Next Article in Journal
Beyond Carcinoembryonic Antigens: The Role of CA-125 and CA-199 in Predicting Prognosis of Lung Adenocarcinoma
Next Article in Special Issue
Phase Ia/Ib Study of Afatinib with Capecitabine in Patients with Refractory Solid Tumors and Pancreaticobiliary Cancers
Previous Article in Journal
T-Cell Redirecting Therapies in Multiple Myeloma: Pathogenesis and Management of Toxicities Beyond CRS and ICANS
Previous Article in Special Issue
Preclinical Photodynamic Therapy Targeting Blood Vessels with AGuIX® Theranostic Nanoparticles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor (EGFR) Common Mutations: New Strategies

1
Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
2
Department of Medicine (DME), University of Udine, 33100 Udine, Italy
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(9), 1515; https://doi.org/10.3390/cancers17091515
Submission received: 11 February 2025 / Revised: 23 April 2025 / Accepted: 26 April 2025 / Published: 30 April 2025
(This article belongs to the Collection Combination Therapies in Cancers)

Simple Summary

The prognosis of non-small cell lung cancer with common EGFR mutations remains poor, despite the results obtained with the introduction of tyrosine kinase inhibitors. Disease progression during targeted therapy is linked to the development of resistance mechanisms. This literature review aims to summarize and clarify the main therapeutic strategies developed to address this important unmet medical need, including the latest updated and recently presented data.

Abstract

Epidermal growth factor receptor (EGFR) mutations are described in 10–15% of Caucasian patients and in 50% of Asian patients with non-squamous non-small cell lung cancer (NSCLC). The introduction of tyrosine kinase inhibitors (TKIs) has revolutionized the therapeutic scenario and has changed the natural history of the disease. Despite the results obtained with osimertinib, a third-generation TKI, most patients experience disease progression. The search for new therapeutic strategies both to enhance first-line treatment and to ensure adequate second-line therapies represents an unmet medical need, towards which all efforts are being concentrated. In this review, we describe the main strategies identified to improve the prognosis of patients with EGFR-mutated NSCLC.

1. Introduction

In 2022, lung cancer was the leading cause of cancer incidence and death worldwide [1]. It is typically associated with smoking habits, although its incidence is increasing in non-smokers, particularly women and young people. Epidermal growth factor receptor (EGFR) gene-activating mutations were first identified in non-small cell lung cancer (NSCLC) in 2004 [2]. These mutations are present in 10–15% of cases of non-squamous NSCLC in the Caucasian population but can reach an incidence of 50% in the Asian population [3]. They are usually found in non-smokers, with a higher incidence in young and female patients. EGFR mutations usually involve exons 18–21, which are part of the tyrosine kinase domain. So-called “common mutations”, including del19 and L858R in exon 21, representing about 90% of all EGFR mutations, have shown a marked sensitivity to tyrosine kinase inhibitors (TKIs), whose introduction into the medical armory has significantly changed the prognosis of these patients and the therapeutic approach to lung cancer [4]. Conversely, “uncommon mutations”, which constitute only 5–10% of cases, are represented by hundreds of mutations affecting exons 18–21 and are associated with a worse prognosis, showing extremely heterogeneous behaviors [5]. Among these, exon 20 insertion mutations, accounting for 4–12% of cases of EGFR-mutated NSCLC, are the most frequent and are described especially in the Asian population and in non-smoker patients and are characterized by a poor response (less than 10%) to TKIs [6]. Other uncommon mutations are represented by G719X (exon 18), S768I (exon 20), and L861Q (exon 21), described with a prevalence of ~3%, ~1%, and ~1%, respectively. The presence of compound mutations, the finding of which is increasingly frequent since the introduction of more sophisticated detection methods such as next-generation sequencing (NGS), can further modify the clinical history of the disease [7]. The quality of life and survival results of patients with advanced NSCLC with EGFR common mutations have been drastically altered by the approval of three generations of anti-EGFR tyrosine kinase inhibitors (TKIs) [8]. These medications are characterized by oral delivery and generally modest adverse effects (AEs), primarily cutaneous and gastrointestinal toxicity, providing acceptable management. Gefitinib and erlotinib are the first-generation anti-EGFR TKIs that were first studied in advanced non-small cell lung cancer and later, for the first time, outperformed chemotherapy in the first-line treatment of EGFR-mutated NSCLC [9]. Compared to first-generation TKIs, second-generation TKIs, afatinib and dacomitinib, have demonstrated a better EGFR inhibition because of their irreversible covalent binding mechanism [10,11,12,13]. Although they demonstrated their superiority compared to gefitinib in the LUX-Lung 3-6-7 studies and in the ARCHER study, respectively, achieving a progression-free survival (PFS) of 11 months for afatinib and 14.7 months for dacomitinib, their toxicity, poor handling, and lack of benefit to overall survival (OS) and the advent of osimertinib have severely limited their use in clinical practice. Osimertinib is a third-generation TKI with a better tolerability profile and strong antitumor activity by combining irreversibility with high selectivity over EGFR-mutated receptors [14]. Osimertinib was first used in clinical practice after progression to first- or second-generation TKIs, when resistance mechanisms developed, usually after 9–14 months of treatment. Specifically, in patients with the T790M mutation, it proved to be more beneficial than chemotherapy [15]. In the FLAURA trial, it showed superiority over first-generation EGFR TKIs in frontline treatment, with a median OS of 38.6 months (95% confidence interval [CI], 34.5 to 41.8) versus 31.8 months (hazard ratio (HR), 0.799; 95% CI, 0.641, 0.997), a median PFS of 18.9 months versus 10.2 months (HR, 0.46 [95% CI, 0.37, 0.57]), and an objective response rate (ORR) of 80% (95% CI, 75 to 85%) [16]. Osimertinib has demonstrated higher intracranial activity and a manageable safety profile. Although it is active against numerous resistance mutations that arise following first- and second-generation TKIs, the efficacy of osimertinib is also time-limited, since the onset of several types of resistance mechanisms is described [17]. In particular, the study of resistance mechanisms has allowed us to distinguish three different categories: on-target mechanisms, related to intrinsic additional mutations of EGFR; off-target mechanisms, related to other intracellular pathways [such as the mitogen-activated protein kinase (MAPK) pathway or Phosphoinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) pathway]; and histological transformation [18]. To understand the biological rationale underlying disease progression and to plan the subsequent treatment, the molecular study of the disease through a tissue re-biopsy and/or a liquid biopsy is essential. Tissue re-biopsy, in particular, is indispensable to document histological transformation. Among on-target mechanisms, the T790M mutation is well known: it is a typical mechanism of acquired resistance to first- and second-generation TKIs, being described in up to 50% of cases [19]. This mutation determines a steric conformation that prevents the binding of first- and second-generation TKIs, but not of osimertinib. Other on-target mechanisms are represented by EGFR amplification or by the acquisition of further point mutations such as C797S [20]. Regarding off-target mechanisms, another common resistance mechanism to EGFR-TKIs is the activation of alternative pathways that circumvent EGFR inhibition, such as the Ras–Raf–MEK–ERK cascade or MET amplification (10–15% of cases), which activates the downstream AKT pathway, the crucial signaling pathway for antiapoptosis and cell proliferation [21]. About 2% of cases of resistance to osimertinib and 12% of cases of resistance to first- and second-generation EGFR-TKIs have been identified to have ErbB2 gene amplification [22]. Furthermore, mutations in FGFR, BRAF, KRAS, and PIK3CA may result in resistance to first-, second- and third-generation EGFR-TKIs, giving cancer cells an edge in survival [23]. The possibility to associate to osimertinib a target therapy against these resistance mechanisms can somehow reverse resistance or, in any case, act on intratumoral heterogeneity [24]. Finally, about 2–15% of patients treated with EGFR-TKIs develop a histological transformation from adenocarcinoma to small cell lung cancer (SCLC), or rarely to squamous carcinoma, acquiring resistance to EGFR-TKIs. The transformation into SCLC can be linked to the co-presence of mutations in tumor suppressor genes, such as RB1 or TP53, or to the presence, since the onset of the disease, of different cell clones [25,26,27]. Thus, the emergence of resistance mechanisms poses a significant challenge in clinical practice. For this reason, finding effective second-line therapies and bolstering first-line treatment are also significant research topics (Figure 1). In this literature review, we summarize the main therapeutic strategies developed to improve the prognosis of patients with EGFR-mutated NSCLC, reporting the results of the most significant studies as well as the updated data that have been recently presented.

2. Update on Advanced NSCLC with EGFR Common Mutations

2.1. Strengthening the First-Line Treatment

2.1.1. Combination of TKIs with Chemotherapy

Despite the demonstrated efficacy of osimertinib and its widespread use in clinical practice, most patients experience disease progression [28,29]. One of the mechanisms identified to enhance first-line treatment efficacy and to postpone the onset of resistance mechanisms is the association between osimertinib and chemotherapy (CT). The rationale for this therapeutic option is to combat intratumoral heterogeneity by exploiting a double mechanism of action. Updated data relating to this topic have recently been presented. Several studies have evaluated the effectiveness of the association between chemotherapy and TKIs with different schedules: administered concurrently or sequentially, as maintenance therapy after chemotherapy induction, or intercalated. The most significant results were obtained by simultaneously administering TKI and chemotherapy (Table 1). A recent meta-analysis analyzed four trials, including 1413 patients [30,31,32,33,34]. When EGFR-TKI and chemotherapy were combined, the PFS was considerably higher than when EGFR-TKI was used alone (HR: 0.52 [95%CI: 0.45–0.59]; p < 0.00001). All examined subgroups showed the PFS benefit, but patients with brain metastases showed the greatest benefit (HR: 0.41 [95% CI: 0.33–0.51]; p < 0.00001) when compared to those without (HR: 0.57 [95% CI: 0.44–0.74]; p < 0.0001). Even in the subgroup of patients with ECOG PS 2, which were included in only one trial [32], PFS was better with the combination. Additionally, the combination showed an advantage in terms of OS (HR:0.69 [95% CI: 0.52–0.93]; p = 0.01) and ORR (HR: 1.93 [95% CI: 1.50–2.47]; p < 0.00001) [30]. FLAURA2, an international phase III study, enrolled 557 patients worldwide, confirming data in a non-Asian population [34]. In terms of safety, the combination was up to five times more likely to cause grade 3 or higher adverse effects than EGFR-TKI alone. The primary toxicities were digestive (mucositis, nausea, vomiting, and lack of appetite), hematological (anemia and neutropenia), and fatigue/asthenia, typically more associated with chemotherapy.

