Next Article in Journal
Solid Lipid Nanoparticles Containing Morin: Preparation, Characterization, and Ex Vivo Permeation Studies
Next Article in Special Issue
Potent and Selective Benzothiazole-Based Antimitotics with Improved Water Solubility: Design, Synthesis, and Evaluation as Novel Anticancer Agents
Previous Article in Journal
Increased Bone Marrow Uptake and Accumulation of Very-Late Antigen-4 Targeted Lipid Nanoparticles
Previous Article in Special Issue
Iminosugar-Based Nicotinamide Phosphoribosyltransferase (NAMPT) Inhibitors as Potential Anti-Pancreatic Cancer Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies

1
Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
2
Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, 70124 Bari, Italy
3
Division of Cardiac Surgery, University of Bari, 70124 Bari, Italy
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2023, 15(6), 1604; https://doi.org/10.3390/pharmaceutics15061604
Submission received: 21 April 2023 / Revised: 13 May 2023 / Accepted: 24 May 2023 / Published: 27 May 2023
(This article belongs to the Special Issue Advances in Anticancer Agent)

Abstract

:
Almost 17% of Western patients affected by non-small cell lung cancer (NSCLC) have an activating epidermal growth factor receptor (EGFR) gene mutation. Del19 and L858R are the most-common ones; they are positive predictive factors for EGFR tyrosine kinase inhibitors (TKIs). Currently, osimertinib, a third-generation TKI, is the standard first-line therapy for advanced NSCLC patients with common EGFR mutations. This drug is also administered as a second-line treatment for those patients with the T790M EGFR mutation and previously treated with first- (erlotinib, gefitinib) or second- (afatinib) generation TKIs. However, despite the high clinical efficacy, the prognosis remains severe due to intrinsic or acquired resistance to EGRF-TKIs. Various mechanisms of resistance have been reported including the activation of other signalling pathways, the development of secondary mutations, the alteration of the downstream pathways, and phenotypic transformation. However, further data are needed to achieve the goal of overcoming resistance to EGFR-TKIs, hence the necessity of discovering novel genetic targets and developing new-generation drugs. This review aimed to deepen the knowledge of intrinsic and acquired molecular mechanisms of resistance to EGFR-TKIs and the development of new therapeutic strategies to overcome TKIs’ resistance.

1. Introduction

Non-small cell lung cancer (NSCLC) is the most-frequent cause of cancer-related deaths in the world [1]. Platinum-based chemotherapy was the only therapeutic option for advanced NSCLC patients for many years with a poor prognosis because of a median overall survival (OS) < 12 months [2]. However, the discovery of NSCLC oncogenic drivers led to the development of targeted drugs with an impressive survival benefit for select patients. In particular, the most-important oncogenic drivers are the epidermal growth factor receptor (EGFR) gene mutations [3]. Currently, various EGFR tyrosine kinase inhibitors (EGFR-TKIs) are standard treatment options for patients with activating EGFR gene mutations.

1.1. Epidermal Growth Factor Receptor Pathway in NSCLC

EGFR (ERBb1/HER1) belongs to the HER (ERBb) family with three other members: HER2 (ErbB2), HER3 (ErbB3), and HER4 (ErbB4) [4]. The binding of specific ligands, such as epidermal growth factor (EGF) and transforming growth factor-α (TGF-α), to the EGFR extracellular domain led to receptor dimerisation with other HER family members [5]. Consequently, the autophosphorylation at the receptor key tyrosine residues takes place. In this way, various downstream signalling pathways are activated including the rat sarcoma (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase (MAPK) pathway, the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) pathway, the phospholipase C-protein kinase C (PLC-PKC) pathway, and the janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, which regulate cellular proliferation, survival, and apoptosis [6]. EGFR exons 18 to 24 encode the tyrosine kinase domain of the receptor. EGFR-activating gene mutations are located in exons 18, 19, 20, and 21 (the most-common ones in exons 19 and 21) and are responsible for the constitutive EGFR activation, which leads to cell proliferation [7]. These mutations are present in 10–15% of Caucasian NSCLC patients and 30–50% of Asian ones. They regard a more typically adenocarcinoma histotype, women, non-smokers, or Asian NSCLC patients [3].
The EGFR-activating gene mutations in exons 18, 19, 20, and 21 are classically divided into common (exon 19 deletion, exon 21 L858R point mutation), which correspond to 85–90% and generally confer sensitivity to EGFR-TKIs treatment, and uncommon (rare EGFR mutations and complex EGFR mutations), which account for 10–15% and present variable predictive values, from sensitivity to resistance [4,8]. Moreover, it is also possible to find other EGFR alterations consisting of the combination of EGFR mutations with other EGFR mutations or with one or more mutations of other genes (tumour suppressor gene or oncogene). In some cases, only a small percentage of tumour cells has the specific EGFR mutation. The variable sensitivities to EGFR-TKIs could be explained by the variable tertiary structure of the EGFR protein under the influence of the different EGFR mutations [8].
Table 1 summarises all categories of EGFR alterations.

1.1.1. Common EGFR mutations

Several studies reported that Del19 (45–50%) appears more frequently than L858R (37–40%) in Asian, as well as in Caucasian populations [4].
The Del19 and L858R mutations lead to elevated receptor dimerisation and activity due to the destabilisation of the inactive conformation of the EGFR receptor [9].
While the L858R mutation corresponds to the substitution of leucine to arginine at codon 858 (c.2573T > G, p.L858R), Del19 presents more than 30 variants, and the most-common is delE746_A750 corresponding to 73% of cases, with a deletion of 9 to 24 nucleotides [9]. In 25% of cases, Del19 variants start at position E747. The remaining percentage is known as entitled non-LRE (2%) [5,9].

1.1.2. Uncommon EGFR Mutations

Approximatively 600 EGFR uncommon or rare mutations have been described, accounting for 10–15% of cases, with variable sensitivity to EGFR-TKI treatment [10] and a similar clinical presentation in comparison with common ones. Examples of rare EGFR mutations are some EGFR exon point mutations such as exon 18, G719X; exon 20, S768I; and exon 21, L861Q [4,11].

1.1.3. Other EGFR Alterations

Complex mutations have a prevalence of 5–15% of all EGFR mutations. They are defined by the combination of common and rare EGFR mutations or rare and rare EGFR mutations or common and common ones. The most-frequent EGFR mutations involved in complex mutations are G179X (90%), G709X (75%), and S768I (50%) [12,13,14,15]. The sensitivity to EGFR-TKIs depends on the specific combination of mutations. It is better when one of them is a sensitivity mutation, such as Del19 or L858R, and lower when the combination includes a resistance mutation. For example, E709A + G719C, G787R + L858R, H870R + L858R, and E884K + L858R are sensitive complex mutations, while T790M + L858R is a resistant one [12,13,14,15].
Co-mutations correspond to the combination of EGFR mutation(s) with one or more mutations of another gene (tumour suppressor gene or oncogene) [16]. Co-mutations account for similar prevalence across the common EGFR mutations. Their incidence seems to be correlated with prior treatment. These genetic alterations often are found in several genes such as TP53, RB1, CTNNB1 (β-catenin), NKX2-1, or PI3KCA [17,18,19]. Some co-mutations are correlated with a worse prognosis; for example, TP53 mutations, ATM alterations, PTEN-inactivating mutations, KRAS mutations, and IDH1 mutations are associated with lower clinical results following EGFR-TKI treatment [17].
Subclonal mutations have a low variant allele frequency (VAF), which may be due to the presence of the specific mutation only in a small percentage of tumour cells [20]. All types of EGFR mutations could be subject to these genetic alterations, in particular the resistant ones. For example, a retrospective analysis of the AURA study, the AURA3 trial, and the study performed by the French Cooperative Thoracic Intergroup showed that the T790 mutation was present only in a small proportion of patients with worse clinical outcomes under third-generation EGFR-TKIs [21,22,23,24].

1.2. Clinical Trials

EGFR-TKIs became the standard therapy for advanced EGFR-mutation-positive NSCLC patients after the evaluation of their safety and efficacy in several clinical trials performed in the last decade. Moreover, some clinical studies documented a better prognosis, in terms of progression-free survival (PFS) and OS, for Del19 compared to L858R-mutation NSCLC patients under treatment with EGFR-TKIs [25,26,27].
Table 2 summarises all clinical trials evaluating EGFR-TKIs.

1.2.1. First-Generation EGFR-TKIs: Gefitinib, Erlotinib, and Icotinib

The NEJ002 study compared gefitinib versus carboplatin plus paclitaxel as a first-line therapy for advanced NSCLC patients with a common EGFR mutation [28,29].
The IPASS trial investigated gefitinib with carboplatin plus paclitaxel in the same population of the NEJ002 study [30,31].
WJTOG3405 is a phase 3 study in which common-EGFR-mutation NSCLC patients were randomised between gefitinib and cisplatin plus docetaxel [32,33].
The OPTIMAL trial evaluated erlotinib with respect to standard chemotherapy as a first-line therapy for common-EGFR-mutation NSCLC patients [34,35].
The ENSURE study analysed erlotinib in comparison with gemcitabine plus cisplatin in Asian patients affected by common-EGFR-mutation NSCLC [36].
In the EURTAC trial, erlotinib was compared to standard chemotherapy as a first-line treatment for European patients affected by common-EGFR-mutation NSCLC [37].
The CONVINCE study was designed to evaluate the efficacy and safety of icotinib as a first-line therapy compared to cisplatin/pemetrexed plus pemetrexed maintenance for common-EGFR-mutation NSCLC patients [38].
All these trials reported a significant improvement in terms of PFS, but no statistical difference was seen for OS, maybe because of the high percentage of crossover from standard therapy to the experimental one after disease progression.
Table 2 summarises all the reported clinical trials.

1.2.2. Second-Generation EGFR-TKIs

The LUX-Lung 3 trial evaluated afatinib versus cisplatin plus gemcitabine or pemetrexed for EGFR-mutation NSCLC patients stratified according to mutation type (exon 19 deletion, L858R, or other) [39].
In the LUX-Lung 6 study, common-EGFR-mutation NSCLC patients were randomised between afatinib versus cisplatin plus gemcitabine or pemetrexed [40].
ARCHER 1050 investigated the safety and efficacy of dacomitinib with respect to gefitinib as a first-line treatment of advanced NCSLC patients with a common EGFR mutation [46].
All these trials reported a significant improvement in terms of PFS, but no statistical difference was seen for OS, maybe because of the high percentage of crossover from standard therapy to the experimental one after disease progression.

1.2.3. Third-Generation EGFR-TKI

AURA3 was designed to evaluate the safety and efficacy of osimertinib in comparison with cis/carboplatin plus pemetrexed for advanced NSCLC patients who experienced disease progression after first-line EGFR-TKI therapy and developed the EGFR T790 mutation. Indeed, osimertinib is the third-generation TKI selective for T790M resistance mutations. The authors reported a longer PFS (10.1 versus 4.4 months; HR 0.30; p < 0.001) and OS (26.8 versus 22.5 months; HR 0.87, p = 0.277) for the osimertinib group, although the latter had no significant difference. This is probably due to the high crossover rate from chemotherapy to osimertinib of patients with progressive disease. After crossover adjustment, there was an HR of 0.54 for OS. The ORR was significantly better with osimertinib (71%) than the control group (31%) (odds ratio for OR: 5.39; p < 0.001). Encephalic PFS was also significantly longer for patients treated with osimertinib (8.5 months vs. 4.2 months; HR 0.32) [42].
The FLAURA trial tested osimertinib versus standard EGFR-TKIs (gefitinib or erlotinib) in previously untreated patients with common-EGFR-mutation NSCLC. The results reported PFS and OS significantly longer for the osimertinib group (PFS: 18.9 months vs. 10.2 months; HR 0.46; p < 0.001. OS: 38.6 months vs. 31.8 months; HR 0.80 p = 0.046). The ORR was similar (80% with osimertinib and 76% with standard EGFR-TKIs; odds ratio: 1.27; p = 0.24) [26]. As a consequence of the good results reported in this trial, osimertinib has become the first-line treatment for advanced or metastatic EGFR-mutant-positive NSCLC, regardless of T790M status.
The ADAURA study analysed osimertinib for 3 years as an adjuvant therapy for NSCLC patients with stage IB-IIIA and common EGFR mutations who have or have not previously received adjuvant chemotherapy. The DFS rate was 73% and 38% at 4 years (HR 0.27; p < 0.001) for the overall population in the osimertinib and control group, respectively. As regards CNS disease, at 24 months, 98% and 85% of patients in the experimental and placebo group were alive and did not have central nervous system disease (HR 0.18). The OS results are still immature [43].

1.2.4. EGFR-TKIs Specific for Ins20

ZENITH20-2 is a multicentre, multicohort, open-label phase 2 trial that investigated poziotinib for previously treated advanced NSCLC patients with EGFR exon 20 insertions that demonstrated resistance to approved TKIs. This type of mutation is an oncogenic driver and accounts for 2–5% of NSCLCs. The ORR was 27.8%. The disease control rate (DCR) was 70.0%, and PFS was 5.5 months [44].
Mobocertinib was tested in a phase 1/2, dose-escalation and dose-expansion trial that enrolled pretreated patients with advanced NSCLC and EGFR exon 20 insertions. This drug is a TKI targeting EGFR exon 20 insertions in NSCLC. The ORR was 43%, and PFS was 7.3 months [45].

1.2.5. EGFR-TKI Treatment Combinations as First-Line Therapy

Therapeutic approaches based on treatment combination have been designed with the aim to delay cancer progression by limiting the heterogeneity of resistance mechanisms. In this regard, the combination of an EGFR-TKI (afatinib) with an anti-EGFR antibody (cetuximab) has been evaluated in both preclinical and clinical data with interesting results. In detail, this approach demonstrated overcoming the resistance correlated with the T790M mutation by inducing a degradation of EGFR. On these bases, IFCT-1503 ACE-Lung, a randomised and open-label phase 2 study, has been designed to compare afatinib plus cetuximab versus the afatinib single agent as a first-line therapy of advanced EGFR-mutant NSCLC. However, the study ended early after a futility analysis documented the comparable results between the two groups [47].
There is strong biologic rationale for therapeutic approaches targeting both the vascular endothelial growth factor (VEGF) and EGFR pathways in NSCLC because they are closely related to each other. In detail, VEGF is known to be downregulated by EGFR inhibition through hypoxia-inducible factor-1alpha-dependent and -independent mechanisms. In addition, acquired resistance to EGFR-TKIs is correlated with increased levels of VEGF, and dual-VEGF/EGFR inhibition has demonstrated activity in the case of EGFR-TKI-resistant disease [48,49,50,51]. To this end, some clinical studies evaluated this type of combination. BELIEF is a multicentre, single-arm, phase 2 trial that investigated the safety and efficacy of erlotinib plus bevacizumab for advanced NSCLC patients with common EGFR mutations. Moreover, the enrolled patients were stratified according to the pretreatment presence of the T790M mutation to test the hypothesis that the coexistence of the T790M resistance mutation with another EGFR mutation correlates with a shorter PFS. The overall PFS was 13.2 months; however, the primary endpoint was met only for T790M-positive patients (PFS of 16 months), unlike the T790M-negative group (PFS of 10.5 months) [52].
JO25567 is a multicentre, open-label, randomised, phase 2 study that evaluated the erlotinib single agent or with bevacizumab as a first-line treatment in patients with advanced NSCLC and common EGFR mutations. PFS was 16 months in the experimental group and 9.7 months in the control one (HR 0.54, p = 0.0015). No significant benefit in OS was observed [53].
RELAY is a randomised, double-blind, placebo-controlled, phase 3 trial that compared erlotinib plus ramucirumab with erlotinib monotherapy in patients suffering from untreated common-EGFR-mutated advanced NSCLC and no CNS metastases. PFS was significantly longer in the experimental group (19.4 months versus 12.4 months; HR 0.59, p < 0.0001). The OS data are still immature [54]. Preclinical data about osimertinib in association with chemotherapy (pemetrexed or cisplatin) reported a delay in the occurrence of acquired resistance and long-lasting effects, even after treatment discontinuation. However, efficacy was lower when osimertinib was administered before chemotherapy; a possible explanation could be that the EGFR-TKI-induced G1-phase blockade protects cells from chemotherapy toxicity. Wu et al., in a meta-analysis, investigated the efficacy and safety of EGFR-TKI plus chemotherapy compared to EGFR-TKI alone as a first-line treatment in advanced NSCLC patients with the common EGFR mutation. The combination and concurrent therapy groups experienced a significantly higher OS (p = 0.02 and p = 0.002, respectively) [55].
FLAURA2 is a phase 3 trial that is currently testing the combination of chemotherapy with osimertinib [56].
Table 3 summarises all clinical trials evaluating EGFR-TKI treatment combinations as first-line therapy.

2. Mechanisms of Resistance

Although all generations of EGFR-TKIs have been proven to be very effective for NSCLC with common EGFR mutations, almost 5–25% of these patients do not experience a clinical benefit with these drugs due to intrinsic resistance [57,58]. On the other hand, the major part of patients treated with EGFR-TKIs became resistant to these therapies despite an initial response or stable disease. The various mechanisms of resistance to EGFR-TKIs could be explained by the high molecular heterogeneity of NSCLCs (Figure 1 and Figure 2) [59]. Therefore, deepening the knowledge about the EGFR-TKI resistance mechanisms is one of the most-important aims in order to improve the treatment strategy of these patients.

2.1. Intrinsic Resistance

Patients with intrinsic resistance report an early tumour progression without prior tumour response; some of them respond for a very short period (<3 months) [60].
A possible cause regards the pharmacokinetics. In detail, treatment can fail due to the ineffective drug dose in the target area. This event can occur because of drug competition or the difficulty of first-/second-generation EGFR-TKIs to reach sanctuary localisations, such as the brain [61]. However, intrinsic resistance is often due to the lack of a target dependency (i.e., EGFR exon 20 mutations) or the genes alterations from other pathways (downstream or parallel pathways) [62,63]. Mechanisms of intrinsic resistance have been reported above all in patients with uncommon mutations and, more rarely, with common ones [62,63]. This type of resistance often depends on the presence of a drug-resistant EGFR mutation; the most-important ones are the exon 20 insertions (1–10% of all EGFR mutations) and the T790M EGFR mutation (approximately <1–65% of cases, based on the detection method employed) [64,65,66,67]. The lack of first-/second-generation EGFR-TKIs’ efficacy for the T790M mutation and exon 20 insertions is the reason for the development of third-generation EGFR-TKIs and EGFR-Ins20-specific inhibitors, respectively.

