mutant NSCLC patients. Lin et al. conducted a study on 94 patients and found that various factors such as gender, smoking history, pathology type, EGFR mutation type, brain metastasis, and duration of targeted therapy did not significantly affect patient outcomes. The Eastern Cooperative Oncology Group score was identified as an independent prognostic factor for PFS in advanced NSCLC patients, while both the Eastern Cooperative Oncology Group score and brain metastasis were identified as independent prognostic factors for OS (109). Interstitial lung disease grade was also found to influence the efficacy and prognosis of EGFR-TKIs, with higher grades correlating with a higher risk of early progression (110). Studies on serum tumor markers have shown that negative pro-gastrin-releasing peptide (ProGRP) and neuron-specific enolase (NSE) levels, as well as a history of never smoking, may indicate longer PFS in patients treated with EGFR-TKIs. However, the predictive value of ProGRP and NSE is limited (111). However, additional studies of advanced patients have identified several factors that may affect the benefit of EGFR-TKIs, including EGFR mutation type, gender, performance status and smoking status (112-114).


Discussion

Primary tumor cells are anticipated to be conditionally reprogrammed, and single-cell level drug sensitivity assays can help determine the most effective medication for lung cancer patients (115,116). The selection of optimal medication is based on the overall response of the patient population to the drug, as in the current practice. The urgent need to identify drug resistance mechanisms and develop new targeted drugs is highlighted to overcome the challenge of long-term effective treatment for NSCLC patients with EGFR-TKIs. A comprehensive understanding of drug resistance mechanisms will assist physicians in exploring personalized treatment options, achieving precision therapy, and improving lung cancer patients’ quality of life and survival rates. The type of EGFR mutation can vary during drug therapy, necessitating dynamic long-term patient management. Non-invasive liquid biopsy technology should be employed to monitor patients’ EGFR mutation status continuously, and drug selection should be guided by the mutation status determined through histological biopsy (7). Several drugs currently in development, such as EAI 045, CH 7233163, JBJ-04-125-02, BLU-945, and BLU-701, hold promise for benefiting NSCLC patients in the future. Various therapeutic approaches for advanced EGFR-mutant NSCLC, including combinations of EGFR-TKIs with VEGF inhibitors, MET-TKIs, surgery, chemotherapy, and radiotherapy, have shown varying degrees of efficacy. However, no significant survival advantage has been observed when combined with JAK inhibitors and ICIs.

Conducting research and analysis on EGFR-TKIs to treat advanced EGFR-sensitive mutant NSCLC patients will enhance our understanding of the current landscape in this field and shed light on this specific patient population. This article offers a comprehensive overview of the current status of both monotherapy and combination therapy involving EGFR-TKIs, drawing insights from real-world data obtained from multiple clinical trials. It delves into treatment trends and influential factors and provides recommendations for clinicians on the selective use of medications and novel approaches for assessing the efficacy and prognosis of EGFR-TKIs in advanced NSCLC treatment. The limitations of this paper are objective, and since it is a summary of the current study, some of the findings need to be validated and explored in further large clinical trials.