2.1.2. Combination of TKIs with Antiangiogenic Agents

According to preclinical research, the EGFR and VEGF (vascular endothelial growth factor) signaling pathways work together to promote tumor growth [35]. Based on clinical research, VEGF expression is linked to increased vascularization in NSCLC, and overexpression of VEGF is associated with a poor prognosis in NSCLC. Patients with EGFR-mutated NSCLC have significantly longer OS when their serum VEGF levels are lower (HR 0.277, p = 0.018), and those who have lower serum VEGF levels after treatment have better PFS. As a result, the association of antiangiogenic agents with EGFR TKIs has been assessed in a number of randomized controlled clinical trials (Table 2).
The combination of bevacizumab with first-generation EGFR TKIs has been shown to be able to achieve consistent improvements in PFS but typically no effect on OS. In a meta-analysis published by Zhang et al., seven trials with a total of 1612 patients were analyzed [36,37,38,39,40,41]. Regardless of the type of EGFR mutation or the existence of brain metastases, TKIs in combination with antiangiogenic agents significantly improved ORR (HR, 1.10; 95% CI, 1.01–1.20; p = 0.029) and PFS. Specifically, HR of PFS was 0.58 (95% CI: 0.48–0.71; p < 0.001) in the subgroup with the L858R mutation. The OS following combination treatment was comparable to that after TKI monotherapy. TKIs combined with antiangiogenic drugs showed a markedly greater incidence of grade 3 or higher adverse events in the safety studies. The addition of bevacizumab to osimertinib, on the other hand, has been inconsistent in improving PFS. According to the ETOP meta-analysis assessing the role of bevacizumab added to EGFR TKIs, smoking populations would benefit more than non-smoking ones [42]. In comparison to erlotinib monotherapy, the RELAY phase III study evaluated the combination of erlotinib and ramucirumab (anti-VEGFR2) and found a significant PFS benefit with a median of 19.4 months and HR 0.59 (95% CI, 0.46–0.76), but no OS benefit [40].
Table 2. Main prospective clinical trials of combination of TKI and antiangiogenic agents.
Table 2. Main prospective clinical trials of combination of TKI and antiangiogenic agents.
Clinical TrialPhaseTreatmentMedian PFSMedian OS
Seto et al. [37]2Erlotinib + bevacizumab
vs. Erlotinib
16 months
9 months
47 months
47.4 months
Stinchcombe et al. [38]2Erlotinib + bevacizumab
vs. Erlotinib
17.9 months
13.5 months
32.4 months
50.6 months
Saito et al. [39]3Erlotinib + bevacizumab
vs. Erlotinib
16.9 months
13.3 months
-
Nakagawa et al. [40]3Erlotinib + ramucirunab
vs. Erlotinib
19.4 months
12.4 months
-
Zhou et al. [41]3Erlotinib + bevacizumab
vs. Erlotinib
18 months
11.3 months
-
PFS: progression-free survival; OS: overall survival.

2.1.3. New Third-Generation TKIs

Among the new third-generation TKIs, lazertinib deserves a special mention. It is an irreversible EGFR inhibitor, with similar efficacy to osimertinib in terms of ORR and survival, but with a greater ability to cross the blood–brain barrier, ensuring better intracranial response rates. In addition to the efficacy data, reassuring safety data have emerged from the LASER201 and, more recently, LASER301 studies: in fact, lazertinib is characterized by less toxicity at the skin and cardiological level, with less impact on QT [43,44]. In LASER301, lazertinib significantly extended median PFS across 393 patients compared to gefitinib (20.6 vs. 9.7 months; hazard ratio [HR], 0.45; 95% CI, 0.34 to 0.58; p < 0.001) [44]. In both groups, the ORR was 76% (odds ratio: 0.99; 95% CI: 0.62 to 1.59). With lazertinib, the median duration of response (mDOR) was 19.4 months (95% CI: 16.6 to 24.9) vs. 8.3 months with gefitinib (95% CI: 6.9 to 10.9). At the interim analysis, the overall survival data were immature. Lazertinib and gefitinib had 18-month survival rates of 80% and 72%, respectively (HR, 0.74; 95% CI, 0.51 to 1.08; p = 0.116).
Aumolertinib, another third-generation EGFR-TKI, was evaluated originally after disease progression in patients with locally advanced or metastatic NSCLC who developed an EGFR T790M mutation after progression on first- or second-generation EGFR TKI therapy, in the APOLLO trial [45]. Positive results have also been obtained from phase III research (AENEAS) that compared the safety and effectiveness of aumolertinib to gefitinib, as a first-line treatment for patients with locally progressed or metastatic NSCLC with EGFR mutations [46]. Compared to gefitinib, aumolertinib significantly prolonged PFS (hazard ratio, 0.46; 95% CI, 0.36 to 0.60; p < 0.0001). In contrast to 9.9 months with gefitinib (95% CI, 8.3 to 12.6), the median PFS with aumolertinib was 19.3 months (95% CI, 17.8 to 20.8). In the aumolertinib and gefitinib groups, the ORR and disease control rate (DCR) were comparable (73.8% and 72.1%, respectively; 93.0% and 96.7%, respectively). With aumolertinib, mDoR was 18.1 months (95% CI, 15.2 to not applicable), while with gefitinib, it was 8.3 months (95% CI, 6.9 to 11.1). Patients receiving aumolertinib experienced rash and diarrhea (any grade) in 23.4% and 16.4% of cases, respectively, compared to 41.4% and 35.8% of patients receiving gefitinib. In untreated EGFR-mutant patients with advanced NSCLC, a combination trial of aumolertinib and anlotinib, an oral multitarget antiangiogenic TKI, showed markedly elevated toxicity, indicating that the combined therapy approach was an unacceptable therapeutic decision in this context [47].
Furmonertinib is an oral, once-daily, and widely effective mutant-selective EGFR TKI that targets both common and rare mutations, such as EGFR exon 20 insertion mutations. In the FURLONG trial, furmonertinib showed improved efficacy over gefitinib, when used as a first-line treatment for Chinese patients with EGFR-mutant NSCLC, with an acceptable safety profile, with a PFS of 20.8 months (95% CI 17.8–23.5) in the furmonertinib group and 11.1 months (9.7–12.5) in the gefitinib group (HR 0.44, 95% CI 0.34–0.58; p < 0.0001) [48].

2.1.4. Fourth-Generation TKIs

Among the main mechanisms of on-target resistance to third-generation TKIs, the appearance of a tertiary point mutation such as C797S or the loss of T790M are widely described. In particular, C797S, accountable for about 40% of occurrences of drug resistance, prevent inhibitors from forming a covalent link with the cysteine residue at position 797 on the border of the ATP-binding pocket. It has been demonstrated that when the C797S and T790M mutations are cis (located at the same allele), lung cancer cells are resistant to both the first- and third-generation EGFR-TKIs. However, when the C797S and T790M mutations are trans (located in distinct alleles), lung cancer cells have been demonstrated to respond well to the combined therapy of first- and third-generation EGFR-TKIs [49,50]. Fourth-generation TKIs were developed with the aim of overcoming resistance to third-generation TKIs and to act on the most frequent tertiary mutations, such as C797S.
Most of the studies on the new fourth-generation TKIs are still at an early stage; in some cases, the development of these drugs has suffered a setback, such as for BLU-701 and BLU-945 [51]. BDTX-1535, actually in a phase I/II dose expansion study, has demonstrated to be active in NSCLC patients across nearly all EGFR mutations, including T790M, C797S, L747P, L718Q, and compound mutations [52]. In a highly pre-treated population of 27 NSCLC patients, the ORR was 36%, with the majority of patients exhibiting sustained responses [53,54]. The most frequent side effects were diarrhea (35%, no grade ≥3 observed) and rash (70%, two patients with grade 3, no grade 4).

2.1.5. Anti-EGFR and Anti-MET Drugs: Combination of Lazertinib and Amivantamab

A widely studied possible option for the first-line treatment of patients with EGFR-mutated NSCLC is the combination of amivantamab and lazertinib, designed to enhance the efficacy of third-generation TKIs. Specifically, amivantamab, an EGFR-MET bispecific fully human antibody, can inhibit these two surface cell receptors and strengthens the innate immune system by promoting ADCC (antibody-dependent cellular cytotoxicity) and Fc-dependent trogocytosis. The action of amivantamab on MET enables it to work on cell clones whose resistance is dependent on MET activation [55]. In the phase III MARIPOSA trial, 1074 patients were divided into three arms: lazertinib, osimertinib, and amivantamab–lazertinib [56]. With a median PFS of 23.7 versus 16.6 months and HR of 0.70 (95% CI, 0.58–0.85), the experimental combination arm’s median PFS was considerably better than osimertinib. In patients with or without CNS metastases, the PFS benefit remained constant. Both arms’ ORRs were comparable, at 86% for amivantamab–lazertinib and 85% for osimertinib. Amivantamab–lazertinib was found to be effective in high-risk subgroups, particularly in patients with liver metastases at baseline (median PFS was 18.2 against 11.0 months) and TP53 co-mutations (median PFS was 18.2 versus 12.9 months for osimertinib), according to a secondary analysis from this research [57]. Furthermore, in patients who had detectable circulating tumor DNA (ctDNA) at baseline (HR 0.68 [0.53–0.86]) and in patients who did not clear ctDNA at C3D1 (0.48 [0.27–0.86]), amivantamab–lazertinib prolonged PFS. During the European Lung Cancer Congress 2025, data related to OS and icPFS were shown. Amivantamab+lazertinib showed a statistically significant and clinically relevant improvement in OS compared to osimertinib at a median follow-up of 37.8 months (HR for death, 0.75; 95% CI, 0.61–0.92; p < 0.005). The amivantamab+lazertinib arm’s median OS was not estimable (NE; 95% CI, 42.9–NE), while that of the osimertinib arm was 36.7 months (95% CI, 33.4–41.0). It is predicted that amivantamab+lazertinib will extend median OS by at least 12 months in comparison to osimertinib, assuming an exponential distribution of OS in both arms. Compared to 51% of patients in the osimertinib arm, 60% of patients in the amivantamab+lazertinib arm were still alive after 36 months. Amivantamab + lazertinib demonstrated a clinically meaningful improvement in icPFS with durable responses: the 3-year landmark icPFS was 36% vs. 18% for amivantamab + lazertinib vs. osimertinib [58]. The amivantamab–lazertinib combination has non-negligible toxicity, which is a safety concern [56]. Compared to 43% of patients treated with osimertinib, 75% of patients treated with amivantamab–lazertinib experienced grade ≥ 3 side effects, including infusion-related reactions (IRRs), skin rashes, and toxicities of the nails. In the experimental arm, 59% of patients and 35% of patients had, respectively, a dose reduction or a discontinuation of the drug, compared to 5% and 14% with osimertinib. Amivantamab–lazertinib dramatically raised the incidence of venous thromboembolic events (VTEs) by up to 37% compared to 9% in the control group, particularly in the four months after treatment started. As a result, it was advised to prescribe systematic prophylactic anticoagulation.