2.1.1. Intrinsic Resistance to First-/Second-Generation EGFR-TKIs

Exon 20 insertions correspond to the addition of residues at the N-lobe of EGFR (M766 to C775), while the C-helix (A767 to C775) is their preferential location [68]. This area is fundamental to regulate ATP and EGFR-TKI binding with the consequent activation of the kinase domain through a conformation change [68]. Commonly, exon 20 insertion mutations led to a reduced sensitivity to EGFR-TKIs; however, in vitro studies described that some insertion mutations, such as the insertion EGFR-A763_Y764insFQEA, confer high sensitivity to EGFR-TKIs [69,70]. These data have been confirmed by clinical trials in which NSCLC patients with some types of insertion mutations experienced prolonged periods of disease control under EGFR-TKI treatment [69].
The presence of the EGFR T790M mutation at diagnosis is a rare event, which suggests, in some cases, a germinal EGFR mutation [66,67,71]. It is associated with the worst response to first- or second-generation EGFR-TKIs and poor clinical outcomes [66,67,72]. The EGFR T790M mutation regards exon 20 and consists of a substitution of the threonine at position 790 with a methionine in the ATP-binding pocket of the kinase domain. This change prevents the binding of EGFR-TKIs to the receptor due to a steric hindrance; on the other hand, it leads to an in increased affinity between ATP and EGFR. Therefore, the receptor affinity for ATP becomes greater than that for the drug with a severe reduction in EGFR-TKI activity [73].
Another EGFR mutation that is responsible for intrinsic resistance to EGFR-TKIs in vitro is the variant III (vIII) in-frame deletion of exons 2–7 in the extracellular domain [74]. This mutation is present in almost 5% of human lung squamous cell carcinoma and determines the unsuccessful binding of EGF and other growth factors to EGFRvIII [75,76]. The reason behind the constitutive activation of EGFRvIII and the EGFR-TKI resistance is probably the structural changes of the EGFR protein affecting the ATP pocket and the intracellular domain conformation [74].
Some genetic alterations could occur in NSCLC patients with common EGFR mutations with the consequent reduction of sensitivity to EGFR-TKI therapy. In this regard, BIM is a proapoptotic member of the Bcl-2 family that plays a critical role in apoptosis mediated by EGFR-TKIs [77]. NSCLC patients with deletion polymorphisms or low-to-intermediate levels of BIM mRNA have poor clinical efficacy under EGFR-TKIs [77,78]. Furthermore, low levels of NF1 and the overexpression of RhoB are correlated with poor clinical efficacy [79,80]. Moreover, the plasma detection of TP53 gene co-mutations within two months of EGFR-TKI treatments is related to the worst PFS and OS [81]. CRIPTO1 is a member of the EGF-CFC family; it is a cell membrane protein linked to glycosylphosphatidylinositol. High basal levels of CRIPTO1 lead to a reduced EGFR-TKI sensitivity through the activation of ZEB1 and SRC. In this way, ZEB1 promotes epithelial-to-mesenchymal transition (EMT), while SCR stimulates AKT and MEK signalling [82].
The major part of oncogenic driver mutations in NSCLC is mutually exclusive, although some of them are present simultaneously, such as PI3KCA or TP53 mutations, with some other oncogenic driver mutations [83,84]. If, on the one hand, co-mutations of the PI3KCA and EGFR genes have no clinical impact [83], on the other hand, the co-mutation of EGFR Del19 and non-disruptive TP53 exon 8 is correlated with intrinsic resistance to first-generation EGFR-TKIs [84].
In some cases, pretreatment AXL and CDCP1 RNA overexpression coexist with EGFR mutations and correlate with poor response to first-generation EGFR-TKIs [85] and, likewise, co-alterations in some cell cycle genes or genes of the PI3K, MAPK, and Wnt/β-catenin pathways [16,60].

2.1.2. Intrinsic Resistance to Third-Generation EGFR-TKIs

Although most studies regard resistance to osimertinib during second-line therapy, some literature data report intrinsic resistance when it is administered as a first-line treatment [86]. In this regard, the transformation of NSCLC to SCLC has been considered a possible mechanism of intrinsic resistance [87].
The HER2 and MET genes’ amplification was associated in in vitro studies with reduced sensitivity to third-generation EGFR-TKIs such as osimertinib and rolecitinib [88,89]. The combination of the KRAS G12D mutation and PTEN loss was also detected in NSCLC patients with intrinsic resistance to osimertinib [60]. The worst response to third-generation EGFR-TKIs was reported also in NSCLC patients with the EGFR mutation and CDCP1 or AXL RNA overexpression at baseline [85].

2.2. Acquired Resistance

All the patients treated with EGFR-TKIs experience a progression of disease (PD) after a variable period of treatment. Patients usually develop acquired resistance after 9–12 months of treatment with first-/second-generation EGFR-TKIs, almost 10 months with second-line third-generation EGFR-TKIs [42], and about 19 months with first-line third-generation EGFR-TKIs [27].
Clinical criteria of acquired resistance to EGFR-TKIs in NSCLC patients have been proposed by Jackman et al., although further clinical validation is needed. The criteria by Gandara et al. are based on the type of PD: central nervous system (CNS), systemic, and oligo-progression. In the consideration of the undefined management of NSCLC patients who progressed to EGFR-TKIs because of acquired resistance, this classification could help clinicians establish the best treatment strategy based on the PD patterns [88]. For example, the same treatment with EGFR-TKI could be continued in patients with slow-PD and without clinical deterioration. A similar strategy could be applied for those patients with CNS PD or oligo-PD in association with local treatment to the site of progression (e.g., radiotherapy or surgery) [90].
The comprehension of the mechanisms leading to the acquired resistance is complex due to different aspects such as: (1) the type of EGFR-TKI; (2) the line of treatment with a specific EGFR-TKI; (3) the tumour biology, in particular histology, intrinsic mutability, microenvironment, and the type of initial EGFR mutation.
In the literature, there are several studies regarding the development of acquired resistance to the first-line treatment with first- or second-generation EGFR-TKIs or second-line treatment with third-generation EGFR-TKIs, usually due to the occurrence of the T790M mutation. In contrast, few data have been published about the acquired resistance to first-line osimertinib treatment. However, given the increasingly larger number of patients who will be treated with this drug, it is crucial to deepen the knowledge about the biological mechanisms of EGFR-TKI resistance.
Below, we describe the acquired resistance mechanisms to EGFR-TKIs known today. In detail, they can be classified into EGFR-dependent due to the insurgence of new EGFR mutations and EGFR-independent mechanisms due to the activation of alternative pathways.

2.2.1. EGFR-Dependent Mechanisms: Secondary EGFR Mutations

Acquired resistance based on EGFR-dependent mechanisms is due to the insurgence of secondary and tertiary mutations and/or amplifications of the EGFR gene with the consequent alteration of the receptor aminoacidic structure. Therefore, this leads to a conformational change that can regard the kinase or the ATP-binding pocket of the mutant EGFR, limiting drug accessibility or increasing the ATP affinity.
The incidence of EGFR-dependent acquired resistance is variable based on the type of EGFR-TKI administered and the line of treatment. To be specific, approximately 50% of patients develop this type of resistance under first-/second-generation EGFR-TKIs, 20% of them if they are treated with third-generation EGFR-TKI as a second-line therapy, and 10–15% with first-line third-generation EGFR-TKIs [91].

EGFR T790 Mutation

This is the most-frequent (49–63%) secondary mutation resulting in the insurgence of acquired resistance under treatment with first-/second-generation EGFR-TKIs [92]. Therefore, third-generation inhibitors were specifically designed to target the EGFR T790 mutation.
The EGFR T790M mutation regards exon 20 and consists of a substitution of the threonine at position 790 with a methionine in the ATP-binding pocket of the kinase domain. This change prevents the binding of EGFR-TKIs to the receptor due to a steric hindrance; on the other hand, it leads to an increased affinity between ATP and EGFR. Therefore, the receptor affinity for ATP becomes greater than that for the drug with a severe reduction in EGFR-TKI activity [93].
Some studies have hypothesised that this type of resistance might depend on the selection of pre-existing drug-resistant EGFR-T790M-positive clones during treatment with first-/second-generation EGFR-TKIs or on de novo acquisition of the EGFR-T790M mutation by initially drug-tolerant cells, negative for the EGFR T790M mutation [94]. Experimental data on gefitinib-resistant PC9 cells showed that the early EGFR T790M mutant clones derived from pre-existing EGFR T790M mutated cells were selected for gefitinib treatment. The other theory includes the late de novo occurrence of this type of mutation in drug-tolerant cells due to the prolonged exposure to a first-/second-generation EGFR inhibitor [64]. In an in vitro study, Hata et al. documented the restorations of late-emerging T790M cells’ sensitivity to third-generation EGFR-TKIs thanks to the treatment of tumour cells with navitoclax, an inhibitor of the antiapoptotic factors BCL-2 and BCL-xL [95].
Approximately 43% of NSCLC patients lose the EGFR T790M mutation with the PD [30,64,68]. This event suggests the existence of subclones with the EGFR T790M mutation [88]. Usually, the loss of T790M is associated with the presence of exon 19 deletion (83%) and, only rarely, with the L858R mutation (14%) [68]. Moreover, from a clinical point of view, this event at the time of progression is correlated with the worst clinical outcomes [23,96,97,98]. From a molecular point of view, it is associated with the loss of EGFR dependence and dependence on non-EGFR mechanisms [96].

Tertiary EGFR Mutations: Resistance to Second-Line Third-Generation EGFR-TKI

The AURA3 trial was the first study that showed the insurgence of acquired resistance to second-line osimertinib by means of the employment of cell-free DNA (cfDNA) genomic profiles [96,99]. To be specific, the results demonstrated that about 50% of the NSCLC patients maintained the EGFR T790M mutation, including those that experienced the insurgence of the tertiary EGFR mutation. The authors reported that acquired tertiary EGFR mutations occurred in 21% of cases, and the most-common one (15%) was the EGFR exon 20 C797S mutation [42,100,101]. In contrast, the FLAURA study, in which osimertinib was administered as a first-line therapy, reported a C797S mutation frequency of 7%.
The C797S mutation corresponds to a substitution in the ATP-binding site of a cysteine with a serine at codon 797, resulting in the inability of osimertinib to covalently bond with the mutant EGFR [102]. Moreover, some studies showed that this mutation also prevents the binding of other irreversible third-generation EGFR-TKIs such as olmutinib, rociletinib, and narzatinib to the EGFR active site [103,104]. Interestingly, the allelic context of the C797S mutation can predict the response to subsequent EGFR-TKI therapies. In detail, when NSCLC patients have the T790M and C797S mutations on the same allele (cis-mutations), they experience resistance to all available generations of EGFR-TKIs as a single agent or combined with other drugs [104,105]. On the other hand, when patients have these mutations on different alleles (trans-mutations), they experience sensitivity to first- and second-generation EGFR-TKIs. However, mutations in trans are rare, regarding less than 30% of cases [105,106].
Rare point EGFR mutations in exon 20 have been also identified in the C796 residue such as the G796R (0.56% of patients with lung adenocarcinoma treated with osimertinib), G796S, and G796D mutations, which are adjacent to C797 in exon 20 and can sterically impair the binding of osimertinib to EGFR [103,107,108,109].
L792 exon 20 mutations, including L792H, L792Y, and L792F, consist of the addition of a benzene or imidazole ring to the side chain of L792, resulting in the binding disruption of osimertinib to the EGFR kinase domain [108,110]. This mutation usually is located in cis with T790M, but less frequently, it can also occur in trans with EGFR C796/C797X mutations [105].
The L718Q, L718V, and L798I mutations in exon 18 affect the ATP-binding site of the EGFR kinase domain. Therefore, they determine steric restriction, preventing the binding of osimertinib [97,105,110]. These mutations are responsible for osimertinib resistance independent of the C797 mutation; in fact, they are not co-existent. L718Q/V are associated with sensitivity to first- and second-generation EGFR-TKIs, above all when T790M has been lost [111]. Osimertinib resistance is also caused by the G719A mutation located close to the L718 residue [105].
G724S is a very rare EGFR mutation located in exon 20, usually associated with EGFR exon 19 deletion. This mutation regards the P-loop of the kinase domain interfering with the binding of osimertinib [112,113,114]. However, this altered structure does not confer resistance to second-generation EGFR inhibitors [62].
SV768IL (S768I + V769L) is another rare (3%) mutation of EGFR exon 20 that has been identified in second-line therapy with osimertinib [101].
Rarely, tertiary EGFR mutations such as G724S, L718Q, V834L, and L718V can occur in patients that lost the T790M mutation [97].

Secondary EGFR Mutations: Resistance to First-Line Third-Generation EGFR-TKI

FLAURA was the first study that evaluated resistance to osimertinib as a first-line treatment [27,115]. Other literature data derive from some case reports or small case series [96,116].
This phase 3 trial analysed cfDNA samples through NGS, but no emergent T790M mutation has been detected. This discovery is in line with the well-known activity of osimertinib towards EGFR-sensitising and T790M mutations [115]. In this study, EGFR mutation/amplification was rare (9%), as well as C797S mutation frequency (7%), although it is the most-common mechanism after MET amplification (15%) [101]. S768I or combined EGFR mutation, such as Del19 + G724S (exon18), L718Q + EGFR ex20ins (exon 18 + 20), C797X, or S768I (exon 20), or L718Q + C797S, L718Q + L797S (exon 18 + 20) are very rare secondary mutations, each corresponding to about 1% of cases [11,12,115,117].
Interestingly, this study gave evidence that the mechanisms of resistance to first-line osimertinib depend on EGFR only for a small proportion of cases, and no EGFR T790M mutation was observed. Alternatively, EGFR-dependent mechanisms of acquired resistance are typical for those patients who receive osimertinib as a second-line therapy. Therefore, the T790M mutation will be less common due to the more-frequent use of osimertinib as the first-line therapy.

Rare EGFR Mutations

Although the underlying mechanisms are not well-defined yet, literature data described other rare EGFR point mutations that are responsible for acquired resistance to first-/second-generation EGFR-TKIs and regard less than 10% of NSCLC patients. They include D761Y and L747S (exon 19) or T854A (exon 21), Asp761Tyr, 39 Thr854Ala, and 40 Leu747Ser [71,110,118].
Other rare molecular alterations, such as the β-catenin mutation, have been detected in association with the EGFR T790M mutation [92].

2.2.2. EGFR-Independent Mechanisms: Alternative Pathways

Oncogene Amplification

The second-most-frequent mechanism of acquired resistance corresponds to the activation of alternative pathways. In this regard, MET oncogene amplification (a copy number of the MET gene ≥ 5 or a MET/CEP7 ratio of ≥2) is the most-important one and represents about 5–22% of cases, regardless of the EGFR-TKI generation or line of therapy [92,119,120]. MET is a tyrosine kinase receptor that is activated by the hepatocyte growth factor (HGF). This leads to the activation of the downstream AKT pathway, resulting in cell proliferation [121]. MET amplification results in receptor overexpression with a continuous ErbB3-AKT signalling, which is kept despite the blockade of EGFR [122].
MET amplification is usually seen concurrently with EGFR exon 19 deletion, and it has been detected with or without the loss of the T790M mutation [92,119]. Moreover, MET amplification was observed in association with the C797S EGFR mutation in 7% of cases in the AURA3 trial, but also with CDK6 or BRAF amplification [96]. In some cases, it was reported concurrently with the EGFR mutation before treatment [123].
Moreover, more than 20 oncogenic mutations of MET have been detected, and most of them are germline. However, MET mutations are rare (P97K/Q, I865F) in NSCLC with the most-common ones that affect the semaphorin domain (avoiding the growth factor binding), the juxtamembrane domain (altering the actin cytoskeleton of the cell), and the TK domain (with the consequent constitutive activation of the receptor, even in the absence of its ligand) [101,103,124,125,126].
The HER2 gene encodes the ErbB2 receptor tyrosine kinase [127]. It is responsible for EGFR-TKI resistance by means of alternative activation via the MAPK or PI3K pathway. Although ErbB2 amplification occurs only in 1% of untreated lung adenocarcinoma, it is responsible for 12% of cases of acquired resistance to first-generation TKIs and 5% to second-line treatment with osimertinib, mutually exclusive with the T790M mutation in both cases [128,129].
Furthermore, 2% of NSCLC patients have ErbB2 mutations at exon 20 encoding for the kinase domain of the receptor [128]. The ErbB2 receptor has a strong kinase activity, although a ligand-binding domain has not been identified; therefore, it is activated following the formation of heterodimers with the other members of EGFR family members [130]. On these bases, the wild-type state of ErbB2 is sensible to EGFR-TKIs in consideration of the EGFR-mediated activation of ErbB2. On the other hand, the mutated state of ErbB2 regarding the kinase domain makes it EGFR-independent and become responsible for resistance to EGFR-TKIs.

Rare Gene Mutations

Other rare gene mutations (<1%) have been reported.
KRAS G12S, G12D, G13D, Q61R, and Q61K were described as resistance mutations to second-line osimertinib [23,61,88,96,101].
An in vitro study documented NRAS mutations (and the novel E63K) in NSCLC EGFR-mutated cell lines resistant to first-, second-, and third-generation TKIs [131]. Other experimental data reported that the combination of osimertinib with selumintinib prevents EGFR-TKI resistance [132,133,134].
BRAF V600E was another resistance mutation to second-line treatment that has been identified in 3% of cases in cfDNA [96], concurrently or not, with the T790M mutation [98,135]. An in vitro study documented BRAF V600E NSCLC cell lines as resistant to osimertinib and sensible to the combination of osimertinib with encorafenib (BRAF inhibitor) [135].
PI3KCA mutations/amplifications and PTEN loss lead to an increased PI3K signaling [110]. They are usually present at baseline concurrently with some other driver mutations in NSCLC, although PI3KCA mutations or amplification were also reported at progression after gefitinib and erlotinib in 3–5% of cases [110]. On the other hand, some PI3KCA mutations such as E454K, E452K, R88Q, N345K, and E418K were identified at progression after second-line osimertinib in 4–11% of patients [23,136]. Moreover, an in vitro study showed the PI3KCA E545K mutation as involved in osimertinib resistance [23]. In the AURA3 trial, the co-existence of PI3KCA amplification and HER2 amplification was reported through NGS analysis of cfDNA [96]. PTEN loss was also identified as an acquired mechanism of resistance [137].
AXL gene upregulation with the consequent protein overexpression was described as another mechanism of acquired resistance to EGFR-TKIs [110,138].
Amplifications of genes regulating the cell cycle such as cyclin D1, cyclin D2, cyclin-dependent kinase N2A, cyclin E1, and CDK4/6 were reported in 12% of cfDNA samples after second-line third-generation EGFR-TKI treatment [96].

Gene Fusions

Gene fusions were reported after first-generation EGFR-TKIs very rarely and in 3–10% of patients after second-line osimertinib [139]. They were seen concurrently with the EGFR C797S mutation, MET amplification, or BRAF mutation [96]. The most-common gene fusions regard RET (46%), in particular RET-ERC1, RET-CCDC6, RET-RUFY2, and RET-NCOA4 [101,140]. Second are ALK fusions (26%) such as ALK-EML4 after second-line osimertinib [141], ALK-SPTBN1, and ALK-PLEKHA7 after first-line osimertinib [116,142]. Third are NTRK1 fusions (16%) including NTRK1-TPM3, which has been described in association with the EGFR Del19 and T790M mutations. Fourth are FGFR3 fusions (11%) such as FGFR3-TACC3, which was reported in combination with the EGFR Del19, C797X, and T790M mutations [96].
Other gene fusions have been described regarding ROS1 (ROS1-GOPC), MET (MET-SPECC1L), and BRAF (BRAF-ESYT2, BRAF-AGK, BRAF-BAIAP2L1, BRAF-PCBP2, BRAF-TRIM24, and BRAF-PJA2) [23,139,143,144].

Activation of Cell Receptors

The activation of the insulin-like growth factor 1 receptor (IGF-1R) seems to be related to EGFR-TKI resistance as shown in in vitro studies on lung cancer cell lines resistant to gefitinib or erlotinib [145]. A possible mechanism could depend on IGF-1R activation as a consequence of the heterodimerisation with EGFR after erlotinib treatment [145]. This event leads to the transmission of extracellular survival signals to downstream intra-cellular factors such as MAPK and AKT. On this basis, the combination of IGF-1R inhibitors such as AG1024, α-IR3, or R1507 with EGFR-TKIs might be a new strategy to overcome the resistance [146].
The activation of FGFR2 and 3 could also play a pivotal role in the drug resistance of cancer cells [147].