Conclusions

Although EGFR-TKIs have largely improved the survival and prognosis of patients with EGFR mutations, more efforts are needed to benefit patients in the long term. Developing new EGFR-TKIs and combining other drugs based on EGFR-TKIs for lung cancer treatment will become a hotspot for future research. Overcoming drug resistance mechanisms, finding new targets, and improving efficacy through combination therapy will become the hope for patients with advanced lung cancer.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-637/rc

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-637/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-24-637/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

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


References

  1. Nasim F, Sabath BF, Eapen GA. Lung Cancer. Med Clin North Am 2019;103:463-73. [Crossref] [PubMed]
  2. Aran V, Omerovic J. Current Approaches in NSCLC Targeting K-RAS and EGFR. Int J Mol Sci 2019;20:5701. [Crossref] [PubMed]
  3. Zhang C, Wang L, Li W, et al. Surgical outcomes of stage IV non-small cell lung cancer: a single-center experience. J Thorac Dis 2019;11:5463-73. [Crossref] [PubMed]
  4. Zhou Q, Zhang HL, Jiang LY, et al. Real-world evidence of osimertinib in Chinese patients with EGFR T790M-positive non-small cell lung cancer: a subgroup analysis from ASTRIS study. J Cancer Res Clin Oncol 2023;149:10771-80. [Crossref] [PubMed]
  5. Wu YL, Tsuboi M, John T, et al. A plain language summary of results from the ADAURA study: osimertinib after surgery for patients who have early-stage EGFR-mutated non-small cell lung cancer. Future Oncol 2021;17:4827-35. [Crossref] [PubMed]
  6. Shirley M, Keam SJ. Aumolertinib: A Review in Non-Small Cell Lung Cancer. Drugs 2022;82:577-84. [Crossref] [PubMed]
  7. He J, Huang Z, Han L, et al. Mechanisms and management of 3rd generation EGFR TKI resistance in advanced non small cell lung cancer Int J Oncol 2021;59:90. (Review). [Crossref] [PubMed]
  8. Kaewjanthong P, Sooksai S, Sasano H, et al. Cell-penetrating peptides containing the progesterone receptor polyproline domain inhibits EGF signaling and cell proliferation in lung cancer cells. PLoS One 2022;17:e0264717. [Crossref] [PubMed]
  9. Kawashima Y, Fukuhara T, Saito H, et al. Bevacizumab plus erlotinib versus erlotinib alone in Japanese patients with advanced, metastatic, EGFR-mutant non-small-cell lung cancer (NEJ026): overall survival analysis of an open-label, randomised, multicentre, phase 3 trial. Lancet Respir Med 2022;10:72-82. [Crossref] [PubMed]
  10. Peng P, Gong J, Zhang Y, et al. EGFR-TKIs plus stereotactic body radiation therapy (SBRT) for stage IV Non-small cell lung cancer (NSCLC): A prospective, multicenter, randomized, controlled phase II study. Radiother Oncol 2023;184:109681. [Crossref] [PubMed]
  11. Noronha V, Patil VM, Joshi A, et al. Gefitinib Versus Gefitinib Plus Pemetrexed and Carboplatin Chemotherapy in EGFR-Mutated Lung Cancer. J Clin Oncol 2020;38:124-36. [Crossref] [PubMed]
  12. Yang JC, Shepherd FA, Kim DW, 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;14:933-9. [Crossref] [PubMed]
  13. Kuo SW, Chen PH, Lu TP, et al. Primary Tumor Resection for Stage IV Non-small-cell Lung Cancer Without Progression After First-Line Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor Treatment: A Retrospective Case-Control Study. Ann Surg Oncol 2022;29:4873-84. [Crossref] [PubMed]
  14. Vansteenkiste JF, Van De Kerkhove C, Wauters E, et al. Capmatinib for the treatment of non-small cell lung cancer. Expert Rev Anticancer Ther 2019;19:659-71. [Crossref] [PubMed]
  15. Park JS, Hong MH, Chun YJ, et al. A phase Ib study of the combination of afatinib and ruxolitinib in EGFR mutant NSCLC with progression on EGFR-TKIs. Lung Cancer 2019;134:46-51. [Crossref] [PubMed]
