Open Access

EGFR mutations in non‑small cell lung cancer: Classification, characteristics and resistance to third‑generation EGFR‑tyrosine kinase inhibitors (Review)

  • Authors:
    • Zhe Tian
    • Lilan Cen
    • Feng Wei
    • Jue Dong
    • Yulan Huang
    • Yi Han
    • Zhibo Wang
    • Junhua Deng
    • Yujie Jiang
  • View Affiliations

  • Published online on: June 2, 2025     https://doi.org/10.3892/ol.2025.15121
  • Article Number: 375
  • Copyright: © Tian et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mutations in EGFR (mEGFRs) in non‑small cell lung cancer (NSCLC) are key factors driving tumor development and treatment response. The present article aimed to review the classification, characteristics and molecular mechanisms of resistance to third‑generation EGFR‑tyrosine kinase inhibitors (EGFR‑TKIs) associated with mEGFRs. Activating mutations (such as L858R and exon 19 deletions) are the primary markers of sensitivity to EGFR‑TKIs, while rare mutations (such as G719X and S768I) require individualized treatment strategies. Resistance mechanisms are categorized into EGFR‑dependent (such as T790M and C797S mutations) and ‑independent (bypassing signaling activation, epithelial‑mesenchymal transition, tumor microenvironment remodeling and epigenetic regulation). Third‑generation EGFR‑TKIs (such as osimertinib) markedly improve patient survival by selectively targeting the T790M mutation, but novel resistance mutations, such as C797S, limit their long‑term efficacy. Combination therapies (such as MET proto‑oncogene, receptor tyrosine kinase/EGFR dual‑target inhibitors) and fourth‑generation TKIs (such as BLU‑945) offer novel directions to overcome resistance. Future research should focus on precise subtyping, dynamic monitoring of resistance mechanisms and regulation of the immune microenvironment to advance personalized treatment for NSCLC.

Introduction

Non-small cell lung cancer (NSCLC) represents 80–85% of all lung cancer cases, making it a notable contributor to cancer morbidity and mortality worldwide (1). The incidence of NSCLC is increasing, particularly in countries such as China, where it is the leading cause of cancer-associated mortality (2). Risk factors for NSCLC include tobacco use, family history and environmental exposure, alongside chronic lung diseases such as chronic obstructive pulmonary disease, which further exacerbate the risk (3). The clinical importance of NSCLC lies not only in its high prevalence but also in its poor prognosis, with numerous patients diagnosed at advanced stages when treatment options are limited, and survival rates are low (4).

Mutations in the EGFR gene (mEGFRs) are prevalent in NSCLC, occurring in ~40% of patients in Asian populations and ~20% in non-Asian populations (5). These mutations serve a key role in the pathogenesis of NSCLC, leading to uncontrolled cell proliferation and survival (4). Among mutations, the EGFR T790M mutation is a key driver of resistance to first- and second-generation EGFR-tyrosine kinase inhibitors (TKIs) (6). Understanding the biological role of mEGFRs is crucial, as they not only dictate the therapeutic approach but also influence patient outcomes and survival rates (7).

Third-generation EGFR-TKIs (such as osimertinib) are designed to selectively target both activating mutations [such as L858R and exon 19 deletions (19del)] and the T790M mutation via irreversible binding to the EGFR kinase domain. Their enhanced selectivity for mutant over wild-type EGFR decreases off-target toxicity, making them preferable first-line options for advanced NSCLC. However, the emergence of resistance mutations (such as C797S) is a challenge.

The present review aimed to summarize mEGFRs in NSCLC, the role of third-generation EGFR-TKIs and the challenges associated with resistance mechanisms to highlight the importance of targeted therapies in improving patient outcomes and elucidate potential pathways for overcoming resistance to EGFR-TKIs. Understanding these dynamics is key for optimizing treatment strategies and enhancing the clinical management of patients with NSCLC.

Classification of mEGFRs

Activating mutations (such as L858R and 19del)

Activating mutations in the EGFR gene are key in the pathogenesis of NSCLC (8). The most common of these mutations include the L858R point mutation and 19del (Fig. 1). These mutations lead to constitutive activation of the EGFR signaling pathway, promoting tumor cell proliferation and survival. Studies have demonstrated that patients harboring these mutations respond favorably to first-line EGFR-TKIs, such as gefitinib and erlotinib (9,10). For example, in a retrospective study involving 532 patients, the median PFS for patients with the L858R mutation receiving first-line TKI treatment was 11.4 months, while wild-type patients had a poorer response to TKI (median PFS typically below 3 months) (11).

The presence of 19del is associated with a higher sensitivity to EGFR-TKIs. This association has been reported in several previous studies (12,13), highlighting that patients with NSCLC harboring these specific mutations exhibit improved clinical outcomes when treated with EGFR-TKIs compared with those with other mutations, such as the L858R mutation (14). For example, patients with 19del had markedly longer PFS compared with patients with the L858R mutation, indicating a more favorable response to treatment (14).

Moreover, the coexistence of the T790M mutation, which is known to confer resistance to first-generation TKIs, has been reported to be more prevalent in patients with the L858R mutation compared with patients with 19del (15). This suggests that 19del not only predicts an improved response to initial treatment but may also be less likely to be associated with resistance mutations that complicate therapy (16). In addition, previous research indicates that certain exon 19 variants exhibit decreased ATP-binding affinity, which is associated with increased sensitivity to specific TKIs, such as afatinib (17). Thus, understanding the specific types of activating mutation is essential for tailoring personalized therapeutic strategies in NSCLC.

Rare mutations (such as G719X and S768I)

Rare mEGFRs, such as G719X and S768I, represent a unique subset of mEGFR alterations that influence treatment outcomes. Although less frequent than L858R and 19del, these mutations have been shown to confer sensitivity to EGFR-TKIs, albeit with variable response rates (1820).

Previous studies have highlighted that patients with NSCLC exhibiting G719X and S768I mutations may experience favorable outcomes when treated with second-generation TKIs, suggesting these agents could be optimal choices for this patient population (21,22). For example, the objective response rate (ORR) for patients with G719X mutations treated with afatinib reached 77.8%, with a median progression-free survival (PFS) of 13.8 months, and similar efficacy was observed for the S768I mutation (11). By contrast, the ORR for G719X patients using first-generation TKIs (such as gefitinib or erlotinib) was only 42.9%, with a median PFS of 5.98 months (18). A retrospective study further confirmed that the median PFS for patients treated with afatinib who carry the S768I compound mutation (such as S768I+L858R) reached 9.79 months, significantly better than gefitinib's 4.6 months (p=0.049) (19). Furthermore, the presence of these mutations complicates treatment decisions, as they may coexist with other mutations, leading to variability in response to therapy (23). For example, a previous study demonstrated that patients with lung adenocarcinoma harboring the G719X mutation often present compound mutations, such as G719X combined with S768I or L861Q, which can alter the efficacy of TKIs such as gefitinib and erlotinib (23). A retrospective analysis reported that patients with the G719X mutation alone have a different response rate compared with those with compound mutations involving S768I: The objective response rate for patients with G719X-only mutations is reported to be higher compared with patients with G719X combined with other mutations (24).

A previous study reported a patient with both G719X and S768I mutations who responded well to afatinib, maintaining disease control for an extended period. For example, a case study reported a 67-year-old male patient with G719X and S768I compound mutations. After failing initial chemoradiotherapy, he received afatinib treatment (40 mg/day), achieving partial remission and maintaining disease control for up to 17 months (25). This result is consistent with the conclusions of several multicenter studies: the G719X mutation (especially when present in combination with S768I) is associated with a longer PFS (2628). Specifically, a real-world study in Vietnam showed that patients with the G719X-S768I compound mutation had a median time to treatment failure (TTF) of 23.2 months, significantly better than other mutation types (12.3 months) (26). A multicenter study in Taiwan also confirmed that the G719X mutation (whether as a single mutation or in combination) is an independent favorable predictor of PFS (HR=0.578), with an overall median PFS of 17.3 months (27). Furthermore, a retrospective analysis in Spain further indicated that the objective response rate (ORR) in the compound mutation group (including G719X+S768I) reached 70%, significantly higher than in other mutation subgroups (28). The clinical importance of these rare mutations underscores the necessity for comprehensive genetic testing in patients with lung cancer, as they may benefit from targeted therapies that are typically reserved for more common mutations. Further research is needed to elucidate the mechanisms underlying the differential responses observed in patients with these rare mutations, which may inform future therapeutic development and clinical guidelines (8).

Other mutations (such as T790M and C797S)

The T790M mutation is an acquired alteration in EGFR and occurs in 50–60% of NSCLC cases (29). By contrast, the C797S mutation represents a tertiary resistance mechanism specific to third-generation EGFR-TKIs (such as osimertinib). Both mutations are clinically notable markers of therapeutic resistance, and their detection necessitates tailored treatment strategies (30).