Phenotypic Transformation

In rare cases, a histological transformation from NSCLC (adenocarcinoma) to SCLC was observed [92]. In particular, this event was reported for some patients under EGFR-TKI treatment of first-, second-, and third-generations [101,106,148]. Therefore, it is considered a mechanism of acquired resistance.
A possible explanation of this phenomenon could be the existence of one shared multipotent stem cell for both adenocarcinoma and SCLC. On the other hand, other hypotheses suggest the development of SCLC through the expansion of minor pre-existent cells under the selection pressure of EGFR-TKIs or transdifferentiation from the adenocarcinoma cells [149].
A genomic study showed that transformed SCLC tumour samples retain the original EGFR-activating mutation detected at the baseline biopsy sample. This suggests that the transformed phenotype was not generated by de novo clones, but rather by cancer cells [92].
EMT plays a pivotal role in tumour transformation, invasion, and metastasis [150,151]. It is characterised by an important remodelling of the cell cytoskeleton due to the loss of polarity and cell–cell contacts by the epithelial cell layers. Therefore, these cells acquire a mesenchymal phenotype through the loss of E-cadherin and overexpression of mesenchymal proteins such as fibronectin, vimentin, and N-cadherin [152]. EMT is also characterised by AXL upregulation, which is considered a novel mechanism of acquired resistance to EGFR-TKI in NSCLCs. In this regard, pre-clinical studies documented that, on the one hand, the pharmacological inhibition of AXL reduced tumour cells’ proliferation and invasion and, on the other hand, increased cancers cells’ chemosensitivity [138].
The activation of the Hedgehog (Hh) pathway is implicated in tumourigenesis, metastatisation, and treatment resistance in various types of human tumours. Recent findings about EMT documented that gene amplification of SMO, a Hh receptor, is another mechanism of acquired resistance to first-generation EGFR-TKIs in EGFR-mutant NSCLC cells [153]. In particular, preclinical results showed that Hh-mediated EGFR-TKI resistance matched with the mesenchymal transformation of EGFR-mutated NSCLC cells [154]. In addition, these preclinical data were confirmed by the results of clinical studies on patients with EGFR-mutant NSCLC under treatment with EGFR-TKIs. In detail, the authors noticed the co-amplification of SMO and MET genes in tumour samples taken as the clinical evidence of EGFR-TKI resistance in 2 of the 16 patients.
Preclinical models of acquired resistance to EGFR-TKIs in EGFR-mutated NSCLC cells showed that the concomitant inhibition of both SMO and MET led to significant antiproliferative and proapoptotic effects, as well as the loss of the mesenchymal phenotype, suggesting new combination strategies [154].
These data confirmed that the upregulation of the Hh pathway results in EGFR-TKI resistance through EMT induction, while the inhibition of this signalling pathway increased EGFR-TKI sensitivity [154].

3. Future Perspectives

Cancer’s heterogeneity favours the occurrence of molecular resistance mechanisms, hence the difficulty in developing new effective treatment strategies. On the bases of the known resistance mechanisms, platinum-based chemotherapy, targeted therapies, and immunotherapy, or their combinations (Table 4), correspond to the potential therapeutic regimens for patients who progressed on EGFR-TKIs. The reassessment of tumour genetic alterations through tissue biopsy and/or liquid biopsy becomes of supreme importance to define the mechanisms of resistance and to guide the therapeutic strategy. Indeed, a specific treatment will be administered to patients if effective therapies currently exist. Conversely, preclinical studies and clinical trials will be considered to offer new therapeutic strategies. Figure 3 summarises the procedures after the occurrence of resistance.

3.1. Other Third-Generation EGFR-TKIs

In addition to osimertinib, other third-generation EGFR-TKIs have been evaluated in NSCLC patients with T790M and EGFR-activating mutations.
Almonertinib has an optimised structure compared to osimertinib. It has been tested in a phase 2 study on 244 NSCLC patients with EGFR T790M who experienced PD to the previous EGFR-TKI line. The results showed a DCR of 93.4% and an ORR of 68.9%, as well as a PFS of 12.3 months. Moreover, this drug provided an important encephalic response rate for those patients with CNS metastases: the DCR and ORR were 91.3% and 60.9%, respectively, while CNS PFS was 10.8 months [170].
Alflutinib inhibits both EGFR-sensitive mutations and acquired EGFR-T790M-positive NSCLC. This drug led to an ORR of 73.6% and mPFS of 7.6 months in a phase 2b in which patients with EGFR-T790M-mutated NSCLC were enrolled [171].
Lazertinib is another third-generation EGFR-TKI that showed an important systemic and intracranial activity on EGFR-T790M-positive NSCLC patients in a phase 1/2 study [172]. Currently, a phase 3 clinical trial is ongoing to compare lazertininb and gefitinib as a first-line therapy in NSCLC patients with EGFR-sensitising mutations.
Abivertinib is an irreversible third-generation EGFR-TKI that selectively targets EGFR-sensitising mutations and the T790M mutation through the formation of a covalent bond with C797 in the ATP-binding pocket. In a phase 2 clinical trial, it led to a DCR of 88.0% and an ORR of 52.2% [173,174].
Nazartinib is a covalent and irreversible EGFR-TKI targeting the exon 19 deletion, L858R, and T790M mutations. Preliminary results from a multicentre, open-label, phase 1/2 trial reported an ORR of 44% and a DCR of 91% with a median duration of response of 9.2 months and an excellent safety profile [175]. Ongoing clinical trials have been evaluating nazartinib combined with capmatinib in patients with advanced NSCLC with EGFR L858R, ex19del, or T790M in various lines of therapy including T790M-negative or treatment-naive patients (NCT02335944).
Olmutinib has potent inhibitory activity against L858R-/T790M-mutant NSCLC cells (44). A single-arm, open-label, phase 1/2 trial analysed olmutinib for EGFR-T790M-positive patients showing an ORR of 55% and a PFS of 6.9 months [176,177]. However, the safety profile of this drug is unfavourable relative to osimertinib, in particular due to skin toxicity including toxic epidermolysis necrosis and Stevens–Johnson syndrome.
Naquotinib is a small irreversible TKI that binds at C797 and targets the common EGFR mutations including T790M while sparing wild-type EGFR. This drug was tested in a phase 1 trial as a second-line therapy for T790M-positive patients, showing an ORR of 31% and a median PFS of 6.0 months [178]. On the other hand, the first-line naquotinib resulted in an ORR of 52%, DCR of 94%, and PFS of 11.3 months. However, the phase 3 clinical trial of naquotinib was closed due to limited predicted efficacy and toxicity [177].
Mavelertinib has a potent cellular inhibitory activity against EGFR L858R, the EGFR del19 mutation, EGFR L858R/T790M, and EGFR del19/T790M, while sparring WT EGFR. A phase 1 study tested this drug with an ORR of 42.3% and a DCR of 65,4% [176,179,180]. Further analyses will evaluate the efficacy and safety of mavelertininb plus palbociclib, according to preclinical data demonstrating synergy (NCT02349633) [179,180].

3.2. Therapeutic Options in T790M-cis-C797S Mutations

As mentioned above, if the C797S and T790M mutations are in trans, the cancer will be sensitive to the combination therapy of first- and third-generation EGFR-TKIs, while if they are in cis, EGFR-TKIs alone or in combination will be ineffective [111,181]. For the latter, alternative treatment strategies have been described. A preclinical study and a case report documented an interesting activity of brigatinib, a dual ALK and EGFR inhibitor, when administered in combination with cetuximab (anti-EGFR antibody), for NSCLC patients with T790M/cis-C797S EGFR mutations [182,183]. Moreover, a retrospective study showed that this combination led to an ORR of 60% and a median PFS of 14 months with respect to an ORR of 10% and a median PFS of 3 months for those patients treated with chemotherapy. Other combinations have been described in some case reports for patients with C797S and T790M mutations in cis with promising results [184,185]. The latter suggest that the combination of EGFR-TKIs with anti-VEGFR might be a promising therapy for this subset of patients. Further clinical trials should evaluate an ICI plus platinum-based doublet chemotherapy since it exhibited efficacy against T790M-cis-C797S [186]. The administration of osimertinib plus first-/second-generation EGFR-TKIs could be effective in cases of in trans C797X and T790M mutations, as well as the first-/second-generation EGFR-TKIs in cases with the C997X and without the T790M mutation [111,181].
However, currently, no standard therapeutic regimens are recommended for this set of patients.

3.3. Next-Generation EGFR Allosteric Inhibitors

Various therapeutic strategies have been studied to solve the issue of C797S-mediated resistance to osimertinib. In particular, several next-generation EGFR allosteric inhibitors have been developed [91,187,188,189]. They bind to EGFR at a site away from the tyrosine kinase domain, which bypasses the C797S-mediated resistance mechanism [190].
EAI045 selectively changes the space configuration of mutated EGFR and hinders its binding to EGFR ligands. In this way, it blocks the phosphorylation and the downstream signalling pathway, including p-STATs, p-AKT, and p-ERK1/2 [187]. In vitro and in vivo data reported a remarkable synergistic effect on Ba/F3 cells with triple mutants (L858R/ C797S/T790M) from EAI045 plus cetuximab [187,190].
Another next-generation EGFR allosteric inhibitor is JBJ-04-125-02, which exhibited, in vitro and in vivo, an increased apoptosis of NSCLC cells with EGFR triple mutants when combined with osimertinib than JBJ-04-125-02 or osimertinib as a single agent [188,191].
Unlike EAI045, CH7233163 proved to inhibit the growth of NSCLC cells with the Del19/T790M/C797S triple EGFR mutants [192].
BLU-701 is another fourth-generation EGFR-TKI against the C797S-resistance mutation that demonstrated antitumour activity in a PC9-cell-line-derived tumour xenograft model [193,194]. In addition, a phase 1/2 clinical study (SYMPHONY trial) is testing BLU-945 against the EGFR-sensitised mutation/T790M/C797S [195,196,197,198].
Despite these new drugs having demonstrated a potent activity on osimertinib-resistant NSCLC cells in preclinical studies, most of them have not been tested in clinical trials yet.

3.4. Osimertinib Plus MET Inhibitors

As mentioned above, MET amplification corresponds to the most-frequent EGFR-independent mechanism of osimertinib resistance (5–24%) [23,199,200]. Therefore, some MET-TKIs have been tested in MET-amplification NSCLC patients to overcome this type of resistance to osimertinib.
In a phase 1b trial (NCT02143466), savolitinib demonstrated an ORR of 33% and a median PFS of 5.4 months when administered with osimertinib [91,155]. The SACHI phase 3 trial is ongoing with the aim to compare this combination with pemetrexed plus platinum. Interestingly, the TATTON study was designed to analyse the safety of osimertinib plus selumetinib (MEK1/2 inhibitor), savolitinib (MET inhibitor), or durvalumab (anti-PD-L1 monoclonal antibody) in advanced EGFR-mutant NSCLC patients who progressed on a previous EGFR-TKI line. The interim results showed that osimertinib combined with savolitinib had acceptable toxicity and favourable antitumour activity [133,155]. Capmatinib, another MET inhibitor, showed an ORR of 27% in combination with gefitinib [91,156]. GEOMETRY-E is an ongoing phase 3 clinical trial testing the combination of capmatinib plus osimertinib compared to platinum–pemetrexed doublet chemotherapy as a second-line therapy for advanced NSCLC [201]. In the INSIGHT trial, tepotinib, a MET inhibitor, in association with gefitinib, documented an mPFS of 4.9 months versus the 4.4 months of the chemotherapy group (HR 0.67, 90% CI 0.35–1.28); mOS was 17.3 months in the combination group versus 18.7 months in the chemotherapy group (15.9–20.7; HR 0.69, 0.34–1.41) [91,157].
Amivantanab is a bispecific EGFR and MET antibody that is under evaluation in combination with lazertinib, another third-generation EGFR-TKI, in a phase 3 clinical study [158]. Some case reports and a retrospective analysis revealed important clinical and radiographic responses in EGFR-mutation-positive NSCLC patients treated with crizotinib plus osimertinib [202,203,204].

3.5. Osimertinib Plus CDK4/6 Inhibitors

Cyclin-dependent kinase (CDK) inhibitors, such as palbociclib and abemaciclib, act through the reduction of CDK-induced phosphorylation of downstream Rb [205,206,207]. Thus, this class of drug can inhibit the switch from the G1 to S phase of the cell cycle [205]. Experimental data reported as the combination of CDK4/6 inhibitors plus osimertinib led to an increased proportion of cells in the G1 phase and the block of the proliferation of osimertinib-resistant cells [205,206]. In addition, other studies revealed that CDK7 inhibitors might be effective to counter EMT-mediated resistance to osimertinib [208]. Two clinical trials are investigating the clinical efficacy of CDK4/6 inhibitors (abemaciclib or lerociclib) plus osimertinib in EGFR-mutation-positive metastatic NSCLC patients who progressed on osimertinib therapy in NCT04545710 and NCT03455829, respectively.

3.6. EGFR-TKIs Plus Anti-EGFR Antibodies

Preclinical data reported that the combination of the dual inhibition with an EGFR-TKI and anti-EGFR antibody was characterised by an elevated activity on EGFR L858R/T790M erlotinib-resistant tumours [209]. Phase 1b/2 clinical trials evaluated afatinib plus cetuximab, afatinib plus nimotuzumab, or osimertinib plus necitumumab for patients affected by EGFR-mutant NSCLC with acquired resistance to gefitinib or erlotinib, regardless of the EGFR T790M mutation [159,160,210,211]. These studies showed an important clinical activity and a safe profile of toxicity. Furthermore, another study investigated necitumumab, an anti-EGFR monoclonal antibody, in combination with chemotherapy versus chemotherapy alone as the first-line therapy. The results reported a longer OS (11.5 months vs. 9.9 months) for advanced squamous NSCLC patients in the experimental group [161]. Finally, several clinical trials (NCT03944772, NCT04285671, and NCT02496663) are testing the efficacy of necitumumab plus osimertinib in NSCLC with progression on EGFR-TKIs, including osimertinib.

3.7. Third-Generation EGFR-TKI Plus Bcl-2 Inhibitor

A phase 1b trial is ongoing with the aim to test osimertinib in combination with navitoclax in 27 patients with disease progression under an anti-EGFR-TKI [162]. Navitoclax is a dual-inhibitor of Bcl-2 and BCL-xL, which induces BIM (a proapoptotic protein from Bcl-2 family components) by decreasing the capacity of BCL-xL to neutralise BIM and facilitates Bax and Bak to initiate caspases, leading to cell death. In the expansion cohort, the ORR was 100% and the PFS was 16.8 months.

3.8. Chemotherapy

Despite several resistance mechanisms to osimertinib having been defined, 30–50% of them are still unknown. For the latter, no targeted therapy options exist; therefore, platinum-based chemotherapy is the best choice of treatment, especially in the case of SCLC transformation. In particular, a retrospective analysis reported an important activity of platinum–etoposide for SCLC-transformed patients (ORR of 54%, mPFS of 3.4 months, OS of 10.9 months). No efficacy was observed for those patients that underwent ICIs [212]. Another retrospective study showed that chemotherapy provides a better survival (25.0 versus 11.8 months) than a non-chemotherapy strategy in advanced NSCLC patients who progressed under first-line therapy with Osimertinib and without a molecular target [213].
However, it is controversial if the continuing administration of EGFR-TKIs during chemotherapy could provide a benefit. To this end, the IMPRESS phase 3 trial evaluated the efficacy and safety for patients who continued gefitinib in addition to cisplatin and pemetrexed after progression on first-line gefitinib treatment with respect to chemotherapy alone [163,214]. The experimental group experienced a similar PFS (5.4 vs. 5.4 months), but worse OS (13.4 vs. 19.5 months). On the other hand, the NEJ009 phase 3 trial showed that the combination of gefitinib plus carboplatin and pemetrexed led to a longer PFS (20.9 versus 11.9 months) and OS (49.0 vs. 38.5 months) than gefitinib as a single agent [164,215]. Therefore, the association therapy with gefitinib and chemotherapy provides opposite results in the different treatment settings. This is probably due to gene alterations in NSCLC patients after therapy with EGFR-TKIs. However, limited clinical data have been published about the efficacy of continuing osimertinib in addition to chemotherapy for those patients who previously received osimertinib as a first-line therapy. A randomised phase 2 clinical trial evaluated osimertinib plus carboplatin–pemetrexed compared to osimertinib alone for EGFR-mutated NSCLC patients with T790M in the second-line setting. The results reported a median PFS of 15.8 months versus 14.6 months and an ORR of 71.4% versus 53.6% in the control group and experimental group, respectively [165]. On the other hand, the combination of osimertinib with chemotherapy did not provide a survival benefit in the second-line setting, similar to gefitinib. Currently, some clinical studies are ongoing to further confirm these results. To be specific, the FLAME and COMPEL trials are enrolling EGFR-mutant NSCLC patient to compare osimertinib plus chemotherapy with osimertinib alone. Furthermore, the FLAURA2 phase 3 trial is evaluating first-line osimertinib plus platinum-based chemotherapy compared to osimertinib as a single agent [56].

3.9. Immunotherapy

Several phase 3 clinical trials reported very favourable results derived from the administration of ICIs for NSCLC patients without targetable mutations and many other cancers [216]. On the other hand, immunotherapy led to a limited benefit in NSCLC patients harbouring EGFR alterations [217,218]. In this regard, the IMMUNOTARGET registry revealed that immune monotherapy determined a low ORR and PFS among EGFR-mutated NSCLC patients, 12% and 2.1 months, respectively [219]. In addition, the single agent pembrolizumab provided limited efficacy in the subset of EGFR-mutated NSCLC patients who did not previously receive EGFR-TKI therapy, even in the case of PD-L1 expression of more than 50% [220]. A higher incidence of hyperprogressive disease (HPD) was observed with the administration of ICI monotherapy in NSCLC patients harbouring EGFR alterations [221,222]. On these bases, ICI monotherapy is not considered an appropriate therapeutic option for this subset of patients. The explanation for this lower sensitivity to anti-PD-1/-PD-L1 treatment has not been well defined yet, but it might be correlated with the low tumour mutational burden (TMB) of NSCLC with targetable mutations, with consequent weak immunogenicity [223].
In consideration of the limited benefit of ICI monotherapy in EGFR-mutated NSCLC patients, combination strategies have been evaluated. Recently, experimental data showed that PD-L1 expression and increased tumour-infiltrating lymphocyte density in the tumour microenvironment are correlated with the EGFR T790M mutation and response to the anti-PD1 antibody [224,225]. These results led to the evaluation of PD-1 blockade plus EGFR-TKI treatment for EGFR-driven NSCLC. In detail, the combinations of nivolumab and erlotinib and of durvalumab and osimertinib were associated with an important efficacy, but, at the same time, severe toxicity including interstitial pneumonitis, interstitial lung disease, and liver enzyme elevation [166,167,226,227,228,229,230,231,232]. Therefore, this type of combination did not demonstrate promising results due to the high incidence of Grade 3/4 adverse events (AEs). Interestingly, Schoenfeld et al. revealed that sequential ICI followed by osimertinib was associated with severe immune-associated AEs, unlike patients who received osimertinib followed by ICI [229]. Therefore, caution should be taken when osimertinib is administered to patients who recently received ICIs.
Recent studies are evaluating the combination of immunotherapy plus chemotherapy and antiangiogenetic agents for EGFR-mutant NSCLC patients intending to increase sensitivity to immunotherapy by modulating the tumour microenvironment [91]. In this regard, the IMpower150 phase 3 trial tested the combination of atezolizumab, carboplatin, paclitaxel, and bevacizumab with respect to the same chemotherapy plus bevacizumab, in non-squamous NSCLC patients, including those with EGFR mutations who could have received prior EGFR-TKIs. This study showed a significant improvement in OS and the PFS for the experimental group [168]. Similarly, promising results derived from another clinical trial in which the combination of atezolizumab, bevacizumab, carboplatin, and pemetrexed was evaluated for advanced EGFR-mutated NSCLC patients who progressed on an EGFR-TKI [233]. The ORIENT-31 is a phase 3 clinical trial that is evaluating sintilimab plus IBI305 (bevacizumab biosimilar), cisplatin, and pemetrexed compared to chemotherapy alone in EGFR-mutated NSCLC patients after EGFR-TKI failure. Interim results revealed a significantly longer PFS (6.9 months vs. 4.3 months) for the experimental group [169].
Other combination treatments are under investigation.