  16. Zhao H, Yao W, Min X, et al. Apatinib Plus Gefitinib as First-Line Treatment in Advanced EGFR-Mutant NSCLC: The Phase III ACTIVE Study (CTONG1706). J Thorac Oncol 2021;16:1533-46. [Crossref] [PubMed]
  17. Piccirillo MC, Bonanno L, Garassino MC, et al. Addition of Bevacizumab to Erlotinib as First-Line Treatment of Patients With EGFR-Mutated Advanced Nonsquamous NSCLC: The BEVERLY Multicenter Randomized Phase 3 Trial. J Thorac Oncol 2022;17:1086-97. [Crossref] [PubMed]
  18. Zhou C, Wu YL, Chen G, et al. Final overall survival results from a randomised, phase III study of erlotinib versus chemotherapy as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer (OPTIMAL, CTONG-0802). Ann Oncol 2015;26:1877-83. [Crossref] [PubMed]
  19. Shi YK, Wang L, Han BH, et al. First-line icotinib versus cisplatin/pemetrexed plus pemetrexed maintenance therapy for patients with advanced EGFR mutation-positive lung adenocarcinoma (CONVINCE): a phase 3, open-label, randomized study. Ann Oncol 2017;28:2443-50. [Crossref] [PubMed]
  20. Rosell R, Carcereny E, Gervais R, 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;13:239-46. [Crossref] [PubMed]
  21. Yang JC, Sequist LV, Geater SL, et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol 2015;16:830-8. [Crossref] [PubMed]
  22. Wu YL, Zhou C, Hu CP, 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;15:213-22. [Crossref] [PubMed]
  23. Mok TS, Cheng Y, Zhou X, et al. Improvement in Overall Survival in a Randomized Study That Compared Dacomitinib With Gefitinib in Patients With Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. J Clin Oncol 2018;36:2244-50. [Crossref] [PubMed]
  24. Yang JC, Wu YL, Schuler M, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015;16:141-51. [Crossref] [PubMed]
  25. Lu S, Dong X, Jian H, et al. AENEAS: A Randomized Phase III Trial of Aumolertinib Versus Gefitinib as First-Line Therapy for Locally Advanced or MetastaticNon-Small-Cell Lung Cancer With EGFR Exon 19 Deletion or L858R Mutations. J Clin Oncol 2022;40:3162-71. [Crossref] [PubMed]
  26. Shi Y, Chen G, Wang X, et al. Furmonertinib (AST2818) versus gefitinib as first-line therapy for Chinese patients with locally advanced or metastatic EGFR mutation-positive non-small-cell lung cancer (FURLONG): a multicentre, double-blind, randomised phase 3 study. Lancet Respir Med 2022;10:1019-28. [Crossref] [PubMed]
  27. Ahn MJ, Han JY, Lee KH, et al. Lazertinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: results from the dose escalation and dose expansion parts of a first-in-human, open-label, multicentre, phase 1-2 study. Lancet Oncol 2019;20:1681-90. [Crossref] [PubMed]
  28. Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N Engl J Med 2020;382:41-50. [Crossref] [PubMed]
  29. Cho BC, Han JY, Kim SW, et al. A Phase 1/2 Study of Lazertinib 240 mg in Patients With Advanced EGFR T790M-Positive NSCLC After Previous EGFR Tyrosine Kinase Inhibitors. J Thorac Oncol 2022;17:558-67. [Crossref] [PubMed]
  30. Park BJ, Shim HS, Lee CY, et al. Genetic Analysis and Operative Outcomes in Patients with Oncogene-Driven Advanced NSCLC Treated with Cytoreductive Surgery as a Component of Local Consolidative Therapy. Cancers (Basel) 2021;13:2549. [Crossref] [PubMed]
  31. Saito H, Fukuhara T, Furuya N, et al. Erlotinib plus bevacizumab versus erlotinib alone in patients with EGFR-positive advanced non-squamous non-small-cell lung cancer (NEJ026): interim analysis of an open-label, randomised, multicentre, phase 3 trial. Lancet Oncol 2019;20:625-35. [Crossref] [PubMed]
  32. Park K, Tan EH, O'Byrne K, et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol 2016;17:577-89. [Crossref] [PubMed]
  33. Wu YL, Cheng Y, Zhou X, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol 2017;18:1454-66. [Crossref] [PubMed]