Characteristics of mEGFRs

Association between mutation frequency and clinical features

The association between mEGFR frequency and clinical features in NSCLC has been a subject of extensive research. Several studies have demonstrated that the prevalence of mEGFRs is notably influenced by demographic and clinical characteristics such as age, sex, smoking status and histological subtype (11,15,18). For example, a study involving 388 patients with NSCLC demonstrated that mEGFRs are more prevalent in younger patients, particularly those aged <80 years, with a mutation rate of 49.6% compared with 24.1% in older patients (31). This suggests that age may serve a key role in the mutation landscape of NSCLC. Additionally, female patients had a higher incidence of mEGFRs, aligning with findings from other cohorts that report a strong association between female sex and mEGFR status (32).

Smoking status is an important factor influencing mEGFR frequency. Research has consistently reported that mEGFRs are more common in never-smokers compared with smokers (26,33). For example, a cohort study from India reported a notable association between mEGFRs and non-smoking status, with EGFR mutations being statistically significant among non-smokers compared to smokers (58.7% vs. 18.4%) (32). This trend is echoed in other studies, reinforcing the hypothesis that smoking may be negatively associated with the likelihood of mEGFRs in patients with NSCLC (34,35).

Histological subtype also affects mEGFR frequency. Adenocarcinoma, the most common subtype of NSCLC, is associated with a higher rate of EGFR mutations compared with squamous cell carcinoma. In 697 Chinese patients, mEGFRs were detected in 52.9% of adenocarcinomas, while 14.5% of squamous cell carcinomas exhibited similar mutations (36). This highlights the value of histological classification in understanding the mutation profile of NSCLC.

Association between mutation type and prognosis

The types of mEGFR impact the prognosis of patients with lung adenocarcinoma. For example, a meta-analysis indicated that patients with the 19del mutation exhibit a higher objective response rate and longer PFS compared with those with the L858R mutation in exon 21 (37). This suggests the 19del mutation may serve as a more effective clinical marker for predicting responses to EGFR-TKIs in patients with NSCLC (37). Furthermore, the presence of concomitant mutations alongside the primary mEGFRs also impacts prognosis. Patients with both 19del and L858R mutations had different sensitivity to EGFR-TKIs, with 19del showing a more favorable response (38).

Moreover, the prognostic value of TP53 mutations in conjunction with mEGFRs has been explored: TP53 mutations are associated with worse overall survival in advanced NSCLC, particularly when coupled with specific mEGFR types (39). This highlights the complexity of the molecular landscape in NSCLC and the need for comprehensive genetic profiling to improve prediction of patient outcomes.

Mechanisms influencing the tumor microenvironment (TME)

The role of mEGFR in NSCLC extends beyond its function as a driver of tumor growth; it notably influences the TME. The TME is a complex ecosystem composed of several cell types, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs) and immune cells, which interact dynamically with cancer cells (40,41). A key mechanism by which mEGFR affects the TME is the modulation of CAFs. CAFs promote tumor progression and drug resistance by creating an immunosuppressive environment. Studies have demonstrated that mEGFR signaling enhances the recruitment and activation of CAFs, leading to increased secretion of pro-tumorigenic factors that further support cancer cell survival and proliferation (42,43). This not only facilitates tumor growth but also contributes to the remodeling of the extracellular matrix (ECM), which can hinder the efficacy of therapeutic agents. The aberrant deposition of ECM components (such as collagen and laminin) establishes a physical barrier that impedes drug penetration, limiting the efficacy of EGFR-TKIs such as osimertinib in tumor cores (44). Concurrently, EGFR activation upregulates MMP2/MMP9, which not only degrade the basement membrane to facilitate metastasis but also liberate ECM-sequestered growth factors (e.g., TGF-β and VEGF) to reactivate PI3K/AKT and MAPK signaling pathways, thereby bypassing targeted therapy (45,46). Integrin-mediated interactions with remodeled ECM components (e.g., fibronectin) further activate FAK/SRC/STAT3 survival cascades, fostering cell adhesion-mediated drug resistance (44).

Moreover, mEGFR influences the polarization of TAMs within the TME. Elevated levels of mEGFRs are associated with an increase in M2-polarized macrophages, which are known to support tumor growth and metastasis (47). This shift in macrophage polarization is mediated by several signaling pathways, including the STAT5A-indoleamine 2,3-dioxygenase 1 axis, which promotes an immunosuppressive environment in NSCLC (48). By enhancing the presence of M2 macrophages, mEGFR contributes to immune evasion, allowing tumor cells to escape detection and destruction by the immune system.

Additionally, the interaction between mEGFR and immune checkpoint molecules, such as programmed death-ligand 1 (PD-L1), is key in shaping the TME. mEGFR signaling can upregulate PD-L1 expression on tumor cells, which inhibits T cell activation and proliferation, further exacerbating the immunosuppressive landscape of the TME (49). This dynamic interaction highlights the potential for combining mEGFR inhibitors with immune checkpoint inhibitors to enhance antitumor immunity and improve patient outcomes.

Mechanism of action of third-generation EGFR-TKIs

Characteristics of drugs targeting mEGFRs

Drugs targeting mEGFRs are a cornerstone in the treatment of NSCLC (Fig. 2). These targeted therapies, primarily TKIs, have shown notable efficacy in patients harboring specific mEGFRs, such as 19del and the L858R point mutation. A notable TKI is gefitinib, which was one of the first agents approved for mEGFR-positive NSCLC by the US FDA. It works by competitively inhibiting the ATP-binding site of the EGFR TK domain, thereby blocking downstream signaling pathways that promote tumor growth and survival (50). Gefitinib can significantly inhibit the phosphorylation of ERK1/2 in the MAPK/ERK pathway, leading to downregulation of Cyclin D1 expression and cell cycle arrest (51). At the same time, gefitinib promotes the upregulation of the pro-apoptotic protein Bim and activates Caspase-3/7 by inhibiting the phosphorylation of AKT at the Ser473 site in the PI3K/AKT/mTOR pathway, inducing apoptosis in tumor cells (52). In addition, gefitinib can block the abnormal activation of the JAK2/STAT3 pathway, reducing STAT3 nuclear translocation, thereby inhibiting the secretion of the angiogenic factor VEGF and the expression of the tumor stem cell marker CD133 (53). Erlotinib is another TKI used in combination with chemotherapy to enhance treatment efficacy (54). Afatinib, a second-generation TKI, has been developed to irreversibly bind to EGFR, providing a broader spectrum of activity against mEGFRs (50).

Despite the initial success of these therapies, resistance to mEGFR-targeted treatments remains a challenge. Mechanisms of resistance can include secondary mEGFRs, such as T790M, which can diminish the effectiveness of first-generation TKIs (55). This has led to the development of third-generation inhibitors, such as osimertinib, which are designed to target both the primary mEGFRs and the T790M mutation (55). In addition to TKIs, combination therapies that may enhance the efficacy of mEGFR-targeted treatments. For example, combining TKIs with immune checkpoint inhibitors or other targeted agents may provide synergistic effects and overcome resistance mechanisms (56). Furthermore, the identification of predictive biomarkers, such as specific serum metabolites, may aid in stratifying patients who are most likely to benefit from mEGFR-targeted therapy (54).

Comparison of third- and second-generation EGFR-TKIs

Second-generation EGFR-TKIs, such as afatinib and dacomitinib, have demonstrated improved efficacy compared with first-generation agents such as gefitinib and erlotinib (57). However, the introduction of third-generation EGFR-TKIs, particularly osimertinib, has advanced treatment options, especially for patients who develop resistance due to the T790M mutation (58,59). Third-generation EGFR-TKIs are designed to selectively target both the activating mutations and the T790M mutation, providing a clinical advantage over second-generation agents (60). For example, osimertinib has shown a median overall survival of 38.6 months in patients with NSCLC, which was significantly better than the control group (31.8 months) (58). Furthermore, third-generation inhibitors have a more favorable safety profile, with fewer adverse effects compared with their predecessors (61).

Despite these advancements, resistance to third-generation TKIs is a challenge, with the emergence of tertiary mutations such as C797S (62). This has prompted research into fourth-generation EGFR-TKIs aimed at overcoming these novel resistance mechanisms (63). By contrast, second-generation TKIs, while effective, often lead to resistance through several pathways, including the activation of alternative signaling routes (64).

Association between drug selectivity and resistance

The relationship between drug selectivity and resistance in the context of EGFR-TKIs is complex and multifaceted. Third-generation TKIs, such as osimertinib, are designed with enhanced selectivity for mutant over wild-type EGFR, which decreases the likelihood of adverse effects associated with off-target inhibition (65). However, the emergence of resistance is a notable clinical challenge. Studies indicate that while the selective pressure exerted by these drugs can initially lead to substantial tumor regression, it can also promote the selection of resistant clones that harbor secondary mutations, such as C797S or alterations in the MET proto-oncogene, receptor tyrosine kinase (MET) pathway (66,67). This underscores the importance of understanding the genetic landscape of tumors when administering third-generation TKIs. Additionally, combination therapies that target multiple pathways may be necessary to mitigate resistance and prolong the effectiveness of treatment. Ongoing research aims to identify biomarkers that can predict resistance and tailor treatment strategies, thereby improving outcomes for patients with mEGFR-NSCLC (68,69).