4. Conclusions

EGFR-TKIs are considered the gold standard treatment for patients affected by EGFR-mutated advanced NSCLC since these medicines have provided a remarkable improvement in clinical outcomes. Although osimertinib is the first-line therapy for these patients thanks to its potent activity and favourable safety profile, as well as early-generation EGFR-TKIs, resistance inevitably occurs due to various mechanisms. The reassessment of molecular alterations thorough tissue biopsy and/or liquid biopsy become of supreme importance to define the mechanisms of resistance and to guide therapeutic strategy. Indeed, targeted therapies, immunotherapy, and chemotherapy are providing interesting results for NSCLC patients harbouring EGFR mutations and with osimertinib resistance. Moreover, an increasing number of clinical trials is evaluating the efficacy and safety of several combination regimens.
Resistance to EGFR-TKIs hampers the correct management of EGFR-mutated NSCLC patients; therefore, the development of novel drugs to overcome this issue is fundamental. In addition, further studies are necessary to deepen knowledge about resistance mechanisms to osimertinib since a considerable proportion of them are still unknow. Indeed, in-depth knowledge of the resistance mechanisms would make it possible to individualise the treatment strategies, providing an improvement in the quality of life and a survival benefit of NSCLC patients.

Author Contributions

Conceptualisation, C.L. and P.F.; methodology, F.M.M.; software, M.L.I.; validation, A.N.S., M.P., and P.D.S.; formal analysis, C.G.; investigation, C.G.; resources, C.L. and F.A.; data curation, P.F. and M.L.; writing—original draft preparation, C.L.; writing—review and editing, P.F.; visualisation, F.M.M.; supervision, P.F.; project administration, C.L.; funding acquisition, A.N.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bade, B.C.; Dela Cruz, C.S. Lung Cancer 2020: Epidemiology, Etiology, and Prevention. Clin. Chest Med. 2020, 41, 1–24. [Google Scholar] [CrossRef]
  2. Schabath, M.B.; Cote, M.L. Cancer Progress and Priorities: Lung Cancer. Cancer Epidemiol. Biomark. Prev. 2019, 28, 1563–1579. [Google Scholar] [CrossRef] [PubMed]
  3. Chan, B.A.; Hughes, B.G. Targeted therapy for non-small cell lung cancer: Current standards and the promise of the future. Transl. Lung Cancer Res. 2015, 4, 36–54. [Google Scholar] [CrossRef] [PubMed]
  4. Leduc, C.; Merlio, J.P.; Besse, B.; Blons, H.; Debieuvre, D.; Bringuier, P.P.; Monnet, I.; Rouquette, I.; Fraboulet-Moreau, S.; Lemoine, A.; et al. Clinical and molecular characteristics of non-small-cell lung cancer (NSCLC) harboring EGFR mutation: Results of the nationwide French Cooperative Thoracic Intergroup (IFCT) program. Ann. Oncol. 2017, 28, 2715–2724. [Google Scholar] [CrossRef] [PubMed]
  5. Liu, X.; Wang, P.; Zhang, C.; Ma, Z. Epidermal growth factor receptor (EGFR): A rising star in the era of precision medicine of lung cancer. Oncotarget 2017, 8, 50209–50220. [Google Scholar] [CrossRef] [PubMed]
  6. Paez, J.G.; Jänne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR Mutations in Lung Cancer: Correlation with Clinical Response to Gefitinib Therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef]
  7. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating Mutations in the Epidermal Growth Factor Receptor Underlying Responsiveness of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef]
  8. Bílek, O.; Holánek, M.; Berkovcová, J.; Horký, O.; Kazda, T.; Čoupková, H.; Špelda, S.; Kristková, L.; Zvaríková, M.; Podhorec, J.; et al. Uncommon EGFR Mutations in Non-Small Cell Lung Cancer and Their Impact on the Treatment. Klin. Onkol. Cas. Ceske A Slov. Onkol. Spol. 2019, 32, 6–12. [Google Scholar] [CrossRef]
  9. Li, W.Q.; Cui, J.W. Non-small cell lung cancer patients with ex19del or exon 21 L858R mutation: Distinct mechanisms, different efficacies to treatments. J. Cancer Res. Clin. Oncol. 2020, 146, 2329–2338. [Google Scholar] [CrossRef]
  10. Russo, A.; Franchina, T.; Ricciardi, G.; Battaglia, A.; Picciotto, M.; Adamo, V. Heterogeneous Responses to Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors (TKIs) in Patients with Uncommon EGFR Mutations: New Insights and Future Perspectives in this Complex Clinical Scenario. Int. J. Mol. Sci. 2019, 20, 1431. [Google Scholar] [CrossRef]
  11. Beau-Faller, M.; Prim, N.; Ruppert, A.M.; Nanni-Metéllus, I.; Lacave, R.; Lacroix, L.; Escande, F.; Lizard, S.; Pretet, J.L.; Rouquette, I.; et al. Rare EGFR exon 18 and exon 20 mutations in non-small-cell lung cancer on 10 117 patients: A multicentre observational study by the French ERMETIC-IFCT network. Ann. Oncol. 2014, 25, 126–131. [Google Scholar] [CrossRef] [PubMed]
  12. Harrison, P.T.; Vyse, S.; Huang, P.H. Rare epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer. Semin. Cancer Biol. 2020, 61, 167–179. [Google Scholar] [CrossRef] [PubMed]
  13. Kobayashi, S.; Canepa, H.M.; Bailey, A.S.; Nakayama, S.; Yamaguchi, N.; Goldstein, M.A.; Huberman, M.S.; Costa, D.B. Compound EGFR mutations and response to EGFR tyrosine kinase inhibitors. J. Thorac. Oncol. 2013, 8, 45–51. [Google Scholar] [CrossRef]
  14. Lin, Y.T.; Tsai, T.H.; Wu, S.G.; Liu, Y.N.; Yu, C.J.; Shih, J.Y. Complex EGFR mutations with secondary T790M mutation confer shorter osimertinib progression-free survival and overall survival in advanced non-small cell lung cancer. Lung Cancer 2020, 145, 1–9. [Google Scholar] [CrossRef]
  15. Normanno, N.; De Luca, A.; Bianco, C.; Strizzi, L.; Mancino, M.; Maiello, M.R.; Carotenuto, A.; De Feo, G.; Caponigro, F.; Salomon, D.S. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene 2006, 366, 2–16. [Google Scholar] [CrossRef] [PubMed]
  16. Blakely, C.M.; Watkins, T.B.K.; Wu, W.; Gini, B.; Chabon, J.J.; McCoach, C.E.; McGranahan, N.; Wilson, G.A.; Birkbak, N.J.; Olivas, V.R.; et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat. Genet. 2017, 49, 1693–1704. [Google Scholar] [CrossRef]
  17. Blons, H.; Oudart, J.B.; Merlio, J.P.; Debieuvre, D.; de Fraipont, F.; Audigier-Valette, C.; Escande, F.; Hominal, S.; Bringuier, P.P.; Fraboulet-Moreau, S.; et al. PTEN, ATM, IDH1 mutations and MAPK pathway activation as modulators of PFS and OS in patients treated by first line EGFR TKI, an ancillary study of the French Cooperative Thoracic Intergroup (IFCT) Biomarkers France project. Lung Cancer 2021, 151, 69–75. [Google Scholar] [CrossRef]
  18. Canale, M.; Petracci, E.; Delmonte, A.; Bronte, G.; Chiadini, E.; Ludovini, V.; Dubini, A.; Papi, M.; Baglivo, S.; De Luigi, N.; et al. Concomitant TP53 Mutation Confers Worse Prognosis in EGFR-Mutated Non-Small Cell Lung Cancer Patients Treated with TKIs. J. Clin. Med. 2020, 9, 1047. [Google Scholar] [CrossRef]
  19. Skoulidis, F.; Heymach, J.V. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat. Rev. Cancer 2019, 19, 495–509. [Google Scholar] [CrossRef]
  20. Kohsaka, S.; Petronczki, M.; Solca, F.; Maemondo, M. Tumor clonality and resistance mechanisms in EGFR mutation-positive non-small-cell lung cancer: Implications for therapeutic sequencing. Future Oncol. 2019, 15, 637–652. [Google Scholar] [CrossRef]
  21. Oxnard, G.R.; Thress, K.S.; Alden, R.S.; Lawrance, R.; Paweletz, C.P.; Cantarini, M.; Yang, J.C.; Barrett, J.C.; Jänne, P.A. Association Between Plasma Genotyping and Outcomes of Treatment with Osimertinib (AZD9291) in Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2016, 34, 3375–3382. [Google Scholar] [CrossRef] [PubMed]
  22. Vaclova, T.; Grazini, U.; Ward, L.; O’Neill, D.; Markovets, A.; Huang, X.; Chmielecki, J.; Hartmaier, R.; Thress, K.S.; Smith, P.D.; et al. Clinical impact of subclonal EGFR T790M mutations in advanced-stage EGFR-mutant non-small-cell lung cancers. Nat. Commun. 2021, 12, 1780. [Google Scholar] [CrossRef] [PubMed]
  23. Oxnard, G.R.; Hu, Y.; Mileham, K.F.; Husain, H.; Costa, D.B.; Tracy, P.; Feeney, N.; Sholl, L.M.; Dahlberg, S.E.; Redig, A.J.; et al. Assessment of Resistance Mechanisms and Clinical Implications in Patients with EGFR T790M-Positive Lung Cancer and Acquired Resistance to Osimertinib. JAMA Oncol. 2018, 4, 1527–1534. [Google Scholar] [CrossRef]
  24. Beau-Faller, M.; Pencreach, E.; Leduc, C.; Blons, H.; Merlio, J.-P.; Bringuier, P.-P.; de Fraipont, F.; Escande, F.; Lemoine, A.; Ouafik, L.H.; et al. Independent prognostic value of ultra-sensitive quantification of tumor pre-treatment T790M subclones in EGFR mutated non-small cell lung cancer (NSCLC) treated by first/second generation TKI, depends on variant allele frequency (VAF): Results of the French cooperative thoracic intergroup (IFCT) biomarkers France project. Lung Cancer 2020, 140, 19–26. [Google Scholar] [CrossRef]
  25. Kobayashi, Y.; Mitsudomi, T. Not all epidermal growth factor receptor mutations in lung cancer are created equal: Perspectives for individualized treatment strategy. Cancer Sci. 2016, 107, 1179–1186. [Google Scholar] [CrossRef]
  26. Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
  27. 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]
  28. Inoue, A.; Kobayashi, K.; Maemondo, M.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin-paclitaxel for chemo-naïve non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002). Ann. Oncol. 2013, 24, 54–59. [Google Scholar] [CrossRef]
  29. Miyauchi, E.; Inoue, A.; Kobayashi, K.; Maemondo, M.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Saijo, Y.; Yoshizawa, H.; et al. Efficacy of chemotherapy after first-line gefitinib therapy in EGFR mutation-positive advanced non-small cell lung cancer-data from a randomized Phase III study comparing gefitinib with carboplatin plus paclitaxel (NEJ002). Jpn. J. Clin. Oncol. 2015, 45, 670–676. [Google Scholar] [CrossRef]
  30. Fukuoka, M.; Wu, Y.L.; Thongprasert, S.; Sunpaweravong, P.; Leong, S.S.; Sriuranpong, V.; Chao, T.Y.; Nakagawa, K.; Chu, D.T.; Saijo, N.; et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J. Clin. Oncol. 2011, 29, 2866–2874. [Google Scholar] [CrossRef]
  31. Mok, T.S.; Wu, Y.-L.; Thongprasert, S.; Yang, C.-H.; Chu, D.-T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or Carboplatin–Paclitaxel in Pulmonary Adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957. [Google Scholar] [CrossRef]
  32. Mitsudomi, T.; Morita, S.; Yatabe, Y.; Negoro, S.; Okamoto, I.; Tsurutani, J.; Seto, T.; Satouchi, M.; Tada, H.; Hirashima, T.; et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): An open label, randomised phase 3 trial. Lancet Oncol. 2010, 11, 121–128. [Google Scholar] [CrossRef]
  33. Yoshioka, H.; Shimokawa, M.; Seto, T.; Morita, S.; Yatabe, Y.; Okamoto, I.; Tsurutani, J.; Satouchi, M.; Hirashima, T.; Atagi, S.; et al. Final overall survival results of WJTOG3405, a randomized phase III trial comparing gefitinib versus cisplatin with docetaxel as the first-line treatment for patients with stage IIIB/IV or postoperative recurrent EGFR mutation-positive non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1978–1984. [Google Scholar] [CrossRef]
  34. Zhou, C.; Wu, Y.L.; Chen, G.; Feng, J.; Liu, X.Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): A multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011, 12, 735–742. [Google Scholar] [CrossRef]
  35. Zhou, C.; Wu, Y.L.; Chen, G.; Feng, J.; Liu, X.Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; et al. Final overall survival results from a randomised, phase III study of erlotinib versus chemotherapy as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer (OPTIMAL, CTONG-0802). Ann. Oncol. 2015, 26, 1877–1883. [Google Scholar] [CrossRef] [PubMed]
  36. Wu, Y.L.; Zhou, C.; Liam, C.K.; Wu, G.; Liu, X.; Zhong, Z.; Lu, S.; Cheng, Y.; Han, B.; Chen, L.; et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: Analyses from the phase III, randomized, open-label, ENSURE study. Ann. Oncol. 2015, 26, 1883–1889. [Google Scholar] [CrossRef] [PubMed]
  37. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012, 13, 239–246. [Google Scholar] [CrossRef] [PubMed]
  38. Shi, Y.K.; Wang, L.; Han, B.H.; Li, W.; Yu, P.; Liu, Y.P.; Ding, C.M.; Song, X.; Ma, Z.Y.; Ren, X.L.; et al. First-line icotinib versus cisplatin/pemetrexed plus pemetrexed maintenance therapy for patients with advanced EGFR mutation-positive lung adenocarcinoma (CONVINCE): A phase 3, open-label, randomized study. Ann. Oncol. 2017, 28, 2443–2450. [Google Scholar] [CrossRef]
  39. Yang, J.C.; Wu, Y.L.; Schuler, M.; Sebastian, M.; Popat, S.; Yamamoto, N.; Zhou, C.; Hu, C.P.; O’Byrne, K.; Feng, J.; et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): Analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015, 16, 141–151. [Google Scholar] [CrossRef]
  40. Wu, Y.L.; Zhou, C.; Hu, C.P.; Feng, J.; Lu, S.; Huang, Y.; Li, W.; Hou, M.; Shi, J.H.; Lee, K.Y.; et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): An open-label, randomised phase 3 trial. Lancet Oncol. 2014, 15, 213–222. [Google Scholar] [CrossRef]
  41. Mok, T.S.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Chawla, A.; Rosell, R.; Corral, J.; Migliorino, M.R.; et al. Updated Overall Survival in a Randomized Study Comparing Dacomitinib with Gefitinib as First-Line Treatment in Patients with Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. Drugs 2021, 81, 257–266. [Google Scholar] [CrossRef] [PubMed]
  42. Mok, T.S.; Wu, Y.-L.; Ahn, M.-J.; Garassino, M.C.; Kim, H.R.; Ramalingam, S.S.; Shepherd, F.A.; He, Y.; Akamatsu, H.; Theelen, W.S.M.E.; et al. Osimertinib or Platinum–Pemetrexed in EGFR T790M–Positive Lung Cancer. N. Engl. J. Med. 2017, 376, 629–640. [Google Scholar] [CrossRef]
  43. Wu, Y.-L.; Tsuboi, M.; He, J.; John, T.; Grohe, C.; Majem, M.; Goldman, J.W.; Laktionov, K.; Kim, S.-W.; Kato, T.; et al. Osimertinib in Resected EGFR-Mutated Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 1711–1723. [Google Scholar] [CrossRef] [PubMed]
  44. Le, X.; Cornelissen, R.; Garassino, M.; Clarke, J.M.; Tchekmedyian, N.; Goldman, J.W.; Leu, S.Y.; Bhat, G.; Lebel, F.; Heymach, J.V.; et al. Poziotinib in Non-Small-Cell Lung Cancer Harboring HER2 Exon 20 Insertion Mutations after Prior Therapies: ZENITH20-2 Trial. J. Clin. Oncol. 2022, 40, 710–718. [Google Scholar] [CrossRef] [PubMed]
  45. Zhou, C.; Ramalingam, S.S.; Kim, T.M.; Kim, S.W.; Yang, J.C.; Riely, G.J.; Mekhail, T.; Nguyen, D.; Garcia Campelo, M.R.; Felip, E.; et al. Treatment Outcomes and Safety of Mobocertinib in Platinum-Pretreated Patients with EGFR Exon 20 Insertion-Positive Metastatic Non-Small Cell Lung Cancer: A Phase 1/2 Open-label Nonrandomized Clinical Trial. JAMA Oncol. 2021, 7, e214761. [Google Scholar] [CrossRef]
  46. 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]
  47. Cortot, A.B.; Madroszyk, A.; Giroux-Leprieur, E.; Molinier, O.; Quoix, E.; Bérard, H.; Otto, J.; Rault, I.; Moro-Sibilot, D.; Raimbourg, J.; et al. First-Line Afatinib plus Cetuximab for EGFR-Mutant Non-Small Cell Lung Cancer: Results from the Randomized Phase II IFCT-1503 ACE-Lung Study. Clin. Cancer Res. 2021, 27, 4168–4176. [Google Scholar] [CrossRef]
  48. Laface, C.; Fedele, P.; Maselli, F.M.; Ambrogio, F.; Foti, C.; Molinari, P.; Ammendola, M.; Lioce, M.; Ranieri, G. Targeted Therapy for Hepatocellular Carcinoma: Old and New Opportunities. Cancers 2022, 14, 4028. [Google Scholar] [CrossRef]