  34. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:113-25. [Crossref] [PubMed]
  35. Yi L, Fan J, Qian R, et al. Efficacy and safety of osimertinib in treating EGFR-mutated advanced NSCLC: A meta-analysis. Int J Cancer 2019;145:284-94. [Crossref] [PubMed]
  36. Leonetti A, Sharma S, Minari R, et al. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer 2019;121:725-37. [Crossref] [PubMed]
  37. Piper-Vallillo AJ, Sequist LV, Piotrowska Z. Emerging Treatment Paradigms for EGFR-Mutant Lung Cancers Progressing on Osimertinib: A Review. J Clin Oncol 2020; Epub ahead of print. [Crossref] [PubMed]
  38. Winther-Larsen A, Nissen PH, Jakobsen KR, et al. Genetic polymorphism in the epidermal growth factor receptor gene predicts outcome in advanced non-small cell lung cancer patients treated with erlotinib. Lung Cancer 2015;90:314-20. [Crossref] [PubMed]
  39. Cooper AJ, Sequist LV, Lin JJ. Third-generation EGFR and ALK inhibitors: mechanisms of resistance and management. Nat Rev Clin Oncol 2022;19:499-514. [Crossref] [PubMed]
  40. Yang Z, Yang J, Chen Y, et al. Acquired EGFR L718V Mutation as the Mechanism for Osimertinib Resistance in a T790M-Negative Non-Small-Cell Lung Cancer Patient. Target Oncol 2019;14:369-74. [Crossref] [PubMed]
  41. Kelly RJ, Shepherd FA, Krivoshik A, et al. A phase III, randomized, open-label study of ASP8273 versus erlotinib or gefitinib in patients with advanced stage IIIB/IV non-small-cell lung cancer. Ann Oncol 2019;30:1127-33. [Crossref] [PubMed]
  42. Wang S, Song Y, Liu D. EAI045: The fourth-generation EGFR inhibitor overcoming T790M and C797S resistance. Cancer Lett 2017;385:51-4. [Crossref] [PubMed]
  43. Jia Y, Yun CH, Park E, et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature 2016;534:129-32. [Crossref] [PubMed]
  44. Kashima K, Kawauchi H, Tanimura H, et al. CH7233163 Overcomes Osimertinib-Resistant EGFR-Del19/T790M/C797S Mutation. Mol Cancer Ther 2020;19:2288-97. [Crossref] [PubMed]
  45. To C, Jang J, Chen T, et al. Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor. Cancer Discov 2019;9:926-43. [Crossref] [PubMed]
  46. Eno MS, Brubaker JD, Campbell JE, et al. Discovery of BLU-945, a Reversible, Potent, and Wild-Type-Sparing Next-Generation EGFR Mutant Inhibitor for Treatment-Resistant Non-Small-Cell Lung Cancer. J Med Chem 2022;65:9662-77. [Crossref] [PubMed]
  47. Laudadio E, Mangano L, Minnelli C. Chemical Scaffolds for the Clinical Development of Mutant-Selective and Reversible Fourth-Generation EGFR-TKIs in NSCLC. ACS Chem Biol 2024;19:839-54. [Crossref] [PubMed]
  48. Ferrara N, Adamis AP. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov 2016;15:385-403. [Crossref] [PubMed]
  49. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer 2013;13:871-82. [Crossref] [PubMed]
  50. Nilsson MB, Robichaux J, Herynk MH, et al. Altered Regulation of HIF-1α in Naive- and Drug-Resistant EGFR-Mutant NSCLC: Implications for a Vascular Endothelial Growth Factor-Dependent Phenotype. J Thorac Oncol 2021;16:439-51. [Crossref] [PubMed]
  51. Le X, Nilsson M, Goldman J, et al. Dual EGFR-VEGF Pathway Inhibition: A Promising Strategy for Patients With EGFR-Mutant NSCLC. J Thorac Oncol 2021;16:205-15. [Crossref] [PubMed]
  52. Larkins E, Scepura B, Blumenthal GM, et al. U.S. Food and Drug Administration Approval Summary: Ramucirumab for the Treatment of Metastatic Non-Small Cell Lung Cancer Following Disease Progression On or After Platinum-Based Chemotherapy. Oncologist 2015;20:1320-5. [Crossref] [PubMed]
  53. Chen YC, Chen JH, Hsieh FI. Major adverse cardiovascular events of vascular endothelial growth factor tyrosine kinase inhibitors among patients with different malignancy: A systemic review and network meta-analysis. J Chin Med Assoc 2024;87:48-57. [PubMed]