2.1.6. Role of Radiotherapy

Local ablative treatments, and in particular radiotherapy, may also play a role in the first-line treatment of patients with EGFR-mutated NSCLC, both to treat any residual disease after a good partial response to TKIs, and to be used upfront together with TKIs, in case of oligometastatic disease. For this second objective, the phase II SINDAS study was designed, which evaluated the addition of stereotactic body radiation therapy (SBRT) to first-generation TKIs in first-line therapy, demonstrating that the incorporation of upfront local therapy via radiotherapy, in contrast to the use of a first-line TKI alone, resulted in a statistically significant enhancement in PFS (20.2 months versus 12.5 months (p < 0.001) and OS (25.5 months versus 17.4 months (p < 0.001) for patients with EGFR-mutated NSCLC [59]. In the TKI with RT arm, treatment resulted in a 6% incidence of symptomatic grade 3–4 pneumonitis and no grade 5 occurrences. On this topic, the results of the phase II NORTHSTAR study are expected, in which local ablative therapies such as surgery or radiotherapy are evaluated on residual disease in combination with osimertinib, after 6–12 weeks of TKIs [60].

2.2. Strengthening Second-Line Treatment

Following the development of progression after first-line treatment, it becomes essential to search for the resistance mechanisms responsible for progression, to guide subsequent therapeutic choices. In case of oligoprogression, corresponding to the appearance or evolution of tumor lesions at a few limited locations, that can be treated with local therapy, such as radiotherapy or surgery, and the indication to continue TKIs beyond progression is widely shared [61,62]. The results of the phase II LAT-FLOSI and phase II/III HALT studies should help to evaluate treatment with SBRT as a new standard option, in cases of oligoprogression under EGFR TKIs [63,64]. In case of systemic progression, the current standard of care is represented by platinum- and pemetrexed-based chemotherapy, even if with a limited efficacy [65]. In fact, platinum–pemetrexed yields an ORR of 27–42.9% and median PFS of 4.4–5.6 months. A tissue re-biopsy and/or liquid biopsy are necessary for the molecular analysis of the disease to comprehend the biological reasoning behind its progression and to plan the subsequent course of treatment.

2.2.1. Combination of Anti-EGFR and Anti-MET Drugs in the Second-Line Treatment

The finding in 10–15% of cases of alterations in the MET gene, in particular amplification, has led to the development of several studies based on the combination of anti-EGFR and anti-MET drugs (Table 3). In the phase Ib TATTON trial, which included not only patients with MET amplification but also with MET overexpression, the combination of osimertinib and savolitinib, an oral, potent, and highly selective MET TKI, produced a median PFS of 5.5 months and an ORR of 33% [66,67]. The phase II ORCHARD trial, designed to identify the mechanisms underlying first-line osimertinib resistance and investigate strategies for overcoming acquired resistance, demonstrated, in the subgroup of 20 patients with MET alterations, the efficacy of the association of osimertinib–savolitinib [68]. The significance of MET status was further demonstrated by the ongoing phase II SAVANNAH’s interim results [69]. MET overexpression (IHC 3+) found in ≥90% of tumor cells and/or MET copy number ≥10 (fluorescence in situ hybridization—FISH) were used to identify the high MET-expressing group (34% of the population). The median PFS for this subgroup was 7.1 months, and the ORR was 49%. By contrast, there was no indication of efficacy in the remaining sample (ORR 9%, mPFS 2.8 months). Based on the outcome of the previously mentioned studies, the phase III SAFFRON study is currently comparing osimertinib plus savolitinib versus conventional chemotherapy, in patients with overexpressed and/or amplified MET. In INSIGHT 2, a phase II trial for patients with MET amplification following osimertinib as first-line therapy, 128 patients received oral tepotinib plus osimertinib once daily [70]. The ORR was 50.0%, the median OS was 17.8 months, the median PFS was 5.6 months, while the mDoR was 8.5 months. With an 8.3% ORR, tepotinib monotherapy had modest antitumor efficacy, supporting the advantages of EGFR and MET combination inhibition. This result implies that, rather than focusing only on MET, managing both oncogenic drivers is essential after NSCLC cells develop osimertinib resistance through MET amplification. Peripheral oedema, decreased appetite, extended electrocardiogram QT interval, and pneumonitis were the most frequent treatment-related grade 3 or worse side effects. Specifically, 13% of patients experienced serious treatment-related side events.

2.2.2. Combination of Lazertinib and Amivantamab in the Second-Line Treatment

With bispecific antibodies, the possibility of acting on both EGFR and MET is exploited. The lazertinib–amivantamab combination was originally tested in the second-line treatment. In the CHRYSALIS trial, specifically in cohort E, an ORR of 36% and a median PFS of 4.9 months were reached, with a better response in patients with high EGFR/MET expression [71]. In the CHRYSALIS2 trial, patients with MET overexpression achieved an ORR of 61% vs. 14% of patients without MET overexpression [72,73] The results of these studies led to the design of MARIPOSA-2, a phase III international trial. In this study, 657 patients with disease progression during therapy with osimertinib as the most recent line of treatment were included [74]. When compared to CT alone (median PFS of 4.2 months), PFS was considerably higher in both experimental arms, with a median PFS of 8.3 months for amivantamab–lazertinib plus CT (A-L-CT) and 6.3 months for amivantamab plus CT (A-CT). The ORR was 64% for A-CT, 63% for A-L-CT, and 36% for CT. When compared to chemotherapy alone (8.3 months), intracranial PFS improved similarly in both experimental arms (median of 12.5 months). OS data are not available yet. In both experimental arms, the number of grade ≥3 adverse events (AEs) increased significantly, reaching 92% (A-L-CT), 72% (A-CT), and 48% (CT). In 34% of cases (A-L-CT), 18% of cases (A-CT), and 4% of cases (CT), treatment was discontinued permanently. In the regimens comprising amivantamab, hematologic toxicities (including neutropenia, thrombocytopenia, anemia, and leucopenia) were the most common adverse events. Additionally, 58% and 56% of patients receiving A-CT and A-L-CT, respectively, experienced infusion-related reactions (IRRs), whose incidence can be reduced with adequate steroid-based premedication. Even in the subcutaneous (SC) formulation, amivantamab has been shown to be effective in the phase III PALOMA-3 trial, in association with lazertinib [75]. When compared to intravenous (IV) amivantamab, SC treatment demonstrated similar pharmacokinetics and a noninferior ORR (30.1%), with a positive trend in PFS (6.1 vs. 4.3 months), a longer median DOR (11.2 vs. 8.3 months for IV) and a better OS (HR 0.62 [0.42–0.92]). With SC amivantamab, lower IRR (13% vs. 66% with IV) and VTE rates (9% vs. 14% with IV) were recorded.

2.2.3. Combination of Anti-EGFR and Other Target Therapies

While the combination of dabrafenib and trametinib is recommended in the treatment of BRAF V600E-mutated NSCLC in the main guidelines, in the event of the appearance of a BRAFV600E mutation as a resistance mechanism after osimertinib progression, there is currently disagreement over the best course of action for concurrently addressing BRAF and EGFR in lung cancer. Case reports have been reported in the literature with evidence of the efficacy of the association between osimertinib and anti-BRAF therapy [76,77,78]. In particular, in a phase 2 clinical trial, patients with BRAF V600E-mutant lung cancer were found to benefit clinically from vemurafenib, a BRAF inhibitor authorized for the treatment of BRAF V600E-mutant melanoma [79].
In the second-line context, ALK rearrangement as an osimertinib resistance mechanism is uncommon (<1%). There are very few case studies in the literature on suitable treatment approaches for these situations [80,81,82]. Further studies are required to provide evidence of the combination’s efficacy, safety, and optimal administration method (concurrent vs. alternative).