Mechanisms of resistance

EGFR-dependent resistance

The secondary mEGFR T790M is caused by the substitution of threonine with methionine in the tyrosine kinase domain of EGFR. This mutation increases the affinity of the ATP binding site, leading to spatial hindrance that interferes with the binding of TKIs, resulting in resistance (70). The C797S mutation is caused by the substitution of cysteine with serine at the C797 site of EGFR. This mutation eliminates the covalent binding site between TKIs and EGFR, rendering third-generation TKIs such as osimertinib ineffective (71). In addition to the T790M and C797S mutations, the L718Q mutation is also noteworthy. Studies have demonstrated that the L718Q mutation can coexist with other mutations such as C797S, further enhancing resistance to osimertinib (72,73). To address these challenges, researchers are developing fourth-generation EGFR-TKIs, such as BLU-945, specifically targeting T790M/C797S mutations. These novel inhibitors overcome resistance issues through higher selectivity and specificity (74). Additionally, dual-target inhibitors such as aurora kinase B (AURKB)/EGFR inhibitors enhance therapeutic effects and delay the onset of resistance by simultaneously targeting multiple signaling pathways (MAPK and PI3K pathways) (8). Highly selective allosteric inhibitors represent another promising strategy. These inhibitors suppress EGFR activity by altering its conformation without directly competing with the ATP binding site, potentially decreasing the impact on wild-type EGFR and lowering toxicity. This strategy shows promise in overcoming resistance caused by EGFR T790M and C797S mutations (75).

EGFR amplification compensates for the inhibitory effects of EGFR-TKIs by upregulating EGFR levels, leading to resistance to EGFR-TKIs. This resistance mechanism has been reported in multiple studies (76,77). EGFR amplification can counteract the inhibitory effects of TKIs by enhancing the activity of the EGFR signaling pathway. For example, amplification of the wild-type EGFR allele is sufficient to confer acquired resistance to selective mutant EGFR-TKIs (78). This indicates that even in the context of mEGFRs, signaling from wild-type EGFR serves a notable role in resistance. Secondly, EGFR amplification is not limited to the EGFR signaling pathway but may also involve the activation of other bypass signaling pathways. For example, MET amplification and MET receptor activation are also mechanisms of EGFR-TKI resistance (79). The activation of these bypass signaling pathways maintains cell proliferation and survival through different mechanisms, such as cross-activation of receptor TKs. Additionally, EGFR amplification may interact with other genetic alterations to enhance resistance. For example, in patients with advanced NSCLC, EGFR amplification coexists with the T790M mutation, and when treated with third-generation EGFR-TKIs (such as Osimertinib), patients experience improved PFS compared to those without acquired EGFR amplification (80). This suggests that EGFR amplification may synergize with other resistance mechanisms, affecting treatment outcomes.

Non-EGFR-dependent resistance

A mechanism involved in non-EGFR-dependent resistance is bypass signal activation. In addition to the T790M mEGFR, MET gene amplification is an important mechanism of resistance to first- or second-generation EGFR-TKIs (81). In certain studies (81,82), MET amplification has been identified as a key mechanism of acquired resistance to third-generation EGFR-TKIs (such as osimertinib), particularly when used as first-line treatment. In NSCLC with mEGFRs, MET amplification or protein overactivation may lead to escape from EGFR inhibition via the PI3K/AKT/mTOR pathway (83). To overcome this resistance, previous research suggests combining osimertinib with MET or MEK inhibitors (81). Furthermore, HER2 amplification is also regarded as an acquired resistance mechanism to EGFR inhibition (84). First, HER2 amplification can mediate resistance by enhancing the activity of the EGFR signaling pathway. Both EGFR and HER2 belong to the ErbB receptor family, and the overexpression or amplification of HER2 can activate downstream signaling pathways by forming heterodimers, thereby counteracting the inhibitory effects of EGFR-TKIs (84). Secondly, HER2 amplification may also promote resistance by affecting other signaling pathways. For example, studies have found that HER2 amplification can activate the PI3K/Akt signaling pathway, which plays a key role in cell proliferation and survival (85). Previous studies have reported that HER2 amplification and mEGFR (T790M) are mutually exclusive, revealing a mechanism of resistance to EGFR-TKIs and providing a rationale for assessing HER2 status and potentially targeting HER2 (76,84). Although there are currently limited standard treatment options for HER2 abnormalities, previous studies have demonstrated that the third-generation TKI osimertinib exhibits strong antitumor efficacy in lung cancer with HER2 abnormalities, providing a strong basis for future clinical trials (86,87).

Another mechanism involved in non-EGFR-dependent resistance is epithelial-mesenchymal transition (EMT). In NSCLC, zinc finger E-box binding homeobox 1 (ZEB1) and Twist family BHLH transcription factor 1 (TWIST1) are key transcription factors that serve important roles in the EMT process. ZEB1 is a notable regulator of EMT, promoting cell mesenchymalization by inhibiting the expression of E-cadherin, thereby enhancing the invasive and migratory ability of cells (88). Studies have demonstrated that the upregulation of ZEB1 is associated with the development of TKI resistance, especially in mEGFR NSCLC cells (89,90). Additionally, ZEB1 promotes the occurrence of EMT through interactions with other signaling pathways, such as the TGF-β signaling pathway (91). TWIST1 is also an important regulator of EMT, facilitating EMT by regulating genes (such as E-cadherin) associated with cytoskeletal reorganization and cell adhesion (92). Furthermore, TWIST1 enhances the effects of EMT through interactions with other signaling molecules, such as STAT3 (93).

In response to EMT-mediated resistance, researchers have proposed potential therapeutic strategies. For example, inhibiting the EMT-associated transcription factor TWIST1 enhances the efficacy of EGFR-TKIs (92). Studies have demonstrated that genetic silencing of TWIST1 or the use of TWIST1 inhibitors can suppress the growth of mEGFR NSCLC and induce apoptosis, thereby overcoming resistance (92,94). Additionally, glycogen synthase kinase-3 (GSK-3) inhibitors reverse EMT-associated resistance, particularly in resistant cells with a mesenchymal phenotype, where GSK-3 inhibitors markedly inhibit cell proliferation and induce apoptosis (95). Another strategy is the combined use of EGFR and fibroblast growth factor receptor (FGFR) inhibitors to suppress the survival and proliferation of resistant cells in mEGFR NSCLC. Dual inhibition of EGFR and FGFR can suppress proliferation of resistant cells over an extended period, preventing the development of fully resistant cancer (96). Dual inhibition of EGFR and FGFR can suppress cancer cell proliferation by blocking the cross-activation of these two signaling pathways. For example, in NSCLC, resistance to EGFR inhibitors is often associated with the activation of the FGFR signaling pathway. By simultaneously inhibiting EGFR and FGFR, cancer cell proliferation and survival is reduced (97). Secondly, dual inhibition of EGFR and FGFR can enhance apoptosis by inhibiting downstream signaling pathways such as the MEK/ERK and mTOR pathways. Studies have shown that feedback activation of EGFR and FGFR limits the efficacy of single FGFR inhibitors, while inhibition of EGFR can enhance the effects of FGFR inhibitors, leading to more significant apoptosis and tumor shrinkage (97). Additionally, dual inhibition of EGFR and FGFR can limit the development of resistance by reducing the frequency of tumor stem cells. Dual inhibition of EGFR and FGFR can decrease the frequency of tumor-initiating cells, thereby enhancing the response to radiotherapy and other treatments (98). These findings provide a theoretical basis for developing novel treatment regimens and offer new insight for overcoming EMT-mediated resistance.

TME remodeling is also involved in non-EGFR-dependent resistance. TAMs serve an important role in the TME, as they weaken the therapeutic effect of EGFR-TKIs by inhibiting the activation of CD8+ T cells (99). In addition, the ECM in the TME is an important factor in EGFR-TKI resistance. Even in the absence of genetic changes, ECM can immediately confer resistance to EGFR-TKIs in tumor cells through its interaction with integrin β1. This effect is dose-dependent and reversible, suggesting that targeting the ECM and integrin β1 may be a potential strategy for treating resistance (100). Moreover, immune evasion mechanisms in the TME may lead to EGFR-TKI resistance. EGFR-TKI resistance may promote immune evasion by upregulating the expression of PD-L1. Studies have found that the expression of PD-L1 is significantly increased in NSCLC cells resistant to EGFR-TKIs, which may facilitate tumor immune evasion by inhibiting lymphocyte activation and cytotoxicity (101,102). EGFR-TKI resistance may enhance the immunosuppressive microenvironment by inhibiting the anti-tumor function of CD8+ T cells. Furthermore, RNA methylation modifications may also play a role in the immune evasion associated with EGFR-TKI resistance. Research indicates that in EGFR-TKI resistant NSCLC, the inhibition of the m6A RNA demethylase ALKBH5 leads to the upregulation of CD47, thereby triggering immunosuppression (103). This mechanism may promote tumor immune evasion by inhibiting the phagocytic activity of dendritic cells and the cytotoxicity of CD8+ T cells. Finally, EGFR-TKI resistance may also reshape the tumor microenvironment from a non-inflammatory state to an inflammatory state, thereby affecting the infiltration and function of immune cells. Studies have found that after EGFR-TKI treatment, pro-inflammatory signals such as interferon-γ and inflammatory responses are significantly enriched in tumor samples, which may be associated with upregulation of effector cells after resistance (104).

Epigenetic regulation also serves a key role in EGFR-TKI resistance, including changes in histone modification, DNA methylation and non-coding RNA (ncRNA), which provide potential therapeutic targets and biomarkers for overcoming drug resistance (105). In the mechanisms of EGFR-independent resistance, epigenetic regulation affects the survival and proliferation of cancer cells through several pathways. For example, the loss of the histone methyltransferase enhancer of zeste homolog 2 (EZH2) is associated with EGFR-TKI resistance, as it influences cell proliferation and resistance by regulating the expression and phosphorylation of MET (106). Additionally, changes in the expression of ncRNAs, such as nc886. In the study of the HCC827 NSCLC cell line, it was found that the expression of nc886 is not related to EMT (epithelial-mesenchymal transition), but the CRISPR/Cas9-mediated disruption of its sequence can increase the sensitivity of cells to TKIs. Furthermore, the disruption of the nc886 sequence hinders the activation of MET RTK, which is a mechanism of TKI resistance with EMT as the endpoint (107). To address the resistance induced by these epigenetic regulations, researchers have proposed various therapeutic strategies: Combination therapy with PI3K/AKT inhibitors and EGFR-TKIs breaks the MET-AKT-EZH2 feedback loop, thereby enhancing tumor suppression effects (106). Furthermore, targeted therapies against ncRNAs are potential strategies to overcome resistance by modulating their expression or function to restore drug sensitivity (107).

Clinical applications and future research directions

Personalized treatment strategies for mEGFRs

The advent of targeted therapies has revolutionized the management of NSCLC, particularly in patients harboring mEGFRs. These mutations are key in dictating the responsiveness to TKIs, such as gefitinib and erlotinib. Personalized treatment strategies that consider the specific type of mEGFR are key for optimizing therapeutic outcomes. For example, 19del and the L858R point mutation exhibit distinct responses to TKIs, necessitating tailored approaches (108). Recent studies have underscored the importance of comprehensive genomic profiling prior to treatment initiation, which can guide clinicians in selecting the most effective agent and dosage for individual patients, thereby enhancing efficacy and minimizing adverse effects (109,110). Furthermore, Combination therapy, where TKIs are paired with immunotherapies or chemotherapy, aims to overcome resistance mechanisms that often develop during treatment (111). The integration of artificial intelligence in analyzing patient data and predicting treatment responses presents a promising avenue for future research, potentially leading to more refined and effective personalized treatment protocols (112).

Advances in the development of novel targeted drugs

Previous studies have highlighted the importance of understanding the molecular mechanisms of NSCLC to identify potential therapeutic targets (113,114). For example, the discovery of mEGFRs and mutations in genes such as anaplastic lymphoma kinase (115) and KRAS (116) has led to the development of targeted agents that specifically inhibit these oncogenic drivers. The implementation of these therapies has shown promising results in clinical trials, demonstrating improved response rate and survival outcomes compared with conventional treatment (117,118). Furthermore, the integration of immunotherapy into the treatment paradigm harnesses the immune system to combat cancer cells, offering hope for patients with advanced disease (119).

Despite these advancements, challenges remain in the clinical application of targeted therapies for NSCLC. A notable hurdle is the development of resistance to these agents, which can occur through various mechanisms, including secondary mutations and activation of alternative signaling pathways. Understanding these resistance mechanisms is key for developing strategies to overcome them, such as combination therapies that target multiple pathways simultaneously (120,121). Additionally, the identification of robust predictive biomarkers is key for patient stratification, ensuring that individuals most likely to benefit from specific therapies are selected for treatment (121,122).

There is a need for clinical trials to evaluate the efficacy of new targeted agents and combination therapy, particularly in biomarker-selected populations. Secondly, the exploration of novel molecular targets and therapeutic strategies, including the potential repurposing of existing drugs, may provide additional avenues for treatment (123,124). Lastly, enhancing the understanding of the TME and its role in cancer progression is key for developing innovative therapies that effectively combat NSCLC (125).

Conclusion

mEGFRs serve a key role in the molecular classification and treatment of NSCLC. Activating mutations (L858R/19del) are sensitive to EGFR-TKIs but prone to secondary T790M/C797S resistance; rare mutations (G719X/S768I) require second-generation TKIs or combination therapy. mEGFRs are also involved in complex resistance mechanisms. EGFR-dependent resistance (such as T790M/C797S) diminishes TKI efficacy through steric hindrance or covalent binding site disruption, whereas EGFR-independent resistance (MET/HER2 amplification, EMT, TME remodeling) relies on bypass activation or immune evasion, necessitating multi-target intervention (104). The present review highlighted the innovative treatment strategies for NSCLC. Fourth-generation TKIs (BLU-945) and dual-target inhibitors (AURKB/EGFR) target resistant mutations and combining immune checkpoint inhibitors or epigenetic modulators (such as histone deacetylase inhibitors) can reverse resistance (126). Future studies should integrate liquid biopsy, artificial intelligence classification and multi-omics data to achieve dynamic monitoring of resistance mechanisms. In addition, evaluating the immune interactions of TME and developing new targeted drugs (such as CD47 inhibitors) may be key to precision treatment for NSCLC.

Acknowledgements

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 81860021), Guangxi Natural Science Foundation (grant no. 2021GXNSFAA325003), Baise Regional Multimorbidity Joint Special Plan [grant no. BaiKe Zi (2022) no. 41], Guangxi University Young and Middle-aged Teachers Research Basic Ability Promotion Project (grant no. 2023KY0573), Guangxi Medical and Health Key Discipline Construction Project and Baise Scientific Research and Technology Development Program Project (grant no. Baike no. 20232086).

Availability of data and materials

Not applicable.

Authors' contributions

ZT, LC and FW conceived the study, performed the literature review and wrote the manuscript. JueD, YuH and YiH performed the literature review and revised the manuscript. ZW and JunD wrote the manuscript. YJ reviewed the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Suster DI and Mino-Kenudson M: Molecular pathology of primary non-small cell lung cancer. Arch Med Res. 51:784–798. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Chen P, Liu Y, Wen Y and Zhou C: Non-small cell lung cancer in China. Cancer Commun (Lond). 42:937–970. 2022. View Article : Google Scholar : PubMed/NCBI

3 

Houghton AM: Common mechanisms linking chronic obstructive pulmonary disease and lung cancer. Ann Am Thorac Soc. 15 (Suppl 4):S273–S277. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Duréndez-Sáez E, Torres-Martinez S, Calabuig-Fariñas S, Meri-Abad M, Ferrero-Gimeno M and Camps C: Exosomal microRNAs in non-small cell lung cancer. Transl Cancer Res. 10:3128–3139. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Gelatti ACZ, Drilon A and Santini FC: Optimizing the sequencing of tyrosine kinase inhibitors (TKIs) in epidermal growth factor receptor (EGFR) mutation-positive non-small cell lung cancer (NSCLC). Lung Cancer. 137:113–122. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Mosca M, Conci N, Di Federico A, Tateo V, Favorito V, Zappi A, Gelsomino F and De Giglio A: First-generation epidermal growth factor receptor inhibitors plus antiangiogenic drugs versus third-generation epidermal growth factor receptor inhibitors in advanced non-small-cell lung cancer: A meta-analysis. JCO Precis Oncol. 7:e23000732023. View Article : Google Scholar : PubMed/NCBI

7 

Qu F, Zhou Y and Yu W: A review of research progress on mechanisms and overcoming strategies of acquired osimertinib resistance. Anticancer Drugs. 33:e76–e83. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Kurup S, Gesinski D, Assaad K and Reynolds A: Design, synthesis, and evaluation of dual EGFR/AURKB inhibitors as anticancer agents for non-small cell lung cancer. Bioorg Med Chem Lett. 100:1296122024. View Article : Google Scholar : PubMed/NCBI

9 

Costa DB, Nguyen KSH, Cho BC, Sequist LV, Jackman DM, Riely GJ, Yeap BY, Halmos B, Kim JH, Jänne PA, et al: Effects of erlotinib in EGFR mutated non-small cell lung cancers with resistance to gefitinib. Clin Cancer Res. 14:7060–7067. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Maione P, Rossi A, Bareschino M, Sacco PC, Schettino C, Casaluce F, Sgambato A and Gridelli C: Irreversible EGFR inhibitors in the treatment of advanced NSCLC. Curr Pharm Des. 20:3894–3900. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Choi YW and Choi JH: Does the efficacy of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor differ according to the type of EGFR mutation in non-small cell lung cancer? Korean J Intern Med. 32:422–428. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Li J, Qu L, Wei X, Gao H, Wang W, Qin H, Tang C, Guo W, Wang H and Liu X: Clinical observation of EGFR-TKI as a first-line therapy on advanced non-small cell lung cancer. Zhongguo Fei Ai Za Zhi. 15:299–304. 2012.(In Chinese). PubMed/NCBI

13 

Vidal ÓJ: Afatinib as first-line therapy in mutation-positive EGFR. Results by type of mutation. Med Clin (Barc). 146 (Suppl 1):S12–S18. 2016.(In Spanish). View Article : Google Scholar

14 

Won YW, Han JY, Lee GK, Park SY, Lim KY, Yoon KA, Yun T, Kim HT and Lee JS: Comparison of clinical outcome of patients with non-small-cell lung cancer harbouring epidermal growth factor receptor exon 19 or exon 21 mutations. J Clin Pathol. 64:947–952. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Yu JY, Yu SF, Wang SH, Bai H, Zhao J, An TT, Duan JC and Wang J: Clinical outcomes of EGFR-TKI treatment and genetic heterogeneity in lung adenocarcinoma patients with EGFR mutations on exons 19 and 21. Chin J Cancer. 35:302016. View Article : Google Scholar : PubMed/NCBI

16 

Chen LY, Molina-Vila MA, Ruan SY, Su KY, Liao WY, Yu KL, Ho CC, Shih JY, Yu CJ, Yang JC, et al: Coexistence of EGFR T790M mutation and common activating mutations in pretreatment non-small cell lung cancer: A systematic review and meta-analysis. Lung Cancer. 94:46–53. 2016. View Article : Google Scholar : PubMed/NCBI

17 

van Alderwerelt van Rosenburgh IK, Lu DM, Grant MJ, Stayrook SE, Phadke M, Walther Z, Goldberg SB, Politi K, Lemmon MA, Ashtekar KD and Tsutsui Y: Biochemical and structural basis for differential inhibitor sensitivity of EGFR with distinct exon 19 mutations. Nat Commun. 13:67912022. View Article : Google Scholar : PubMed/NCBI

18 

Xu J, Jin B, Chu T, Dong X, Yang H, Zhang Y, Wu D, Lou Y, Zhang X, Wang H and Han B: EGFR tyrosine kinase inhibitor (TKI) in patients with advanced non-small cell lung cancer (NSCLC) harboring uncommon EGFR mutations: A real-world study in China. Lung Cancer. 96:87–92. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Park S, Lee SY, Kim D, Sim YS, Ryu JS, Choi J, Lee SH, Ryu YJ, Lee JH and Chang JH: Comparison of epidermal growth factor receptor tyrosine kinase inhibitors for patients with lung adenocarcinoma harboring different epidermal growth factor receptor mutation types. BMC Cancer. 21:522021. View Article : Google Scholar : PubMed/NCBI

20 

Singh V, Nambirajan A, Malik PS, Thulkar S, Pandey RM, Luthra K, Arava S, Ray R, Mohan A and Jain D: Spectrum of uncommon and compound epidermal growth factor receptor mutations in non-small-cell lung carcinomas with treatment response and outcome analysis: A study from India. Lung Cancer. 149:53–60. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Chiu CH, Yang CT, Shih JY, Huang MS, Su WC, Lai RS, Wang CC, Hsiao SH, Lin YC, Ho CL, et al: Epidermal growth factor receptor tyrosine Kinase inhibitor treatment response in advanced lung adenocarcinomas with G719X/L861Q/S768I mutations. J Thorac Oncol. 10:793–799. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Watanabe M, Oizumi S, Kiuchi S, Yamada N, Yokouchi H, Fukumoto S and Harada M: The effectiveness of afatinib in a patient with advanced lung adenocarcinoma harboring rare G719X and S768I mutations. Intern Med. 57:993–996. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Leventakos K, Kipp BR, Rumilla KM, Winters JL, Yi ES and Mansfield AS: S768I mutation in EGFR in patients with lung cancer. J Thorac Oncol. 11:1798–1801. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Zhu X, Bai Q, Lu Y, Qi P, Ding J, Wang J and Zhou X: Response to tyrosine kinase inhibitors in lung adenocarcinoma with the rare epidermal growth factor receptor mutation S768I: A retrospective analysis and literature review. Target Oncol. 12:81–88. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Kutsuzawa N, Takahashi F, Tomomatsu K, Obayashi S, Takeuchi T, Takihara T, Hayama N, Oguma T, Aoki T and Asano K: Successful treatment of a patient with lung adenocarcinoma harboring compound EGFR gene mutations, G719X and S768I, with afatinib. Tokai J Exp Clin Med. 45:113–116. 2020.PubMed/NCBI

26 

Pham VL, Le TA, Pham CP, Hoa Nguyen TT, Do AT, Nguyen TK, Nguyen MH, Thu Hoang TA, Hao Vuong DT, Tam Nguyen DN, et al: Real-world analysis of afatinib as a first-line treatment for patients with advanced stage non-small-cell lung cancer with uncommon EGFR mutations: A multicenter study in Vietnam. Ther Adv Med Oncol. 16:175883592412429722024. View Article : Google Scholar : PubMed/NCBI

27 

Hsu PC, Lee SH, Chiu LC, Lee CS, Wu CE, Kuo SC, Ju JS, Huang AC, Li SH, Ko HW, et al: Afatinib in untreated stage IIIB/IV lung adenocarcinoma with major uncommon epidermal growth factor receptor (EGFR) mutations (G719X/L861Q/S768I): A multicenter observational study in Taiwan. Target Oncol. 18:195–207. 2023. View Article : Google Scholar : PubMed/NCBI

28 

Moran T, Taus A, Arriola E, Aguado C, Dómine M, Rueda AG, Calles A, Cedrés S, Viñolas N, Isla D, et al: Clinical activity of afatinib in patients with non-small-cell lung cancer harboring uncommon EGFR mutations: A Spanish retrospective multicenter study. Clin Lung Cancer. 21:428–436.e2. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Lee SH, Kim EY, Kim A and Chang YS: Clinical implication and usefulness of de novo EGFR T790M mutation in lung adenocarcinoma with EGFR-tyrosine kinase inhibitor sensitizing mutation. Cancer Biol Ther. 21:741–748. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Barsouk A, Elghawy O, Heidlauf A, Yu C, Wang L, Yang D, Kurian M, Goel K, Rushkin L, Anran Huang A, et al: Real-world outcomes of atypical EGFR-mutated metastatic non-small cell lung cancer (mNSCLC) treated with osimertinib (osi) vs afatinib or erlotinib. Lung Cancer. 195:1079262024. View Article : Google Scholar : PubMed/NCBI

31 

Nishii T, Yokose T, Miyagi Y, Daigo Y, Ito H, Isaka T, Imai K, Murakami S, Kondo T, Saito H, et al: Clinicopathological features and EGFR gene mutation status in elderly patients with resected non-small-cell lung cancer. BMC Cancer. 14:6102014. View Article : Google Scholar : PubMed/NCBI

32 

Thomas R, Balaram G, Varayathu H, Ghorpade SN, Kowsik PV, Dharman B, Thomas BE, Ramaswamy V, Nanjaiah T, Patil S, et al: Molecular epidemiology and clinical characteristics of epidermal growth factor receptor mutations in NSCLC: A single-center experience from India. J Cancer Res Ther. 19:1398–1406. 2023. View Article : Google Scholar : PubMed/NCBI

33 

Zhang T, Wan B, Zhao Y, Li C, Liu H, Lv T, Zhan P and Song Y: Treatment of uncommon EGFR mutations in non-small cell lung cancer: New evidence and treatment. Transl Lung Cancer Res. 8:302–316. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Wen L, Wang S, Xu W, Xu X, Li M, Zhang Y, Du X and Liu S: Value of serum tumor markers for predicting EGFR mutations in non-small cell lung cancer patients. Ann Diagn Pathol. 49:1516332020. View Article : Google Scholar : PubMed/NCBI

35 

Kim EY, Kim A, Lee G, Lee H and Chang YS: Different mutational characteristics of the subsets of EGFR-tyrosine kinase inhibitor sensitizing mutation-positive lung adenocarcinoma. BMC Cancer. 18:12212018. View Article : Google Scholar : PubMed/NCBI

36 

Lai Y, Zhang Z, Li J, Sun D, Zhou Y, Jiang T, Han Y, Huang L, Zhu Y, Li X and Yan X: EGFR mutations in surgically resected fresh specimens from 697 consecutive Chinese patients with non-small cell lung cancer and their relationships with clinical features. Int J Mol Sci. 14:24549–24559. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Wang H, Huang J, Yu X, Han S, Yan X, Sun S and Zhu X: Different efficacy of EGFR tyrosine kinase inhibitors and prognosis in patients with subtypes of EGFR-mutated advanced non-small cell lung cancer: A meta-analysis. J Cancer Res Clin Oncol. 140:1901–1909. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Liang H, Li C, Zhao Y, Zhao S, Huang J, Cai X, Cheng B, Xiong S, Li J, Wang W, et al: Concomitant mutations in EGFR 19Del/L858R mutation and their association with response to EGFR-TKIs in NSCLC patients. Cancer Manag Res. 12:8653–8662. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Jiao XD, Qin BD, You P, Cai J and Zang YS: The prognostic value of TP53 and its correlation with EGFR mutation in advanced non-small cell lung cancer, an analysis based on cBioPortal data base. Lung Cancer. 123:70–75. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Timperi E, Croizer H, Khantakova D, Rana M, Molgora M, Guerriero JL, Mechta-Grigoriou F and Romano E: At the interface of tumor-associated macrophages and fibroblasts: Immune-suppressive networks and emerging exploitable targets. Clin Cancer Res. 30:5242–5251. 2024. View Article : Google Scholar : PubMed/NCBI

41 

Tajaldini M, Saeedi M, Amiriani T, Amiriani AH, Sedighi S, Mohammad Zadeh F, Dehghan M, Jahanshahi M, Zanjan Ghandian M, Khalili P, et al: Cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs); where do they stand in tumorigenesis and how they can change the face of cancer therapy? Eur J Pharmacol. 928:1750872022. View Article : Google Scholar : PubMed/NCBI

42 

Suzuki J, Tsuboi M and Ishii G: Cancer-associated fibroblasts and the tumor microenvironment in non-small cell lung cancer. Expert Rev Anticancer Ther. 22:169–182. 2022. View Article : Google Scholar : PubMed/NCBI

43 

Garvey CM, Lau R, Sanchez A, Sun RX, Fong EJ, Doche ME, Chen O, Jusuf A, Lenz HJ, Larson B and Mumenthaler SM: Anti-EGFR therapy induces EGF secretion by cancer-associated fibroblasts to confer colorectal cancer chemoresistance. Cancers (Basel). 12:13932020. View Article : Google Scholar : PubMed/NCBI

44 

Morgillo F, Woo JK, Kim ES, Hong WK and Lee HY: Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Res. 66:10100–10111. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Mitsudomi T and Yatabe Y: Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS J. 277:301–308. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Hirsch FR, Varella-Garcia M, Bunn PA Jr, Di Maria MV, Veve R, Bremmes RM, Barón AE, Zeng C and Franklin WA: Epidermal growth factor receptor in non-small-cell lung carcinomas: Correlation between gene copy number and protein expression and impact on prognosis. J Clin Oncol. 21:3798–3807. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Madeddu C, Donisi C, Liscia N, Lai E, Scartozzi M and Macciò A: EGFR-mutated non-small cell lung cancer and resistance to immunotherapy: Role of the tumor microenvironment. Int J Mol Sci. 23:64892022. View Article : Google Scholar : PubMed/NCBI

48 

Yang Y, Zheng X, Ni P, Li D, Dan Q, Wang X, Wang Y, Sun Y, Liu K, Dong Z and Ge H: Targeting the STAT5A/IDO1 axis overcomes radioresistance and reverses the immunosuppressive tumor microenvironment in NSCLC. Int J Oncol. 62:122023. View Article : Google Scholar : PubMed/NCBI

49 

Lin A, Wei T, Meng H, Luo P and Zhang J: Role of the dynamic tumor microenvironment in controversies regarding immune checkpoint inhibitors for the treatment of non-small cell lung cancer (NSCLC) with EGFR mutations. Mol Cancer. 18:1392019. View Article : Google Scholar : PubMed/NCBI

50 

Burotto M, Ali SA and O'sullivan Coyne G: Class act: Safety comparison of approved tyrosine kinase inhibitors for non-small-cell lung carcinoma. Expert Opin Drug Saf. 14:97–110. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Zhang Y, Liu H, Liu X and Lang L: Gefitinib induces apoptosis in NSCLC cells by promoting glutaminolysis and inhibiting the MEK/ERK signaling pathway. Discov Med. 36:836–841. 2024. View Article : Google Scholar : PubMed/NCBI

52 

Zhao ZQ, Yu ZY, Li J and Ouyang XN: Gefitinib induces lung cancer cell autophagy and apoptosis via blockade of the PI3K/AKT/mTOR pathway. Oncol Lett. 12:63–68. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Li Q, Zhang D, Chen X, He L, Li T, Xu X and Li M: Nuclear PKM2 contributes to gefitinib resistance via upregulation of STAT3 activation in colorectal cancer. Sci Rep. 5:160822015. View Article : Google Scholar : PubMed/NCBI

54 

Han X, Luo R, Wang L, Zhang L, Wang T, Zhao Y, Xiao S, Qiao N, Xu C, Ding L, et al: Potential predictive value of serum targeted metabolites and concurrently mutated genes for EGFR-TKI therapeutic efficacy in lung adenocarcinoma patients with EGFR sensitizing mutations. Am J Cancer Res. 10:4266–4286. 2020.PubMed/NCBI

55 

Lee SH, Kim EY, Kim T and Chang YS: Compared to plasma, bronchial washing fluid shows higher diagnostic yields for detecting EGFR-TKI sensitizing mutations by ddPCR in lung cancer. Respir Res. 21:1422020. View Article : Google Scholar : PubMed/NCBI

56 

Masui K, Gini B, Wykosky J, Zanca C, Mischel PS, Furnari FB and Cavenee WK: A tale of two approaches: Complementary mechanisms of cytotoxic and targeted therapy resistance may inform next-generation cancer treatments. Carcinogenesis. 34:725–738. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Hou B, Lu X, Gao DC, Liu QX, Zhou D, Zheng H and Dai JG: REPORT-clinical outcomes of using second-versus first-generation EGFR-tkis for the first-line treatment of advanced NSCLC patients with EGFR mutations: A meta-analysis. Pak J Pharm Sci. 34:1459–1468. 2021.PubMed/NCBI

58 

Du X, Yang B, An Q, Assaraf YG, Cao X and Xia J: Acquired resistance to third-generation EGFR-TKIs and emerging next-generation EGFR inhibitors. Innovation (Camb). 2:1001032021.PubMed/NCBI

59 

Tan CS, Kumarakulasinghe NB, Huang YQ, Ang YLE, Choo JRE, Goh BC and Soo RA: Third generation EGFR TKIs: Current data and future directions. Mol Cancer. 17:292018. View Article : Google Scholar : PubMed/NCBI

60 

Pirker R: Third-generation epidermal growth factor receptor tyrosine kinase inhibitors in advanced nonsmall cell lung cancer. Curr Opin Oncol. 28:115–121. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Wang S and Li J: Second-generation EGFR and ErbB tyrosine kinase inhibitors as first-line treatments for non-small cell lung cancer. Onco Targets Ther. 12:6535–6548. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Wang S, Tsui ST, Liu C, Song Y and Liu D: EGFR C797S mutation mediates resistance to third-generation inhibitors in T790M-positive non-small cell lung cancer. J Hematol Oncol. 9:592016. View Article : Google Scholar : PubMed/NCBI

63 

Scalvini L, Castelli R, La Monica S, Tiseo M and Alfieri R: Fighting tertiary mutations in EGFR-driven lung-cancers: Current advances and future perspectives in medicinal chemistry. Biochem Pharmacol. 190:1145432021. View Article : Google Scholar : PubMed/NCBI

64 

Nagano T, Tachihara M and Nishimura Y: Mechanism of resistance to epidermal growth factor receptor-tyrosine kinase inhibitors and a potential treatment strategy. Cells. 7:2122018. View Article : Google Scholar : PubMed/NCBI

65 

Shi P, Zhang S, Zhu L, Qian G, Ren H, Ramalingam SS, Chen M and Sun SY: The third-generation EGFR inhibitor, osimertinib, promotes c-FLIP degradation, enhancing apoptosis including TRAIL-induced apoptosis in NSCLC cells with activating EGFR mutations. Transl Oncol. 12:705–713. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Venkatesan S, Swanton C, Taylor BS and Costello JF: Treatment-induced mutagenesis and selective pressures sculpt cancer evolution. Cold Spring Harb Perspect Med. 7:a0266172017. View Article : Google Scholar : PubMed/NCBI

67 

Vander Velde R, Yoon N, Marusyk V, Durmaz A, Dhawan A, Miroshnychenko D, Lozano-Peral D, Desai B, Balynska O, Poleszhuk J, et al: Resistance to targeted therapies as a multifactorial, gradual adaptation to inhibitor specific selective pressures. Nat Commun. 11:23932020. View Article : Google Scholar : PubMed/NCBI

68 

Li K, Peng ZY, Wang R, Li X, Du N, Liu DP, Zhang J, Zhang YF, Ma L, Sun Y, et al: Enhancement of TKI sensitivity in lung adenocarcinoma through m6A-dependent translational repression of Wnt signaling by circ-FBXW7. Mol Cancer. 22:1032023. View Article : Google Scholar : PubMed/NCBI

69 

Pang LL, Zhuang WT, Huang YH, Liao J, Li MZ, Lv Y, Zhang L and Fang WF: Uncommon de novo EGFRT790M-Mutant NSCLC characterized with unique genetic features: Clinical response and acquired resistance to the third-generation EGFR-TKIs treatment. Lung Cancer. 190:1075282024. View Article : Google Scholar : PubMed/NCBI

70 

Denis MG, Vallée A and Théoleyre S: EGFR T790M resistance mutation in non-small-cell lung carcinoma. Clin Chim Acta. 444:81–85. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Shaikh M, Shinde Y, Pawara R, Noolvi M, Surana S, Ahmad I and Patel H: Emerging approaches to overcome acquired drug resistance obstacles to osimertinib in non-small-cell lung cancer. J Med Chem. 65:1008–1046. 2022. View Article : Google Scholar : PubMed/NCBI

72 

Nishino M, Suda K, Kobayashi Y, Ohara S, Fujino T, Koga T, Chiba M, Shimoji M, Tomizawa K, Takemoto T and Mitsudomi T: Effects of secondary EGFR mutations on resistance against upfront osimertinib in cells with EGFR-activating mutations in vitro. Lung Cancer. 126:149–155. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Ding K, Peng Z and Xu Y: Triplet therapy overcomes 3rd-EGFR TKI-resistant EGFR-L858R/T790M/C797S in trans and in cis/L718Q mutation. Genes Dis. 12:1014082024. View Article : Google Scholar : PubMed/NCBI

74 

Lim SM, Schalm SS, Lee EJ, Park S, Conti C, Millet YA, Woessner R, Zhang Z, Tavera-Mendoza LE, Stevison F, et al: BLU-945, a potent and selective next-generation EGFR TKI, has antitumor activity in models of osimertinib-resistant non-small-cell lung cancer. Ther Adv Med Oncol. 16:175883592412806892024. View Article : Google Scholar : PubMed/NCBI

75 

Ahmadi A, Mohammadnejadi E and Razzaghi-Asl N: Gefitinib derivatives and drug-resistance: A perspective from molecular dynamics simulations. Comput Biol Med. 163:1072042023. View Article : Google Scholar : PubMed/NCBI

76 

Ercan D, Zejnullahu K, Yonesaka K, Xiao Y, Capelletti M, Rogers A, Lifshits E, Brown A, Lee C, Christensen JG, et al: Amplification of EGFR T790M causes resistance to an irreversible EGFR inhibitor. Oncogene. 29:2346–2356. 2010. View Article : Google Scholar : PubMed/NCBI

77 

Kubo T, Yamamoto H, Lockwood WW, Valencia I, Soh J, Peyton M, Jida M, Otani H, Fujii T, Ouchida M, et al: MET gene amplification or EGFR mutation activate MET in lung cancers untreated with EGFR tyrosine kinase inhibitors. Int J Cancer. 124:1778–1784. 2009. View Article : Google Scholar : PubMed/NCBI

78 

Nukaga S, Yasuda H, Tsuchihara K, Hamamoto J, Masuzawa K, Kawada I, Naoki K, Matsumoto S, Mimaki S, Ikemura S, et al: Amplification of EGFR wild-type alleles in non-small cell lung cancer cells confers acquired resistance to mutation-selective EGFR tyrosine kinase inhibitors. Cancer Res. 77:2078–2089. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Presutti D, Santini S, Cardinali B, Papoff G, Lalli C, Samperna S, Fustaino V, Giannini G and Ruberti G: MET gene amplification and MET receptor activation are not sufficient to predict efficacy of combined MET and EGFR inhibitors in EGFR TKI-resistant NSCLC cells. PLoS One. 10:e01433332015. View Article : Google Scholar : PubMed/NCBI

80 

Zhang Y, Xu Y, Xu J, Zhong H, Xia J and Zhong R: Osimertinib for EGFR-mutant NSCLC patients with acquired T790M and EGFR amplification after first-generation EGFR-TKI resistance. Cancer Sci. 116:753–763. 2025. View Article : Google Scholar : PubMed/NCBI

81 

Zhang Z, Yang S and Wang Q: Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer. Biomark Res. 7:272019. View Article : Google Scholar : PubMed/NCBI

82 

Nie N, Li J, Zhang J, Dai J, Liu Z, Ding Z, Wang Y, Zhu M, Hu C, Han R, et al: First-line osimertinib in patients with EGFR-mutated non-small cell lung cancer: Effectiveness, resistance mechanisms, and prognosis of different subsequent treatments. Clin Med Insights Oncol. 16:117955492211347352022. View Article : Google Scholar : PubMed/NCBI

83 

Turke AB, Zejnullahu K, Wu YL, Song Y, Dias-Santagata D, Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al: Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell. 17:77–88. 2010. View Article : Google Scholar : PubMed/NCBI

84 

Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E, Ohashi K, Janjigian YY, Spitzler PJ, Melnick MA, et al: HER2 amplification: a potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFR T790M mutation. Cancer Discov. 2:922–933. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Ding D, Zhang J, Luo Z, Wu H, Lin Z, Liang W and Xue X: Analysis of the lncRNA-miRNA-mRNA network reveals a potential regulatory mechanism of EGFR-TKI resistance in NSCLC. Front Genet. 13:8513912022. View Article : Google Scholar : PubMed/NCBI

86 

Liu S, Li S, Hai J, Wang X, Chen T, Quinn MM, Gao P, Zhang Y, Ji H, Cross DAE and Wong KK: Targeting HER2 aberrations in non-small cell lung cancer with osimertinib. Clin Cancer Res. 24:2594–2604. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Jonsdottir G, Smith M, Wood S, Hejleh TA and Furqan M: Activity of osimertinib in a patient with stage IV non-small cell lung cancer harboring HER2 exon 19, p.L755P mutation: Case report. Transl Lung Cancer Res. 12:927–932. 2023. View Article : Google Scholar : PubMed/NCBI

88 

Rastogi I, Rajanna S, Webb A, Chhabra G, Foster B, Webb B and Puri N: Mechanism of c-Met and EGFR tyrosine kinase inhibitor resistance through epithelial mesenchymal transition in non-small cell lung cancer. Biochem Biophys Res Commun. 477:937–944. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Soucheray M, Capelletti M, Pulido I, Kuang Y, Paweletz CP, Becker JH, Kikuchi E, Xu C, Patel TB, Al-Shahrour F, et al: Intratumoral heterogeneity in EGFR-mutant NSCLC results in divergent resistance mechanisms in response to EGFR tyrosine kinase inhibition. Cancer Res. 75:4372–4383. 2015. View Article : Google Scholar : PubMed/NCBI

90 

Lu Y, Liu Y, Oeck S, Zhang GJ, Schramm A and Glazer PM: Hypoxia induces resistance to EGFR inhibitors in lung cancer cells via upregulation of FGFR1 and the MAPK pathway. Cancer Res. 80:4655–4667. 2020. View Article : Google Scholar : PubMed/NCBI

91 

Comaills V, Kabeche L, Morris R, Buisson R, Yu M, Madden MW, LiCausi JA, Boukhali M, Tajima K, Pan S, et al: Genomic instability is induced by persistent proliferation of cells undergoing epithelial-to-mesenchymal transition. Cell Rep. 17:2632–2647. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Yochum ZA, Cades J, Wang H, Chatterjee S, Simons BW, O'Brien JP, Khetarpal SK, Lemtiri-Chlieh G, Myers KV, Huang EH, et al: Targeting the EMT transcription factor TWIST1 overcomes resistance to EGFR inhibitors in EGFR-mutant non-small-cell lung cancer. Oncogene. 38:656–670. 2019. View Article : Google Scholar : PubMed/NCBI

93 

Chen K, Xu J, Tong YL, Yan JF, Pan Y, Wang WJ, Zheng L, Zheng XX, Hu C, Hu X, et al: Rab31 promotes metastasis and cisplatin resistance in stomach adenocarcinoma through Twist1-mediated EMT. Cell Death Dis. 14:1152023. View Article : Google Scholar : PubMed/NCBI

94 

Burns TF, Dobromilskaya I, Murphy SC, Gajula RP, Thiyagarajan S, Chatley SN, Aziz K, Cho YJ, Tran PT and Rudin CM: Inhibition of TWIST1 leads to activation of oncogene-induced senescence in oncogene-driven non-small cell lung cancer. Mol Cancer Res. 11:329–338. 2013. View Article : Google Scholar : PubMed/NCBI

95 

Fukuda K, Takeuchi S, Arai S, Kita K, Tanimoto A, Nishiyama A and Yano S: Glycogen synthase kinase-3 inhibition overcomes epithelial-mesenchymal transition-associated resistance to osimertinib in EGFR-mutant lung cancer. Cancer Sci. 111:2374–2384. 2020. View Article : Google Scholar : PubMed/NCBI

96 

Raoof S, Mulford IJ, Frisco-Cabanos H, Nangia V, Timonina D, Labrot E, Hafeez N, Bilton SJ, Drier Y, Ji F, et al: Targeting FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer. Oncogene. 38:6399–6413. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Wu Q, Zhen Y, Shi L, Vu P, Greninger P, Adil R, Merritt J, Egan R, Wu MJ, Yin X, et al: EGFR inhibition potentiates FGFR inhibitor therapy and overcomes resistance in FGFR2 fusion-positive cholangiocarcinoma. Cancer Discov. 12:1378–1395. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Hu S, Fu W, Li T, Yuan Q, Wang F, Lv G, Lv Y, Fan X, Shen Y, Lin F, et al: Antagonism of EGFR and Notch limits resistance to EGFR inhibitors and radiation by decreasing tumor-initiating cell frequency. Sci Transl Med. 9:eaag03392017. View Article : Google Scholar : PubMed/NCBI

99 

Lin Z, Wang Q, Jiang T, Wang W and Zhao JJ: Targeting tumor-associated macrophages with STING agonism improves the antitumor efficacy of osimertinib in a mouse model of EGFR-mutant lung cancer. Front Immunol. 14:10772032023. View Article : Google Scholar : PubMed/NCBI

100 

Wang Y, Zhang T, Guo L, Ren T and Yang Y: Stromal extracellular matrix is a microenvironmental cue promoting resistance to EGFR tyrosine kinase inhibitors in lung cancer cells. Int J Biochem Cell Biol. 106:96–106. 2019. View Article : Google Scholar : PubMed/NCBI

101 

Peng S, Wang R, Zhang X, Ma Y, Zhong L, Li K, Nishiyama A, Arai S, Yano S and Wang W: EGFR-TKI resistance promotes immune escape in lung cancer via increased PD-L1 expression. Mol Cancer. 18:1652019. View Article : Google Scholar : PubMed/NCBI

102 

Hsu KH, Huang YH, Tseng JS, Chen KC, Ku WH, Su KY, Chen JJW, Chen HW, Yu SL, Yang TY and Chang GC: High PD-L1 expression correlates with primary resistance to EGFR-TKIs in treatment naïve advanced EGFR-mutant lung adenocarcinoma patients. Lung Cancer. 127:37–43. 2019. View Article : Google Scholar : PubMed/NCBI

103 

Zhang W, Wang J, Liang J, He Z, Wang K and Lin H: RNA methylation of CD47 mediates tumor immunosuppression in EGFR-TKI resistant NSCLC. Br J Cancer. 132:569–579. 2025. View Article : Google Scholar : PubMed/NCBI

104 

Chen Q, Xia L, Wang J, Zhu S, Wang J, Li X, Yu Y, Li Z, Wang Y, Zhu G and Lu S: EGFR-mutant NSCLC may remodel TME from non-inflamed to inflamed through acquiring resistance to EGFR-TKI treatment. Lung Cancer. 192:1078152024. View Article : Google Scholar : PubMed/NCBI

105 

Yao X, Gao C, Sun C, Chen ZS and Zhuang J: Epigenetic code underlying EGFR-TKI resistance in non-small cell lung cancer: Elucidation of mechanisms and perspectives on therapeutic strategies. Drug Discov Today. 30:1043212025. View Article : Google Scholar : PubMed/NCBI

106 

Quan C, Chen Y, Wang X, Yang D, Wang Q, Huang Y, Petersen RB, Liu X, Zheng L, Li Y and Huang K: Loss of histone lysine methyltransferase EZH2 confers resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Cancer Lett. 495:41–52. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Bui VNV, Daugaard TF, Sorensen BS and Nielsen AL: Expression of the non-coding RNA nc886 facilitates the development of tyrosine kinase inhibitor resistance in EGFR-mutated non-small-cell lung cancer cells. Biochem Biophys Res Commun. 731:1503952024. View Article : Google Scholar : PubMed/NCBI

108 

Hong W, Wu Q, Zhang J and Zhou Y: Prognostic value of EGFR 19-del and 21-L858R mutations in patients with non-small cell lung cancer. Oncol Lett. 18:3887–3895. 2019.PubMed/NCBI

109 

Soleimani-Meigooni DN and Rabinovici GD: Tau PET visual reads: Research and clinical applications and future directions. J Nucl Med. 64:822–824. 2023. View Article : Google Scholar : PubMed/NCBI

110 

George RS, Htoo A, Cheng M, Masterson TM, Huang K, Adra N, Kaimakliotis HZ, Akgul M and Cheng L: Artificial intelligence in prostate cancer: Definitions, current research, and future directions. Urol Oncol. 40:262–270. 2022. View Article : Google Scholar : PubMed/NCBI

111 

Luo Q, Zhang L, Luo C and Jiang M: Emerging strategies in cancer therapy combining chemotherapy with immunotherapy. Cancer Lett. 454:191–203. 2019. View Article : Google Scholar : PubMed/NCBI

112 

Rahmat K, Mumin NA, Hamid MTR, Hamid SA and Ng WL: MRI breast: Current imaging trends, clinical applications, and future research directions. Curr Med Imaging. 18:1347–1361. 2022. View Article : Google Scholar : PubMed/NCBI

113 

Bajbouj K, Al-Ali A, Ramakrishnan RK, Saber-Ayad M and Hamid Q: Histone modification in NSCLC: Molecular mechanisms and therapeutic targets. Int J Mol Sci. 22:117012021. View Article : Google Scholar : PubMed/NCBI

114 

Reungwetwattana T, Weroha SJ and Molina JR: Oncogenic pathways, molecularly targeted therapies, and highlighted clinical trials in non-small-cell lung cancer (NSCLC). Clin Lung Cancer. 13:252–266. 2012. View Article : Google Scholar : PubMed/NCBI

115 

Chia PL, Dobrovic A, Dobrovic A and John T: Prevalence and natural history of ALK positive non-small-cell lung cancer and the clinical impact of targeted therapy with ALK inhibitors. Clin Epidemiol. 6:423–432. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Hsu J, Annunziata JF, Burns E, Bernicker EH, Olsen RJ and Thomas JS: Molecular signatures of KRAS-mutated lung adenocarcinoma: Analysis of concomitant EGFR, ALK, STK11, and PD-L1 status. Clin Pathol. 15:2632010X2211020542022. View Article : Google Scholar : PubMed/NCBI

117 

Gkolfinopoulos S and Mountzios G: Beyond EGFR and ALK: Targeting rare mutations in advanced non-small cell lung cancer. Ann Transl Med. 6:1422018. View Article : Google Scholar : PubMed/NCBI

118 

Hirsch FR, Suda K, Wiens J and Bunn PA Jr: New and emerging targeted treatments in advanced non-small-cell lung cancer. Lancet. 388:1012–1024. 2016. View Article : Google Scholar : PubMed/NCBI

119 

Weishan H, Donglin Z, Guangmei D, Wenya L, Fasheng W and Jibing C: Immunoradiotherapy for NSCLC: Mechanisms, clinical outcomes, and future directions. Clin Transl Oncol. 26:1063–1076. 2024. View Article : Google Scholar : PubMed/NCBI

120 

Selzer E: Impact of molecular targets in cancer drug development: Historical influence and future perspectives. Expert Rev Clin Pharmacol. 3:161–163. 2010. View Article : Google Scholar : PubMed/NCBI

121 

Amaravadi RK, Kimmelman AC and Debnath J: Targeting autophagy in cancer: Recent advances and future directions. Cancer Discov. 9:1167–1181. 2019. View Article : Google Scholar : PubMed/NCBI

122 

Black A and Morris D: Personalized medicine in metastatic non-small-cell lung cancer: Promising targets and current clinical trials. Curr Oncol. 19 (Suppl 1):S73–S85. 2012. View Article : Google Scholar : PubMed/NCBI

123 

Saxena A, Becker D, Preeshagul I, Lee K, Katz E and Levy B: Therapeutic effects of repurposed therapies in non-small cell lung cancer: What is old is new again. Oncologist. 20:934–945. 2015. View Article : Google Scholar : PubMed/NCBI

124 

Harmey D, Griffin PR and Kenny PJ: Development of novel pharmacotherapeutics for tobacco dependence: Progress and future directions. Nicotine Tob Res. 14:1300–1318. 2012. View Article : Google Scholar : PubMed/NCBI

125 

Ciardiello D, Elez E, Tabernero J and Seoane J: Clinical development of therapies targeting TGFβ: Current knowledge and future perspectives. Ann Oncol. 31:1336–1349. 2020. View Article : Google Scholar : PubMed/NCBI

126 

Kannan K and Mohan S: Targeting exon mutations in NSCLC: Clinical insights into LAG-3, TIM-3 pathways, and advances in fourth-generation EGFR-TKIs. Med Oncol. 42:1962025. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 30 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian Z, Cen L, Wei F, Dong J, Huang Y, Han Y, Wang Z, Deng J and Jiang Y: EGFR mutations in non‑small cell lung cancer: Classification, characteristics and resistance to third‑generation EGFR‑tyrosine kinase inhibitors (Review). Oncol Lett 30: 375, 2025.
APA
Tian, Z., Cen, L., Wei, F., Dong, J., Huang, Y., Han, Y. ... Jiang, Y. (2025). EGFR mutations in non‑small cell lung cancer: Classification, characteristics and resistance to third‑generation EGFR‑tyrosine kinase inhibitors (Review). Oncology Letters, 30, 375. https://doi.org/10.3892/ol.2025.15121
MLA
Tian, Z., Cen, L., Wei, F., Dong, J., Huang, Y., Han, Y., Wang, Z., Deng, J., Jiang, Y."EGFR mutations in non‑small cell lung cancer: Classification, characteristics and resistance to third‑generation EGFR‑tyrosine kinase inhibitors (Review)". Oncology Letters 30.2 (2025): 375.
Chicago
Tian, Z., Cen, L., Wei, F., Dong, J., Huang, Y., Han, Y., Wang, Z., Deng, J., Jiang, Y."EGFR mutations in non‑small cell lung cancer: Classification, characteristics and resistance to third‑generation EGFR‑tyrosine kinase inhibitors (Review)". Oncology Letters 30, no. 2 (2025): 375. https://doi.org/10.3892/ol.2025.15121