  54. Duch P, Díaz-Valdivia N, Gabasa M, et al. Aberrant TIMP-1 production in tumor-associated fibroblasts drives the selective benefits of nintedanib in lung adenocarcinoma. Cancer Sci 2024;115:1505-19. [Crossref] [PubMed]
  55. Fan Q, Wu G, Chen M, et al. Cediranib ameliorates portal hypertensive syndrome via inhibition of VEGFR-2 signaling in cirrhotic rats. Eur J Pharmacol 2024;964:176278. [Crossref] [PubMed]
  56. Zhou Q, Xu CR, Cheng Y, et al. Bevacizumab plus erlotinib in Chinese patients with untreated, EGFR-mutated, advanced NSCLC (ARTEMIS-CTONG1509): A multicenter phase 3 study. Cancer Cell 2021;39:1279-1291.e3. [Crossref] [PubMed]
  57. Nishio M, Paz-Ares L, Reck M, et al. RELAY, Ramucirumab Plus Erlotinib (RAM+ERL) in Untreated Metastatic EGFR-Mutant NSCLC (EGFR+ NSCLC): Association Between TP53 Status and Clinical Outcome. Clin Lung Cancer 2023;24:415-28. [Crossref] [PubMed]
  58. Nakagawa K, Garon EB, Seto T, et al. Ramucirumab plus erlotinib in patients with untreated, EGFR-mutated, advanced non-small-cell lung cancer (RELAY): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2019;20:1655-69. [Crossref] [PubMed]
  59. Yu HA, Schoenfeld AJ, Makhnin A, et al. Effect of Osimertinib and Bevacizumab on Progression-Free Survival for Patients With Metastatic EGFR-Mutant Lung Cancers: A Phase 1/2 Single-Group Open-Label Trial. JAMA Oncol 2020;6:1048-54. [Crossref] [PubMed]
  60. Akamatsu H, Teraoka S, Morita S, et al. Phase I/II Study of Osimertinib With Bevacizumab in EGFR-mutated, T790M-positive Patients With Progressed EGFR-TKIs: West Japan Oncology Group 8715L (WJOG8715L). Clin Lung Cancer 2019;20:e492-4. [Crossref] [PubMed]
  61. Xin P, Xu X, Deng C, et al. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol 2020;80:106210. [Crossref] [PubMed]
  62. Su Q, Banks E, Bebernitz G, et al. Discovery of (2R)-N-[3-[2-[(3-Methoxy-1-methyl-pyrazol-4-yl)amino]pyrimidin-4-yl]-1H-indol-7-yl]-2-(4-methylpiperazin-1-yl)propenamide (AZD4205) as a Potent and Selective Janus Kinase 1 Inhibitor. J Med Chem 2020;63:4517-27. [Crossref] [PubMed]
  63. Rivas S, Marín A, Samtani S, et al. MET Signaling Pathways, Resistance Mechanisms, and Opportunities for Target Therapies. Int J Mol Sci 2022;23:13898. [Crossref] [PubMed]
  64. Lai GGY, Lim TH, Lim J, et al. Clonal MET Amplification as a Determinant of Tyrosine Kinase Inhibitor Resistance in Epidermal Growth Factor Receptor-Mutant Non-Small-Cell Lung Cancer. J Clin Oncol 2019;37:876-84. [Crossref] [PubMed]
  65. Liu X, Wang Q, Yang G, et al. A novel kinase inhibitor, INCB28060, blocks c-MET-dependent signaling, neoplastic activities, and cross-talk with EGFR and HER-3. Clin Cancer Res 2011;17:7127-38. [Crossref] [PubMed]
  66. Baltschukat S, Engstler BS, Huang A, et al. Capmatinib (INC280) Is Active Against Models of Non-Small Cell Lung Cancer and Other Cancer Types with Defined Mechanisms of MET Activation. Clin Cancer Res 2019;25:3164-75. [Crossref] [PubMed]
  67. Lara MS, Holland WS, Chinn D, et al. Preclinical Evaluation of MET Inhibitor INC-280 With or Without the Epidermal Growth Factor Receptor Inhibitor Erlotinib in Non-Small-Cell Lung Cancer. Clin Lung Cancer 2017;18:281-5. [Crossref] [PubMed]
  68. Wu YL, Zhang L, Kim DW, et al. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients With EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer. J Clin Oncol 2018;36:3101-9. [Crossref] [PubMed]
  69. Yang H, Miao Y, Yu Z, et al. Cell adhesion molecules and immunotherapy in advanced non-small cell lung cancer: Current process and potential application. Front Oncol 2023;13:1107631. [Crossref] [PubMed]
  70. Zhou F, Zhou C. Chemotherapy Should Be Combined With Checkpoint Inhibitors in the Treatment of Patients With Stage IV EGFR-Mutant NSCLC Whose Disease Has Progressed on All Available Tyrosine Kinase Inhibitors. J Thorac Oncol 2021;16:1622-6. [Crossref] [PubMed]