2.2.4. Role of Antibody-Drug Conjugates (ADCs)

There is considerable interest in exploring the role of ADCs in NSCLC with actionable genomic alterations after failure of traditional treatments. The increased human epidermal growth factor receptor 3 (HER3) signaling in NSCLC patients who become resistant to EGFR TKIs has been an interesting finding [83]. Research has consistently demonstrated that elevated HER3 expression is linked to more aggressive NSCLC with worse outcomes, indicating its potential as a prognostic marker. Also, overexpression of heregulin (HRG), HER3’s ligand, adds to the disease’s aggressiveness, by increasing tumor proliferation and metastatic potential. HRG-mediated HER3 activation results in downstream PI3K/AKT pathway signaling, which enhances cell survival and confers resistance to EGFR-targeted treatments. Patritumab–deruxtecan is an ADC in which a human monoclonal antibody directed against HER3 is linked to deruxtecan, a topoisomerase I inhibitor. In the HERTHENA-lung01 phase II trial, 275 patients with EGFR common mutations who had previously had platinum-based chemotherapy and EGFR TKIs received patritumab–deruxtecan at a dose of 5.6 mg/kg every three weeks [84]. A median OS of 11.9 months, a median PFS of 5.5 months, and an ORR of 29% have been reported. Furthermore, in 30 patients with brain metastases who had not received previous radiation therapy, the intracranial ORR was 33%. Forty-five percent of patients experienced grade ≥ 3 adverse events, with hematological toxicity and interstitial lung disease (ILD) described as the most frequently reported adverse events. The phase III HERTHENA-Lung02 study will provide further information on the role of patritumab–deruxtecan compared to standard chemotherapy after TKI failure [85]. A further promising ADC is datopotamab–deruxtecan (Dato-Dxd), in which a trophoblast cell surface antigen 2 (TROP2) directed monoclonal antibody is linked to a topoisomerase I inhibitor payload via a cleavable linker. In the phase III TROPION-Lung01 trial, Dato-DXd dramatically increased PFS compared to docetaxel in patients with advanced or metastatic NSCLC, in patients with non-squamous histology, and, in particular, with actionable genomic alterations (AGAs) [86]. The TROPION-Lung05 phase II trial evaluated the safety and clinical activity of Dato-DXd at a dose of 6 mg/kg once every 3 weeks in patients with advanced/metastatic NSCLC with AGAs progressing during or after targeted therapy and platinum-based chemotherapy [87]. Overall, of 137 treated patients, 56.9% had EGFR mutations. In this population, the confirmed ORR was 43.6% (95% CI, 32.4 to 55.3), the median DOR was 7.0 months (95% CI, 4.2 to 9.8), the overall DCR was 82.1% (95% CI, 71.7 to 89.8), and the median PFS was 5.8 months (95% CI, 5.4 to 8.3). Additionally, 28.5% of patients experienced treatment-related adverse events (TRAEs) of grade ≥3, with stomatitis reported as the most prevalent TRAEs. Five patients (3.6%) developed ILD/pneumonitis as a result of the treatment, and one patient (0.7%) sustained a grade 5 AE.

2.2.5. Role of Immunotherapy

The role of immunotherapy after progression to TKIs in patients with EGFR-mutated NSCLC is still somewhat controversial [88]. The phosphorylated extracellular signal-regulated kinases 1/2/phosphorylated c-Jun (p-ERK1/2/p-c-Jun) signaling pathway and inhibition of Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) are two mechanisms through which EGFR activation appears to induce programmed death-ligand 1 (PD-L1) expression, resulting in T-cell apoptosis and immune response suppression [89,90]. A pooled analysis of 15 studies revealed that PD-L1 expression was lower in EGFR-mutant NSCLCs [91]. Furthermore, EGFR mutations were linked to a lower tumor mutational burden (TMB) in comparison to wild-type EGFR, offering a scientific justification and a possible explanation for this subgroup’s poor immunotherapy response [92]. Several prospective and retrospective studies have failed to demonstrate a significant benefit of immunotherapy as a single agent compared with chemotherapy in previously treated EGFR-mutated NSCLC [88]. Numerous studies have assessed the possible advantages of a combination with chemotherapy in EGFR-mutated NSCLC. Nivolumab + chemotherapy did not significantly enhance PFS compared to chemotherapy alone, according to CheckMate 722 (median PFS 5.6 vs. 5.4 months, HR 0.75, p = 0.0528) [93]. In TKI-resistant EGFR-mutant metastatic non-squamous NSCLC, the most recent phase III data from the KEYNOTE-789 trial, evaluating the safety and effectiveness of combining pembrolizumab with chemotherapy, have demonstrated that pembrolizumab with chemotherapy did not significantly enhance PFS or OS over chemotherapy alone [94]. In patients with EGFR-mutated NSCLC, the IMpower 150 study showed encouraging efficacy for a combination of immune chemotherapy and bevacizumab [95]. In 22 patients with EGFR-mutated NSCLC after TKI failure, an open-label, single-arm, phase II trial assessed the immune cell profile and effectiveness of the modified regimen consisting of atezolizumab, bevacizumab (7.5 mg/kg), and chemotherapy [96]. The DCR was 100% while the ORR was 42.9%, with 6.3 months of median PFS. The ORR (75 vs. 23.1%; p = 0.032) and PFS (14.0 vs. 6.1 months; p = 0.022) were substantially greater in patients with PD-L1 expression ≥1% than in those with PD-L1 expression <1%. The phase III ORIENT-31 trial compared sintilimab, an antibody anti-PD1, plus the bevacizumab biosimilar IBI305 plus pemetrexed–cisplatin versus sintilimab plus pemetrexed–cisplatin or chemotherapy alone, in pretreated patients with a sensitizing EGFR mutation [97]. When compared to chemotherapy alone, sintilimab plus chemotherapy increased PFS (median 5.5 months [95% CI 4.5–6.1] vs. 4.3 months [4.1–5.3]), and sintilimab plus IBI305 plus chemotherapy maintained a significant PFS benefit (median 7.2 months [95% CI 6.6–9.3). In contrast to 19.2 months for chemotherapy alone, the median OS was 21.1 months (95% CI 17.5–23.9) for the sintilimab-plus-IBI305-plus-chemotherapy group and 20.5 months for the sintilimab-plus-chemotherapy group. Regarding safety, treatment-related adverse events of grade 3 or worse happened in 56% of patients in the sintilimab-plus-IBI305-plus-chemotherapy group, 41% of patients in the sintilimab-plus-chemotherapy group, and 49% of patients in the chemotherapy-alone group. In this area, encouraging data have been derived from studies evaluating ivonescimab, a bispecific antibody targeting both PD-1 and VEGF. In patients whose disease progresses following EGFR-TKI treatment, the HARMONi-A trial, a double-blind, placebo-controlled, randomized phase III study, compares the effectiveness of ivonescimab with chemotherapy versus chemotherapy alone [98]. When ivonescimab was added to chemotherapy, the primary endpoint, PFS, improved significantly, with a median of 7.1 months compared to 4.8 months for the placebo (HR, 0.46 [95% CI, 0.34–0.62]; p < 0.001). Patients with brain metastases and those subjected to osimertinib also benefited from PFS. Data on survival are not yet mature. These findings support earlier research indicating that VEGF suppression is necessary for anti-PD(L)-1 and chemotherapy to be beneficial in EGFR-mutated patients.

3. Conclusions

Almost two decades have passed since the discovery of the first EGFR mutations in NSCLC. As understanding has grown throughout the years, the natural history of this disease has evolved. Specifically, there have been numerous developments from a therapeutic and diagnostic perspective. Regarding the diagnosis, next-generation sequencing technology allows the simultaneous testing of numerous predictive and prognostic biomarkers with high sensitivity and specificity. Specifically, it enables the identification of multiple EGFR mutations and other oncogenic molecular targets; it also makes it possible to identify the mechanisms of resistance that may arise during therapy, guiding subsequent therapeutic choices. In terms of therapeutic advancements, the clinical history of EGFR-mutated NSCLC has been completely transformed by the introduction of TKIs. To overcome resistance mechanisms and improve survival and quality of life, novel TKIs with increased selectivity and greater blood–brain barrier penetration have been investigated. Notwithstanding the good responses recorded, most patients undergo disease progression. The emergence of resistance mechanisms has led to the search for new therapeutic strategies to improve the prognosis of patients. In this literature review, we have reported the main studies designed with the aim of developing therapeutic strategies to improve the prognosis of EGFR-mutated NSCLC. Despite the achieved results, there are several critical points to consider. Specifically, if it is true that improving the first line of treatment leads to greater results, this presents a toxicity issue because, often, there is also an increase in adverse effects. The recently announced data, in particular, about the combination of amivantamab–lazertinib and osimertinib–CT in addition to standard osimertinib, have the potential to revolutionize clinical practice. Due to the increased risk of side effects, combination therapies are undoubtedly not suitable for every patient and must be customized based on the individual’s features. Osimertinib monotherapy may still be beneficial for patients who are elderly, have a modest disease burden, or have a poor performance status (PS). Conversely, combinations of osimertinib–CT or amivantamab–TKIs can be used in younger patients, with a good PS or with a higher burden of disease or in the presence of prognostic commutations of greater aggressiveness. Increased toxicity is by no means a minor issue. Also, the use of antiangiogenic drugs, like bevacizumab or ivonescimab, in patients who are more likely to experience bleeding is also limited by the possibility of increased adverse effects, such as bleeding or thromboembolic events, hypertension, or renal toxicity. The introduction of new drugs, such as ADCs or bispecific antibodies, has also brought with it the need to deal with new side effects, different from those known related to drugs used in clinical practice. Additionally, once first-line combination therapies are employed, the restricted availability of drugs for succeeding lines is another crucial factor. Although novel medications like ADCs have shown promising outcomes, their position in the therapeutic algorithm is still unclear, and further research is required to fully realize their potential. Moreover, comparing different studies is quite challenging because of the significant variables involved. In conclusion, for patients with EGFR-mutated NSCLC, there is a plethora of therapeutic options, and many more could arrive in the future, both in the first line and in the subsequent lines. It is therefore essential to be guided by the molecular re-evaluation of the disease, through a tissue biopsy or liquid biopsy, to identify the resistance mechanism and then choose a targeted therapy or propose enrollment in clinical trials.

Author Contributions

B.S., E.B., E.D.C. and A.B. conceived the review; B.S. provided data curation; B.S. wrote the manuscript; A.D.C., B.S., M.B. and M.S. provided review and editing; B.S. and A.B. supervised the manuscript; S.T. created figures through biorender.com; B.S. created all the tables. All authors have read and agreed to the published version of the manuscript.

Funding

The authors received no financial support for the research.