  49. Ranieri, G.; Laface, C.; Laforgia, M.; De Summa, S.; Porcelli, M.; Macina, F.; Ammendola, M.; Molinari, P.; Lauletta, G.; Di Palo, A.; et al. Bevacizumab Plus FOLFOX-4 Combined with Deep Electro-Hyperthermia as First-line Therapy in Metastatic Colon Cancer: A Pilot Study. Front. Oncol. 2020, 10, 590707. [Google Scholar] [CrossRef]
  50. Laface, C.; Laforgia, M.; Zito, A.F.; Loisi, D.; Zizzo, N.; Tamma, R.; Gadaleta, C.D.; Porcelli, M.; Currò, G.; Ammendola, M.; et al. Chymase-positive Mast cells correlate with tumor angiogenesis: First report in pancreatic cancer patients. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 6862–6873. [Google Scholar] [CrossRef]
  51. Laforgia, M.; Calabrò, C.; Scattone, A.; Laface, C.; Porcelli, M.; Gadaleta, C.D.; Nardulli, P.; Ranieri, G. Pharmacotherapy in Mast Cell Leukemia. Expert Opin. Pharmacother. 2020, 21, 1059–1069. [Google Scholar] [CrossRef]
  52. Rosell, R.; Dafni, U.; Felip, E.; Curioni-Fontecedro, A.; Gautschi, O.; Peters, S.; Massutí, B.; Palmero, R.; Aix, S.P.; Carcereny, E.; et al. Erlotinib and bevacizumab in patients with advanced non-small-cell lung cancer and activating EGFR mutations (BELIEF): An international, multicentre, single-arm, phase 2 trial. Lancet Respir. Med. 2017, 5, 435–444. [Google Scholar] [CrossRef] [PubMed]
  53. Yamamoto, N.; Seto, T.; Nishio, M.; Goto, K.; Yamamoto, N.; Okamoto, I.; Yamanaka, T.; Tanaka, M.; Takahashi, K.; Fukuoka, M. Erlotinib plus bevacizumab vs erlotinib monotherapy as first-line treatment for advanced EGFR mutation-positive non-squamous non-small-cell lung cancer: Survival follow-up results of the randomized JO25567 study. Lung Cancer 2021, 151, 20–24. [Google Scholar] [CrossRef]
  54. Nakagawa, K.; Garon, E.B.; Seto, T.; Nishio, M.; Ponce Aix, S.; 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]
  55. Wu, Q.; Luo, W.; Li, W.; Wang, T.; Huang, L.; Xu, F. First-Generation EGFR-TKI Plus Chemotherapy Versus EGFR-TKI Alone as First-Line Treatment in Advanced NSCLC with EGFR Activating Mutation: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front. Oncol. 2021, 11, 598265. [Google Scholar] [CrossRef] [PubMed]
  56. Planchard, D.; Feng, P.H.; Karaseva, N.; Kim, S.W.; Kim, T.M.; Lee, C.K.; Poltoratskiy, A.; Yanagitani, N.; Marshall, R.; Huang, X.; et al. Osimertinib plus platinum-pemetrexed in newly diagnosed epidermal growth factor receptor mutation-positive advanced/metastatic non-small-cell lung cancer: Safety run-in results from the FLAURA2 study. ESMO Open 2021, 6, 100271. [Google Scholar] [CrossRef] [PubMed]
  57. 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]
  58. Voldborg, B.R.; Damstrup, L.; Spang-Thomsen, M.; Poulsen, H.S. Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann. Oncol. 1997, 8, 1197–1206. [Google Scholar] [CrossRef]
  59. Dong, R.F.; Zhu, M.L.; Liu, M.M.; Xu, Y.T.; Yuan, L.L.; Bian, J.; Xia, Y.Z.; Kong, L.Y. EGFR mutation mediates resistance to EGFR tyrosine kinase inhibitors in NSCLC: From molecular mechanisms to clinical research. Pharmacol. Res. 2021, 167, 105583. [Google Scholar] [CrossRef]
  60. Santoni-Rugiu, E.; Melchior, L.C.; Urbanska, E.M.; Jakobsen, J.N.; Stricker, K.; Grauslund, M.; Sørensen, J.B. Intrinsic resistance to EGFR-Tyrosine Kinase Inhibitors in EGFR-Mutant Non-Small Cell Lung Cancer: Differences and Similarities with Acquired Resistance. Cancers 2019, 11, 923. [Google Scholar] [CrossRef]
  61. Yu, H.A.; Arcila, M.E.; Rekhtman, N.; Sima, C.S.; Zakowski, M.F.; Pao, W.; Kris, M.G.; Miller, V.A.; Ladanyi, M.; Riely, G.J. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin. Cancer Res. 2013, 19, 2240–2247. [Google Scholar] [CrossRef] [PubMed]
  62. Jin, Y.; Shi, X.; Zhao, J.; He, Q.; Chen, M.; Yan, J.; Ou, Q.; Wu, X.; Shao, Y.W.; Yu, X. Mechanisms of primary resistance to EGFR targeted therapy in advanced lung adenocarcinomas. Lung Cancer 2018, 124, 110–116. [Google Scholar] [CrossRef] [PubMed]
  63. Morgillo, F.; Bareschino, M.A.; Bianco, R.; Tortora, G.; Ciardiello, F. Primary and acquired resistance to anti-EGFR targeted drugs in cancer therapy. Differ. Res. Biol. Divers. 2007, 75, 788–799. [Google Scholar] [CrossRef] [PubMed]
  64. Morgillo, F.; Della Corte, C.M.; Fasano, M.; Ciardiello, F. Mechanisms of resistance to EGFR-targeted drugs: Lung cancer. ESMO Open 2016, 1, e000060. [Google Scholar] [CrossRef]
  65. Reita, D.; Pabst, L.; Pencreach, E.; Guérin, E.; Dano, L.; Rimelen, V.; Voegeli, A.-C.; Vallat, L.; Mascaux, C.; Beau-Faller, M. Molecular Mechanism of EGFR-TKI Resistance in EGFR-Mutated Non-Small Cell Lung Cancer: Application to Biological Diagnostic and Monitoring. Cancers 2021, 13, 4926. [Google Scholar] [CrossRef]
  66. Attili, I.; Karachaliou, N.; Conte, P.; Bonanno, L.; Rosell, R. Therapeutic approaches for T790M mutation positive non-small-cell lung cancer. Expert Rev. Anticancer. Ther. 2018, 18, 1021–1030. [Google Scholar] [CrossRef]
  67. Chen, L.Y.; Molina-Vila, M.A.; Ruan, S.Y.; Su, K.Y.; Liao, W.Y.; Yu, K.L.; Ho, C.C.; Shih, J.Y.; Yu, C.J.; Yang, J.C.; 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 2016, 94, 46–53. [Google Scholar] [CrossRef]
  68. Eck, M.J.; Yun, C.-H. Structural and mechanistic underpinnings of the differential drug sensitivity of EGFR mutations in non-small cell lung cancer. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2010, 1804, 559–566. [Google Scholar] [CrossRef]
  69. De Pas, T.; Toffalorio, F.; Manzotti, M.; Fumagalli, C.; Spitaleri, G.; Catania, C.; Delmonte, A.; Giovannini, M.; Spaggiari, L.; de Braud, F.; et al. Activity of Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors in Patients with Non-small Cell Lung Cancer Harboring Rare Epidermal Growth Factor Receptor Mutations. J. Thorac. Oncol. 2011, 6, 1895–1901. [Google Scholar] [CrossRef]
  70. Costa, D.B. More Than Just an Oncogene Translocation and a Kinase Inhibitor: Kevin’s Story. J. Clin. Oncol. 2011, 30, 110–112. [Google Scholar] [CrossRef]
  71. Pao, W.; Girard, N. New driver mutations in non-small-cell lung cancer. Lancet Oncol. 2011, 12, 175–180. [Google Scholar] [CrossRef] [PubMed]
  72. Costa, C.; Molina, M.A.; Drozdowskyj, A.; Giménez-Capitán, A.; Bertran-Alamillo, J.; Karachaliou, N.; Gervais, R.; Massuti, B.; Wei, J.; Moran, T.; et al. The Impact of EGFR T790M Mutations and BIM mRNA Expression on Outcome in Patients with EGFR-Mutant NSCLC Treated with Erlotinib or Chemotherapy in the Randomized Phase III EURTAC Trial. Clin. Cancer Res. 2014, 20, 2001–2010. [Google Scholar] [CrossRef]
  73. Soejima, K.; Yasuda, H.; Hirano, T. Osimertinib for EGFR T790M mutation-positive non-small cell lung cancer. Expert Rev. Clin. Pharmacol. 2017, 10, 31–38. [Google Scholar] [CrossRef]
  74. Ji, H.; Zhao, X.; Yuza, Y.; Shimamura, T.; Li, D.; Protopopov, A.; Jung, B.L.; McNamara, K.; Xia, H.; Glatt, K.A.; et al. Epidermal growth factor receptor variant III mutations in lung tumorigenesis and sensitivity to tyrosine kinase inhibitors. Proc. Natl. Acad. Sci. USA 2006, 103, 7817–7822. [Google Scholar] [CrossRef] [PubMed]
  75. Okamoto, I.; Kenyon, L.C.; Emlet, D.R.; Mori, T.; Sasaki, J.; Hirosako, S.; Ichikawa, Y.; Kishi, H.; Godwin, A.K.; Yoshioka, M.; et al. Expression of constitutively activated EGFRvIII in non-small cell lung cancer. Cancer Sci. 2003, 94, 50–56. [Google Scholar] [CrossRef]
  76. Sasaki, H.; Kawano, O.; Endo, K.; Yukiue, H.; Yano, M.; Fujii, Y. EGFRvIII mutation in lung cancer correlates with increased EGFR copy number. Oncol. Rep. 2007, 17, 319–323. [Google Scholar] [CrossRef] [PubMed]
  77. Faber, A.C.; Corcoran, R.B.; Ebi, H.; Sequist, L.V.; Waltman, B.A.; Chung, E.; Incio, J.; Digumarthy, S.R.; Pollack, S.F.; Song, Y.; et al. BIM Expression in Treatment-Naïve Cancers Predicts Responsiveness to Kinase Inhibitors. Cancer Discov. 2011, 1, 352–365. [Google Scholar] [CrossRef] [PubMed]
  78. Murakami, H.; Nokihara, H.; Shimizu, T.; Seto, T.; Keating, A.; Krivoshik, A.; Uegaki, K.; Morita, S.; Nakagawa, K.; Fukuoka, M. 9LBA Antitumor activity of ASP8273, an irreversible mutant selective EGFR-TKI, in NSCLC patients with tumors harboring EGFR activating mutations and T790M resistance mutation. Eur. J. Cancer 2014, 50, 198. [Google Scholar] [CrossRef]
  79. de Bruin, E.C.; Cowell, C.; Warne, P.H.; Jiang, M.; Saunders, R.E.; Melnick, M.A.; Gettinger, S.; Walther, Z.; Wurtz, A.; Heynen, G.J.; et al. Reduced NF1 expression confers resistance to EGFR inhibition in lung cancer. Cancer Discov. 2014, 4, 606–619. [Google Scholar] [CrossRef] [PubMed]
  80. Calvayrac, O.; Mazières, J.; Figarol, S.; Marty-Detraves, C.; Raymond-Letron, I.; Bousquet, E.; Farella, M.; Clermont-Taranchon, E.; Milia, J.; Rouquette, I.; et al. The RAS-related GTPase RHOB confers resistance to EGFR-tyrosine kinase inhibitors in non-small-cell lung cancer via an AKT-dependent mechanism. EMBO Mol. Med. 2017, 9, 238–250. [Google Scholar] [CrossRef]
  81. Tsui, D.W.Y.; Murtaza, M.; Wong, A.S.C.; Rueda, O.M.; Smith, C.G.; Chandrananda, D.; Soo, R.A.; Lim, H.L.; Goh, B.C.; Caldas, C.; et al. Dynamics of multiple resistance mechanisms in plasma DNA during EGFR-targeted therapies in non-small cell lung cancer. EMBO Mol. Med. 2018, 10, e7945. [Google Scholar] [CrossRef]
  82. Park, K.-S.; Raffeld, M.; Moon, Y.W.; Xi, L.; Bianco, C.; Pham, T.; Lee, L.C.; Mitsudomi, T.; Yatabe, Y.; Okamoto, I.; et al. CRIPTO1 expression in EGFR-mutant NSCLC elicits intrinsic EGFR-inhibitor resistance. J. Clin. Investig. 2014, 124, 3003–3015. [Google Scholar] [CrossRef] [PubMed]
  83. Eng, J.; Woo, K.M.; Sima, C.S.; Plodkowski, A.; Hellmann, M.D.; Chaft, J.E.; Kris, M.G.; Arcila, M.E.; Ladanyi, M.; Drilon, A. Impact of Concurrent PIK3CA Mutations on Response to EGFR Tyrosine Kinase Inhibition in EGFR-Mutant Lung Cancers and on Prognosis in Oncogene-Driven Lung Adenocarcinomas. J. Thorac. Oncol. 2015, 10, 1713–1719. [Google Scholar] [CrossRef] [PubMed]
  84. Canale, M.; Petracci, E.; Delmonte, A.; Chiadini, E.; Dazzi, C.; Papi, M.; Capelli, L.; Casanova, C.; De Luigi, N.; Mariotti, M.; et al. Impact of TP53 Mutations on Outcome in EGFR-Mutated Patients Treated with First-Line Tyrosine Kinase Inhibitors. Clin. Cancer Res. 2017, 23, 2195–2202. [Google Scholar] [CrossRef]
  85. Karachaliou, N.; Chaib, I.; Cardona, A.F.; Berenguer, J.; Bracht, J.W.P.; Yang, J.; Cai, X.; Wang, Z.; Hu, C.; Drozdowskyj, A.; et al. Common Co-activation of AXL and CDCP1 in EGFR-mutation-positive Non-smallcell Lung Cancer Associated with Poor Prognosis. EBioMedicine 2018, 29, 112–127. [Google Scholar] [CrossRef] [PubMed]
  86. Michels, S.; Heydt, C.; van Veggel, B.; Deschler-Baier, B.; Pardo, N.; Monkhorst, K.; Rüsseler, V.; Stratmann, J.; Griesinger, F.; Steinhauser, S.; et al. Genomic Profiling Identifies Outcome-Relevant Mechanisms of Innate and Acquired Resistance to Third-Generation Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Therapy in Lung Cancer. JCO Precis. Oncol. 2019, 3, PO.18.00210. [Google Scholar] [CrossRef]
  87. Minari, R.; Bordi, P.; Del Re, M.; Facchinetti, F.; Mazzoni, F.; Barbieri, F.; Camerini, A.; Comin, C.E.; Gnetti, L.; Azzoni, C.; et al. Primary resistance to osimertinib due to SCLC transformation: Issue of T790M determination on liquid re-biopsy. Lung Cancer 2018, 115, 21–27. [Google Scholar] [CrossRef]
  88. Jackman, D.; Pao, W.; Riely, G.J.; Engelman, J.A.; Kris, M.G.; Jänne, P.A.; Lynch, T.; Johnson, B.E.; Miller, V.A. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J. Clin. Oncol. 2010, 28, 357–360. [Google Scholar] [CrossRef]
  89. Ortiz-Cuaran, S.; Scheffler, M.; Plenker, D.; Dahmen, I.; Scheel, A.H.; Fernandez-Cuesta, L.; Meder, L.; Lovly, C.M.; Persigehl, T.; Merkelbach-Bruse, S.; et al. Heterogeneous Mechanisms of Primary and Acquired Resistance to Third-Generation EGFR Inhibitors. Clin. Cancer Res. 2016, 22, 4837–4847. [Google Scholar] [CrossRef]
  90. Gandara, D.R.; Li, T.; Lara, P.N.; Kelly, K.; Riess, J.W.; Redman, M.W.; Mack, P.C. Acquired Resistance to Targeted Therapies Against Oncogene-Driven Non–Small-Cell Lung Cancer: Approach to Subtyping Progressive Disease and Clinical Implications. Clin. Lung Cancer 2014, 15, 1–6. [Google Scholar] [CrossRef]
  91. Passaro, A.; Jänne, P.A.; Mok, T.; Peters, S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat. Cancer 2021, 2, 377–391. [Google Scholar] [CrossRef] [PubMed]
  92. Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl. Med. 2011, 3, 75ra26. [Google Scholar] [CrossRef]
  93. Yun, C.-H.; Mengwasser, K.E.; Toms, A.V.; Woo, M.S.; Greulich, H.; Wong, K.-K.; Meyerson, M.; Eck, M.J. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl. Acad. Sci. USA 2008, 105, 2070–2075. [Google Scholar] [CrossRef]
  94. Oxnard, G.R.; Arcila, M.E.; Sima, C.S.; Riely, G.J.; Chmielecki, J.; Kris, M.G.; Pao, W.; Ladanyi, M.; Miller, V.A. Acquired Resistance to EGFR Tyrosine Kinase Inhibitors in EGFR-Mutant Lung Cancer: Distinct Natural History of Patients with Tumors Harboring the T790M Mutation. Clin. Cancer Res. 2011, 17, 1616–1622. [Google Scholar] [CrossRef] [PubMed]
  95. Hata, A.N.; Niederst, M.J.; Archibald, H.L.; Gomez-Caraballo, M.; Siddiqui, F.M.; Mulvey, H.E.; Maruvka, Y.E.; Ji, F.; Bhang, H.-e.C.; Krishnamurthy Radhakrishna, V.; et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 2016, 22, 262–269. [Google Scholar] [CrossRef] [PubMed]
  96. Chmielecki, J.; Mok, T.; Wu, Y.L.; Han, J.Y.; Ahn, M.J.; Ramalingam, S.S.; John, T.; Okamoto, I.; Yang, J.C.; Shepherd, F.A.; et al. Analysis of acquired resistance mechanisms to osimertinib in patients with EGFR-mutated advanced non-small cell lung cancer from the AURA3 trial. Nat. Commun. 2023, 14, 1071. [Google Scholar] [CrossRef]
  97. Fang, W.; Gan, J.; Huang, Y.; Zhou, H.; Zhang, L. Acquired EGFR L718V Mutation and Loss of T790M-Mediated Resistance to Osimertinib in a Patient with NSCLC Who Responded to Afatinib. J. Thorac. Oncol. 2019, 14, e274–e275. [Google Scholar] [CrossRef]
  98. Lin, C.C.; Shih, J.Y.; Yu, C.J.; Ho, C.C.; Liao, W.Y.; Lee, J.H.; Tsai, T.H.; Su, K.Y.; Hsieh, M.S.; Chang, Y.L.; et al. Outcomes in patients with non-small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: A genomic study. Lancet Respir. Med. 2018, 6, 107–116. [Google Scholar] [CrossRef]
  99. Papadimitrakopoulou, V.A.; Han, J.Y.; Ahn, M.J.; Ramalingam, S.S.; Delmonte, A.; Hsia, T.C.; Laskin, J.; Kim, S.W.; He, Y.; Tsai, C.M.; et al. Epidermal growth factor receptor mutation analysis in tissue and plasma from the AURA3 trial: Osimertinib versus platinum-pemetrexed for T790M mutation-positive advanced non-small cell lung cancer. Cancer 2020, 126, 373–380. [Google Scholar] [CrossRef]
  100. Lu, X.; Yu, L.; Zhang, Z.; Ren, X.; Smaill, J.B.; Ding, K. Targeting EGFR(L858R/T790M) and EGFR(L858R/T790M/C797S) resistance mutations in NSCLC: Current developments in medicinal chemistry. Med. Res. Rev. 2018, 38, 1550–1581. [Google Scholar] [CrossRef]
  101. Schoenfeld, A.J.; Chan, J.M.; Kubota, D.; Sato, H.; Rizvi, H.; Daneshbod, Y.; Chang, J.C.; Paik, P.K.; Offin, M.; Arcila, M.E.; et al. Tumor Analyses Reveal Squamous Transformation and Off-Target Alterations As Early Resistance Mechanisms to First-line Osimertinib in EGFR-Mutant Lung Cancer. Clin. Cancer Res. 2020, 26, 2654–2663. [Google Scholar] [CrossRef] [PubMed]
  102. Zhou, W.; Ercan, D.; Chen, L.; Yun, C.-H.; Li, D.; Capelletti, M.; Cortot, A.B.; Chirieac, L.; Iacob, R.E.; Padera, R.; et al. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature 2009, 462, 1070–1074. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, Z.; Yang, N.; Ou, Q.; Xiang, Y.; Jiang, T.; Wu, X.; Bao, H.; Tong, X.; Wang, X.; Shao, Y.W.; et al. Investigating Novel Resistance Mechanisms to Third-Generation EGFR Tyrosine Kinase Inhibitor Osimertinib in Non-Small Cell Lung Cancer Patients. Clin. Cancer Res. 2018, 24, 3097–3107. [Google Scholar] [CrossRef] [PubMed]
  104. 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]
  105. Arulananda, S.; Do, H.; Musafer, A.; Mitchell, P.; Dobrovic, A.; John, T. Combination Osimertinib and Gefitinib in C797S and T790M EGFR-Mutated Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 1728–1732. [Google Scholar] [CrossRef]
  106. 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]
  107. Li, Y.; Mao, T.; Wang, J.; Zheng, H.; Hu, Z.; Cao, P.; Yang, S.; Zhu, L.; Guo, S.; Zhao, X.; et al. Toward the next generation EGFR inhibitors: An overview of osimertinib resistance mediated by EGFR mutations in non-small cell lung cancer. Cell Commun. Signal. 2023, 21, 71. [Google Scholar] [CrossRef]
  108. Ou, S.I.; Cui, J.; Schrock, A.B.; Goldberg, M.E.; Zhu, V.W.; Albacker, L.; Stephens, P.J.; Miller, V.A.; Ali, S.M. Emergence of novel and dominant acquired EGFR solvent-front mutations at Gly796 (G796S/R) together with C797S/R and L792F/H mutations in one EGFR (L858R/T790M) NSCLC patient who progressed on osimertinib. Lung Cancer 2017, 108, 228–231. [Google Scholar] [CrossRef]
  109. Zheng, D.; Hu, M.; Bai, Y.; Zhu, X.; Lu, X.; Wu, C.; Wang, J.; Liu, L.; Wang, Z.; Ni, J.; et al. EGFR G796D mutation mediates resistance to osimertinib. Oncotarget 2017, 8, 49671–49679. [Google Scholar] [CrossRef]
  110. Lim, S.M.; Syn, N.L.; Cho, B.C.; Soo, R.A. Acquired resistance to EGFR targeted therapy in non-small cell lung cancer: Mechanisms and therapeutic strategies. Cancer Treat. Rev. 2018, 65, 1–10. [Google Scholar] [CrossRef]
  111. Ercan, D.; Choi, H.G.; Yun, C.H.; Capelletti, M.; Xie, T.; Eck, M.J.; Gray, N.S.; Jänne, P.A. EGFR Mutations and Resistance to Irreversible Pyrimidine-Based EGFR Inhibitors. Clin. Cancer Res. 2015, 21, 3913–3923. [Google Scholar] [CrossRef]
  112. Zhang, Y.; He, B.; Zhou, D.; Li, M.; Hu, C. Newly emergent acquired EGFR exon 18 G724S mutation after resistance of a T790M specific EGFR inhibitor osimertinib in non-small-cell lung cancer: A case report. OncoTargets Ther. 2019, 12, 51–56. [Google Scholar] [CrossRef]
  113. Oztan, A.; Fischer, S.; Schrock, A.B.; Erlich, R.L.; Lovly, C.M.; Stephens, P.J.; Ross, J.S.; Miller, V.; Ali, S.M.; Ou, S.I.; et al. Emergence of EGFR G724S mutation in EGFR-mutant lung adenocarcinoma post progression on osimertinib. Lung Cancer 2017, 111, 84–87. [Google Scholar] [CrossRef]
  114. Brown, B.P.; Zhang, Y.K.; Westover, D.; Yan, Y.; Qiao, H.; Huang, V.; Du, Z.; Smith, J.A.; Ross, J.S.; Miller, V.A.; et al. On-target Resistance to the Mutant-Selective EGFR Inhibitor Osimertinib Can Develop in an Allele-Specific Manner Dependent on the Original EGFR-Activating Mutation. Clin. Cancer Res. 2019, 25, 3341–3351. [Google Scholar] [CrossRef] [PubMed]
  115. Gray, J.E.; Okamoto, I.; Sriuranpong, V.; Vansteenkiste, J.; Imamura, F.; Lee, J.S.; Pang, Y.K.; Cobo, M.; Kasahara, K.; Cheng, Y.; et al. Tissue and Plasma EGFR Mutation Analysis in the FLAURA Trial: Osimertinib versus Comparator EGFR Tyrosine Kinase Inhibitor as First-Line Treatment in Patients with EGFR-Mutated Advanced Non-Small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 6644–6652. [Google Scholar] [CrossRef] [PubMed]
  116. Ríos-Hoyo, A.; Moliner, L.; Arriola, E. Acquired Mechanisms of Resistance to Osimertinib—The Next Challenge. Cancers 2022, 14, 1931. [Google Scholar] [CrossRef]
  117. Kohsaka, S.; Nagano, M.; Ueno, T.; Suehara, Y.; Hayashi, T.; Shimada, N.; Takahashi, K.; Suzuki, K.; Takamochi, K.; Takahashi, F.; et al. A method of high-throughput functional evaluation of EGFR gene variants of unknown significance in cancer. Sci. Transl. Med. 2017, 9, eaan6566. [Google Scholar] [CrossRef] [PubMed]
  118. Chmielecki, J.; Foo, J.; Oxnard, G.R.; Hutchinson, K.; Ohashi, K.; Somwar, R.; Wang, L.; Amato, K.R.; Arcila, M.; Sos, M.L.; et al. Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling. Sci. Transl. Med. 2011, 3, 90ra59. [Google Scholar] [CrossRef] [PubMed]
  119. Engelman, J.A.; Jänne, P.A. Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clin. Cancer Res. 2008, 14, 2895–2899. [Google Scholar] [CrossRef]
  120. Weingertner, N.; Meyer, N.; Voegeli, A.C.; Guenot, D.; Renaud, S.; Massard, G.; Falcoz, P.E.; Olland, A.; Mennecier, B.; Gaub, M.P.; et al. Correlation between MET protein expression and MET gene copy number in a Caucasian cohort of non-small cell lung cancers according to the new IASLC/ATS/ERS classification. Pathology 2015, 47, 320–328. [Google Scholar] [CrossRef]
  121. Drilon, A.; Cappuzzo, F.; Ou, S.I.; Camidge, D.R. Targeting MET in Lung Cancer: Will Expectations Finally Be MET? J. Thorac. Oncol. 2017, 12, 15–26. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. Schildhaus, H.U.; Schultheis, A.M.; Rüschoff, J.; Binot, E.; Merkelbach-Bruse, S.; Fassunke, J.; Schulte, W.; Ko, Y.D.; Schlesinger, A.; Bos, M.; et al. MET amplification status in therapy-naïve adeno- and squamous cell carcinomas of the lung. Clin. Cancer Res. 2015, 21, 907–915. [Google Scholar] [CrossRef]
  124. Awad, M.M.; Oxnard, G.R.; Jackman, D.M.; Savukoski, D.O.; Hall, D.; Shivdasani, P.; Heng, J.C.; Dahlberg, S.E.; Jänne, P.A.; Verma, S.; et al. MET Exon 14 Mutations in Non-Small-Cell Lung Cancer Are Associated with Advanced Age and Stage-Dependent MET Genomic Amplification and c-Met Overexpression. J. Clin. Oncol. 2016, 34, 721–730. [Google Scholar] [CrossRef] [PubMed]
  125. Huang, C.; Zou, Q.; Liu, H.; Qiu, B.; Li, Q.; Lin, Y.; Liang, Y. Management of Non-small Cell Lung Cancer Patients with MET Exon 14 Skipping Mutations. Curr. Treat. Options Oncol. 2020, 21, 33. [Google Scholar] [CrossRef] [PubMed]
  126. Sankar, K.; Gadgeel, S.M.; Qin, A. Molecular therapeutic targets in non-small cell lung cancer. Expert Rev. Anticancer. Ther. 2020, 20, 647–661. [Google Scholar] [CrossRef]
  127. Ren, S.; Wang, J.; Ying, J.; Mitsudomi, T.; Lee, D.H.; Wang, Z.; Chu, Q.; Mack, P.C.; Cheng, Y.; Duan, J.; et al. Consensus for HER2 alterations testing in non-small-cell lung cancer. ESMO Open 2022, 7, 100395. [Google Scholar] [CrossRef]
  128. Takezawa, K.; Pirazzoli, V.; Arcila, M.E.; Nebhan, C.A.; Song, X.; de Stanchina, E.; Ohashi, K.; Janjigian, Y.Y.; Spitzler, P.J.; Melnick, M.A.; et al. HER2 amplification: A potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov. 2012, 2, 922–933. [Google Scholar] [CrossRef] [PubMed]
  129. Planchard, D.; Loriot, Y.; André, F.; Gobert, A.; Auger, N.; Lacroix, L.; Soria, J.C. EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann. Oncol. 2015, 26, 2073–2078. [Google Scholar] [CrossRef]
  130. Fan, W.; Tang, Z.; Yin, L.; Morrison, B.; Hafez-Khayyata, S.; Fu, P.; Huang, H.; Bagai, R.; Jiang, S.; Kresak, A.; et al. MET-independent lung cancer cells evading EGFR kinase inhibitors are therapeutically susceptible to BH3 mimetic agents. Cancer Res. 2011, 71, 4494–4505. [Google Scholar] [CrossRef] [PubMed]
  131. Eberlein, C.A.; Stetson, D.; Markovets, A.A.; Al-Kadhimi, K.J.; Lai, Z.; Fisher, P.R.; Meador, C.B.; Spitzler, P.; Ichihara, E.; Ross, S.J.; et al. Acquired Resistance to the Mutant-Selective EGFR Inhibitor AZD9291 Is Associated with Increased Dependence on RAS Signaling in Preclinical Models. Cancer Res. 2015, 75, 2489–2500. [Google Scholar] [CrossRef]
  132. Chen, W.; Yu, D.; Sun, S.Y.; Li, F. Nanoparticles for co-delivery of osimertinib and selumetinib to overcome osimertinib-acquired resistance in non-small cell lung cancer. Acta Biomater. 2021, 129, 258–268. [Google Scholar] [CrossRef] [PubMed]
  133. Oxnard, G.R.; Yang, J.C.; Yu, H.; Kim, S.W.; Saka, H.; Horn, L.; Goto, K.; Ohe, Y.; Mann, H.; Thress, K.S.; et al. TATTON: A multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 2020, 31, 507–516. [Google Scholar] [CrossRef] [PubMed]
  134. Yang, J.C.; Ohe, Y.; Chiu, C.H.; Ou, X.; Cantarini, M.; Jänne, P.A.; Hartmaier, R.J.; Ahn, M.J. Osimertinib plus Selumetinib in EGFR-Mutated Non-Small Cell Lung Cancer after Progression on EGFR-TKIs: A Phase Ib, Open-Label, Multicenter Trial (TATTON Part B). Clin. Cancer Res. 2022, 28, 4222–4231. [Google Scholar] [CrossRef]
  135. Ho, C.C.; Liao, W.Y.; Lin, C.A.; Shih, J.Y.; Yu, C.J.; Yang, J.C. Acquired BRAF V600E Mutation as Resistant Mechanism after Treatment with Osimertinib. J. Thorac. Oncol. 2017, 12, 567–572. [Google Scholar] [CrossRef]
  136. Zhang, Z.; Zhang, M.; Liu, H.; Yin, W. AZD9291 promotes autophagy and inhibits PI3K/Akt pathway in NSCLC cancer cells. J. Cell. Biochem. 2019, 120, 756–767. [Google Scholar] [CrossRef] [PubMed]
  137. Lee, J.; Kim, H.S.; Lee, B.; Kim, H.K.; Sun, J.M.; Ahn, J.S.; Ahn, M.J.; Park, K.; Lee, S.H. Genomic landscape of acquired resistance to third-generation EGFR tyrosine kinase inhibitors in EGFR T790M-mutant non-small cell lung cancer. Cancer 2020, 126, 2704–2712. [Google Scholar] [CrossRef]
  138. Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; Abdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef]
  139. Vojnic, M.; Kubota, D.; Kurzatkowski, C.; Offin, M.; Suzawa, K.; Benayed, R.; Schoenfeld, A.J.; Plodkowski, A.J.; Poirier, J.T.; Rudin, C.M.; et al. Acquired BRAF Rearrangements Induce Secondary Resistance to EGFR therapy in EGFR-Mutated Lung Cancers. J. Thorac. Oncol. 2019, 14, 802–815. [Google Scholar] [CrossRef]
  140. Aldea, M.; Andre, F.; Marabelle, A.; Dogan, S.; Barlesi, F.; Soria, J.C. Overcoming Resistance to Tumor-Targeted and Immune-Targeted Therapies. Cancer Discov. 2021, 11, 874–899. [Google Scholar] [CrossRef]
  141. Offin, M.; Somwar, R.; Rekhtman, N.; Benayed, R.; Chang, J.C.; Plodkowski, A.; Lui, A.J.W.; 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. [Google Scholar] [CrossRef]
  142. Schrock, A.B.; Zhu, V.W.; Hsieh, W.S.; Madison, R.; Creelan, B.; Silberberg, J.; Costin, D.; Bharne, A.; Bonta, I.; Bosemani, T.; et al. Receptor Tyrosine Kinase Fusions and BRAF Kinase Fusions are Rare but Actionable Resistance Mechanisms to EGFR Tyrosine Kinase Inhibitors. J. Thorac. Oncol. 2018, 13, 1312–1323. [Google Scholar] [CrossRef]
  143. Nelson, A.W.; Schrock, A.B.; Pavlick, D.C.; Ali, S.M.; Atkinson, E.C.; Chachoua, A. Novel SPECC1L-MET Fusion Detected in Circulating Tumor DNA in a Patient with Lung Adenocarcinoma following Treatment with Erlotinib and Osimertinib. J. Thorac. Oncol. 2019, 14, e27–e29. [Google Scholar] [CrossRef] [PubMed]
  144. Piotrowska, Z.; Isozaki, H.; Lennerz, J.K.; Gainor, J.F.; Lennes, I.T.; Zhu, V.W.; Marcoux, N.; Banwait, M.K.; Digumarthy, S.R.; Su, W.; et al. Landscape of Acquired Resistance to Osimertinib in EGFR-Mutant NSCLC and Clinical Validation of Combined EGFR and RET Inhibition with Osimertinib and BLU-667 for Acquired RET Fusion. Cancer Discov. 2018, 8, 1529–1539. [Google Scholar] [CrossRef] [PubMed]
  145. Morgillo, F.; Woo, J.K.; Kim, E.S.; Hong, W.K.; Lee, H.Y. Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Res. 2006, 66, 10100–10111. [Google Scholar] [CrossRef] [PubMed]
  146. Choi, Y.J.; Rho, J.K.; Jeon, B.S.; Choi, S.J.; Park, S.C.; Lee, S.S.; Kim, H.R.; Kim, C.H.; Lee, J.C. Combined inhibition of IGFR enhances the effects of gefitinib in H1650: A lung cancer cell line with EGFR mutation and primary resistance to EGFR-TK inhibitors. Cancer Chemother. Pharmacol. 2010, 66, 381–388. [Google Scholar] [CrossRef]
  147. 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]
  148. Li, L.; Wang, H.; Li, C.; Wang, Z.; Zhang, P.; Yan, X. Transformation to small-cell carcinoma as an acquired resistance mechanism to AZD9291: A case report. Oncotarget 2017, 8, 18609–18614. [Google Scholar] [CrossRef]
  149. Sutherland, K.D.; Proost, N.; Brouns, I.; Adriaensen, D.; Song, J.Y.; Berns, A. Cell of origin of small cell lung cancer: Inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung. Cancer Cell 2011, 19, 754–764. [Google Scholar] [CrossRef]
  150. Thiery, J.P. Epithelial-mesenchymal transitions in development and pathologies. Curr. Opin. Cell Biol. 2003, 15, 740–746. [Google Scholar] [CrossRef]
  151. Kang, Y.; Massagué, J. Epithelial-mesenchymal transitions: Twist in development and metastasis. Cell 2004, 118, 277–279. [Google Scholar] [CrossRef]
  152. Yauch, R.L.; Januario, T.; Eberhard, D.A.; Cavet, G.; Zhu, W.; Fu, L.; Pham, T.Q.; Soriano, R.; Stinson, J.; Seshagiri, S.; et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin. Cancer Res. 2005, 11, 8686–8698. [Google Scholar] [CrossRef] [PubMed]
  153. Della Corte, C.M.; Bellevicine, C.; Vicidomini, G.; Vitagliano, D.; Malapelle, U.; Accardo, M.; Fabozzi, A.; Fiorelli, A.; Fasano, M.; Papaccio, F.; et al. SMO Gene Amplification and Activation of the Hedgehog Pathway as Novel Mechanisms of Resistance to Anti-Epidermal Growth Factor Receptor Drugs in Human Lung Cancer. Clin. Cancer Res. 2015, 21, 4686–4697. [Google Scholar] [CrossRef] [PubMed]
  154. Bai, X.Y.; Zhang, X.C.; Yang, S.Q.; An, S.J.; Chen, Z.H.; Su, J.; Xie, Z.; Gou, L.Y.; Wu, Y.L. Blockade of Hedgehog Signaling Synergistically Increases Sensitivity to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small-Cell Lung Cancer Cell Lines. PLoS ONE 2016, 11, e0149370. [Google Scholar] [CrossRef] [PubMed]
  155. Sequist, L.V.; Han, J.Y.; Ahn, M.J.; Cho, B.C.; Yu, H.; Kim, S.W.; Yang, J.C.; 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 Oncology 2020, 21, 373–386. [Google Scholar] [CrossRef] [PubMed]
  156. Wu, Y.L.; Zhang, L.; Kim, D.W.; Liu, X.; Lee, D.H.; Yang, J.C.; Ahn, M.J.; Vansteenkiste, J.F.; Su, W.C.; Felip, E.; et al. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients with EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3101–3109. [Google Scholar] [CrossRef]
  157. Wu, Y.L.; Cheng, Y.; Zhou, J.; Lu, S.; Zhang, Y.; Zhao, J.; Kim, D.W.; Soo, R.A.; Kim, S.W.; Pan, H.; et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): An open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir. Med. 2020, 8, 1132–1143. [Google Scholar] [CrossRef] [PubMed]
  158. Cho, B.C.; Felip, E.; Hayashi, H.; Thomas, M.; Lu, S.; Besse, B.; Sun, T.; Martinez, M.; Sethi, S.N.; Shreeve, S.M.; et al. MARIPOSA: Phase 3 study of first-line amivantamab + lazertinib versus osimertinib in EGFR-mutant non-small-cell lung cancer. Future Oncol. 2022, 18, 639–647. [Google Scholar] [CrossRef]
  159. Janjigian, Y.Y.; Smit, E.F.; Groen, H.J.; Horn, L.; Gettinger, S.; Camidge, D.R.; Riely, G.J.; Wang, B.; Fu, Y.; Chand, V.K.; et al. Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations. Cancer Discov. 2014, 4, 1036–1045. [Google Scholar] [CrossRef]
  160. Lee, J.Y.; Sun, J.M.; Lim, S.H.; Kim, H.S.; Yoo, K.H.; Jung, K.S.; Song, H.N.; Ku, B.M.; Koh, J.; Bae, Y.H.; et al. A Phase Ib/II Study of Afatinib in Combination with Nimotuzumab in Non-Small Cell Lung Cancer Patients with Acquired Resistance to Gefitinib or Erlotinib. Clin. Cancer Res. 2016, 22, 2139–2145. [Google Scholar] [CrossRef]
  161. Thatcher, N.; Hirsch, F.R.; Luft, A.V.; Szczesna, A.; Ciuleanu, T.E.; Dediu, M.; Ramlau, R.; Galiulin, R.K.; Bálint, B.; Losonczy, G.; et al. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): An open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015, 16, 763–774. [Google Scholar] [CrossRef]
  162. Bertino, E.M.; Gentzler, R.D.; Clifford, S.; Kolesar, J.; Muzikansky, A.; Haura, E.B.; Piotrowska, Z.; Camidge, D.R.; Stinchcombe, T.E.; Hann, C.; et al. Phase IB Study of Osimertinib in Combination with Navitoclax in EGFR-mutant NSCLC Following Resistance to Initial EGFR Therapy (ETCTN 9903). Clin. Cancer Res. 2021, 27, 1604–1611. [Google Scholar] [CrossRef]
  163. Mok, T.S.K.; Kim, S.-W.; Wu, Y.-L.; Nakagawa, K.; Yang, J.-J.; Ahn, M.-J.; Wang, J.; Yang, J.C.-H.; Lu, Y.; Atagi, S.; et al. Gefitinib Plus Chemotherapy Versus Chemotherapy in Epidermal Growth Factor Receptor Mutation–Positive Non–Small-Cell Lung Cancer Resistant to First-Line Gefitinib (IMPRESS): Overall Survival and Biomarker Analyses. J. Clin. Oncol. 2017, 35, 4027–4034. [Google Scholar] [CrossRef] [PubMed]
  164. 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]
  165. Tanaka, K.; Asahina, H.; Kishimoto, J.; Miyata, Y.; Uchida, T.; Watanabe, K.; Hamai, K.; Harada, T.; Tsubata, Y.; Sugawara, S.; et al. Osimertinib versus osimertinib plus chemotherapy for non-small cell lung cancer with EGFR (T790M)-associated resistance to initial EGFR inhibitor treatment: An open-label, randomised phase 2 clinical trial. Eur. J. Cancer 2021, 149, 14–22. [Google Scholar] [CrossRef] [PubMed]
  166. Gettinger, S.; Hellmann, M.D.; Chow, L.Q.M.; Borghaei, H.; Antonia, S.; Brahmer, J.R.; Goldman, J.W.; Gerber, D.E.; Juergens, R.A.; Shepherd, F.A.; et al. Nivolumab Plus Erlotinib in Patients with EGFR-Mutant Advanced NSCLC. J. Thorac. Oncol. 2018, 13, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
  167. Yang, J.C.; Shepherd, F.A.; Kim, D.W.; Lee, G.W.; Lee, J.S.; Chang, G.C.; Lee, S.S.; Wei, Y.F.; Lee, Y.G.; Laus, G.; et al. Osimertinib Plus Durvalumab versus Osimertinib Monotherapy in EGFR T790M-Positive NSCLC following Previous EGFR TKI Therapy: CAURAL Brief Report. J. Thorac. Oncol. 2019, 14, 933–939. [Google Scholar] [CrossRef]
  168. 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]
  169. Lu, S.; Wu, L.; Jian, H.; Chen, Y.; Wang, Q.; Fang, J.; Wang, Z.; Hu, Y.; Sun, M.; Han, L.; et al. Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): First interim results from a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2022, 23, 1167–1179. [Google Scholar] [CrossRef]
  170. 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. Abstract CT190: A multicenter, open-label, single-arm, phase II study: The third generation EGFR tyrosine kinase inhibitor almonertinib for pretreated EGFR T790M-positive locally advanced or metastatic non-small cell lung cancer (APOLLO). Cancer Res. 2020, 80, CT190. [Google Scholar] [CrossRef]
  171. Shi, Y.; Hu, X.; Zhang, S.; Lv, D.; Zhang, Y.; Yu, Q.; Wu, L.; Liu, L.; Wang, X.; Ma, Z.; et al. Efficacy and safety of alflutinib (AST2818) in patients with T790M mutation-positive NSCLC: A phase IIb multicenter single-arm study. J. Clin. Oncol. 2020, 38, 9602. [Google Scholar] [CrossRef]
  172. Ahn, M.-J.; Han, J.-Y.; Lee, K.H.; Kim, S.-W.; Kim, D.-W.; Lee, Y.-G.; Cho, E.K.; Kim, J.-H.; Lee, G.-W.; Lee, J.-S.; et al. Lazertinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: Results from the dose escalation and dose expansion parts of a first-in-human, open-label, multicentre, phase 1–2 study. Lancet Oncol. 2019, 20, 1681–1690. [Google Scholar] [CrossRef] [PubMed]
  173. Ma, Y.; Zheng, X.; Zhao, H.; Fang, W.; Zhang, Y.; Ge, J.; Wang, L.; Wang, W.; Jiang, J.; Chuai, S.; et al. First-in-Human Phase I Study of AC0010, a Mutant-Selective EGFR Inhibitor in Non–Small Cell Lung Cancer: Safety, Efficacy, and Potential Mechanism of Resistance. J. Thorac. Oncol. 2018, 13, 968–977. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, Y.L.; Zhou, Q.; Liu, X.; Zhang, L.; Zhou, J.; Wu, L.; An, T.; Cheng, Y.; Zheng, X.; Hu, B.; et al. MA16.06 Phase I/II Study of AC0010, Mutant-Selective EGFR Inhibitor, in Non-Small Cell Lung Cancer (NSCLC) Patients with EGFR T790M Mutation. J. Thorac. Oncol. 2017, 12, S437–S438. [Google Scholar] [CrossRef]
  175. Tan, D.S.W.; Kim, S.W.; Ponce Aix, S.; Sequist, L.V.; Smit, E.F.; Yang, J.C.H.; Hida, T.; Toyozawa, R.; Felip, E.; Wolf, J.; et al. Nazartinib for treatment-naive EGFR-mutant non-small cell lung cancer: Results of a phase 2, single-arm, open-label study. Eur. J. Cancer 2022, 172, 276–286. [Google Scholar] [CrossRef]
  176. Nagasaka, M.; Zhu, V.W.; Lim, S.M.; Greco, M.; Wu, F.; Ou, S.I. Beyond Osimertinib: The Development of Third-Generation EGFR Tyrosine Kinase Inhibitors For Advanced EGFR+ NSCLC. J. Thorac. Oncol. 2021, 16, 740–763. [Google Scholar] [CrossRef]
  177. Park, K.; Jӓnne, P.A.; Kim, D.W.; Han, J.Y.; Wu, M.F.; Lee, J.S.; Kang, J.H.; Lee, D.H.; Cho, B.C.; Yu, C.J.; et al. Olmutinib in T790M-positive non-small cell lung cancer after failure of first-line epidermal growth factor receptor-tyrosine kinase inhibitor therapy: A global, phase 2 study. Cancer 2021, 127, 1407–1416. [Google Scholar] [CrossRef]
  178. Murakami, H.; Nokihara, H.; Hayashi, H.; Seto, T.; Park, K.; Azuma, K.; Tsai, C.M.; Yang, J.C.; Nishio, M.; Kim, S.W.; et al. Clinical activity of ASP8273 in Asian patients with non-small-cell lung cancer with EGFR activating and T790M mutations. Cancer Sci. 2018, 109, 2852–2862. [Google Scholar] [CrossRef]
  179. Cho, B.C.; Goldberg, S.B.; Kim, D.W.; Socinski, M.A.; Burns, T.F.; Lwin, Z.; Pathan, N.; Ma, W.D.; Masters, J.C.; Cossons, N.; et al. A phase 1b/2 study of PF-06747775 as monotherapy or in combination with Palbociclib in patients with epidermal growth factor receptor mutant advanced non-small cell lung cancer. Expert Opin. Investig. Drugs 2022, 31, 747–757. [Google Scholar] [CrossRef]
  180. Wang, S.; Cang, S.; Liu, D. Third-generation inhibitors targeting EGFR T790M mutation in advanced non-small cell lung cancer. J. Hematol. Oncol. 2016, 9, 34. [Google Scholar] [CrossRef]
  181. Rangachari, D.; To, C.; Shpilsky, J.E.; VanderLaan, P.A.; Kobayashi, S.S.; Mushajiang, M.; Lau, C.J.; Paweletz, C.P.; Oxnard, G.R.; Jänne, P.A.; et al. EGFR-Mutated Lung Cancers Resistant to Osimertinib through EGFR C797S Respond to First-Generation Reversible EGFR Inhibitors but Eventually Acquire EGFR T790M/C797S in Preclinical Models and Clinical Samples. J. Thorac. Oncol. 2019, 14, 1995–2002. [Google Scholar] [CrossRef]
  182. Wang, X.; Zhou, L.; Yin, J.C.; Wu, X.; Shao, Y.W.; Gao, B. Lung Adenocarcinoma Harboring EGFR 19del/C797S/T790M Triple Mutations Responds to Brigatinib and Anti-EGFR Antibody Combination Therapy. J. Thorac. Oncol. 2019, 14, e85–e88. [Google Scholar] [CrossRef] [PubMed]
  183. Uchibori, K.; Inase, N.; Araki, M.; Kamada, M.; Sato, S.; Okuno, Y.; Fujita, N.; Katayama, R. Brigatinib combined with anti-EGFR antibody overcomes osimertinib resistance in EGFR-mutated non-small-cell lung cancer. Nat. Commun. 2017, 8, 14768. [Google Scholar] [CrossRef] [PubMed]
  184. Yang, Y.; Xu, H.; Ma, L.; Yang, L.; Yang, G.; Zhang, S.; Ai, X.; Zhang, S.; Wang, Y. Possibility of brigatinib-based therapy, or chemotherapy plus anti-angiogenic treatment after resistance of osimertinib harboring EGFR T790M-cis-C797S mutations in lung adenocarcinoma patients. Cancer Med. 2021, 10, 8328–8337. [Google Scholar] [CrossRef]
  185. Wang, Y.; Yang, N.; Zhang, Y.; Li, L.; Han, R.; Zhu, M.; Feng, M.; Chen, H.; Lizaso, A.; Qin, T.; et al. Effective Treatment of Lung Adenocarcinoma Harboring EGFR Activating Mutation, T790M, and cis-C797S Triple Mutations by Brigatinib and Cetuximab Combination Therapy. J. Thorac. Oncol. 2020, 15, 1369–1375. [Google Scholar] [CrossRef]
  186. Song, Z.; Ren, G.; Hu, L.; Wang, X.; Song, J.; Jia, Y.; Zhao, G.; Zang, A.; Du, H.; Sun, Y.; et al. Two case reports of non-small cell lung cancer patients harboring acquired EGFR T790M-cis-C797S benefit from immune checkpoint inhibitor combined with platinum-based doublet chemotherapy. Ann. Transl. Med. 2022, 10, 719. [Google Scholar] [CrossRef] [PubMed]
  187. Wang, S.; Song, Y.; Liu, D. EAI045: The fourth-generation EGFR inhibitor overcoming T790M and C797S resistance. Cancer Lett. 2017, 385, 51–54. [Google Scholar] [CrossRef]
  188. To, C.; Jang, J.; Chen, T.; Park, E.; Mushajiang, M.; De Clercq, D.J.H.; Xu, M.; Wang, S.; Cameron, M.D.; Heppner, D.E.; et al. Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor. Cancer Discov. 2019, 9, 926–943. [Google Scholar] [CrossRef] [PubMed]
  189. Schalm, S.S.; Dineen, T.; Lim, S.M.; Park, C.W.; Hsieh, J.; Woessner, R.; Zhang, Z.; Wilson, K.; Eno, M.; Wilson, D.; et al. 1296P BLU-945, a highly potent and selective 4th generation EGFR TKI for the treatment of EGFR T790M/C797S resistant NSCLC. Ann. Oncol. 2020, 31, S839. [Google Scholar] [CrossRef]
  190. Jia, Y.; Yun, C.-H.; Park, E.; Ercan, D.; Manuia, M.; Juarez, J.; Xu, C.; Rhee, K.; Chen, T.; Zhang, H.; et al. Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature 2016, 534, 129–132. [Google Scholar] [CrossRef]
  191. To, C.; Beyett, T.S.; Jang, J.; Feng, W.W.; Bahcall, M.; Haikala, H.M.; Shin, B.H.; Heppner, D.E.; Rana, J.K.; Leeper, B.A.; et al. An allosteric inhibitor against the therapy-resistant mutant forms of EGFR in non-small cell lung cancer. Nat. Cancer 2022, 3, 402–417. [Google Scholar] [CrossRef] [PubMed]
  192. Kashima, K.; Kawauchi, H.; Tanimura, H.; Tachibana, Y.; Chiba, T.; Torizawa, T.; Sakamoto, H. CH7233163 Overcomes Osimertinib-Resistant EGFR-Del19/T790M/C797S Mutation. Mol. Cancer Ther. 2020, 19, 2288–2297. [Google Scholar] [CrossRef]
  193. Conti, C.; Campbell, J.; Woessner, R.; Guo, J.; Timsit, Y.; Iliou, M.; Wardwell, S.; Davis, A.; Chicklas, S.; Hsieh, J.; et al. Abstract 1262: BLU-701 is a highly potent, brain-penetrant and WT-sparing next-generation EGFR TKI for the treatment of sensitizing (ex19del, L858R) and C797S resistance mutations in metastatic NSCLC. Cancer Res. 2021, 81, 1262. [Google Scholar] [CrossRef]
  194. Tavera, L.; Schalm, S.; Campbell, J.; Guo, J.; Medendorp, C.; Chen, M.; Albayya, F.; Dineen, T.; Zhang, Z.; Iliou, M.; et al. Abstract 3328: Antitumor activity of BLU-945 and BLU-701 as single agents and in combination in EGFR L858R-driven models of NSCLC. Cancer Res. 2022, 82, 3328. [Google Scholar] [CrossRef]
  195. Eno, M.S.; Brubaker, J.D.; Campbell, J.E.; De Savi, C.; Guzi, T.J.; Williams, B.D.; Wilson, D.; Wilson, K.; Brooijmans, N.; Kim, J.; et al. Discovery of BLU-945, a Reversible, Potent, and Wild-Type-Sparing Next-Generation EGFR Mutant Inhibitor for Treatment-Resistant Non-Small-Cell Lung Cancer. J. Med. Chem. 2022, 65, 9662–9677. [Google Scholar] [CrossRef] [PubMed]
  196. Spigel, D.; Goto, K.; Camidge, D.R.; Elamin, Y.; de Langen, A.J.; Leighl, N.B.; Minchom, A.; Piotrowska, Z.; Planchard, D.; Reckamp, K.; et al. Abstract P230: A phase 1/2 study of BLU-945, a highly potent and selective inhibitor of epidermal growth factor receptor (EGFR) resistance mutations, in patients with EGFR-mutant non-small cell lung cancer (NSCLC). Mol. Cancer Ther. 2021, 20, P230. [Google Scholar] [CrossRef]
  197. Lim, S.M.; Park, C.W.; Zhang, Z.; Woessner, R.; Dineen, T.; Stevison, F.; Hsieh, J.; Eno, M.; Wilson, D.; Campbell, J.; et al. Abstract 1467: BLU-945, a fourth-generation, potent and highly selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) with intracranial activity, demonstrates robust in vivo antitumor activity in models of osimertinib-resistant non-small cell lung cancer (NSCLC). Cancer Res. 2021, 81, 1467. [Google Scholar] [CrossRef]
  198. Shum, E.; Elamin, Y.; Reckamp, K.L.; Piotrowska, Z.; Rotow, J.; Tan, D.S.; Goto, K.; Parepally, J.; Albayya, F.; Louie-Gao, M.; et al. Abstract CT184: Emerging evidence of activity of BLU-945 in patients with advanced EGFR-mutant NSCLC utilizing circulating tumor DNA (ctDNA) in the phase 1/2 SYMPHONY study. Cancer Res. 2022, 82, CT184. [Google Scholar] [CrossRef]
  199. Le, X.; Puri, S.; Negrao, M.V.; Nilsson, M.B.; Robichaux, J.; Boyle, T.; Hicks, J.K.; Lovinger, K.L.; Roarty, E.; Rinsurongkawong, W.; et al. Landscape of EGFR-Dependent and -Independent Resistance Mechanisms to Osimertinib and Continuation Therapy Beyond Progression in EGFR-Mutant NSCLC. Clin. Cancer Res. 2018, 24, 6195–6203. [Google Scholar] [CrossRef]
  200. Piper-Vallillo, A.J.; Sequist, L.V.; Piotrowska, Z. Emerging Treatment Paradigms for EGFR-Mutant Lung Cancers Progressing on Osimertinib: A Review. J. Clin. Oncol. 2020, 38, 2926–2936. [Google Scholar] [CrossRef]
  201. Wu, Y.-L.; Han, J.-Y.; Kato, T.; Barlesi, F.; Garon, E.B.; Cappuzzo, F.; Shibata, Y.; Smith, N.; Khanna, S.; Belli, R.; et al. Capmatinib plus osimertinib versus platinum-pemetrexed doublet chemotherapy as second-line therapy in patients with stage IIIb/IIIc or IV EGFR-mutant, T790M-negative NSCLC harboring MET amplification. J. Clin. Oncol. 2022, 40, TPS9153. [Google Scholar] [CrossRef]
  202. Deng, L.; Kiedrowski, L.A.; Ravera, E.; Cheng, H.; Halmos, B. Response to Dual Crizotinib and Osimertinib Treatment in a Lung Cancer Patient with MET Amplification Detected by Liquid Biopsy Who Acquired Secondary Resistance to EGFR Tyrosine Kinase Inhibition. J. Thorac. Oncol. 2018, 13, e169–e172. [Google Scholar] [CrossRef] [PubMed]
  203. York, E.R.; Varella-Garcia, M.; Bang, T.J.; Aisner, D.L.; Camidge, D.R. Tolerable and Effective Combination of Full-Dose Crizotinib and Osimertinib Targeting MET Amplification Sequentially Emerging after T790M Positivity in EGFR-Mutant Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, e85–e88. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, Y.; Tian, P.; Xia, L.; Li, L.; Han, R.; Zhu, M.; Lizaso, A.; Qin, T.; Li, M.; Yu, B.; et al. The clinical efficacy of combinatorial therapy of EGFR-TKI and crizotinib in overcoming MET amplification-mediated resistance from prior EGFR-TKI therapy. Lung Cancer 2020, 146, 165–173. [Google Scholar] [CrossRef] [PubMed]
  205. La Monica, S.; Fumarola, C.; Cretella, D.; Bonelli, M.; Minari, R.; Cavazzoni, A.; Digiacomo, G.; Galetti, M.; Volta, F.; Mancini, M.; et al. Efficacy of the CDK4/6 Dual Inhibitor Abemaciclib in EGFR-Mutated NSCLC Cell Lines with Different Resistance Mechanisms to Osimertinib. Cancers 2021, 13, 6. [Google Scholar] [CrossRef]
  206. Qin, Q.; Li, X.; Liang, X.; Zeng, L.; Wang, J.; Sun, L.; Zhong, D. CDK4/6 inhibitor palbociclib overcomes acquired resistance to third-generation EGFR inhibitor osimertinib in non-small cell lung cancer (NSCLC). Thorac. Cancer 2020, 11, 2389–2397. [Google Scholar] [CrossRef]
  207. Perrone, M.; Giuliani, F.; Sanna, V.; Bruno, S.; Melaccio, A.; Santoro, A.N.; Laface, C.; Maselli, F.M.; Iaia, M.L.; Guarini, C.; et al. Advances in pharmacotherapies that target the cell cycle for treatment of breast cancer: Where are we at today? Expert Opin. Pharmacother. 2023, 24, 887–900. [Google Scholar] [CrossRef]
  208. Ji, W.; Choi, Y.J.; Kang, M.-H.; Sung, K.J.; Kim, D.H.; Jung, S.; Choi, C.-M.; Lee, J.C.; Rho, J.K. Efficacy of the CDK7 Inhibitor on EMT-Associated Resistance to 3rd Generation EGFR-TKIs in Non-Small Cell Lung Cancer Cell Lines. Cells 2020, 9, 2596. [Google Scholar] [CrossRef]
  209. Regales, L.; Gong, Y.; Shen, R.; de Stanchina, E.; Vivanco, I.; Goel, A.; Koutcher, J.A.; Spassova, M.; Ouerfelli, O.; Mellinghoff, I.K.; et al. Dual targeting of EGFR can overcome a major drug resistance mutation in mouse models of EGFR mutant lung cancer. J. Clin. Investig. 2009, 119, 3000–3010. [Google Scholar] [CrossRef]
  210. van Veggel, B.; de Langen, A.J.; Hashemi, S.M.S.; Monkhorst, K.; Heideman, D.A.M.; Thunnissen, E.; Smit, E.F. Afatinib and Cetuximab in Four Patients with EGFR Exon 20 Insertion–Positive Advanced NSCLC. J. Thorac. Oncol. 2018, 13, 1222–1226. [Google Scholar] [CrossRef]
  211. Fang, W.; Huang, Y.; Gan, J.; Shao, Y.W.; Zhang, L. Durable Response of Low-Dose Afatinib plus Cetuximab in an Adenocarcinoma Patient with a Novel EGFR Exon 20 Insertion Mutation. J. Thorac. Oncol. 2019, 14, e220–e221. [Google Scholar] [CrossRef]
  212. Marcoux, N.; Gettinger, S.N.; O’Kane, G.; Arbour, K.C.; Neal, J.W.; Husain, H.; Evans, T.L.; Brahmer, J.R.; Muzikansky, A.; Bonomi, P.D.; et al. EGFR-Mutant Adenocarcinomas That Transform to Small-Cell Lung Cancer and Other Neuroendocrine Carcinomas: Clinical Outcomes. J. Clin. Oncol. 2018, 37, 278–285. [Google Scholar] [CrossRef]
  213. Mu, Y.; Hao, X.; Yang, K.; Ma, D.; Wang, S.; Xu, Z.; Li, J.; Xing, P. Clinical Modality of Resistance and Subsequent Management of Patients with Advanced Non-small Cell Lung Cancer Failing Treatment with Osimertinib. Target. Oncol. 2019, 14, 335–342. [Google Scholar] [CrossRef] [PubMed]
  214. Soria, J.-C.; Wu, Y.-L.; Nakagawa, K.; Kim, S.-W.; Yang, J.-J.; Ahn, M.-J.; Wang, J.; Yang, J.C.-H.; Lu, Y.; Atagi, S.; et al. Gefitinib plus chemotherapy versus placebo plus chemotherapy in EGFR-mutation-positive non-small-cell lung cancer after progression on first-line gefitinib (IMPRESS): A phase 3 randomised trial. Lancet Oncol. 2015, 16, 990–998. [Google Scholar] [CrossRef]
  215. Hosomi, Y.; Morita, S.; Sugawara, S.; Kato, T.; Fukuhara, T.; Gemma, A.; Takahashi, K.; Fujita, Y.; Harada, T.; Minato, K.; et al. Gefitinib alone versus Gefitinib Plus Chemotherapy for Non–Small-Cell Lung Cancer with Mutated Epidermal Growth Factor Receptor: NEJ009 Study. J. Clin. Oncol. 2019, 38, 115–123. [Google Scholar] [CrossRef]
  216. Laface, C.; Ranieri, G.; Maselli, F.M.; Ambrogio, F.; Foti, C.; Ammendola, M.; Laterza, M.; Cazzato, G.; Memeo, R.; Mastrandrea, G.; et al. Immunotherapy and the Combination with Targeted Therapies for Advanced Hepatocellular Carcinoma. Cancers 2023, 15, 654. [Google Scholar] [CrossRef]
  217. Herbst, R.S.; Baas, P.; Kim, D.-W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.-Y.; Molina, J.; Kim, J.-H.; Arvis, C.D.; Ahn, M.-J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
  218. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed]
  219. Mazieres, J.; Drilon, A.; Lusque, A.; Mhanna, L.; Cortot, A.B.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  220. Lisberg, A.; Cummings, A.; Goldman, J.W.; Bornazyan, K.; Reese, N.; Wang, T.; Coluzzi, P.; Ledezma, B.; Mendenhall, M.; Hunt, J.; et al. A Phase II Study of Pembrolizumab in EGFR-Mutant, PD-L1+, Tyrosine Kinase Inhibitor Naïve Patients with Advanced NSCLC. J. Thorac. Oncol. 2018, 13, 1138–1145. [Google Scholar] [CrossRef] [PubMed]
  221. Wang, X.; Wang, F.; Zhong, M.; Yarden, Y.; Fu, L. The biomarkers of hyperprogressive disease in PD-1/PD-L1 blockage therapy. Mol. Cancer 2020, 19, 81. [Google Scholar] [CrossRef]
  222. Kim, C.G.; Kim, K.H.; Pyo, K.H.; Xin, C.F.; Hong, M.H.; Ahn, B.C.; Kim, Y.; Choi, S.J.; Yoon, H.I.; Lee, J.G.; et al. Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1104–1113. [Google Scholar] [CrossRef]
  223. To, K.K.W.; Fong, W.; Cho, W.C.S. Immunotherapy in Treating EGFR-Mutant Lung Cancer: Current Challenges and New Strategies. Front. Oncol. 2021, 11, 635007. [Google Scholar] [CrossRef] [PubMed]
  224. Lau, S.C.M.; Fares, A.F.; Le, L.W.; Mackay, K.M.; Soberano, S.; Chan, S.W.; Smith, E.; Ryan, M.; Tsao, M.S.; Bradbury, P.A.; et al. Subtypes of EGFR- and HER2-Mutant Metastatic NSCLC Influence Response to Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 253–259. [Google Scholar] [CrossRef] [PubMed]
  225. Haratani, K.; Hayashi, H.; Tanaka, T.; Kaneda, H.; Togashi, Y.; Sakai, K.; Hayashi, K.; Tomida, S.; Chiba, Y.; Yonesaka, K.; et al. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann. Oncol. 2017, 28, 1532–1539. [Google Scholar] [CrossRef]
  226. Rizvi, N.A.; Chow, L.Q.M.; Borghaei, H.; Shen, Y.; Harbison, C.; Alaparthy, S.; Chen, A.C.; Gettinger, S.N. Safety and response with nivolumab (anti-PD-1; BMS-936558, ONO-4538) plus erlotinib in patients (pts) with epidermal growth factor receptor mutant (EGFR MT) advanced NSCLC. J. Clin. Oncol. 2014, 32, 8022. [Google Scholar] [CrossRef]
  227. Oshima, Y.; Tanimoto, T.; Yuji, K.; Tojo, A. EGFR–TKI-Associated Interstitial Pneumonitis in Nivolumab-Treated Patients with Non–Small Cell Lung Cancer. JAMA Oncol. 2018, 4, 1112–1115. [Google Scholar] [CrossRef]
  228. Gibbons, D.L.; Chow, L.Q.; Kim, D.W.; Kim, S.W.; Yeh, T.; Song, X.; Jiang, H.; Taylor, R.; Karakunnel, J.; Creelan, B. 57O Efficacy, safety and tolerability of MEDI4736 (durvalumab [D]), a human IgG1 anti-programmed cell death-ligand-1 (PD-L1) antibody, combined with gefitinib (G): A phase I expansion in TKI-naïve patients (pts) with EGFR mutant NSCLC. J. Thorac. Oncol. 2016, 11, S79. [Google Scholar] [CrossRef]
  229. Schoenfeld, A.J.; Arbour, K.C.; Rizvi, H.; Iqbal, A.N.; Gadgeel, S.M.; Girshman, J.; Kris, M.G.; Riely, G.J.; Yu, H.A.; Hellmann, M.D. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann. Oncol. 2019, 30, 839–844. [Google Scholar] [CrossRef]
  230. Hsu, P.-C.; Jablons, D.M.; Yang, C.-T.; You, L. Epidermal Growth Factor Receptor (EGFR) Pathway, Yes-Associated Protein (YAP) and the Regulation of Programmed Death-Ligand 1 (PD-L1) in Non-Small Cell Lung Cancer (NSCLC). Int. J. Mol. Sci. 2019, 20, 3821. [Google Scholar] [CrossRef]
  231. Kaira, K.; Kagamu, H. Drastic Response of Re-challenge of EGFR-TKIs Immediately After Nivolumab Therapy in EGFR-TKI–Resistant Patients. J. Thorac. Oncol. 2019, 14, e135–e136. [Google Scholar] [CrossRef] [PubMed]
  232. Kaira, K.; Kobayashi, K.; Shiono, A.; Yamaguchi, O.; Hashimoto, K.; Mouri, A.; Shinomiya, S.; Miura, Y.; Imai, H.; Kagamu, H. Effectiveness of EGFR-TKI rechallenge immediately after PD-1 blockade failure. Thorac. Cancer 2021, 12, 864–873. [Google Scholar] [CrossRef] [PubMed]
  233. Lam, T.C.; Tsang, K.C.; Choi, H.C.; Lee, V.H.; Lam, K.O.; Chiang, C.L.; So, T.H.; Chan, W.W.; Nyaw, S.F.; Lim, F.; et al. Combination atezolizumab, bevacizumab, pemetrexed and carboplatin for metastatic EGFR mutated NSCLC after TKI failure. Lung Cancer 2021, 159, 18–26. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mutations of resistance to EGFR-TKIs, according to the generation of TKIs and the line of therapy.
Figure 1. Mutations of resistance to EGFR-TKIs, according to the generation of TKIs and the line of therapy.
Pharmaceutics 15 01604 g001
Figure 2. EGFR signaling pathway and EGFR-TKIs’ resistance mechanism.
Figure 2. EGFR signaling pathway and EGFR-TKIs’ resistance mechanism.
Pharmaceutics 15 01604 g002
Figure 3. Summary of the procedures after osimertinib resistance. Tissue biopsy and/or liquid biopsy should be performed to identify the mechanisms of resistance to guide treatment. Therapeutic strategies are comprised of targeted therapy, chemotherapy, and immunotherapy or combination regimens. In the case of a lack of valid targets, clinical trials should be considered.
Figure 3. Summary of the procedures after osimertinib resistance. Tissue biopsy and/or liquid biopsy should be performed to identify the mechanisms of resistance to guide treatment. Therapeutic strategies are comprised of targeted therapy, chemotherapy, and immunotherapy or combination regimens. In the case of a lack of valid targets, clinical trials should be considered.
Pharmaceutics 15 01604 g003
Table 1. EGFR mutations: common, uncommon, and other alterations.
Table 1. EGFR mutations: common, uncommon, and other alterations.
EGFR MutationsSpecific AlterationFrequencyClinical CharacteristicsResponse to
EGFR-TKIs
Gefitinib/ErlotinibAfatinibOsimertininb
CommonExon 19 deletion45–50%Adenocarcinoma
Female gender
Never smoker status
Asian ethnicity
SensitiveSensitiveSensitive
Exon 21 L858R
point mutation
37–40%SensitiveSensitiveSensitive
Uncommon
or
rare
Exon 18G719X3%Adenocarcinoma
Male gender
Smoker history
Asian ethnicity
IntermediateSensitiveInter./sensitive
E709X0.3%E709K IntermediateSensitiveSensitive
Del180.3%IntermediateSensitiveSensitive
Exon 19Ins19<0.6%IntermediateInter./Sens.Inter./Sens.
Exon 20Ins20>5.8%ResistantResistant/Sens.Inter./Sens.
S768I<1.5%IntermediateInter./Sens.Sensitive
Exon 21L861Q0.9–3%IntermediateInter./Sens.Inter./Sens.
Other EGFR
alterations
  • Complex mutations are a combination between:
    -
    common and rare mutations
    -
    rare and rare mutations
    -
    common and common mutations
5–15%Depend on
the specific
combination
of mutations
Depend on
the specific
combination
of mutations
  • Co-mutations are combinations of EGFR mutation(s) with one or more mutations of another gene
(tumour suppressor gene or oncogene)
3–6%
  • Subclonal mutations: the presence of the specific mutation only in a small percentage of tumour cells
Variable
Table 2. Clinical trials evaluating all generations of EGFR-TKIs.
Table 2. Clinical trials evaluating all generations of EGFR-TKIs.
Generation
EGFR-TKI
Clinical TrailEGFR StatusComparisonResults
First-generationNEJ002 [28,29]Common mutationsGefitinib vs. carboplatin plus paclitaxel* PFS: 10.8 vs. 5.4 mo.
** OS: 27.7 vs. 26.6 mo.
IPASS [30,31]Common mutationsGefitinib vs. carboplatin plus paclitaxel* PFS: 9.5 vs. 6.3 mo.
** OS: 18.8 vs. 17.4 mo.
WJTOG3405 [32,33]Common mutationsGefitinib vs. cisplatin plus docetaxel* PFS: 9.2 vs. 6.3 mo.
** OS: 34.9 vs. 37.3 mo.
OPTIMAL [34,35]Common mutationsErlotinib vs. gemcitabine plus carboplatin* PFS: 13.1 vs. 4.6 mo.
** OS: 22.8 vs. 27.2 mo.
ENSURE [36]Common mutationsErlotinib vs. gemcitabine plus cisplatin* PFS: 11 vs. 5.5 mo.
** OS: 26.3 vs. 25.5 mo.
EUTARC [37]Common mutationsErlotinib vs. cisplatin plus docetaxel or
gemcitabine
* PFS: 9.7 vs. 5.2 mo.
** OS: 22.8 vs. 27.2 mo.
CONVINCE [38]Common mutationsIcotinib vs. cisplatin plus pemetrexed* PFS: 11.2 vs. 7.9 mo.
** OS: 30.5 vs. 32.1 mo.
Second-generationLUX-Lung 3 [39]Common mutationsAfatinib vs. cisplatin plus gemcitabine or
pemetrexed
* PFS: 13.6 vs. 6.9 mo.
** OS: 28.2 vs. 28.2 mo.
LUX-Lung 6 [40]Common mutationsAfatinib vs. cisplatin plus gemcitabine or
pemetrexed
* PFS: 11 vs. 5.6 mo.
** OS: 23.1 vs. 23.5 mo.
ARCHER 1050 [40,41]Common mutations +/− T790MDacotinib vs. gefitinib* PFS: 14.7 vs. 9.2 mo.
** OS: 34.1 vs. 27 mo.
Third-generationAURA3 [42]T790M mutationOsimertinib vs. cis/carboplatin plus pemetrexed after first-line EGFR-TKI therapy* PFS: 10.1 vs. 4.4 mo.
** OS: 26.8 vs. 22.5 mo.
FLAURA [26]Common mutationsOsimertinib vs. gefitinib or erlotinib* PFS: 18.9 vs. 10.2 mo.
** OS: 38.6 vs. 31.8 mo.
ADAURA [43]Common mutationsOsimertinib for 3 years as adjuvant therapy* 4ys-DFS: 73% vs. 38%
EGFR-TKIs
specific for Ins20
ZENITH20-2 [44]Exon 20 insertionsPoziotininbORR: 27.8%
DCR: 70%
PFS: 5.5 mo.
EXCLAIM [45]Exon 20 insertionsMobocertinibORR: 43%
PFS: 7.3 mo.
*: statistically significant difference; **: no statistically significant difference.
Table 3. Clinical trials evaluating EGFR-TKI treatment combinations.
Table 3. Clinical trials evaluating EGFR-TKI treatment combinations.
Clinical TrailEGFR StatusComparisonResults
IFCT-1503
ACE-Lung [47]
Exon 19 deletions, L858R, G719X, L861Q, and S768I mutations or exon 19 insertionsAfatinib plus cetuximab vs. afatinibEnded for futility
BELIEF [52]Common mutations +/− T790MErlotinib plus bevacizumabPFS in ITT: 13.2 mo.
PFS in T790M-positive group: 16 mo.
PFS in the T790M-negative group: 10.5 mo.
JO25567 [53]Common mutationsErlotinib single agent or with bevacizumab* PFS: 16 vs. 9.7
** OS: 47.0 vs. 47.4 mo.
RELAY [54]Common mutationsErlotinib plus ramucirumab vs. erlotinib* PFS: 19.4 vs. 12.4 mo.
OS: immature data
FLAURA2 [56]Common mutations +/− T790MChemotherapy plus osimertinibOngoing
*: statistically significant difference; **: no statistically significant difference.
Table 4. Early-phase combinations for EGFR-mutated NSCLC patients with osimertinib resistance.
Table 4. Early-phase combinations for EGFR-mutated NSCLC patients with osimertinib resistance.
Clinical TrailPhaseArm(s)Endpoint
NCT02143466 [155]1bOsimertinib + savolitinibORR: 33%
PFS: 5.4 mo.
SAVANNAH2Osimertinib + savolitinibORR (ongoing)
SACHI3Osimertinib + savolitinib vs. pemetrexed + platinumPFS (ongoing)
TATTON [133]1bOsimertinib plus: selumetinib (MEK1/2 inhibitor), savolitinib (MET-TKI), or durvalumab (anti-PD-L1).Safety and tollerability
INC280 [156]1b/2Capmatinib plus gefitinibORR: 27%
GEOMETRY-E3Capmatinib + osimertinib vs. pemetrexed + platinumDLT, PFS (ongoing)
INSIGHT [157]1b/2Tepotinib plus gefitinib vs. standard platinum chemotherapyPFS: 4.9 vs. 4.4 mo.
OS: 17.3 vs. 18.7 mo.
MARIPOSA [158]3Amivantanab + lazertinib vs. osimertinibPFS (ongoing)
NCT045457102Osimertinib + abemaciclibPFS
NCT034558291/2Osimertinib + lerociclibDLT, RP2D, AEs, PFS
NCT01090011 [159]1bAfatinib + cetuximabORR: 30%
PFR: 5 mo.
NCT01861223 [160]1b/2Afatinib + nimotuzumabORR: 23%
PFS: 4.3 mo.
OS: 11.7 mo.
SQUIRE [161]3Necitumumab + gemcitabine + cisplatin vs. gemcitabine + cisplatinOS: 11.5 vs. 9.9 mo.
NCT02520778 [162]1bOsimertinib plus navitoclaxORR: 100%
PFS: 16.8 mo.
IMPRESS [163]3Gefitinib + cisplatin + pemetrexed vs. cisplatin + pemetrexedOS: 13.4 vs. 19.5 mo.
PFS: 5.4 vs. 5.4 mo.
NEJ009 [164]3Gefitinib vs. gefitinib + carboplatin+ pemetrexedOS: 49 vs. 38.5 mo.
PFS: 20.9 vs. 11.9 mo.
jRCTs071180062 [165]2Osimertinib + carboplatin + pemetrexed vs. osimertinibPFS: 15.8 vs. 14.6 mo.
ORR: 71.4% vs. 53.6%
FLAME2Osimertinib plus chemotherapy vs. osimertinibPFS (ongoing)
COMPEL2Osimertinib plus chemotherapy vs. osimertinibPFS (ongoing)
FLAURA 2 [56]3Osimertinib + platinum–pemetrexed vs. platinum–pemetrexedPFS (ongoing)
OS (ongoing)
CheckMate 012 [166]1Nivolumab + erlotinibORR:15%
CAURAL [167]3Osimertinib + durvalumab vs. osimertinibAEs
IMpower150 [168]3Atezolizumab plus bevacizumab plus carboplatin plus paclitaxel vs. bevacizumab plus carboplatin plus paclitaxelOS: 19.5 vs. 14.7 mo.
ORIENT-31 [169]3Sintilimab + bevacizumab biosimilar IBI305 + chemotherapy vs. chemotherapy alonePFS: 6.9 vs. 4.3 mo.
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

Laface, C.; Maselli, F.M.; Santoro, A.N.; Iaia, M.L.; Ambrogio, F.; Laterza, M.; Guarini, C.; De Santis, P.; Perrone, M.; Fedele, P. The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies. Pharmaceutics 2023, 15, 1604. https://doi.org/10.3390/pharmaceutics15061604

AMA Style

Laface C, Maselli FM, Santoro AN, Iaia ML, Ambrogio F, Laterza M, Guarini C, De Santis P, Perrone M, Fedele P. The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies. Pharmaceutics. 2023; 15(6):1604. https://doi.org/10.3390/pharmaceutics15061604

Chicago/Turabian Style

Laface, Carmelo, Felicia Maria Maselli, Anna Natalizia Santoro, Maria Laura Iaia, Francesca Ambrogio, Marigia Laterza, Chiara Guarini, Pierluigi De Santis, Martina Perrone, and Palma Fedele. 2023. "The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies" Pharmaceutics 15, no. 6: 1604. https://doi.org/10.3390/pharmaceutics15061604

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