  71. Chen N, Fang W, Zhan J, 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;10:910-23. [Crossref] [PubMed]
  72. Gettinger S, Hellmann MD, Chow LQM, et al. Nivolumab Plus Erlotinib in Patients With EGFR-Mutant Advanced NSCLC. J Thorac Oncol 2018;13:1363-72. [Crossref] [PubMed]
  73. Lee CK, Man J, Lord S, et al. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non-Small Cell Lung Cancer-A Meta-Analysis. J Thorac Oncol 2017;12:403-7. [Crossref] [PubMed]
  74. Akamatsu H, Murakami E, Oyanagi J, et al. Immune-Related Adverse Events by Immune Checkpoint Inhibitors Significantly Predict Durable Efficacy Even in Responders with Advanced Non-Small Cell Lung Cancer. Oncologist 2020;25:e679-83. [Crossref] [PubMed]
  75. Yang Z, Tam KY. Combination Strategies Using EGFR-TKi in NSCLC Therapy: Learning from the Gap between Pre-Clinical Results and Clinical Outcomes. Int J Biol Sci 2018;14:204-16. [Crossref] [PubMed]
  76. Galvani E, Peters GJ, Giovannetti E. Thymidylate synthase inhibitors for non-small cell lung cancer. Expert Opin Investig Drugs 2011;20:1343-56. [Crossref] [PubMed]
  77. Cheng Y, Murakami H, Yang PC, et al. Randomized Phase II Trial of Gefitinib With and Without Pemetrexed as First-Line Therapy in Patients With Advanced Nonsquamous Non-Small-Cell Lung Cancer With Activating Epidermal Growth Factor Receptor Mutations. J Clin Oncol 2016;34:3258-66. [Crossref] [PubMed]
  78. Pan C, Duan H, Wu Y, et al. Inhibition of DNA PK by gefitinib causes synergism between gefitinib and cisplatin in NSCLC. Int J Oncol 2020;57:939-55. [Crossref] [PubMed]
  79. Gu W, Zhang H, Lu Y, et al. EGFR-TKI Combined with Pemetrexed versus EGFR-TKI Monotherapy in Advanced EGFR-mutated NSCLC: A Prospective, Randomized, Exploratory Study. Cancer Res Treat 2023;55:841-50. [Crossref] [PubMed]
  80. Jänne PA, Planchard D, Kobayashi K, et al. CNS Efficacy of Osimertinib With or Without Chemotherapy in Epidermal Growth Factor Receptor-Mutated Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2024;42:808-20. [Crossref] [PubMed]
  81. Hou X, Li M, Wu G, et al. Gefitinib Plus Chemotherapy vs Gefitinib Alone in Untreated EGFR-Mutant Non-Small Cell Lung Cancer in Patients With Brain Metastases: The GAP BRAIN Open-Label, Randomized, Multicenter, Phase 3 Study. JAMA Netw Open 2023;6:e2255050. [Crossref] [PubMed]
  82. Lou Y, Xu J, Zhang Y, et al. Chemotherapy Plus EGFR-TKI as First-Line Treatment Provides Better Survival for Advanced EGFR-Positive Lung Adenocarcinoma Patients: Updated Data and Exploratory In Vitro Study. Target Oncol 2020;15:175-84. [Crossref] [PubMed]
  83. Hosomi Y, Morita S, Sugawara S, et al. Gefitinib Alone Versus Gefitinib Plus Chemotherapy for Non-Small-Cell Lung Cancer With Mutated Epidermal Growth Factor Receptor: NEJ009 Study. J Clin Oncol 2020;38:115-23. [Crossref] [PubMed]
  84. Lin J, Li M, Chen S, et al. Efficacy and Safety of First-Generation EGFR-TKIs Combined with Chemotherapy for Treatment-Naïve Advanced Non-Small-Cell Lung Cancer Patients Harboring Sensitive EGFR Mutations: A Single-Center, Open-Label, Single-Arm, Phase II Clinical Trial. J Inflamm Res 2021;14:2557-67. [Crossref] [PubMed]
  85. Wu Q, Luo W, Li W, et al. First-Generation EGFR-TKI Plus Chemotherapy Versus EGFR-TKI Alone as First-Line Treatment in Advanced NSCLC With EGFR Activating Mutation: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Oncol 2021;11:598265. [Crossref] [PubMed]
  86. Li X, Cai W, Yang G, et al. Comprehensive Analysis of EGFR-Mutant Abundance and Its Effect on Efficacy of EGFR TKIs in Advanced NSCLC with EGFR Mutations. J Thorac Oncol 2017;12:1388-97. [Crossref] [PubMed]
  87. Yan X, Wang H, Li P, et al. Efficacy of first-line treatment with epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) alone or in combination with chemotherapy for advanced non-small cell lung cancer (NSCLC) with low-abundance mutation. Lung Cancer 2019;128:6-12. [Crossref] [PubMed]
  88. Sugawara S, Oizumi S, Minato K, et al. Randomized phase II study of concurrent versus sequential alternating gefitinib and chemotherapy in previously untreated non-small cell lung cancer with sensitive EGFR mutations: NEJ005/TCOG0902. Ann Oncol 2015;26:888-94. [Crossref] [PubMed]
  89. Chang Q, Xu J, Qiang H, et al. EGFR Tyrosine Kinase Inhibitor (TKI) Combined With Concurrent or Sequential Chemotherapy for Patients With Advanced Lung Cancer and Gradual Progression After First-Line EGFR-TKI Therapy: A Randomized Controlled Study. Clin Lung Cancer 2021;22:e395-404. [Crossref] [PubMed]
  90. Mu Y, Hao X, Yang K, et al. Clinical Modality of Resistance and Subsequent Management of Patients with Advanced Non-small Cell Lung Cancer Failing Treatment with Osimertinib. Target Oncol 2019;14:335-42. [Crossref] [PubMed]
  91. Shang K, Huang H, Xu Y, et al. Efficacy and safety analyses of epidermal growth factor receptor tyrosine kinase inhibitors combined with chemotherapy in the treatment of advanced non-small-cell lung cancer with an EGFR/TP53 co-mutation. BMC Cancer 2022;22:1295. [Crossref] [PubMed]
  92. Joo JW, Hong MH, Shim HS. Clinical characteristics of T790M-positive lung adenocarcinoma after resistance to epidermal growth factor receptor-tyrosine kinase inhibitors with an emphasis on brain metastasis and survival. Lung Cancer 2018;121:12-7. [Crossref] [PubMed]
  93. Yen CT, Wu WJ, Chen YT, et al. Surgical resection of brain metastases prolongs overall survival in non-small-cell lung cancer. Am J Cancer Res 2021;11:6160-72. [PubMed]
  94. Yu X, Fan Y. Effect of pemetrexed on brain metastases from nonsmall cell lung cancer with wild-type and unknown EGFR status. Medicine (Baltimore) 2019;98:e14110. [Crossref] [PubMed]
  95. Iyengar P, Wardak Z, Gerber DE, et al. Consolidative Radiotherapy for Limited Metastatic Non-Small-Cell Lung Cancer: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2018;4:e173501. [Crossref] [PubMed]
  96. Gomez DR, Tang C, Zhang J, et al. Local Consolidative Therapy Vs. Maintenance Therapy or Observation for Patients With Oligometastatic Non-Small-Cell Lung Cancer: Long-Term Results of a Multi-Institutional, Phase II, Randomized Study. J Clin Oncol 2019;37:1558-65. [Crossref] [PubMed]
  97. Wang XS, Bai YF, Verma V, et al. Randomized Trial of First-Line Tyrosine Kinase Inhibitor With or Without Radiotherapy for Synchronous Oligometastatic EGFR-Mutated Non-Small Cell Lung Cancer. J Natl Cancer Inst 2023;115:742-8. [Crossref] [PubMed]
  98. Khan TM, Verbus EA, Gandhi S, et al. Osimertinib, Surgery, and Radiation Therapy in Treating Patients with Stage IIIB or IV Non-Small Cell Lung Cancer with EGFR Mutations (NORTHSTAR). Ann Surg Oncol 2022;29:4688-9. [Crossref] [PubMed]
  99. Al-Halabi H, Sayegh K, Digamurthy SR, et al. Pattern of Failure Analysis in Metastatic EGFR-Mutant Lung Cancer Treated with Tyrosine Kinase Inhibitors to Identify Candidates for Consolidation Stereotactic Body Radiation Therapy. J Thorac Oncol 2015;10:1601-7. [Crossref] [PubMed]
  100. Sorensen BS, Wu L, Wei W, et al. Monitoring of epidermal growth factor receptor tyrosine kinase inhibitor-sensitizing and resistance mutations in the plasma DNA of patients with advanced non-small cell lung cancer during treatment with erlotinib. Cancer 2014;120:3896-901. [Crossref] [PubMed]
  101. David EA, Clark JM, Cooke DT, et al. The Role of Thoracic Surgery in the Therapeutic Management of Metastatic Non-Small Cell Lung Cancer. J Thorac Oncol 2017;12:1636-45. [Crossref] [PubMed]
  102. Hanagiri T, Takenaka M, Oka S, et al. Results of a surgical resection for patients with stage IV non--small-cell lung cancer. Clin Lung Cancer 2012;13:220-4. [Crossref] [PubMed]
  103. Collaud S, Stahel R, Inci I, et al. Survival of patients treated surgically for synchronous single-organ metastatic NSCLC and advanced pathologic TN stage. Lung Cancer 2012;78:234-8. [Crossref] [PubMed]
  104. Tang YJ, Chang JW, Chang CF, et al. Impact of T790M Mutation Status on Later-Line Osimertinib Treatment in Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2022;14:5095. [Crossref] [PubMed]
  105. Liu J, Itchins M, Nagrial A, et al. Relationship between PD-L1 expression and outcome in EGFR-mutant lung cancer patients treated with EGFR tyrosine kinase inhibitors. Lung Cancer 2021;155:28-33. [Crossref] [PubMed]
  106. Liu J, Xiang Y, Fang T, et al. Advances in the Diagnosis and Treatment of Advanced Non-Small-Cell Lung Cancer With EGFR Exon 20 Insertion Mutation. Clin Lung Cancer 2024;25:100-8. [Crossref] [PubMed]
  107. Bazhenova L, Minchom A, Viteri S, et al. Comparative clinical outcomes for patients with advanced NSCLC harboring EGFR exon 20 insertion mutations and common EGFR mutations. Lung Cancer 2021;162:154-61. [Crossref] [PubMed]
  108. Wang F, Diao XY, Zhang X, et al. Identification of genetic alterations associated with primary resistance to EGFR-TKIs in advanced non-small-cell lung cancer patients with EGFR sensitive mutations. Cancer Commun (Lond) 2019;39:7. [Crossref] [PubMed]
  109. Lin JH, Lin D, Xu L, et al. The association between clinical prognostic factors and epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) efficacy in advanced non-small-cell lung cancer patients: a retrospective assessment of 94 cases with EGFR mutations. Oncotarget 2017;8:3412-21. [Crossref] [PubMed]
  110. Zhang XY, Cao R, Guo YJ, et al. Impact of pulmonary interstitial lesions on efficacy and prognosis of EGFR-TKI-treated advanced non-small cell lung cancers. J Thorac Dis 2020;12:839-48. [Crossref] [PubMed]
  111. Dong J, Tong S, Shi X, et al. Progastrin-Releasing Peptide Precursor and Neuron-Specific Enolase Predict the Efficacy of First-Line Treatment with Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors Among Non-Small-Cell Lung Cancer Patients Harboring EGFR Mutations. Cancer Manag Res 2021;12:13607-16. [Crossref] [PubMed]
  112. Lee CK, Wu YL, Ding PN, et al. Impact of Specific Epidermal Growth Factor Receptor (EGFR) Mutations and Clinical Characteristics on Outcomes After Treatment With EGFR Tyrosine Kinase Inhibitors Versus Chemotherapy in EGFR-Mutant Lung Cancer: A Meta-Analysis. J Clin Oncol 2015;33:1958-65. [Crossref] [PubMed]
  113. Okuma Y, Shintani Y, Sekine I, et al. Efficacy of Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors in Metastatic Non-Small Cell Lung Cancer Patients with Poor Performance Status and Epidermal Growth Factor Receptor Mutations: Findings from the Japanese Lung Cancer Registry Database. Clin Lung Cancer 2024;25:336-346.e2. [Crossref] [PubMed]
  114. Lassalle S, Hofman V, Heeke S, et al. Targeted Assessment of the EGFR Status as Reflex Testing in Treatment-Naive Non-Squamous Cell Lung Carcinoma Patients: A Single Laboratory Experience (LPCE, Nice, France). Cancers (Basel) 2020;12:955. [Crossref] [PubMed]
  115. Zhong M, Fu L. Culture and application of conditionally reprogrammed primary tumor cells. Gastroenterol Rep (Oxf) 2020;8:224-33. [Crossref] [PubMed]
  116. Yang C, Yang C, Yarden Y, et al. The prospects of tumor chemosensitivity testing at the single-cell level. Drug Resist Updat 2021;54:100741. [Crossref] [PubMed]
Cite this article as: Kuang L, Wang P, Zhou L, Li Y. Strategies and influencing factors for the treatment of advanced non-small cell lung cancer based on epidermal growth factor receptor tyrosine kinase inhibitors: a narrative review. Transl Cancer Res 2024;13(9):5123-5140. doi: 10.21037/tcr-24-637

Download Citation