Acknowledgments

This work was supported by the Italian Ministry of Health (Ricerca Corrente); no grant provided. Figures were realized with biorender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. LoPiccolo, J.; Gusev, A.; Christiani, D.C.; Jänne, P.A. Lung cancer in patients who have never smoked—An emerging disease. Nat. Rev. Clin. Oncol. 2024, 21, 121–146. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Castellanos, E.; Feld, E.; Horn, L. Driven by Mutations: The Predictive Value of Mutation Subtype in EGFR -Mutated Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 612–623. [Google Scholar] [CrossRef] [PubMed]
  4. Hendriks, L.; Kerr, K.; Menis, J.; Mok, T.; Nestle, U.; Passaro, A.; Peters, S.; Planchard, D.; Smit, E.; Solomon, B.; et al. Oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023, 34, 339–357. [Google Scholar] [CrossRef] [PubMed]
  5. Gristina, V.; Malapelle, U.; Galvano, A.; Pisapia, P.; Pepe, F.; Rolfo, C.; Tortorici, S.; Bazan, V.; Troncone, G.; Russo, A. The significance of epidermal growth factor receptor uncommon mutations in non-small cell lung cancer: A systematic review and critical appraisal. Cancer Treat. Rev. 2020, 85, 101994. [Google Scholar] [CrossRef] [PubMed]
  6. Passaro, A.; Mok, T.; Peters, S.; Popat, S.; Ahn, M.-J.; de Marinis, F. Recent Advances on the Role of EGFR Tyrosine Kinase Inhibitors in the Management of NSCLC with Uncommon, Non Exon 20 Insertions, EGFR Mutations. J. Thorac. Oncol. 2021, 16, 764–773. [Google Scholar] [CrossRef] [PubMed]
  7. Kim, E.Y.; Na Cho, E.; Park, H.S.; Hong, J.Y.; Lim, S.; Youn, J.P.; Hwang, S.Y.; Chang, Y.S. Compound EGFR mutation is frequently detected with co-mutations of actionable genes and associated with poor clinical outcome in lung adenocarcinoma. Cancer Biol. Ther. 2016, 17, 237–245. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  8. Lee, D.H. Treatments for EGFR-mutant non-small cell lung cancer (NSCLC): The road to a success, paved with failures. Pharmacol. Ther. 2017, 174, 1–21. [Google Scholar] [CrossRef] [PubMed]
  9. Lee, C.K.; Davies, L.; Wu, Y.-L.; Mitsudomi, T.; Inoue, A.; Rosell, R.; Zhou, C.; Nakagawa, K.; Thongprasert, S.; Fukuoka, M.; et al. Gefitinib or Erlotinib vs Chemotherapy for EGFR Mutation-Positive Lung Cancer: Individual Patient Data Meta-Analysis of Overall Survival. JNCI J. Natl. Cancer Inst. 2017, 109, djw279. [Google Scholar] [CrossRef] [PubMed]
  10. Park, K.; Tan, E.-H.; O’Byrne, K.; Zhang, L.; Boyer, M.; Mok, T.; Hirsh, V.; Yang, J.C.-H.; Lee, K.H.; Lu, S.; 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, 577–589. [Google Scholar] [CrossRef] [PubMed]
  11. Paz-Ares, L.; Tan, E.-H.; O’byrne, K.; Zhang, L.; Hirsh, V.; Boyer, M.; Yang, J.-H.; Mok, T.; Lee, K.H.; Lu, S.; et al. Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: Overall survival data from the phase IIb LUX-Lung 7 trial. Ann. Oncol. 2017, 28, 270–277. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Wu, Y.-L.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Tsuji, F.; Linke, R.; Rosell, R.; Corral, J.; 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, 1454–1466. [Google Scholar] [CrossRef] [PubMed]
  13. Cheng, Y.; Mok, T.S.; Zhou, X.; Lu, S.; Zhou, Q.; Zhou, J.; Du, Y.; Yu, P.; Liu, X.; Hu, C.; et al. Safety and efficacy of first-line dacomitinib in Asian patients with EGFR mutation-positive non-small cell lung cancer: Results from a randomized, open-label, phase 3 trial (ARCHER 1050). Lung Cancer 2021, 154, 176–185. [Google Scholar] [CrossRef]
  14. Bertoli, E.; De Carlo, E.; Del Conte, A.; Stanzione, B.; Revelant, A.; Fassetta, K.; Spina, M.; Bearz, A. Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It? Int. J. Mol. Sci. 2022, 23, 6936. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. Yang, J.C.-H.; Ahn, M.-J.; Kim, D.-W.; Ramalingam, S.S.; Sequist, L.V.; Su, W.-C.; Kim, S.-W.; Kim, J.-H.; Planchard, D.; Felip, E.; et al. Osimertinib in Pretreated T790M-Positive Advanced Non–Small-Cell Lung Cancer: AURA Study Phase II Extension Component. J. Clin. Oncol. 2017, 35, 1288–1296. [Google Scholar] [CrossRef] [PubMed]
  16. Soria, J.-C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef] [PubMed]
  17. Huang, L.; Fu, L. Mechanisms of resistance to EGFR tyrosine kinase inhibitors. Acta Pharm. Sin. B 2015, 5, 390–401. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Favorito, V.; Ricciotti, I.; De Giglio, A.; Fabbri, L.; Seminerio, R.; Di Federico, A.; Gariazzo, E.; Costabile, S.; Metro, G. Non-small cell lung cancer: An update on emerging EGFR-targeted therapies. Expert Opin. Emerg. Drugs 2024, 29, 139–154. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, Z.-F.; Ren, S.-X.; Li, W.; Gao, G.-H. Frequency of the acquired resistant mutation T790 M in non-small cell lung cancer patients with active exon 19Del and exon 21 L858R: A systematic review and meta-analysis. BMC Cancer 2018, 18, 148. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  20. Zalaquett, Z.; Hachem, M.C.R.; Kassis, Y.; Hachem, S.; Eid, R.; Kourie, H.R.; Planchard, D. Acquired resistance mechanisms to osimertinib: The constant battle. Cancer Treat. Rev. 2023, 116, 102557. [Google Scholar] [CrossRef] [PubMed]
  21. Coleman, N.; Hong, L.; Zhang, J.; Heymach, J.; Hong, D.; Le, X. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open 2021, 6, 100319. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Ohashi, K.; Sequist, L.V.; Arcila, M.E.; Moran, T.; Chmielecki, J.; Lin, Y.-L.; Pan, Y.; Wang, L.; de Stanchina, E.; Shien, K.; et al. Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF gene mutations but lack mutations in KRAS, NRAS, or MEK1. Proc. Natl. Acad. Sci. USA 2012, 109, E2127–E2133. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Ware, K.E.; Marshall, M.E.; Heasley, L.R.; Marek, L.; Hinz, T.K.; Hercule, P.; Helfrich, B.A.; Doebele, R.C.; Heasley, L.E. Rapidly Acquired Resistance to EGFR Tyrosine Kinase Inhibitors in NSCLC Cell Lines through De-Repression of FGFR2 and FGFR3 Expression. PLoS ONE 2010, 5, e14117. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Pecci, F.; Cantini, L.; Metro, G.; Ricciuti, B.; Lamberti, G.; Farooqi, A.A.; Berardi, R. Non-small-cell lung cancer: How to manage EGFR-mutated disease. Drugs Context 2022, 11, 2022-4-1. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Niederst, M.J.; Sequist, L.V.; Poirier, J.T.; Mermel, C.H.; Lockerman, E.L.; Garcia, A.R.; Katayama, R.; Costa, C.; Ross, K.N.; Moran, T.; et al. RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer. Nat. Commun. 2015, 6, 6377. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Offin, M.; Chan, J.M.; Tenet, M.; Rizvi, H.A.; Shen, R.; Riely, G.J.; Rekhtman, N.; Daneshbod, Y.; Quintanal-Villalonga, A.; Penson, A.; et al. Concurrent RB1 and TP53 Alterations Define a Subset of EGFR-Mutant Lung Cancers at risk for Histologic Transformation and Inferior Clinical Outcomes. J. Thorac. Oncol. 2019, 14, 1784–1793. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  27. Lee, J.-K.; Lee, J.; Kim, S.; Kim, S.; Youk, J.; Park, S.; An, Y.; Keam, B.; Kim, D.-W.; Heo, D.S.; et al. Clonal History and Genetic Predictors of Transformation Into Small-Cell Carcinomas From Lung Adenocarcinomas. J. Clin. Oncol. 2017, 35, 3065–3074. [Google Scholar] [CrossRef] [PubMed]
  28. Du, X.; Yang, B.; An, Q.; Assaraf, Y.G.; Cao, X.; Xia, J. Acquired resistance to third-generation EGFR-TKIs and emerging next-generation EGFR inhibitors. Innovation 2021, 2, 100103. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Schmid, S.; Li, J.J.; Leighl, N.B. Mechanisms of osimertinib resistance and emerging treatment options. Lung Cancer 2020, 147, 123–129. [Google Scholar] [CrossRef] [PubMed]
  30. Landre, T.; Assié, J.-B.; Chouahnia, K.; Guetz, G.D.; Auliac, J.-B.; Chouaïd, C. First-line concomitant EGFR-TKI + chemotherapy versus EGFR-TKI alone for advanced EGFR-mutated NSCLC: A meta-analysis of randomized phase III trials. Expert Rev. Anticancer. Ther. 2024, 24, 775–780. [Google Scholar] [CrossRef] [PubMed]
  31. Miyauchi, E.; Morita, S.; Nakamura, A.; Hosomi, Y.; Watanabe, K.; Ikeda, S.; Seike, M.; Fujita, Y.; Minato, K.; Ko, R.; et al. Updated Analysis of NEJ009: Gefitinib-Alone Versus Gefitinib Plus Chemotherapy for Non–Small-Cell Lung Cancer with Mutated EGFR. J. Clin. Oncol. 2022, 40, 3587–3592. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Noronha, V.; Patil, V.M.; Joshi, A.; Menon, N.; Chougule, A.; Mahajan, A.; Janu, A.; Purandare, N.; Kumar, R.; More, S.; et al. Gefitinib Versus Gefitinib Plus Pemetrexed and Carboplatin Chemotherapy in EGFR-Mutated Lung Cancer. J. Clin. Oncol. 2020, 38, 124–136. [Google Scholar] [CrossRef] [PubMed]
  33. Hou, X.; Li, M.; Wu, G.; Feng, W.; Su, J.; Jiang, H.; Jiang, G.; Chen, J.; Zhang, B.; You, Z.; 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. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Planchard, D.; Jänne, P.A.; Cheng, Y.; Yang, J.C.-H.; Yanagitani, N.; Kim, S.-W.; Sugawara, S.; Yu, Y.; Fan, Y.; Geater, S.L.; et al. Osimertinib with or without Chemotherapy in EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2023, 389, 1935–1948. [Google Scholar] [CrossRef] [PubMed]
  35. Lichtenberger, B.M.; Tan, P.K.; Niederleithner, H.; Ferrara, N.; Petzelbauer, P.; Sibilia, M. Autocrine VEGF Signaling Synergizes with EGFR in Tumor Cells to Promote Epithelial Cancer Development. Cell 2010, 140, 268–279. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, S.; Li, S.; Liu, J.; Yang, C.; Zhang, L.; Bao, H.; Cheng, Y. Comparative Efficacy and Safety of TKIs Alone or in Combination with Antiangiogenic Agents in Advanced EGFR-Mutated NSCLC as the First-Line Treatment: A Systematic Review and Meta-Analysis. Clin. Lung Cancer 2021, 23, 159–169. [Google Scholar] [CrossRef] [PubMed]
  37. Seto, T.; Kato, T.; Nishio, M.; Goto, K.; Atagi, S.; Hosomi, Y.; Hida, T.; Maemondo, M.; Nakagawa, K.; Nagase, S.; et al. Erlotinib alone or with bevacizumab as first-line therapy in patients with advanced non-squamous non-small-cell lung cancer harbouring EGFR mutations (JO25567): An open-label, randomised, multicentre, phase 2 study. Lancet Oncol. 2014, 15, 1236–1244. [Google Scholar] [CrossRef] [PubMed]
  38. Stinchcombe, T.E.; Jänne, P.A.; Wang, X.; Bertino, E.M.; Weiss, J.; Bazhenova, L.; Gu, L.; Lau, C.; Paweletz, C.; Jaslowski, A.; et al. Effect of Erlotinib Plus Bevacizumab vs Erlotinib Alone on Progression-Free Survival in Patients with Advanced EGFR-Mutant Non–Small Cell Lung Cancer. JAMA Oncol. 2019, 5, 1448–1455. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. Saito, H.; Fukuhara, T.; Furuya, N.; Watanabe, K.; Sugawara, S.; Iwasawa, S.; Tsunezuka, Y.; Yamaguchi, O.; Okada, M.; Yoshimori, K.; 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, 625–635. [Google Scholar] [CrossRef] [PubMed]
  40. Nakagawa, K.; Garon, E.B.; Seto, T.; Nishio, M.; Aix, S.P.; Paz-Ares, L.; Chiu, C.-H.; Park, K.; Novello, S.; Nadal, E.; 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, 1655–1669. [Google Scholar] [CrossRef] [PubMed]
  41. Zhou, Q.; Xu, C.-R.; Cheng, Y.; Liu, Y.-P.; Chen, G.-Y.; Cui, J.-W.; Yang, N.; Song, Y.; Li, X.-L.; Lu, S.; 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. [Google Scholar] [CrossRef] [PubMed]
  42. Dafni, U.; Peters, S.; Tsourti, Z.; Zygoura, P.; Vervita, K.; Han, J.-Y.; De Castro, J.; Coate, L.; Früh, M.; Hashemi, S.; et al. Impact of smoking status on the relative efficacy of the EGFR TKI/angiogenesis inhibitor combination therapy in advanced NSCLC—A systematic review and meta-analysis. ESMO Open 2022, 7, 100507. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Han, J.-Y.; Ahn, M.-J.; Lee, K.H.; Lee, Y.-G.; Kim, D.-W.; Min, Y.J.; Kim, S.-W.; Cho, E.K.; Kim, J.-H.; Lee, G.-W.; et al. Updated overall survival and ctDNA analysis in patients with EGFR T790M-positive advanced non-small cell lung cancer treated with lazertinib in the phase 1/2 LASER201 study. BMC Med. 2024, 22, 428. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Cho, B.C.; Ahn, M.-J.; Kang, J.H.; Soo, R.A.; Reungwetwattana, T.; Yang, J.C.-H.; Cicin, I.; Kim, D.-W.; Wu, Y.-L.; Lu, S.; et al. Lazertinib Versus Gefitinib as First-Line Treatment in Patients with EGFR-Mutated Advanced Non–Small-Cell Lung Cancer: Results From LASER301. J. Clin. Oncol. 2023, 41, 4208–4217. [Google Scholar] [CrossRef] [PubMed]
  45. Lu, S.; Wang, Q.; Zhang, G.; Dong, X.; Yang, C.-T.; Song, Y.; Chang, G.-C.; Lu, Y.; Pan, H.; Chiu, C.-H.; et al. Efficacy of Aumolertinib (HS-10296) in Patients with Advanced EGFR T790M+ NSCLC: Updated Post-National Medical Products Administration Approval Results From the APOLLO Registrational Trial. J. Thorac. Oncol. 2022, 17, 411–422. [Google Scholar] [CrossRef] [PubMed]
  46. Lu, S.; Dong, X.; Jian, H.; Chen, J.; Chen, G.; Sun, Y.; Ji, Y.; Wang, Z.; Shi, J.; Lu, J.; 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, 3162–3171. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Li, T.; Chang, K.; Qiu, X.; Lai, Z.; Luo, Y.; Chen, J.; Lv, W.; Lin, Z.; Pei, X.; Wu, X.; et al. A pilot study of anlotinib with third-generation epidermal growth factor receptor tyrosine kinase inhibitors in untreated EGFR-mutant patients with advanced non-small cell lung cancer. Transl. Lung Cancer Res. 2023, 12, 1256–1263. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  48. Shi, Y.; Chen, G.; Wang, X.; Liu, Y.; Wu, L.; Hao, Y.; Liu, C.; Zhu, S.; Zhang, X.; Li, Y.; 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, 1019–1028. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, Z.; Yang, J.-J.; Huang, J.; Ye, J.-Y.; Zhang, X.-C.; Tu, H.-Y.; Han-Zhang, H.; Wu, Y.-L. Lung Adenocarcinoma Harboring EGFR T790M and In Trans C797S Responds to Combination Therapy of First- and Third-Generation EGFR TKIs and Shifts Allelic Configuration at Resistance. J. Thorac. Oncol. 2017, 12, 1723–1727. [Google Scholar] [CrossRef] [PubMed]
  50. Niederst, M.J.; Hu, H.; Mulvey, H.E.; Lockerman, E.L.; Garcia, A.R.; Piotrowska, Z.; Sequist, L.V.; Engelman, J.A. The Allelic Context of the C797S Mutation Acquired upon Treatment with Third-Generation EGFR Inhibitors Impacts Sensitivity to Subsequent Treatment Strategies. Clin. Cancer Res. 2015, 21, 3924–3933. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  51. Mansour, M.A.; AboulMagd, A.M.; Abbas, S.H.; Abdel-Rahman, H.M.; Abdel-Aziz, M. Insights into fourth generation selective inhibitors of (C797S) EGFR mutation combating non-small cell lung cancer resistance: A critical review. RSC Adv. 2023, 13, 18825–18853. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Dempke, W.C.M.; Fenchel, K. Targeting C797S mutations and beyond in non-small cell lung cancer—A mini-review. Transl. Cancer Res. 2024, 13, 6540–6549. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Johnson, M.L.; Henry, J.T.; Spira, A.I.; Battiste, J.; Alnahhas, I.; Ahluwalia, M.S.; Barve, M.A.; Edenfield, W.J.; Nam, D.-H.; Eathiraj, S.; et al. A phase 1 study to assess BDTX-1535, an oral EGFR inhibitor, in patients with glioblastoma or non-small-cell lung cancer. J. Clin. Oncol. 2023, 41, TPS9156. [Google Scholar] [CrossRef]
  54. Wrigley, N. BDTX-1535 Yields Radiographic Responses in Small EGFR+ NSCLC Population. Available online: https://cancernetwork.com (accessed on 27 August 2024).
  55. Wespiser, M.; Swalduz, A.; Pérol, M. Treatment sequences in EGFR mutant advanced NSCLC. Lung Cancer 2024, 194, 107895. [Google Scholar] [CrossRef] [PubMed]
  56. Cho, B.C.; Lu, S.; Felip, E.; Spira, A.I.; Girard, N.; Lee, J.-S.; Lee, S.-H.; Ostapenko, Y.; Danchaivijitr, P.; Liu, B.; et al. Amivantamab plus Lazertinib in Previously Untreated EGFR -Mutated Advanced NSCLC. N. Engl. J. Med. 2024, 391, 1486–1498. [Google Scholar] [CrossRef] [PubMed]
  57. Felip, E.; Cho, B.; Gutiérrez, V.; Alip, A.; Besse, B.; Lu, S.; Spira, A.; Girard, N.; Califano, R.; Gadgeel, S.; et al. Amivantamab plus lazertinib versus osimertinib in first-line EGFR-mutant advanced non-small-cell lung cancer with biomarkers of high-risk disease: A secondary analysis from MARIPOSA. Ann. Oncol. 2024, 35, 805–816. [Google Scholar] [CrossRef] [PubMed]
  58. Yang, J.-H.; Kim, Y.; Lee, S.-H.; Liu, B.; Ostapenko, Y.; Lu, S.; Alip, A.; Korbenfeld, E.; Dias, J.; Danchaivijitr, P.; et al. 4O: Amivantamab plus lazertinib vs osimertinib in first-line (1L) EGFR-mutant (EGFRm) advanced NSCLC: Final overall survival (OS) from the phase III MARIPOSA study. J. Thorac. Oncol. 2025, 20, S6–S8. [Google Scholar] [CrossRef]
  59. Wang, X.-S.; Bai, Y.-F.; Verma, V.; Yu, R.-L.; Tian, W.; Ao, R.; Deng, Y.; Zhu, X.-Q.; Liu, H.; Pan, H.-X.; et al. Randomized Trial of First-Line Tyrosine Kinase Inhibitor with or Without Radiotherapy for Synchronous Oligometastatic EGFR-Mutated Non-Small Cell Lung Cancer. JNCI J. Natl. Cancer Inst. 2022, 115, 742–748. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Khan, T.M.; Verbus, E.A.; Gandhi, S.; Heymach, J.V.; Hernandez, J.M.; Elamin, Y.Y. 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, 4688–4689. [Google Scholar] [CrossRef] [PubMed]
  61. Park, K.; Yu, C.-J.; Kim, S.-W.; Lin, M.-C.; Sriuranpong, V.; Tsai, C.-M.; Lee, J.-S.; Kang, J.-H.; Chan, K.C.A.; Perez-Moreno, P.; et al. First-Line Erlotinib Therapy Until and Beyond Response Evaluation Criteria in Solid Tumors Progression in Asian Patients with Epidermal Growth Factor Receptor Mutation–Positive Non–Small-Cell Lung Cancer: The ASPIRATION Study. JAMA Oncol. 2016, 2, 305–308. [Google Scholar] [CrossRef] [PubMed]
  62. Zheng, L.; Wang, Y.; Xu, Z.; Yang, Q.; Zhu, G.; Liao, X.-Y.; Chen, X.; Zhu, B.; Duan, Y.; Sun, J. Concurrent EGFR-TKI and Thoracic Radiotherapy as First-Line Treatment for Stage IV Non-Small Cell Lung Cancer Harboring EGFR Active Mutations. Oncologist 2019, 24, 1031–e612. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Mavrikios, A.; Remon, J.; Quevrin, C.; Mercier, O.; Tselikas, L.; Botticella, A.; Nicolas, E.; Deutsch, E.; Besse, B.; Planchard, D.; et al. Local control strategies for management of NSCLC with oligoprogressive disease. Cancer Treat. Rev. 2023, 120, 102621. [Google Scholar] [CrossRef] [PubMed]
  64. McDonald, F.; Hanna, G. Oligoprogressive Oncogene-addicted Lung Tumours: Does Stereotactic Body Radiotherapy Have a Role? Introducing the HALT Trial. Clin. Oncol. 2017, 30, 1–4. [Google Scholar] [CrossRef] [PubMed]
  65. Passaro, A.; Leighl, N.; Blackhall, F.; Popat, S.; Kerr, K.; Ahn, M.; Arcila, M.; Arrieta, O.; Planchard, D.; de Marinis, F.; et al. ESMO expert consensus statements on the management of EGFR mutant non-small-cell lung cancer. Ann. Oncol. 2022, 33, 466–487. [Google Scholar] [CrossRef] [PubMed]
  66. Hartmaier, R.J.; Markovets, A.A.; Ahn, M.J.; Sequist, L.V.; Han, J.-Y.; Cho, B.C.; Yu, H.A.; Kim, S.-W.; Yang, J.C.-H.; Lee, J.-S.; et al. Osimertinib + Savolitinib to Overcome Acquired MET-Mediated Resistance in Epidermal Growth Factor Receptor–Mutated, MET-Amplified Non–Small Cell Lung Cancer: TATTON. Cancer Discov. 2022, 13, 98–113. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Sequist, L.V.; Han, J.-Y.; Ahn, M.-J.; Cho, B.C.; Yu, H.; Kim, S.-W.; Yang, J.C.-H.; Lee, J.S.; Su, W.-C.; Kowalski, D.; et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: Interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef] [PubMed]
  68. Yu, H.A.; Goldberg, S.B.; Le, X.; Piotrowska, Z.; Goldman, J.W.; De Langen, A.J.; Okamoto, I.; Cho, B.C.; Smith, P.; Mensi, I.; et al. Biomarker-Directed Phase II Platform Study in Patients with EGFR Sensitizing Mutation-Positive Advanced/Metastatic Non-Small Cell Lung Cancer Whose Disease Has Progressed on First-Line Osimertinib Therapy (ORCHARD). Clin. Lung Cancer 2021, 22, 601–606. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  69. Ahn, M.J.; Marinis, F.D.; Bonanno, L.; Cho, B.C.; Kim, T.M.; Cheng, S.; Novello, S.; Proto, C.; Kim, S.-W.; Lee, J.S.; et al. EP08.02-140 MET Biomarker-based Preliminary Efficacy Analysis in SAVANNAH: Savolitinib+ osimertinib in EGFRm NSCLC post-Osimertinib. J. Thorac. Oncol. 2022, 17, S469–S470. [Google Scholar] [CrossRef]
  70. Wu, Y.-L.; Guarneri, V.; Voon, P.J.; Lim, B.K.; Yang, J.-J.; Wislez, M.; Huang, C.; Liam, C.K.; Mazieres, J.; Tho, L.M.; et al. Tepotinib plus osimertinib in patients with EGFR-mutated non-small-cell lung cancer with MET amplification following progression on first-line osimertinib (INSIGHT 2): A multicentre, open-label, phase 2 trial. Lancet Oncol. 2024, 25, 989–1002, Erratum in Lancet Oncol. 2024, 25, e472. [Google Scholar] [CrossRef] [PubMed]
  71. Cho, B.C.; Kim, D.-W.; Spira, A.I.; Gomez, J.E.; Haura, E.B.; Kim, S.-W.; Sanborn, R.E.; Cho, E.K.; Lee, K.H.; Minchom, A.; et al. Amivantamab plus lazertinib in osimertinib-relapsed EGFR-mutant advanced nonsmall cell lung cancer: A phase 1 trial. Nat. Med. 2023, 29, 2577–2585. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Besse, B.; Baik, C.S.; Marmarelis, M.E.; Sabari, J.K.; Goto, K.; Shu, C.A.; Lee, J.-S.; Ou, S.-H.I.; Cho, B.C.; Waqar, S.N.; et al. Predictive biomarkers for treatment with amivantamab plus lazertinib among EGFR-mutated NSCLC in the post-osimertinib setting: Analysis of tissue IHC and ctDNA NGS. J. Clin. Oncol. 2023, 41, 9013. [Google Scholar] [CrossRef]
  73. Besse, B.; Goto, K.; Wang, Y.; Lee, S.-H.; Marmarelis, M.E.; Ohe, Y.; Caro, R.B.; Kim, D.-W.; Lee, J.-S.; Cousin, S.; et al. Amivantamab Plus Lazertinib in Patients with EGFR-Mutant NSCLC After Progression on Osimertinib and Platinum-Based Chemotherapy: Results From CHRYSALIS-2 Cohort A. J. Thorac. Oncol. 2025; Online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  74. Passaro, A.; Wang, J.; Wang, Y.; Lee, S.-H.; Melosky, B.; Shih, J.-Y.; Azuma, K.; Juan-Vidal, O.; Cobo, M.; Felip, E.; et al. Amivantamab plus chemotherapy with and without lazertinib in EGFR-mutant advanced NSCLC after disease progression on osimertinib: Primary results from the phase III MARIPOSA-2 study. Ann. Oncol. 2023, 35, 77–90. [Google Scholar] [CrossRef] [PubMed]
  75. Leighl, N.B.; Akamatsu, H.; Lim, S.M.; Cheng, Y.; Minchom, A.R.; Marmarelis, M.E.; Sanborn, R.E.; Yang, J.C.-H.; Liu, B.; John, T.; et al. Subcutaneous Versus Intravenous Amivantamab, Both in Combination with Lazertinib, in Refractory Epidermal Growth Factor Receptor–Mutated Non–Small Cell Lung Cancer: Primary Results From the Phase III PALOMA-3 Study. J. Clin. Oncol. 2024, 42, 3593–3605. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  76. Huang, Y.; Gan, J.; Guo, K.; Deng, Y.; Fang, W. Acquired BRAF V600E Mutation Mediated Resistance to Osimertinib and Responded to Osimertinib, Dabrafenib, and Trametinib Combination Therapy. J. Thoracic Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2019, 14, e236–e237. [Google Scholar] [CrossRef] [PubMed]
  77. Dagogo-Jack, I.; Piotrowska, Z.; Cobb, R.; Banwait, M.; Lennerz, J.K.; Hata, A.N.; Digumarthy, S.R.; Sequist, L.V. Response to the Combination of Osimertinib and Trametinib in a Patient with EGFR-Mutant NSCLC Harboring an Acquired BRAF Fusion. J. Thorac. Oncol. 2019, 14, e226–e228. [Google Scholar] [CrossRef] [PubMed]
  78. Xie, Z.; Gu, Y.; Xie, X.; Lin, X.; Ouyang, M.; Qin, Y.; Zhang, J.; Lizaso, A.; Chen, S.; Zhou, C. Lung Adenocarcinoma Harboring Concomitant EGFR Mutations and BRAF V600E Responds to a Combination of Osimertinib and Vemurafenib to Overcome Osimertinib Resistance. Clin. Lung Cancer 2021, 22, e390–e394. [Google Scholar] [CrossRef] [PubMed]
  79. Subbiah, V.; Gervais, R.; Riely, G.; Hollebecque, A.; Blay, J.-Y.; Felip, E.; Schuler, M.; Gonçalves, A.; Italiano, A.; Keedy, V.; et al. Efficacy of Vemurafenib in Patients with Non–Small-Cell Lung Cancer with BRAF V600 Mutation: An Open-Label, Single-Arm Cohort of the Histology-Independent VE-BASKET Study. JCO Precis. Oncol. 2019, 3, PO.18.00266. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  80. Batra, U.; Sharma, M.; Amrith, B.; Mehta, A.; Jain, P. EML4-ALK Fusion as a Resistance Mechanism to Osimertinib and Its Successful Management with Osimertinib and Alectinib: Case Report and Review of the Literature. Clin. Lung Cancer 2020, 21, E597–E600. [Google Scholar] [CrossRef] [PubMed]
  81. Offin, M.; Somwar, R.; Rekhtman, N.; Benayed, R.; Chang, J.C.; Plodkowski, A.; Lui, A.J.; Eng, J.; Rosenblum, M.; Li, B.T.; et al. Acquired ALK and RET Gene Fusions as Mechanisms of Resistance to Osimertinib in EGFR -Mutant Lung Cancers. JCO Precis. Oncol. 2018, 2, PO.18.00126. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  82. Hu, Y.; Xiao, L.; Yang, N.; Zhang, Y. Tyrosine kinase inhibitor acquired resistance mechanism alternates between EGFR and ALK in a lung adenocarcinoma patient. Thorac. Cancer 2019, 10, 1252–1255. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Chen, Q.; Jia, G.; Zhang, X.; Ma, W. Targeting HER3 to overcome EGFR TKI resistance in NSCLC. Front. Immunol. 2024, 14, 1332057, Erratum in Front. Immunol. 2024, 15, 1376045. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Yu, H.A.; Goto, Y.; Hayashi, H.; Felip, E.; Yang, J.C.-H.; Reck, M.; Yoh, K.; Lee, S.-H.; Paz-Ares, L.; Besse, B.; et al. HERTHENA-Lung01, a Phase II Trial of Patritumab Deruxtecan (HER3-DXd) in Epidermal Growth Factor Receptor–Mutated Non–Small-Cell Lung Cancer After Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy and Platinum-Based Chemotherapy. J. Clin. Oncol. 2023, 41, 5363–5375. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  85. Mok, T.; Jänne, P.A.; Nishio, M.; Novello, S.; Reck, M.; Steuer, C.; Wu, Y.-L.; Fougeray, R.; Fan, P.-D.; Meng, J.; et al. HERTHENA-Lung02: Phase III study of patritumab deruxtecan in advanced EGFR -mutated NSCLC after a third-generation EGFR TKI. Future Oncol. 2023, 20, 969–980. [Google Scholar] [CrossRef] [PubMed]
  86. Ahn, M.-J.; Tanaka, K.; Paz-Ares, L.; Cornelissen, R.; Girard, N.; Pons-Tostivint, E.; Baz, D.V.; Sugawara, S.; Cobo, M.; Pérol, M.; et al. Datopotamab Deruxtecan Versus Docetaxel for Previously Treated Advanced or Metastatic Non–Small Cell Lung Cancer: The Randomized, Open-Label Phase III TROPION-Lung01 Study. J. Clin. Oncol. 2025, 43, 260–272. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  87. Sands, J.; Sands, J.; Sands, J.; Ahn, M.-J.; Ahn, M.-J.; Ahn, M.-J.; Lisberg, A.; Lisberg, A.; Lisberg, A.; Cho, B.C.; et al. Datopotamab Deruxtecan in Advanced or Metastatic Non–Small Cell Lung Cancer with Actionable Genomic Alterations: Results From the Phase II TROPION-Lung05 Study. J. Clin. Oncol. 2025, 43, 1254–1265. [Google Scholar] [CrossRef] [PubMed]
  88. Foffano, L.; Bertoli, E.; Bortolot, M.; Torresan, S.; De Carlo, E.; Stanzione, B.; Del Conte, A.; Puglisi, F.; Spina, M.; Bearz, A. Immunotherapy in Oncogene-Addicted NSCLC: Evidence and Therapeutic Approaches. Int. J. Mol. Sci. 2025, 26, 583. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Chen, N.; Fang, W.; Zhan, J.; Hong, S.; Tang, Y.; Kang, S.; Zhang, Y.; He, X.; Zhou, T.; Qin, T.; et al. Upregulation of PD-L1 by EGFR Activation Mediates the Immune Escape in EGFR-Driven NSCLC: Implication for Optional Immune Targeted Therapy for NSCLC Patients with EGFR Mutation. J. Thorac. Oncol. 2015, 10, 910–923. [Google Scholar] [CrossRef] [PubMed]
  90. Lin, K.; Cheng, J.; Yang, T.; Li, Y.; Zhu, B. EGFR-TKI down-regulates PD-L1 in EGFR mutant NSCLC through inhibiting NF-κB. Biochem. Biophys. Res. Commun. 2015, 463, 95–101, Erratum in Biochem. Biophys. Res. Commun. 2022, 629, 189. [Google Scholar] [CrossRef] [PubMed]
  91. Dong, Z.-Y.; Zhang, J.-T.; Liu, S.-Y.; Su, J.; Zhang, C.; Xie, Z.; Zhou, Q.; Tu, H.-Y.; Xu, C.-R.; Yan, L.-X.; et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. OncoImmunology 2017, 6, e1356145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  92. Toki, M.I.; Mani, N.; Smithy, J.W.; Liu, Y.; Altan, M.; Wasserman, B.; Tuktamyshov, R.; Schalper, K.; Syrigos, K.N.; Rimm, D.L. Immune Marker Profiling and Programmed Death Ligand 1 Expression Across NSCLC Mutations. J. Thorac. Oncol. 2018, 13, 1884–1896. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  93. Mok, T.; Nakagawa, K.; Park, K.; Ohe, Y.; Girard, N.; Kim, H.R.; Wu, Y.-L.; Gainor, J.; Lee, S.-H.; Chiu, C.-H.; et al. Nivolumab Plus Chemotherapy in Epidermal Growth Factor Receptor–Mutated Metastatic Non–Small-Cell Lung Cancer After Disease Progression on Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors: Final Results of CheckMate 722. J. Clin. Oncol. 2024, 42, 1252–1264. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  94. Yang, J.C.-H.; Lee, D.H.; Lee, J.-S.; Fan, Y.; de Marinis, F.; Iwama, E.; Inoue, T.; Rodríguez-Cid, J.; Zhang, L.; Yang, C.-T.; et al. Phase III KEYNOTE-789 Study of Pemetrexed and Platinum with or Without Pembrolizumab for Tyrosine Kinase Inhibitor–Resistant, EGFR –Mutant, Metastatic Nonsquamous Non–Small Cell Lung Cancer. J. Clin. Oncol. 2024, 42, 4029–4039. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  95. Reck, M.; Mok, T.S.K.; Nishio, M.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): Key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir. Med. 2019, 7, 387–401. [Google Scholar] [CrossRef] [PubMed]
  96. Wu, S.; Ho, C.; Yang, J.C.; Yu, S.; Lin, S.; Liao, B.; Yang, C.; Lin, Y.; Yu, C.; Chuang, Y.; et al. Atezolizumab, bevacizumab, pemetrexed and platinum for EGFR-mutant NSCLC patients after EGFR TKI failure: A phase II study with immune cell profile analysis. Clin. Transl. Med. 2024, 15, e70149. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  97. Lu, S.; Wu, L.; Jian, H.; Cheng, Y.; Wang, Q.; Fang, J.; Wang, Z.; Hu, Y.; Han, L.; Sun, M.; et al. Sintilimab plus chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer with disease progression after EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): Second interim analysis from a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Respir. Med. 2023, 11, 624–636. [Google Scholar] [CrossRef] [PubMed]
  98. HARMONi-A Study Investigators; Fang, W.; Zhao, Y.; Luo, Y.; Yang, R.; Huang, Y.; He, Z.; Zhao, H.; Li, M.; Li, K.; et al. Ivonescimab Plus Chemotherapy in Non–Small Cell Lung Cancer with EGFR Variant: A Randomized Clinical Trial. JAMA 2024, 332, 561–570. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Figure 1. Main new strategies in EGFR-mutated NSCLC: novel approaches for EGFR-mutated NSCLC involve either using new therapies or improving existing ones by adding other drugs.
Figure 1. Main new strategies in EGFR-mutated NSCLC: novel approaches for EGFR-mutated NSCLC involve either using new therapies or improving existing ones by adding other drugs.
Cancers 17 01515 g001
Table 1. Main clinical studies of combination of TKIs and chemotherapy.
Table 1. Main clinical studies of combination of TKIs and chemotherapy.
Clinical TrialTreat
Ments
PTS (n)ORRMedian icPFSMedian PFSMedian OS
GAP-Brain [33]Gefitinib + ChT vs. Gefitinib16180.0% (95% CI, 71.0–89.0%) vs. 64.2% (95% CI, 53.5–74.9%) 15.6 mo (95% CI, 14.3–16.9) vs. 9.1 mo (95% CI, 8.0–10.2)16.3 (95% CI, 14.4–18.2) vs. 9.5 mo (95% CI, 8.3–10.8)35.0 vs. 28.9 mo; hazard ratio, 0.65; 95% CI, 0.43–0.99
NEJ009 [31]Gefitinib + ChT vs. Gefitinib345--20.9 (95% CI, 18.0–24.0) vs. 18.0 mo (95% CI, 16.3–20.7) 49.03 (95% CI, 41.77–56.73) vs. 38.47 mo (95% CI, 31.1–47.1)
Noronha et al. [32] Gefitinib + ChT vs. Gefitinib350--16 (95% CI, 13.5 to 18.5) vs. 8 mo (95% CI, 7.0–9.0)not reached vs. 17 mo (95% CI, 13.5–20.5)
FLAURA-2 [34]Osimertinib + ChT vs. Osimertinib55783% (95% CI, 78–87) vs. 76% (95% CI, 70–80)-25.5 (95% CI, 24.7–NC) vs. 16.7 mo (95% CI, 14.1–21.3)-
PTS: patients; n: number; ChT: chemotherapy; mo: months; ORR: overall response rate; icPFS: intracranial progression-free survival; PFS: progression-free survival; OS: overall survival.
Table 3. Main clinical studies of combination of anti-EGFR and anti-MET agents.
Table 3. Main clinical studies of combination of anti-EGFR and anti-MET agents.
Clinical TrialPhaseORR Median DoRMedian PFSMedian OS
TATTON [63]1b33%-5.5 months-
ORCHARD [64]2----
SAVANNAH [65]2b49%-7.1 months-
INSIGHT2 [66]250%8.5 months5.6 months17.8 months
ORR: overall response rate; DoR: duration of response; PFS: progression-free survival; OS: overall survival.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stanzione, B.; Del Conte, A.; Bertoli, E.; De Carlo, E.; Bortolot, M.; Torresan, S.; Spina, M.; Bearz, A. Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor (EGFR) Common Mutations: New Strategies. Cancers 2025, 17, 1515. https://doi.org/10.3390/cancers17091515

AMA Style

Stanzione B, Del Conte A, Bertoli E, De Carlo E, Bortolot M, Torresan S, Spina M, Bearz A. Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor (EGFR) Common Mutations: New Strategies. Cancers. 2025; 17(9):1515. https://doi.org/10.3390/cancers17091515

Chicago/Turabian Style

Stanzione, Brigida, Alessandro Del Conte, Elisa Bertoli, Elisa De Carlo, Martina Bortolot, Sara Torresan, Michele Spina, and Alessandra Bearz. 2025. "Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor (EGFR) Common Mutations: New Strategies" Cancers 17, no. 9: 1515. https://doi.org/10.3390/cancers17091515

APA Style

Stanzione, B., Del Conte, A., Bertoli, E., De Carlo, E., Bortolot, M., Torresan, S., Spina, M., & Bearz, A. (2025). Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor (EGFR) Common Mutations: New Strategies. Cancers, 17(9), 1515. https://doi.org/10.3390/cancers17091515

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop