Next Article in Journal
Acknowledgment to the Reviewers of Cancers in 2022
Next Article in Special Issue
Antigene MYCN Silencing by BGA002 Inhibits SCLC Progression Blocking mTOR Pathway and Overcomes Multidrug Resistance
Previous Article in Journal
Prognostic Significance of Integrin Subunit Alpha 2 (ITGA2) and Role of Mechanical Cues in Resistance to Gemcitabine in Pancreatic Ductal Adenocarcinoma (PDAC)
Previous Article in Special Issue
Both In Situ and Circulating SLC3A2 Could Be Used as Prognostic Markers for Human Lung Squamous Cell Carcinoma and Lung Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Treatment Strategies for Non-Small Cell Lung Cancer with Common EGFR Mutations: A Review of the History of EGFR TKIs Approval and Emerging Data

by
Julian A. Marin-Acevedo
1,†,
Bruna Pellini
2,3,*,†,
ErinMarie O. Kimbrough
4,
J. Kevin Hicks
5 and
Alberto Chiappori
2,3,*
1
Division of Medical Oncology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
2
Department of Thoracic Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
3
Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
4
Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
5
Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, FL 33612, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(3), 629; https://doi.org/10.3390/cancers15030629
Submission received: 19 December 2022 / Revised: 11 January 2023 / Accepted: 13 January 2023 / Published: 19 January 2023
(This article belongs to the Special Issue Lung Cancer - Molecular Insights and Targeted Therapies)

Abstract

:

Simple Summary

The management of non-small cell lung cancer with a common EGFR mutation has evolved over the past decades. While frontline use of second- or third-generation EGFR tyrosine kinase inhibitors (TKIs) is preferred over first-generation EGFR-TKIs, choosing the ideal agent depends on multiple factors (drug availability, physician comfort, specific EGFR mutation, presence of brain metastasis, etc.). Furthermore, defining subsequent therapies at the time of progression will rely on numerous variables (extent of disease, frontline EGFR TKI generation used, mechanism of resistance, etc.). Consequently, defining an optimal sequencing strategy is both, crucial and challenging. In this review, we present a detailed summary of evidence supporting the use of EGFR TKIs with or without other therapeutic approaches, outline available options at the time of disease progression, summarize investigational strategies, and suggest an approach to therapeutic sequencing in patients with common EGFR mutations.

Abstract

The development of targeted therapies over the past two decades has led to a dramatic change in the management of EGFR-mutant non-small cell lung cancer (NSCLC). While there are currently five approved EGFR tyrosine kinase inhibitors (TKIs) for treating EGFR-mutant NSCLC in the first-line setting, therapy selection after progression on EGFR TKIs remains complex. Multiple groups are investigating novel therapies and drug combinations to determine the optimal therapy and treatment sequence for these patients. In this review, we summarize the landmark trials and history of the approval of EGFR TKIs, their efficacy and tolerability, and the role of these therapies in patients with central nervous system metastasis. We also briefly discuss the mechanisms of resistance to EGFR TKIs, ongoing attempts to overcome resistance and improve outcomes, and finalize by offering treatment sequencing recommendations.

1. Introduction

Sensitizing mutations in the epidermal growth factor receptor (EGFR) gene are one of the most common targetable genomic alterations in non-small cell lung cancer (NSCLC). These can be found in ~15% of lung adenocarcinomas in the United States and 22–64% of lung adenocarcinoma in Asian patients [1,2]. In the past two decades, five EGFR tyrosine kinase inhibitors (TKIs) have become commercially available for the management of advanced NSCLC with common EGFR-sensitizing mutations [i.e., EGFR exon 19 deletions or exon 21 mutations (L858R)]: erlotinib, gefitinib, afatinib, dacomitinib, and osimertinib (Table 1; Figure 1) [3,4,5,6,7,8,9]. These drugs exhibit distinct activity and safety profiles and are divided into first- (e.g., erlotinib, gefitinib), second- (e.g., afatinib, dacomitinib), and third-generation (e.g., osimertinib) TKIs. The second- and third-generation TKIs, afatinib and osimertinib, have demonstrated extended activity against some uncommon EGFR mutations [e.g., T790M (osimertinib), G719X, L861Q, or S768I (afatinib and osimertinib)] [10,11,12,13,14].
The efficacy of the EGFR TKIs has improved with each new generation. First- and second-generation EGFR TKIs have superior response rates (RRs) and progression-free survival (PFS) compared to platinum-doublet chemotherapy (i.e., cisplatin or carboplatin combined with either gemcitabine, pemetrexed, paclitaxel, or docetaxel) [3,4,5,6,15]. Dacomitinib and osimertinib confer improved PFS and overall survival (OS) rates compared to the first-generation EGFR TKIs [7,8,16,17]. Afatinib confers superior PFS and OS compared to platinum-doublet chemotherapy but did not improve OS compared to first-generation EGFR TKIs [6,18,19]. While first-generation EGFR TKIs have been compared head-to-head with second- and third-generation TKIs and have proven to be less effective, second- and third-generation EGFR TKIs have not been compared head-to-head in a prospective clinical trial. Therefore, it is unclear if these drugs confer the same outcomes for patients.
Despite EGFR TKIs improved efficacy over chemotherapy, drug resistance eventually occurs [20]. A wide array of genomic pathways activation and transcriptional remodeling have been reported as mechanisms of resistance [20]. These findings have prompted investigation of novel combinations and therapeutics. In this review, we discuss the data to support the use of EGFR TKIs alone or in combination, describe developing therapies for resistant disease, and propose a treatment sequencing strategy based on the available data.

2. Exploring the Use of EGFR TKIs

2.1. EGFR TKIs versus Platinum-Doublet Chemotherapy

In July 2002, gefitinib became the first EGFR TKI to be approved in the world, specifically in Japan, for advanced NSCLC [21]. Two years later, erlotinib was approved in the US for unselected patients with advanced NSCLC [22,23]. Nine years later it was approved in the first-line setting for the treatment of advanced NSCLC with an EGFR exon 19 del (ex19del) or L858R mutation based on findings of the EURTAC study [3,24]. This phase 3 trial demonstrated improvement in RR (64% vs. 18%) and PFS (9.7 months vs. 5.2 months; p < 0.0001) in the intention-to-treat patient population when compared to a platinum-doublet [3]. Erlotinib was better tolerated, and its use was associated with less serious adverse events (AEs) (6% vs. 20%) (Table 1) [3].
In 2009, the European Medicines Agency (EMA) approved gefitinib for the treatment of advanced EGFR-mutant NSCLC based on the phase 3 clinical trial IPASS [4,25]. Gefitinib showed improved RR (43% vs. 32%) and OS (18.8 vs. 17.4 months; p = 0.109) compared to chemotherapy in a population of light/never-smokers East Asians patients with advanced pulmonary adenocarcinoma [4,26]. Subgroup and post-hoc analyses of this study confirmed that gefitinib conferred a superior RR for all patients with EGFR-mutant NSCLC (71% vs. 47%). Specifically, the RR was 85% vs. 43% for patients with ex19del and 61% vs. 53% for the ones with L858R mutations [4,26]. Following IPASS, several landmark trials have confirmed the superiority of gefitinib over platinum-doublet in the first-line setting for patients with EGFR-mutant NSCLC in East Asian (NEJ-002, WJTOG3405) and White patients (IFUM) (Table 1) [27,28,29,30,31]. In 2015, the U.S. Food and Drugs administration (FDA) approved gefitinib for first-line treatment of metastatic NSCLC [32].
Afatinib was the first second-generation EGFR TKI approved by the FDA in 2013 [33]. It demonstrated improved PFS and RR compared to chemotherapy in the LUX-Lung 3 (PFS 11 vs. 7 months, p = 0.0004; RR 56% vs. 23%) and the LUX-Lung 6 (PFS 14 vs. 6 months, p < 0.0001; RR 67% vs. 23%) clinical trials [5,6,9,34]. Interestingly, in both trials the OS benefit from afatinib was seen among patients with EGFR ex19del mutations but not in those with L858R mutations (Table 1) [6]. Up to 49% of patients receiving afatinib developed serious toxicities, however, cessation of treatment due to side effects was more common among those receiving chemotherapy [5,6,9]. According to the retrospective RealGiDo study, afatinib can be dose-reduced to improve tolerability without affecting efficacy [35].
Osimertinib was initially approved for patients with EGFR T790M mutant-NSCLC who had progressed on or after EGFR TKI therapy [36]. The phase 3 AURA3 trial demonstrated improved RR (71% vs. 31%) and median PFS (10.1 vs. 4.4 months; p < 0.001) in those receiving osimertinib vs. platinum-based chemotherapy [37]. In this study, osimertinib also demonstrated activity against asymptomatic central nervous system (CNS) metastasis while showcasing a safe toxicity profile. Only 23% of patients experienced serious AEs compared to 47% of patients in the chemotherapy group. Osimertinib was only discontinued in 7% of patients [37]. While there was a trend towards improved OS with osimertinib (26.2 vs. 22.5 months), this difference did not reach statistical significance (p = 0.277) [38].

2.2. Second- and Third-Generation EGFR TKIs versus First-Generation EGFR TKIs

The phase 2b LUX-Lung 7 clinical trial evaluated the use of afatinib vs. gefitinib in treatment-naïve patients with advanced EGFR-mutant NSCLC [19]. Afatinib conferred longer PFS (11 vs. 10.9 months; p = 0.017) and time-to-treatment failure (13.7 vs. 11.5 months; p = 0.0073) [19]. There were no differences in OS. The rate of toxicities and discontinuation of therapy were also similar (Table 1) [18,19]. In contrast, a real-world study showed that afatinib improved PFS and 1-year OS rates (16.4 months, p = 0.005; 78.2%; p = 0.004) compared to gefitinib (10.3 months and 69.1%) and erlotinib (12.1 months and 71.6%) [39].
Dacomitinib was approved in 2018 based on the phase 3 ARCHER 1050 trial [8,40,41]. In this study, first-line dacomitinib resulted in improved PFS (15 vs. 9 months; p < 0.0001) and OS (34 vs. 27 months; p = 0.438) compared to gefitinib [8,16]. The toxicities and rate of therapy discontinuation were similar in both groups (Table 1) [8,16].
Osimertinib was approved for the first-line treatment of EGFR-mutant NSCLC in 2018 based on results of the phase 3 FLAURA trial [7,36]. In this study, osimertinib demonstrated similar RR (80% vs. 76%) and disease control rate (DCR: 97% vs. 92%) to first-generation EGFR TKIS, but longer PFS (19 vs. 10 months; p < 0.001) and OS (39 vs. 32 months; p = 0.046) [7,17]. It was also better tolerated than gefitinib or erlotinib (Table 1) [7,17]. Based on these combined findings, osimertinib, afatinib and dacomitinib are preferred over first-generation EGFR TKIs in the front-line setting.

2.3. Second-Generation EGFR TKIs Following First-Generation EGFR TKI Failure

The phase 2 LUX-Lung 4 trial evaluated the use of afatinib in patients who progressed after treatment with erlotinib or gefitinib [42]. The RR was 8% and the PFS was 4.4 months. Approximately 37% of patients experienced a grade ≥3 toxicity and 29% of patients discontinued afatinib due to serious AEs [42]. Given the limited clinical efficacy, afatinib is not used in this setting.

2.4. Third-Generation EGFR TKIs Following First and Second-Generation EGFR TKI Failure

The phase 1/2 AURA trial evaluated the use of osimertinib after progression on gefitinib or erlotinib. The DCR and PFS were 84% and 8.2 months for all-comers, and 95% and 9.6 months for those with a T790M mutation, respectively [43]. The phase 2 AURA2 trial evaluated osimertinib in patients with EGFR T790M, who progressed on any first- or second-generation TKI. The DCR was 92%, RR was 70%, and PFS was 9.9 months. Approximately 34% of patients experienced a grade ≥3 toxicity but only 5% discontinued osimertinib due to an AE [44].
The GioTag observational study evaluated the role of sequencing osimertinib after afatinib failure in patients with EGFR T790M mutations [45]. The OS was 37.6 months for all-comers, 41.6 months for those with co-existing EGFR del19ex mutation, and 44.8 months for Asian patients [45]. A retrospective study from South Korea also demonstrated a role for sequencing osimertinib after afatinib among those with EGFR T790M-mutant NSCLC [46]. In this analysis, the median time on treatment for patients who received osimertinib was 20.8 months while the 2- and 3-year OS rates were 86% and 69%, respectively [46]. Therefore, osimertinib is recommended for patients with EGFR T790M-mutant NSCLC who progress after a first- or second-generation TKI.

2.5. EGFR TKIs in Combination with Anti-Vascular Endothelial Growth Factors

Combination therapies targeting the vascular endothelial growth factor (VEGF), or the VEGF receptor (VEGFR), and EGFR have been studied [47]. The phase 3 NEJ026 trial evaluated erlotinib +/− bevacizumab in Japanese patients with advanced EGFR-mutant NSCLC [48]. Erlotinib plus bevacizumab resulted in improved PFS (16.9 vs. 13.3 months; p = 0.016) without increasing toxicity rates [48]. Similarly, the phase 3 RELAY trial evaluating erlotinib +/− ramucirumab demonstrated an improved PFS in those receiving combination therapy (19 vs. 12 months; p < 0.0001) [49]. Grade ≥3 toxicities and treatment discontinuation, however, were more common in the combination therapy group (Table 1) [49]. While the median OS data is not yet available, there were no differences at 1 and 2 years between the two treatment groups [49].
Afatinib plus bevacizumab were evaluated in a phase 1 clinical trial in Japan achieving a RR of 81% [50]. Similarly, in an observational study from Taiwan using the same combination therapy the RR was 88%, the PFS was 24 months, and the OS was 46 months [51].
Osimertinib in combination with anti-VEGF has also been investigated. A phase 1/2 trial (NCT02803203) evaluated first-line osimertinib with bevacizumab [52]. The RR was 80% and the PFS was 19 months [52]. Approximately 31% of patients discontinued bevacizumab due to toxicity [52]. This combination was also evaluated in Japanese (WJOG 8715L) and European (BOOSTER) patients with EGFR T790M-mutant NSCLC who developed disease progression after a first- or second-generation EGFR TKI [53,54]. These phase 2 trials failed to demonstrate an improvement in PFS compared to osimertinib alone (14 vs. 9 months; p = 0.20 in the WJOG 8715L Trial, and 15 vs.12 months; p = 0.83 in the BOOSTER Trial) [53,54]. Combination therapy resulted in a significantly shorter time to treatment failure (8 vs. 11 months; p = 0.0074) and an increased incidence of grade ≥3 toxicities (47% vs. 18%) [54]. Another phase 2 trial (WJOG9717L) evaluated the use of osimertinib with bevacizumab in patients with common EGFR mutations [55]. The median PFS was similar to osimertinib alone (22.1 vs. 20.2 months; HR 0.862 p = 0.213), while the rate of grade ≥3 toxicities was higher in the combination group (56% vs. 48%) [55]. These combined findings suggest osimertinib plus bevacizumab does not improve outcomes and increases toxicities.
Currently, there are several ongoing studies evaluating bevacizumab or ramucirumab with second- or third-generation EGFR TKIs in the frontline setting (Clinicaltrials.gov: NCT04575415, NCT04148898, NCT03909334, NCT02971501, NCT04181060, accessed on 9 January 2023). The results of these studies are awaited and may affect current treatment paradigms.

2.6. EGFR TKIs in Combination with Chemotherapy

The use of EGFR TKIs with chemotherapy has been investigated in retrospective and prospective studies [56,57,58,59,60,61]. Although the results of these studies are mixed, some suggest that combination therapy may be more beneficial in selected treatment-naïve patients [56,57,58,59,60,61].
Erlotinib plus chemotherapy was evaluated as first-line therapy in patients with advanced/metastatic NSCLC with or without common EGFR mutation in the phase 2 trial CALGB 30406 [56]. This study included 66 patients with a common EGFR mutation. Of these, 33 received erlotinib monotherapy and 33 received erlotinib with paclitaxel and carboplatin. There were no significant differences in PFS for all-comers between the two treatment groups (PFS = 5.0 vs. 6.6 months; p = 0.1988) [56]. The phase 3 IMPRESS trial evaluated chemotherapy +/− gefitinib after disease progression on gefitinib [57]. The PFS was 5.4 months in both groups, however, more toxicities were seen in the combination arm [57]. Two phase 3 trials, one in India (CTRI/2016/08/007149) and one in Japan (NEJ009), evaluated gefitinib in combination with chemotherapy versus gefitinib alone in treatment-naïve patients [58,59]. The combination arms resulted in improved RR, longer PFS and OS. However, grade ≥3 toxicities were more frequent in the combination arms [58,59].
The LUX-Lung 5 trial evaluated afatinib plus chemotherapy vs. single agent chemotherapy in patients with advanced EGFR-mutant NSCLC who had progressed after treatment with first-generation EGFR TKI, chemotherapy, and afatinib [60]. In this phase 3 clinical trial, afatinib plus chemotherapy resulted in improved RR (32.1% vs. 13.2%; p = 0.005) and PFS (5.6 vs. 2.8 months; p = 0.003), however, there was no difference in OS between the two groups [60]. The incidence of grade ≥3 toxicities was higher in the combination group [60].
A retrospective study evaluated osimertinib plus chemotherapy in the third-line setting or beyond [61]. The median duration of treatment was 6.1 months, and the median OS was 10.4 months (95% CI 7.0–13.2 months). According to the authors, the OS was slightly inferior compared to the AURA3 trial likely reflecting that their populations was more heavily pre-treated than AURA3 [61]. Approximately 27% of patients developed grade ≥3 toxicities. The rate of osimertinib discontinuation was 2% [61]. Currently, the use of osimertinib in combination with chemotherapy is being investigated as first-line therapy in EGFR-mutant NSCLC in the phase 3 FLAURA2 trial (NCT04035486) and in patients with detectable EGFR mutations in ctDNA in two phase 2 studies (NCT04410796, NCT05281406). Preliminary data from FLAURA2 suggests that the combination is well tolerated and safe [62].

2.7. EGFR TKIs in Combination with Immunotherapy

The use of first-generation EGFR TKIs in combination with immune checkpoint inhibitors (ICIs) has been investigated in three phase 1 clinical trials in patients with EGFR-mutant NSCLC [63,64,65]. While response to therapy appeared promising, grade ≥3 toxicities were seen in more than 40% of patients and therapy was discontinued in 35% of patients [63,64,65].
Osimertinib was combined with durvalumab in one arm of the phase 1b TATTON trial [66]. The RR was 43% with this combination, however, the treatment arm was terminated because of increased reports of interstitial lung disease (ILD) [66]. The phase 3 CAURAL trial evaluated osimertinib with or without durvalumab in patients with EGFR T790M mutations who had received prior EGFR TKI [67]. The RR was 80% and no grade ≥3 toxicities were seen among the first 14 treated patients. However, recruitment was terminated due to the high rates of ILD reported in the TATTON trial [66,67]. While the combination of EGFR TKIs with ICIs appeared promising, the incidence and severity of AEs seems prohibitive.

3. Mechanisms of Resistance to EGFR TKIs

Cancer cells may develop innate or acquired resistance to EGFR TKI therapy (Table 2). Primary resistance occurs from coexisting uncommon EGFR mutations, mutations in genes other than EGFR, or heterogeneity to TKI response [68]. Acquired resistance is defined as progression on an EGFR TKI in a patient with a common EGFR mutation that achieved significant or durable (≥6 months) clinical response [69]. Acquired resistance mechanisms are further subclassified as EGFR-dependent and -independent, but these can coexist and overlap [70,71].
EGFR-dependent mechanisms lead to an increase in EGFR kinase activity. The most common is the development of the EGFR T790M mutation (“gatekeeper”) which accounts for 25–50% of cases of treatment failure after first- and second-generation EGFR TKIs [7,68,70,72,73,74,75,76,77,78,79]. The EGFR C797X mutations are also commonly seen. Specifically, the EGFR C797S mutation accounts for ~7–8% of cases of osimertinib resistance when used as first-line therapy and 10–26% of cases when used as second-line therapy [80,81]. Other mutations like EGFR L792X, G769X, L718Q, G719A, G724S, or exon 18 variants D761Y, S768I, V769L are less common [77,78,79,80,82,83].
EGFR-independent mechanisms include bypass mechanisms and histologic/phenotypic transformation [70,71,81,84,85,86,87,88,89]. EGFR proteins, as members of the ERBB/HER family, normally interact with other ERBB/HER family members to create dimers that phosphorylate and activate downstream signaling pathways [20]. Bypass mechanisms may be a result of ERBB2/HER2 mutations/amplifications that form EGFR/HER2 dimers or active HER2 molecules with downstream activating effects [20]. MET amplifications can promote persistent HER3 tyrosine kinase activity with downstream activation [20]. Other bypass mechanisms of resistance may result from PIK3CA, BRAF, KRAS, and MET exon 14 skipping mutations, as well as RET and FGFR3 fusions [84,85,86,87,88]. Upregulation of PD-L1 leading to immune escape has also been described [90]. Histologic transformation to small cell lung cancer (SCLC) occurs in 3–14% of NSCLC patients treated with EGFR TKIs [70,71,81,89]. It has been suggested that initial biopsies may fail to capture pre-existing SCLC and that treatment with EGFR TKIs results in regression of the NSCLC component while allowing the SCLC component to progress [91]. Patients with concurrent EGFR/RB1/TP53 mutations seem to be at a particularly high risk of undergoing SCLC histologic transformation [92]. Epithelial-to-mesenchymal transition (EMT) is another mechanism of resistance affecting ~5% of EGFR-TKI resistant tumors [71]. EMT occurs following genetic changes in cancer cells that allow them to transition from having an epithelial to having a mesenchymal phenotype. This transition enables cancer cells to migrate, invade surrounding tissue, and become resistant to therapy [71,93,94].

4. Therapy Following EGFR TKI

The therapy choice after progression on EGFR TKIs varies according to symptoms, metastatic burden, mechanism of resistance, and the class of EGFR TKI used in the front-line setting. In patients who are initially treated with first- or second-generation EGFR TKIs and develop an EGFR T790M mutation, osimertinib is preferred over chemotherapy [37,38,44,61,95]. Patients who develop asymptomatic disease progression or oligoprogression (3 to 5 new metastasis) while on an EGFR TKI, but do not acquire an EGFR T790M mutation, should continue treatment with the same EGFR TKI plus local palliative therapy (surgery or radiation) to sites of active disease [96,97,98]. For symptomatic patients with multiple new metastases after an EGFR TKI without an EGFR T790M mutation, a change in systemic therapy is recommended (Table 3) [98,99,100,101,102,103]. The use of first- or second-generation TKIs after progression on osimertinib is not recommended given the poor disease control and short PFS [104].
Chemoimmunotherapy is considered the standard of care for patients with advanced NSCLC, but studies have often excluded patients with EGFR mutations [99,100,101]. The phase 3 trials IMpower130, IMpower150, and ORIENT-31, included patients with EGFR-mutant NSCLC after progression on EGFR TKIs [102,105,106,107]. IMpower150 evaluated the combination of carboplatin, paclitaxel, atezolizumab, and bevacizumab. This combination resulted in a trend towards improved RR (71% vs. 42%), PFS (10 vs. 7 months; HR 0.61–95% CI 0.36–1.03), and OS (26 vs. 20 months; HR 0.91–95% CI 0.53–1.59) compared to chemotherapy plus bevacizumab alone among patients with EGFR-mutant NSCLC; however, the differences were not statistically significant [105,106]. Further, there was no significant improvement in RR, PFS, or OS in the those receiving chemoimmunotherapy versus chemotherapy plus bevacizumab (Table 3) [106]. Similar results were seen in the IMpower130 trial, where the addition of immunotherapy to chemotherapy did not result in improved PFS or OS among patients with EGFR-mutant NSCLC [102]. The ORIENT-31 trial evaluated the combination of an anti-PD-1 agent (sintilimab), an anti-VEGF agent (IBI305), and chemotherapy in EGFR-mutant NSCLC after EGFR TKI failure [107]. Preliminary results demonstrated an improvement in PFS with chemoimmunotherapy plus anti-VEGF vs. chemotherapy alone (6.9 vs. 4.3 months; p < 0.0001). This improvement was also seen with the use of chemoimmunotherapy compared to chemotherapy alone (5.6 vs. 4.3 months; p < 0.0584) [107]. The safety and efficacy of chemoimmunotherapy in EGFR-mutant NSCLC is being assessed in ongoing phase 2 and 3 trials (NCT03786692, Checkmate 722–NCT02864251).
Expression of PD-L1 seems to play a role in response to immunotherapy alone in EGFR-mutant NSCLC [108,109,110]. A subgroup analysis in the KEYNOTE-001 trial suggested that patients with common EGFR mutations and PD-L1 ≥50% had better RR with pembrolizumab than those with PD-L1 <1% [108]. Similarly, the phase 2 ATLANTIC trial evaluating the use of durvalumab after ≥2 lines of therapy in those with EGFR-mutant NSCLC, demonstrated that patients with PD-L1 ≥25% had improved RR (12% vs. 4%) and OS (13.3 vs. 9.9 months) compared to those with PD-L1 <25% [109,110].

5. Novel EGFR TKIs and Targeted Therapies

Savolitinib, an oral TKI against c-MET, was combined with osimertinib in patients with EGFR-mutant NSCLC with MET amplifications/mutations after progression on osimertinib in the phase 2 trial ORCHARD [111]. The RR was 41% among the 17 evaluable patients, while PFS and OS was not reported [111]. The phase 1b TATTON trial evaluated the combination of osimertinib and savolitinib in patients who had progressed on osimertinib. Combination therapy resulted in a RR of 30% and a PFS of 5.4 months [112]. INSIGHT-2 is an ongoing phase 2 trial evaluating tepotinib (MET inhibitor) plus osimertinib in patients with MET amplification after progression on osimertinib (NCT03940703).
The phase 1 CHRYSALIS trial investigated amivantamab (bi-specific antibody against EGFR and MET) +/− lazertinib (third-generation EGFR TKI) in patients with EGFR-mutant NSCLC who progressed on osimertinib [113]. Preliminary results demonstrated a RR of 19% in the monotherapy group and 36% in those receiving combination therapy [113]. The phase 1 CHRYSALIS-2 trial evaluated amivantamab plus lazertinib in patients with EGFR-mutant NSCLC who progressed on osimertinib and platinum-based chemotherapy. Among the 50 evaluable patients, the RR was 36%, the median duration of response was not reached, and grade ≥3 toxicities mainly included infusion reactions, dermatitis, and hypoalbuminemia [114]. The amivantamab plus lazertinib combination has been moved to phase 3 investigation under the MARIPOSA trial. The latter is comparing frontline amivantamab plus lazertinib versus osimertinib alone in treatment-naïve patients with advanced NSCLC with common EGFR mutations (NCT04487080) [115].
Patritumumab deruxtecan, an anti-HER3 antibody-drug conjugate (ADC), was investigated in a phase 1 clinical trial (U3-1402) in patients with EGFR-mutant NSCLC who progressed on EGFR TKI [116]. The RR was 39% and the PFS was 8.2 months [116]. The phase 2 HERTHENA-Lung01 trial is underway to confirm these findings (NCT04619004).
Datopotamab deruxtecan, an ADC directed against Trop-2, was investigated in the phase 1 TROPION-PanTumor01 trial in patients with advanced NSCLC with an actionable mutation (including EGFR) who had previously progressed after treatment with a TKI and chemotherapy [117]. The RR was 35% and the median duration of response was 9.5 months [117]. The phase 3 TROPION-Lung01 trial is underway to confirm these findings (NCT04656652).

6. Special Considerations: Brain, Liver, and Bone Metastases

Retrospective data suggest that the metastatic pattern does not differ among patients with EGFR-mutant NSCLC compared to those without EGFR mutations [118]. Approximately 25% of patients with advanced EGFR-mutant NSCLC have brain metastases at the time of diagnosis and ~50% develop these within 3 years [119]. The management of brain metastasis may be challenging depending on the location and the number of lesions. Therefore, it is important to select an EGFR TKI with good CNS coverage. Unfortunately, many EGFR TKIs trials excluded patients with brain metastases [4,8,28,49]. A retrospective study assessing first-generation EGFR TKIs demonstrated that up to 12% of patients receiving first-line erlotinib and 18–30% of patients receiving first-line gefitinib developed CNS disease progression [120]. Findings from a retrospective Japanese study suggest that patients treated with erlotinib had a lower chance of developing CNS metastasis than those treated with gefitinib (4.8% vs. 24.5%; p = 0.04) [121].
The LUX-Lung 3 and LUX-Lung 6 trials included patients with asymptomatic brain metastases but did not report the rates of CNS progression [5,9]. This was reported in a cohort study in Taiwan [122]. Approximately 18% (N = 47) of patients treated with front-line afatinib developed CNS progression [122]. Among patients without known CNS metastasis, 11% developed brain metastases, while 33% of patients with known brain metastases had CNS disease progression [122]. The ARCHER 1050 study excluded patients with brain metastasis, however, 0.44% of patient treated with dacomitinib and 4.9% receiving gefitinib developed CNS disease [8,123]. In a series of 14 patients with brain metastases treated with first-line dacomitinib, nearly 86% had improvement of their CNS disease suggesting dacomitinib has CNS activity [124]. The CNS activity of dacomitinib is currently under investigation (NCT04675008).
The FLAURA trial allowed the enrollment of patients with neurologically stable CNS metastasis [7]. Approximately 6% of patients treated with osimertinib had progressive CNS disease versus 15% of patients treated with erlotinib or gefitinib [7]. In patients without known or treated CNS disease, 3% of patients on osimertinib and 7% receiving standard EGFR TKI developed CNS disease [125]. The CNS PFS was longer in those receiving osimertinib compared to standard EGFR TKI (median CNS PFS, not reached vs. 13.9 months; HR, 0.18; p = 0.014) [125]. The CNS RR in those with known brain metastases receiving osimertinib was better than in those receiving standard EGFR TKI (91% vs. 68%) [7,126].
Combination therapy may provide improved control of CNS disease. The RELAY study evaluating erlotinib +/− ramucirumab also excluded patients with brain metastases. In this study, however, only two patients (0.9%) treated with erlotinib plus ramucirumab developed CNS metastasis versus eight patients (3.6%) in the placebo plus erlotinib group [49]. It is unclear which approach between second-, third-generation EGFR TKIs, or combination anti-EGFR/VEGF therapy will result in better CNS outcomes. Osimertinib, however, remains the agent with the strongest evidence supporting its use in prevention and treatment of CNS metastasis.
Liver metastases affect 14–17% of patients with EGFR-mutant NSCLC [118,127]. This incidence is similar to that seen among EGFR wild-type NSCLC, suggesting EGFR mutations do not confer a higher risk for developing liver metastasis [128]. Although the treatment of patients with liver metastasis does not usually differ from patients without liver involvement, outcomes tend to be worse when liver metastases are present, even with the use of EGFR TKIs like osimertinib [128,129,130].
In contrast to liver, bone metastases occur more commonly in EGFR-mutant (40–54%) than in EGFR wild-type NSCLC (32%) [118,131]. Bone metastases seem to be associated with a lower risk of death among patients with EGFR-mutant NSCLC [132]. Furthermore, patients with EGFR mutations and bone metastases appear to have better OS than those without EGFR mutations and bone metastasis [132]. Retrospective data suggest that the use of osimertinib is associated with better clinical outcomes than the use of first- or second-generation EGFR TKIs in patients with this metastatic pattern [130]. Finally, the addition of bisphosphonates to therapy not only prevents skeletal complications but also seems to enhance the effect of EGFR TKIs and improve PFS [132].

7. Treatment Sequencing: A Suggested Approach

The ideal sequencing of EGFR TKIs and other therapies for patients with EGFR-mutant NSCLC remains uncertain. While osimertinib is often incorporated in the treatment of EGFR-mutant NSCLC, the timing of its incorporation remains unclear. The FLAURA trial demonstrated that frontline osimertinib conferred an advantage in PFS and OS when compared to first-generation EGFR TKIs [7]. After first- and second-generation TKI failure due to an acquired T790M mutation, osimertinib also improved outcomes compared to chemotherapy (AURA3 trial) [37]. However, it is unknown whether front-line osimertinib use results in more durable response than sequencing EGFR TKIs as in the AURA 1-3 trials [133,134,135].
Many experts advocate for the use of osimertinib in the front-line setting due to its better tolerability than first- and second-generation EGFR TKIs [134,135], and superior outcomes compared to first-generation EGFR TKIs [5,7,9,134]. However, other experts favor the use of osimertinib after progression on a first- or second-generation EGFR TKI to delay the use of chemotherapy [136,137]. The latter approach has some limitations. At the time of progression on first- or second-generation EGFR TKI, mutation analysis should be pursued, but testing for EGFR and other acquired mutations at progression may not be feasible or readily available [138,139]. According to the Flatiron Health database, evaluating a predominately US-based population, only 30% of patients were tested for EGFR mutations following progression on a first- or second-generation EGFR TKI [139]. Sequencing strategies also assume that patients who progress develop a T790M mutation. This mutation, however, only occurs in 25–50% of patients treated with a first- or second-generation EGFR TKIs [7,68,70,72,73,74,75,76,77,78,79]. A simulation study comparing first-line osimertinib to alternative EGFR TKI sequencing strategies suggests an improvement in PFS among those receiving osimertinib in the first-line setting regardless of the presence of a T790M mutation [135].
Sequencing EGFR TKIs also assumes patients will be fit to receive subsequent therapies, however, ~30% of patients will not be eligible for second-line treatment [139,140,141]. In the FLAURA trial, 35% of patients receiving first-generation EGFR TKIs did not receive second-line therapy [17]. A small retrospective real-world study conducted in three certified lung cancer centers in Germany also found that 30% of patients treated with front-line first- or second-generation EGFR TKIs did not receive second-line therapy due to poor performance status, CNS metastasis, rapid disease progression, or death [138]. Additionally, studies in the United States revealed that 28–30% of patients treated with frontline first- or second-generation EGFR TKI did not receive subsequent therapies [139,141]. Among those who received subsequent treatment, only 23% to 25% received osimertinib [139,141].
As it is difficult to predict the mechanisms of resistance and performance status at the time of disease progression, using upfront osimertinib over sequencing strategies may provide the patients the best outcomes for all-comers. If osimertinib is not available, a second-generation EGFR TKI is also a good choice, especially for those with uncommon EGFR mutations. The prospective APPLE study is evaluating the optimal strategy for osimertinib use (upfront vs. sequential) (NCT02856893) and hopefully will inform future EGFR TKIs sequencing.
The choice of upfront EGFR TKI may also be affected by the specific EGFR mutation. For example, in the LUX-Lung 3 and 6 trials, a subgroup analysis revealed superior OS in patients with EGFR ex19del receiving afatinib compared to chemotherapy [6]. However, there was no significant difference in OS in patients with L858R mutations receiving afatinib or chemotherapy [6]. In the FLAURA trial, patients receiving osimertinib who had an ex19del had improved OS compared to patients with L858R mutations [17]. In the ARCHER 1050 study, however, patients receiving dacomitinib who had L858R mutations only had a trend toward superior OS compared to patients with ex19del [16]. These combined findings suggest that ex19del mutations are associated with better prognosis.
The presence of CNS metastasis may also dictate the choice of front-line therapy. Osimertinib and afatinib are the only EGFR TKIs that have been evaluated in prospective trials in patients with CNS disease on presentation [5,7,9]. Osimertinib demonstrated superior CNS DCR and PFS compared to first-generation EGFR TKIs [7,121,125,126]. While there is no direct comparison with second-generation EGFR TKIs, preclinical data suggest that osimertinib provides improved CNS penetration compared to afatinib [142]. Approximately 11% of patients receiving front-line afatinib developed CNS metastases, while only 3% receiving front-line osimertinib developed CNS metastasis in the FLAURA trial [122,125].

8. Conclusions

Studies have demonstrated that the use of frontline second- and third-generation is preferred over first-generation EGFR TKIs in patients with EGFR-mutant NSCLC. Only osimertinib and dacomitinib have prospective data demonstrating improved PFS and OS over first-generation EGFR TKIs [16,17]. While there are no prospective studies to support an OS benefit of afatinib compared to first-generation EGFR TKIs, real-world evidence does suggest this benefit. Afatinib, osimertinib, and dacomitinib have not been compared head-to-head, therefore there is no strong evidence to support one over the other in the front-line setting. The use of erlotinib plus ramucirumab has also demonstrated good activity compared to erlotinib alone. However, OS data is lacking and patients with CNS metastasis were not included in this study. Furthermore, this approach is more costly and burdensome for patients as it involves infusions every 2 weeks.
When selecting a sequencing strategy for EGFR there are several aspects to consider, including CNS disease at presentation, the type of EGFR mutation at presentation, access to specific drugs, access to genomic testing at the time of progression, mechanisms of resistance, clinical performance status at progression, and provider level of comfort.
Evidence suggests osimertinib has better CNS penetration than afatinib and may provide better clinical outcomes in patients with bone metastasis, therefore we recommend it over other EGFR TKIs in patients with CNS or bone involvement. On the other hand, for patients with uncommon EGFR mutations, although osimertinib is an alternative, afatinib is the most extensively studied drug and the only one approved for this patient population [5,10,34,143].
When progression occurs and EGFR TKI therapy has been exhausted, a preferred therapeutic option is the use of chemoimmunotherapy with bevacizumab or the combination of chemotherapy plus bevacizumab, over chemotherapy alone. For patients who cannot receive an antiangiogenic agent, chemotherapy +/− EGFR TKI continuation should be considered. Immunotherapy alone could be another therapeutic option for selected patients with high PD-L1 levels (e.g., ≥25%) [109,110].
Finally, novel agents and drug combinations have shown promising results in early phase trials (Table 3). Ongoing studies evaluating the ideal sequencing and combination strategies to improve outcomes and overcome EGFR TKI resistance will hopefully inform the optimal treatment sequencing strategy (NCT04811001, NCT04413201, NCT04105153, NCT04035486, NCT03909334 and NCT02789345). Results of these studies are anxiously awaited.

Author Contributions

Conceptualization, J.A.M.-A. and B.P.; methodology, J.A.M.-A. and B.P.; investigation, J.A.M.-A. and B.P.; writing–original draft preparation, J.A.M.-A. and B.P.; writing–review and editing, J.A.M.-A., B.P., E.O.K., J.K.H. and A.C.; visualization, J.A.M.-A., B.P., J.K.H. and A.C.; supervision, B.P. and A.C. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was received for the conduct of this research.

Acknowledgments

Figure 1 was created using BioRender.com, accessed on 8 January 2023.

Conflicts of Interest

Pellini receives research support from Bristol Myers Squibb (to the institution), has received speaker honoraria from BioAscend, Merck, MJH Life Science, Play to Know AG, Grupo Pardini, Foundation Medicine, and has done consulting/advisory board work with Guidepoint, Guardant Health, Illumina, Regeneron, and AstraZeneca. B.P. reports funding from the Bristol Myers Squibb Foundation/the Robert A. Winn Diversity in Clinical Trials Awards Program, outside of the submitted work. Chiappori receives Research support from Bristol Myers Squibb, AstraZeneca, and Novartis. He has received speaker, consultant and/or advisor honoraria from Blueprint, Takeda, Genentech, Pfizer, Merck, Astra Zeneca, Janssen and Jazz. Hicks received research support from OneOme. Marin-Acevedo and Kimbrough disclose no conflicts.

References

  1. D’Angelo, S.P.; Pietanza, M.C.; Johnson, M.L.; Riely, G.J.; Miller, V.A.; Sima, C.S.; Zakowski, M.F.; Rusch, V.W.; Ladanyi, M.; Kris, M.G. Incidence of EGFR exon 19 deletions and L858R in tumor specimens from men and cigarette smokers with lung adenocarcinomas. J. Clin. Oncol. 2011, 29, 2066–2070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Shi, Y.; Au, J.S.; Thongprasert, S.; Srinivasan, S.; Tsai, C.M.; Khoa, M.T.; Heeroma, K.; Itoh, Y.; Cornelio, G.; Yang, P.C. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER). J. Thorac. Oncol. 2014, 9, 154–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. 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]
  4. 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]
  5. 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]
  6. 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] [Green Version]
  7. 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]
  8. 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]
  9. Sequist, L.V.; Yang, J.C.; Yamamoto, N.; O’Byrne, K.; Hirsh, V.; Mok, T.; Geater, S.L.; Orlov, S.; Tsai, C.M.; Boyer, M.; et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J. Clin. Oncol. 2013, 31, 3327–3334. [Google Scholar] [CrossRef] [Green Version]
  10. Yang, J.C.; Schuler, M.; Popat, S.; Miura, S.; Heeke, S.; Park, K.; Marten, A.; Kim, E.S. Afatinib for the Treatment of NSCLC Harboring Uncommon EGFR Mutations: A Database of 693 Cases. J. Thorac. Oncol. 2020, 15, 803–815. [Google Scholar] [CrossRef]
  11. Yang, J.C.; Schuler, M.; Popat, S.; Miura, S.; Park, K.; Passaro, A.; De Marinis, F.; Solca, F.; Marten, A.; Kim, E.S. Afatinib for the Treatment of Non-Small Cell Lung Cancer Harboring Uncommon EGFR Mutations: An Updated Database of 1023 Cases Brief Report. Front. Oncol. 2022, 12, 834704. [Google Scholar] [CrossRef] [PubMed]
  12. Passaro, A.; Mok, T.; Peters, S.; Popat, S.; Ahn, M.J.; De Marinis, F. Recent Advances on the Role of EGFR Tyrosine Kinase Inhibitors in the Management of NSCLC with Uncommon, Non Exon 20 Insertions, EGFR Mutations. J. Thorac. Oncol. 2021, 16, 764–773. [Google Scholar] [CrossRef]
  13. Cho, J.H.; Lim, S.H.; An, H.J.; Kim, K.H.; Park, K.U.; Kang, E.J.; Choi, Y.H.; Ahn, M.S.; Lee, M.H.; Sun, J.M.; et al. Osimertinib for Patients with Non-Small-Cell Lung Cancer Harboring Uncommon EGFR Mutations: A Multicenter, Open-Label, Phase II Trial (KCSG-LU15-09). J. Clin. Oncol. 2020, 38, 488–495. [Google Scholar] [CrossRef] [PubMed]
  14. Song, C.; Yang, X. Osimertinib-Centered Therapy Against Uncommon Epidermal Growth Factor Receptor-Mutated Non-Small-Cell Lung Cancer—A Mini Review. Front. Oncol. 2022, 12, 834585. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Mok, T.S.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Lee, M.; Linke, R.; Rosell, R.; Corral, J.; et al. Improvement in Overall Survival in a Randomized Study That Compared Dacomitinib with Gefitinib in Patients with Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. J. Clin. Oncol. 2018, 36, 2244–2250. [Google Scholar] [CrossRef]
  17. 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]
  18. Paz-Ares, L.; Tan, E.H.; O’Byrne, K.; Zhang, L.; Hirsh, V.; Boyer, M.; Yang, J.C.; Mok, T.; Lee, K.H.; Lu, S.; et al. Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: Overall survival data from the phase IIb LUX-Lung 7 trial. Ann. Oncol. 2017, 28, 270–277. [Google Scholar] [CrossRef]
  19. Park, K.; Tan, E.-H.; O’Byrne, K.; Zhang, L.; Boyer, M.; Mok, T.; Hirsh, V.; Yang, J.C.-H.; Lee, K.H.; Lu, S.; et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): A phase 2B, open-label, randomised controlled trial. Lancet Oncol. 2016, 17, 577–589. [Google Scholar] [CrossRef]
  20. 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]
  21. Hama, R.; Sakaguchi, K. The Gefitinib Story. Available online: https://npojip.org/english/The-gefitinib-story.pdf (accessed on 3 January 2023).
  22. Shepherd, F.A.; Rodrigues Pereira, J.; Ciuleanu, T.; Tan, E.H.; Hirsh, V.; Thongprasert, S.; Campos, D.; Maoleekoonpiroj, S.; Smylie, M.; Martins, R.; et al. Erlotinib in previously treated non-small-cell lung cancer. N. Engl. J. Med. 2005, 353, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Cohen, M.H.; Johnson, J.R.; Chen, Y.F.; Sridhara, R.; Pazdur, R. FDA drug approval summary: Erlotinib (Tarceva) tablets. Oncologist 2005, 10, 461–466. [Google Scholar] [CrossRef] [PubMed]
  24. Drugs.com. Tarceva FDA Approval History. Available online: https://www.drugs.com/history/tarceva.html (accessed on 11 April 2021).
  25. Kazandjian, D.; Blumenthal, G.M.; Yuan, W.; He, K.; Keegan, P.; Pazdur, R. FDA Approval of Gefitinib for the Treatment of Patients with Metastatic EGFR Mutation-Positive Non-Small Cell Lung Cancer. Clin. Cancer Res. 2016, 22, 1307–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. 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]
  27. 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-naive non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002). Ann. Oncol. 2013, 24, 54–59. [Google Scholar] [CrossRef]
  28. 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]
  29. 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] [Green Version]
  30. Douillard, J.Y.; Ostoros, G.; Cobo, M.; Ciuleanu, T.; McCormack, R.; Webster, A.; Milenkova, T. First-line gefitinib in Caucasian EGFR mutation-positive NSCLC patients: A phase-IV, open-label, single-arm study. Br. J. Cancer 2014, 110, 55–62. [Google Scholar] [CrossRef] [Green Version]
  31. Maemondo, M.; Inoue, A.; Kobayashi, K.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N. Engl. J. Med. 2010, 362, 2380–2388. [Google Scholar] [CrossRef] [Green Version]
  32. Drugs.com. Iressa FDA Approval History. Available online: https://www.drugs.com/history/iressa.html (accessed on 1 September 2022).
  33. Drugs.com. Gilotrif FDA Approval History. Available online: https://www.drugs.com/newdrugs/fda-approves-gilotrif-late-stage-non-small-cell-lung-cancer-3851.html (accessed on 11 April 2022).
  34. Yang, J.C.; Sequist, L.V.; Geater, S.L.; Tsai, C.M.; Mok, T.S.; Schuler, M.; Yamamoto, N.; Yu, C.J.; Ou, S.H.; Zhou, C.; et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: A combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol. 2015, 16, 830–838. [Google Scholar] [CrossRef]
  35. Halmos, B.; Tan, E.H.; Soo, R.A.; Cadranel, J.; Lee, M.K.; Foucher, P.; Hsia, T.C.; Hochmair, M.; Griesinger, F.; Hida, T.; et al. Impact of afatinib dose modification on safety and effectiveness in patients with EGFR mutation-positive advanced NSCLC: Results from a global real-world study (RealGiDo). Lung Cancer J. IASLC 2019, 127, 103–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Drugs.com. Tagrisso FDA Approval History. Available online: https://www.drugs.com/history/tagrisso.html (accessed on 13 April 2021).
  37. 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.; et al. Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N. Engl. J. Med. 2017, 376, 629–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Papadimitrakopoulou, V.A.; Mok, T.S.; Han, J.Y.; Ahn, M.J.; Delmonte, A.; Ramalingam, S.S.; Kim, S.W.; Shepherd, F.A.; Laskin, J.; He, Y.; et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann. Oncol. 2020, 31, 1536–1544. [Google Scholar] [CrossRef] [PubMed]
  39. Pluzanski, A.; Krzakowski, M.; Kowalski, D.; Dziadziuszko, R. Real-world clinical outcomes of first-generation and second-generation epidermal growth factor receptor tyrosine kinase inhibitors in a large cohort of European non-small-cell lung cancer patients. ESMO Open 2020, 5, e001011. [Google Scholar] [CrossRef] [PubMed]
  40. Drugs.com. Vizimpro FDA Approval History. Available online: https://www.drugs.com/history/vizimpro.html (accessed on 13 April 2021).
  41. US Food and Drug Administration. FDA Approves Dacomitinib for Metastatic Non-Small Cell Lung Cancer. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-dacomitinib-metastatic-non-small-cell-lung-cancer-0 (accessed on 1 September 2022).
  42. Katakami, N.; Atagi, S.; Goto, K.; Hida, T.; Horai, T.; Inoue, A.; Ichinose, Y.; Koboyashi, K.; Takeda, K.; Kiura, K.; et al. LUX-Lung 4: A phase II trial of afatinib in patients with advanced non-small-cell lung cancer who progressed during prior treatment with erlotinib, gefitinib, or both. J. Clin. Oncol. 2013, 31, 3335–3341. [Google Scholar] [CrossRef] [PubMed]
  43. Janne, P.A.; Yang, J.C.; Kim, D.W.; Planchard, D.; Ohe, Y.; Ramalingam, S.S.; Ahn, M.J.; Kim, S.W.; Su, W.C.; Horn, L.; et al. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N. Engl. J. Med. 2015, 372, 1689–1699. [Google Scholar] [CrossRef]
  44. Goss, G.; Tsai, C.M.; Shepherd, F.A.; Bazhenova, L.; Lee, J.S.; Chang, G.C.; Crino, L.; Satouchi, M.; Chu, Q.; Hida, T.; et al. Osimertinib for pretreated EGFR Thr790Met-positive advanced non-small-cell lung cancer (AURA2): A multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 2016, 17, 1643–1652. [Google Scholar] [CrossRef]
  45. Hochmair, M.J.; Morabito, A.; Hao, D.; Yang, C.T.; Soo, R.A.; Yang, J.C.; Gucalp, R.; Halmos, B.; Marten, A.; Cufer, T. Sequential afatinib and osimertinib in patients with EGFR mutation-positive non-small-cell lung cancer: Final analysis of the GioTag study. Future Oncol. 2020, 16, 2799–2808. [Google Scholar] [CrossRef]
  46. Kim, T.; Jang, T.W.; Choi, C.M.; Kim, M.H.; Lee, S.Y.; Park, C.K.; Chang, Y.S.; Lee, K.Y.; Kim, S.J.; Yang, S.H.; et al. Sequential treatment of afatinib and osimertinib or other regimens in patients with advanced non-small-cell lung cancer harboring EGFR mutations: Results from a real-world study in South Korea. Cancer Med. 2021, 10, 5809–5822. [Google Scholar] [CrossRef]
  47. Byers, L.A.; Heymach, J.V. Dual targeting of the vascular endothelial growth factor and epidermal growth factor receptor pathways: Rationale and clinical applications for non-small-cell lung cancer. Clin. Lung Cancer 2007, 8 (Suppl. 2), S79–S85. [Google Scholar] [CrossRef]
  48. Saito, H.; Fukuhara, T.; Furuya, N.; Watanabe, K.; Sugawara, S.; Iwasawa, S.; Tsunezuka, Y.; Yamaguchi, O.; Okada, M.; Yoshimori, K.; et al. Erlotinib plus bevacizumab versus erlotinib alone in patients with EGFR-positive advanced non-squamous non-small-cell lung cancer (NEJ026): Interim analysis of an open-label, randomised, multicentre, phase 3 trial. Lancet Oncol. 2019, 20, 625–635. [Google Scholar] [CrossRef] [PubMed]
  49. 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] [Green Version]
  50. Ninomiya, T.; Nogami, N.; Kozuki, T.; Harada, D.; Kubo, T.; Ohashi, K.; Kuyama, S.; Kudo, K.; Bessho, A.; Fukamatsu, N.; et al. A phase I trial of afatinib and bevacizumab in chemo-naive patients with advanced non-small-cell lung cancer harboring EGFR mutations: Okayama Lung Cancer Study Group Trial 1404. Lung Cancer J. IASLC 2018, 115, 103–108. [Google Scholar] [CrossRef] [PubMed]
  51. Hsu, P.C.; Huang, C.Y.; Wang, C.C.; Kuo, S.C.; Chu, C.H.; Tung, P.H.; Huang, A.C.; Wang, C.L.; Chiu, L.C.; Fang, Y.F.; et al. The Combination of Afatinib and Bevacizumab in Untreated EGFR-Mutated Advanced Lung Adenocarcinoma: A Multicenter Observational Study. Pharmaceuticals 2020, 13, 331. [Google Scholar] [CrossRef] [PubMed]
  52. Yu, H.A.; Schoenfeld, A.J.; Makhnin, A.; Kim, R.; Rizvi, H.; Tsui, D.; Falcon, C.; Houck-Loomis, B.; Meng, F.; Yang, J.L.; et al. Effect of Osimertinib and Bevacizumab on Progression-Free Survival for Patients with Metastatic EGFR-Mutant Lung Cancers: A Phase 1/2 Single-Group Open-Label Trial. JAMA Oncol. 2020, 6, 1048–1054. [Google Scholar] [CrossRef] [PubMed]
  53. Akamatsu, H.; Toi, Y.; Hayashi, H.; Fujimoto, D.; Tachihara, M.; Furuya, N.; Otani, S.; Shimizu, J.; Katakami, N.; Azuma, K.; et al. Efficacy of Osimertinib Plus Bevacizumab vs Osimertinib in Patients with EGFR T790M-Mutated Non-Small Cell Lung Cancer Previously Treated with Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor: West Japan Oncology Group 8715L Phase 2 Randomized Clinical Trial. JAMA Oncol. 2021, 7, 386–394. [Google Scholar] [CrossRef]
  54. Soo, R.A.; Han, J.Y.; Dafni, U.; Cho, B.C.; Yeo, C.M.; Nadal, E.; Carcereny, E.; de Castro, J.; Sala, M.A.; Bernabe, R.; et al. A randomised phase II study of osimertinib and bevacizumab versus osimertinib alone as second-line targeted treatment in advanced NSCLC with confirmed EGFR and acquired T790M mutations: The European Thoracic Oncology Platform (ETOP 10-16) BOOSTER trial. Ann. Oncol. 2022, 33, 181–192. [Google Scholar] [CrossRef]
  55. Kenmotsu, H.; Wakuda, K.; Mori, K.; Kato, T.; Sugawara, S.; Kirita, K.; Yoneshima, Y.; Azuma, K.; Nishino, K.; Teraoka, S.; et al. Randomized Phase 2 Study of Osimertinib Plus Bevacizumab Versus Osimertinib for Untreated Patients with Nonsquamous NSCLC Harboring EGFR Mutations: WJOG9717L Study. J. Thorac. Oncol. 2022, 17, 1098–1108. [Google Scholar] [CrossRef]
  56. Janne, P.A.; Wang, X.; Socinski, M.A.; Crawford, J.; Stinchcombe, T.E.; Gu, L.; Capelletti, M.; Edelman, M.J.; Villalona-Calero, M.A.; Kratzke, R.; et al. Randomized phase II trial of erlotinib alone or with carboplatin and paclitaxel in patients who were never or light former smokers with advanced lung adenocarcinoma: CALGB 30406 trial. J. Clin. Oncol. 2012, 30, 2063–2069. [Google Scholar] [CrossRef]
  57. Soria, J.C.; Wu, Y.L.; Nakagawa, K.; Kim, S.W.; Yang, J.J.; Ahn, M.J.; Wang, J.; Yang, J.C.; 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]
  58. Noronha, V.; Patil, V.M.; Joshi, A.; Menon, N.; Chougule, A.; Mahajan, A.; Janu, A.; Purandare, N.; Kumar, R.; More, S.; et al. Gefitinib Versus Gefitinib Plus Pemetrexed and Carboplatin Chemotherapy in EGFR-Mutated Lung Cancer. J. Clin. Oncol. 2020, 38, 124–136. [Google Scholar] [CrossRef] [PubMed]
  59. 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. 2020, 38, 115–123. [Google Scholar] [CrossRef] [PubMed]
  60. Schuler, M.; Yang, J.C.; Park, K.; Kim, J.H.; Bennouna, J.; Chen, Y.M.; Chouaid, C.; De Marinis, F.; Feng, J.F.; Grossi, F.; et al. Afatinib beyond progression in patients with non-small-cell lung cancer following chemotherapy, erlotinib/gefitinib and afatinib: Phase III randomized LUX-Lung 5 trial. Ann. Oncol. 2016, 27, 417–423. [Google Scholar] [CrossRef] [PubMed]
  61. White, M.N.; Piotrowska, Z.; Stirling, K.; Liu, S.V.; Banwait, M.K.; Cunanan, K.; Sequist, L.V.; Wakelee, H.A.; Hausrath, D.; Neal, J.W. Combining Osimertinib with Chemotherapy in EGFR-Mutant NSCLC at Progression. Clin. Lung Cancer 2021, 22, 201–209. [Google Scholar] [CrossRef] [PubMed]
  62. Planchard, D.; Feng, P.H.; Karaseva, N.; Kim, S.W.; Kim, T.M.; Lee, C.K.; Poltoratskiy, A.; Yanagitani, N.; Powar, S.; Huang, X.; et al. 1401P Osimertinib plus platinum/pemetrexed in newly-diagnosed EGFR mutation (EGFRm)-positive advanced NSCLC: Safety run-in results from the FLAURA2 study. Ann. Oncol. 2020, 31, S888. [Google Scholar] [CrossRef]
  63. Creelan, B.C.; Yeh, T.C.; Kim, S.W.; Nogami, N.; Kim, D.W.; Chow, L.Q.M.; Kanda, S.; Taylor, R.; Tang, W.; Tang, M.; et al. A Phase 1 study of gefitinib combined with durvalumab in EGFR TKI-naive patients with EGFR mutation-positive locally advanced/metastatic non-small-cell lung cancer. Br. J. Cancer 2021, 124, 383–390. [Google Scholar] [CrossRef]
  64. 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] [Green Version]
  65. Rudin, C.; Cervantes, A.; Dowlati, A.; Besse, B.; Ma, B.; Costa, D.; Schmid, P.; Heist, R.; Villaflor, V.; Sarkar, I.; et al. MA15.02 Long-Term Safety and Clinical Activity Results from a Phase Ib Study of Erlotinib Plus Atezolizumab in Advanced NSCLC. J. Thorac. Oncol. 2018, 13, S407. [Google Scholar] [CrossRef] [Green Version]
  66. 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] [Green Version]
  67. 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]
  68. Gainor, J.F.; Shaw, A.T. Emerging paradigms in the development of resistance to tyrosine kinase inhibitors in lung cancer. J. Clin. Oncol. 2013, 31, 3987–3996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Jackman, D.; Pao, W.; Riely, G.J.; Engelman, J.A.; Kris, M.G.; Janne, 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] [PubMed]
  70. 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] [Green Version]
  71. 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] [Green Version]
  72. 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] [PubMed] [Green Version]
  73. Toyooka, S.; Kiura, K.; Mitsudomi, T. EGFR mutation and response of lung cancer to gefitinib. N. Engl. J. Med. 2005, 352, 2136. [Google Scholar] [CrossRef]
  74. Tanaka, K.; Nosaki, K.; Otsubo, K.; Azuma, K.; Sakata, S.; Ouchi, H.; Morinaga, R.; Wataya, H.; Fujii, A.; Nakagaki, N.; et al. Acquisition of the T790M resistance mutation during afatinib treatment in EGFR tyrosine kinase inhibitor-naive patients with non-small cell lung cancer harboring EGFR mutations. Oncotarget 2017, 8, 68123–68130. [Google Scholar] [CrossRef]
  75. Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Janne, P.A.; Kocher, O.; Meyerson, M.; Johnson, B.E.; Eck, M.J.; Tenen, D.G.; Halmos, B. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
  76. Wu, S.G.; Liu, Y.N.; Tsai, M.F.; Chang, Y.L.; Yu, C.J.; Yang, P.C.; Yang, J.C.; Wen, Y.F.; Shih, J.Y. The mechanism of acquired resistance to irreversible EGFR tyrosine kinase inhibitor-afatinib in lung adenocarcinoma patients. Oncotarget 2016, 7, 12404–12413. [Google Scholar] [CrossRef] [Green Version]
  77. Jotte, R.M.; Spigel, D.R. Advances in molecular-based personalized non-small-cell lung cancer therapy: Targeting epidermal growth factor receptor and mechanisms of resistance. Cancer Med. 2015, 4, 1621–1632. [Google Scholar] [CrossRef]
  78. Balak, M.N.; Gong, Y.; Riely, G.J.; Somwar, R.; Li, A.R.; Zakowski, M.F.; Chiang, A.; Yang, G.; Ouerfelli, O.; Kris, M.G.; et al. Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin. Cancer Res. 2006, 12, 6494–6501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Zhang, H.; Shao, Y.W.; Xia, Y. Responsiveness to Full-Dose Afatinib in a Patient with Lung Adenocarcinoma Harboring EGFR S768I and V769L Mutations. J. Thorac. Oncol. 2019, 14, e25–e27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
  81. Schoenfeld, A.J.; Yu, H.A. The Evolving Landscape of Resistance to Osimertinib. J. Thorac. Oncol. 2020, 15, 18–21. [Google Scholar] [CrossRef]
  82. Thress, K.S.; Paweletz, C.P.; Felip, E.; Cho, B.C.; Stetson, D.; Dougherty, B.; Lai, Z.; Markovets, A.; Vivancos, A.; Kuang, Y.; et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat. Med. 2015, 21, 560–562. [Google Scholar] [CrossRef] [Green Version]
  83. 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 J. IASLC 2017, 108, 228–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. 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] [Green Version]
  85. Ricordel, C.; Friboulet, L.; Facchinetti, F.; Soria, J.C. Molecular mechanisms of acquired resistance to third-generation EGFR-TKIs in EGFR T790M-mutant lung cancer. Ann. Oncol. 2019, 30, 858. [Google Scholar] [CrossRef] [Green Version]
  86. Suzawa, K.; Offin, M.; Schoenfeld, A.J.; Plodkowski, A.J.; Odintsov, I.; Lu, D.; Lockwood, W.W.; Arcila, M.E.; Rudin, C.M.; Drilon, A.; et al. Acquired MET Exon 14 Alteration Drives Secondary Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor in EGFR-Mutated Lung Cancer. JCO Precis. Oncol. 2019, 3, 19. [Google Scholar] [CrossRef]
  87. Romaniello, D.; Marrocco, I.; Belugali Nataraj, N.; Ferrer, I.; Drago-Garcia, D.; Vaknin, I.; Oren, R.; Lindzen, M.; Ghosh, S.; Kreitman, M.; et al. Targeting HER3, a Catalytically Defective Receptor Tyrosine Kinase, Prevents Resistance of Lung Cancer to a Third-Generation EGFR Kinase Inhibitor. Cancers 2020, 12, 2394. [Google Scholar] [CrossRef]
  88. Shields, M.D.; Hicks, J.K.; Boyle, T.A.; Haura, E.B.; Creelan, B.C. Selpercatinib Overcomes CCDC6-RET-Mediated Resistance to Osimertinib. J. Thorac. Oncol. 2021, 16, e15–e17. [Google Scholar] [CrossRef] [PubMed]
  89. 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. 2019, 37, 278–285. [Google Scholar] [CrossRef] [PubMed]
  90. Chen, N.; Fang, W.; Zhan, J.; Hong, S.; Tang, Y.; Kang, S.; Zhang, Y.; He, X.; Zhou, T.; Qin, T.; et al. Upregulation of PD-L1 by EGFR Activation Mediates the Immune Escape in EGFR-Driven NSCLC: Implication for Optional Immune Targeted Therapy for NSCLC Patients with EGFR Mutation. J. Thorac. Oncol. 2015, 10, 910–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Suda, K.; Murakami, I.; Sakai, K.; Mizuuchi, H.; Shimizu, S.; Sato, K.; Tomizawa, K.; Tomida, S.; Yatabe, Y.; Nishio, K.; et al. Small cell lung cancer transformation and T790M mutation: Complimentary roles in acquired resistance to kinase inhibitors in lung cancer. Sci. Rep. 2015, 5, 14447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Offin, M.; Chan, J.M.; Tenet, M.; Rizvi, H.A.; Shen, R.; Riely, G.J.; Rekhtman, N.; Daneshbod, Y.; Quintanal-Villalonga, A.; Penson, A.; et al. Concurrent RB1 and TP53 Alterations Define a Subset of EGFR-Mutant Lung Cancers at risk for Histologic Transformation and Inferior Clinical Outcomes. J. Thorac. Oncol. 2019, 14, 1784–1793. [Google Scholar] [CrossRef] [PubMed]
  93. Raoof, S.; Mulford, I.J.; Frisco-Cabanos, H.; Nangia, V.; Timonina, D.; Labrot, E.; Hafeez, N.; Bilton, S.J.; Drier, Y.; Ji, F.; et al. Targeting FGFR overcomes EMT-mediated resistance in EGFR mutant non-small cell lung cancer. Oncogene 2019, 38, 6399–6413. [Google Scholar] [CrossRef]
  94. Tulchinsky, E.; Demidov, O.; Kriajevska, M.; Barlev, N.A.; Imyanitov, E. EMT: A mechanism for escape from EGFR-targeted therapy in lung cancer. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 29–39. [Google Scholar] [CrossRef]
  95. John, T.; Akamatsu, H.; Delmonte, A.; Su, W.C.; Lee, J.S.; Chang, G.C.; Huang, X.; Jenkins, S.; Wu, Y.L. EGFR mutation analysis for prospective patient selection in AURA3 phase III trial of osimertinib versus platinum-pemetrexed in patients with EGFR T790M-positive advanced non-small-cell lung cancer. Lung Cancer J. IASLC 2018, 126, 133–138. [Google Scholar] [CrossRef]
  96. Yap, T.A.; Macklin-Doherty, A.; Popat, S. Continuing EGFR inhibition beyond progression in advanced non-small cell lung cancer. Eur. J. Cancer 2017, 70, 12–21. [Google Scholar] [CrossRef] [PubMed]
  97. Goto, Y.; Tanai, C.; Yoh, K.; Hosomi, Y.; Sakai, H.; Kato, T.; Kaburagi, T.; Nishio, M.; Kim, Y.H.; Inoue, A.; et al. Continuing EGFR-TKI beyond radiological progression in patients with advanced or recurrent, EGFR mutation-positive non-small-cell lung cancer: An observational study. ESMO Open 2017, 2, e000214. [Google Scholar] [CrossRef] [PubMed]
  98. NCCN Clinical Practice Guidelines in Oncology. Non-Small Cell Lung Cancer NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) 2021, Version 4.2021—March 3, 2021. J. Natl. Compr. Cancer Netw. JNCCN 2021, 2, 94–123. [Google Scholar]
  99. Gandhi, L.; Rodriguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
  100. Paz-Ares, L.; Ciuleanu, T.E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 198–211. [Google Scholar] [CrossRef] [PubMed]
  101. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gumus, M.; Mazieres, J.; Hermes, B.; Cay Senler, F.; Csoszi, T.; Fulop, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef]
  102. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef]
  103. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef]
  104. Choudhury, N.J.; Makhnin, A.; Tobi, Y.Y.; Daly, R.M.; Preeshagul, I.R.; Iqbal, A.N.; Ahn, L.S.; Hayes, S.A.; Heller, G.; Kris, M.G.; et al. Pilot Study of Dacomitinib for Patients with Metastatic EGFR-Mutant Lung Cancers with Disease Progression After Initial Treatment with Osimertinib. JCO Precis. Oncol. 2021, 5, 695–700. [Google Scholar] [CrossRef]
  105. Nogami, N.; Barlesi, F.; Socinski, M.A.; Reck, M.; Thomas, C.A.; Cappuzzo, F.; Mok, T.S.K.; Finley, G.; Aerts, J.G.; Orlandi, F.; et al. IMpower150 Final Exploratory Analyses for Atezolizumab Plus Bevacizumab and Chemotherapy in Key NSCLC Patient Subgroups with EGFR Mutations or Metastases in the Liver or Brain. J. Thorac. Oncol. 2022, 17, 309–323. [Google Scholar] [CrossRef]
  106. Reck, M.; Mok, T.S.K.; Nishio, M.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-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]
  107. Lu, S.; Wu, L.; Jian, H.; Cheng, Y.; Wang, Q.; Fang, J.; Wang, Z.; Hu, Y.; Sun, M.; Han, L.; et al. VP9-2021: ORIENT-31: Phase III study of sintilimab with or without IBI305 plus chemotherapy in patients with EGFR mutated nonsquamous NSCLC who progressed after EGFR-TKI therapy. Ann. Oncol. 2022, 33, 112–113. [Google Scholar] [CrossRef]
  108. Hellmann, M.D.; Garon, E.B.; Gandhi, L.; Hui, R.; Zhang, J.; Rangwala, R.; Rizvi, N.A. MINI03.05 Efficacy of Pembrolizumab in Key Subgroups of Patients with Advanced NSCLC. J. Thorac. Oncol. 2015, 10, S261–S406. [Google Scholar] [CrossRef]
  109. Garassino, M.C.; Cho, B.C.; Kim, J.H.; Mazieres, J.; Vansteenkiste, J.; Lena, H.; Jaime, J.C.; Gray, J.E.; Powderly, J.; Chouaid, C.; et al. Final overall survival and safety update for durvalumab in third- or later-line advanced NSCLC: The phase II ATLANTIC study. Lung Cancer J. IASLC 2020, 147, 137–142. [Google Scholar] [CrossRef] [PubMed]
  110. Garassino, M.C.; Cho, B.-C.; Kim, J.-H.; Mazières, J.; Vansteenkiste, J.; Lena, H.; Corral Jaime, J.; Gray, J.E.; Powderly, J.; Chouaid, C.; et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): An open-label, single-arm, phase 2 study. Lancet Oncol. 2018, 19, 521–536. [Google Scholar] [CrossRef] [PubMed]
  111. Yu, H.A.; Ambrose, H.; Baik, C.; Cho, B.C.; Cocco, E.; Goldberg, S.B.; Goldman, J.W.; Kraljevic, S.; De Langen, A.J.; Okamoto, I.; et al. 1239P ORCHARD osimertinib + savolitinib interim analysis: A biomarker-directed phase II platform study in patients (pts) with advanced non-small cell lung cancer (NSCLC) whose disease has progressed on first-line (1L) osimertinib. Ann. Oncol. 2021, 32, S978–S979. [Google Scholar] [CrossRef]
  112. 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 Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef]
  113. Leighl, N.B.; Shu, C.A.; Minchom, A.; Felip, E.; Cousin, S.; Cho, B.C.; Park, K.; Han, J.Y.; Boyer, M.; Lee, C.K.; et al. 1192MO Amivantamab monotherapy and in combination with lazertinib in post-osimertinib EGFR-mutant NSCLC: Analysis from the CHRYSALIS study. Ann. Oncol. 2021, 32, S951–S952. [Google Scholar] [CrossRef]
  114. Shu, C.A.; Goto, K.; Ohe, Y.; Besse, B.; Lee, S.-H.; Wang, Y.; Griesinger, F.; Yang, J.C.-H.; Felip, E.; Sanborn, R.E.; et al. Amivantamab and lazertinib in patients with EGFR-mutant non–small cell lung (NSCLC) after progression on osimertinib and platinum-based chemotherapy: Updated results from CHRYSALIS-2. J. Clin. Oncol. 2022, 40, 9006. [Google Scholar] [CrossRef]
  115. 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]
  116. Janne, P.A.; Baik, C.; Su, W.C.; Johnson, M.L.; Hayashi, H.; Nishio, M.; Kim, D.W.; Koczywas, M.; Gold, K.A.; Steuer, C.E.; et al. Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR Inhibitor-Resistant, EGFR-Mutated Non-Small Cell Lung Cancer. Cancer Discov. 2022, 12, 74–89. [Google Scholar] [CrossRef]
  117. Garon, E.B.; Johnson, M.L.; Lisberg, A.E.; Spira, A.; Yamamoto, N.; Heist, R.S.; Sands, J.M.; Yoh, K.; Meric-Bernstam, F.; Kitazono, S.; et al. LBA49 Efficacy of datopotamab deruxtecan (Dato-DXd) in patients (pts) with advanced/metastatic (adv/met) non-small cell lung cancer (NSCLC) and actionable genomic alterations (AGAs): Preliminary results from the phase I TROPION-PanTumor01 study. Ann. Oncol. 2021, 32, S1326–S1327. [Google Scholar] [CrossRef]
  118. Beypinar, I.; Demir, H.; Araz, M.; Uysal, M. The relationship between EGFR mutation and metastasis pattern in lung adenocarcinoma. J. Oncol. Sci. 2019, 5, 65–69. [Google Scholar] [CrossRef]
  119. Rangachari, D.; Yamaguchi, N.; VanderLaan, P.A.; Folch, E.; Mahadevan, A.; Floyd, S.R.; Uhlmann, E.J.; Wong, E.T.; Dahlberg, S.E.; Huberman, M.S.; et al. Brain metastases in patients with EGFR-mutated or ALK-rearranged non-small-cell lung cancers. Lung Cancer J. IASLC 2015, 88, 108–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Aiko, N.; Shimokawa, T.; Miyazaki, K.; Misumi, Y.; Agemi, Y.; Ishii, M.; Nakamura, Y.; Yamanaka, T.; Okamoto, H. Comparison of the efficacies of first-generation epidermal growth factor receptor tyrosine kinase inhibitors for brain metastasis in patients with advanced non-small-cell lung cancer harboring EGFR mutations. BMC Cancer 2018, 18, 1012. [Google Scholar] [CrossRef] [PubMed]
  121. Yoshida, K.; Kanda, S.; Shiraishi, H.; Goto, K.; Itahashi, K.; Goto, Y.; Horinouchi, H.; Fujiwara, Y.; Nokihara, H.; Yamamoto, N.; et al. Difference in central nerve system metastasis during gefitinib or erlotinib therapy in patients with EGFR-mutated non-small cell lung cancer: A retrospective study. J. Thorac. Dis. 2019, 11, 1347–1354. [Google Scholar] [CrossRef]
  122. Liang, S.K.; Lee, M.R.; Liao, W.Y.; Ho, C.C.; Ko, J.C.; Shih, J.Y. Prognostic factors of afatinib as a first-line therapy for advanced EGFR mutation-positive lung adenocarcinoma: A real-world, large cohort study. Oncotarget 2018, 9, 23749–23760. [Google Scholar] [CrossRef]
  123. Bergonzini, C.; Leonetti, A.; Tiseo, M.; Giovannetti, E.; Peters, G.J. Is there a role for dacomitinib, a second-generation irreversible inhibitor of the epidermal-growth factor receptor tyrosine kinase, in advanced non-small cell lung cancer? Expert Opin. Pharmacother. 2020, 21, 1287–1298. [Google Scholar] [CrossRef] [Green Version]
  124. Peng, W.; Pu, X.; Jiang, M.; Wang, J.; Li, J.; Li, K.; Xu, Y.; Xu, F.; Chen, B.; Wang, Q.; et al. Dacomitinib induces objective responses in metastatic brain lesions of patients with EGFR-mutant non-small-cell lung cancer: A brief report. Lung Cancer J. IASLC 2021, 152, 66–70. [Google Scholar] [CrossRef]
  125. Reungwetwattana, T.; Nakagawa, K.; Cho, B.C.; Cobo, M.; Cho, E.K.; Bertolini, A.; Bohnet, S.; Zhou, C.; Lee, K.H.; Nogami, N.; et al. CNS Response to Osimertinib Versus Standard Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3290. [Google Scholar] [CrossRef]
  126. Wu, Y.L.; Ahn, M.J.; Garassino, M.C.; Han, J.Y.; Katakami, N.; Kim, H.R.; Hodge, R.; Kaur, P.; Brown, A.P.; Ghiorghiu, D.; et al. CNS Efficacy of Osimertinib in Patients with T790M-Positive Advanced Non-Small-Cell Lung Cancer: Data From a Randomized Phase III Trial (AURA3). J. Clin. Oncol. 2018, 36, 2702–2709. [Google Scholar] [CrossRef]
  127. Castanon, E.; Rolfo, C.; Vinal, D.; Lopez, I.; Fusco, J.P.; Santisteban, M.; Martin, P.; Zubiri, L.; Echeveste, J.I.; Gil-Bazo, I. Impact of epidermal growth factor receptor (EGFR) activating mutations and their targeted treatment in the prognosis of stage IV non-small cell lung cancer (NSCLC) patients harboring liver metastasis. J. Transl. Med. 2015, 13, 257. [Google Scholar] [CrossRef] [Green Version]
  128. Jiang, T.; Cheng, R.; Zhang, G.; Su, C.; Zhao, C.; Li, X.; Zhang, J.; Wu, F.; Chen, X.; Gao, G.; et al. Characterization of Liver Metastasis and Its Effect on Targeted Therapy in EGFR-mutant NSCLC: A Multicenter Study. Clin. Lung Cancer 2017, 18, 631–639.e632. [Google Scholar] [CrossRef] [PubMed]
  129. Walia, P.; Zhan, L.; Schmid, S.; Brown, M.C.; Khan, K.; Garcia, M.; Chowdhury, M.; Herman, J.; Sabouhanian, A.; Strom, E.; et al. P59.17 EGFR Mutation Status, Liver Metastasis, and Overall Survival in Advanced Lung Adenocarcinoma Patients. J. Thorac. Oncol. 2021, 16, S1154–S1155. [Google Scholar] [CrossRef]
  130. Gen, S.; Tanaka, I.; Morise, M.; Koyama, J.; Kodama, Y.; Matsui, A.; Miyazawa, A.; Hase, T.; Hibino, Y.; Yokoyama, T.; et al. Clinical efficacy of osimertinib in EGFR-mutant non-small cell lung cancer with distant metastasis. BMC Cancer 2022, 22, 654. [Google Scholar] [CrossRef] [PubMed]
  131. Kuijpers, C.; Hendriks, L.E.L.; Derks, J.L.; Dingemans, A.C.; Van Lindert, A.S.R.; Van den Heuvel, M.M.; Damhuis, R.A.; Willems, S.M. Association of molecular status and metastatic organs at diagnosis in patients with stage IV non-squamous non-small cell lung cancer. Lung Cancer J. IASLC 2018, 121, 76–81. [Google Scholar] [CrossRef]
  132. Zhang, G.; Cheng, R.; Zhang, Z.; Jiang, T.; Ren, S.; Ma, Z.; Zhao, S.; Zhou, C.; Zhang, J. Bisphosphonates enhance antitumor effect of EGFR-TKIs in patients with advanced EGFR mutant NSCLC and bone metastases. Sci. Rep. 2017, 7, 42979. [Google Scholar] [CrossRef] [Green Version]
  133. Remon, J.; Lopes, G. Upfront osimertinib—Winner takes it all? Nat. Rev. Clin. Oncol. 2020, 17, 202–203. [Google Scholar] [CrossRef]
  134. Passiglia, F.; Raez, L.E.; Rolfo, C. Moving osimertinib to first-line: The right “strategy” in the chessboard of epidermal growth factor receptor-mutated non-small cell lung cancer? J. Thorac. Dis. 2018, 10, S1076–S1080. [Google Scholar] [CrossRef] [Green Version]
  135. Haratake, N.; Misumi, T.; Yamanaka, T.; Seto, T. Optimizing Sequential Treatment with EGFR Tyrosine Kinase Inhibitor with a Simulation of the T790M Mutation Rate in EGFR–Mutated Lung Cancer. J. Thorac. Oncol. 2020, 1, 100085. [Google Scholar] [CrossRef]
  136. Hirsh, V.; Singh, J. Optimal sequencing strategies in the treatment of EGFR mutation-positive non-small cell lung cancer: Clinical benefits and cost-effectiveness. Am. J. Health Syst. Pharm. 2020, 77, 1466–1476. [Google Scholar] [CrossRef]
  137. Girard, N. Optimizing outcomes and treatment sequences in EGFR mutation-positive non-small-cell lung cancer: Recent updates. Future Oncol. 2019, 15, 2983–2997. [Google Scholar] [CrossRef] [Green Version]
  138. Roeper, J.; Falk, M.; Schatz, S.; Tiemann, M.; Sackmann, S.; Ukena, D.; Wessler, C.; Wiest, G.; Heukamp, L.; Friesinger, F. Risk of Not Receiving 2nd Line Therapy is High in EGFR mt+ pts: Real World Data of Certified Lung Cancer Centers on Treatment Sequence in EGFR mt+ pts. J. Thorac. Oncol. 2018, 13, S494–S495. [Google Scholar] [CrossRef]
  139. Chiang, A.; Fernandes, A.; Pavilack, M.; Wu, J.; Laliberte, F.; Duh, M.S.; Chehab, N.; Subramanian, J. Real World Biomarker Testing and Treatment Patterns in Patients with Advanced NSCLC Receiving EGFR-TKIs. J. Thorac. Oncol. 2018, 13, S410–S411. [Google Scholar] [CrossRef] [Green Version]
  140. Cuppens, K.; Lodewyckx, L.; Demedts, I.; Decoster, L.; Colinet, B.; Deschepper, K.; Janssens, A.; Galdermans, D.; Pieters, T.; Group, R.S. Real-World Treatment Patterns, Epidermal Growth Factor Receptor (EGFR) Testing and Outcomes in EGFR-Mutated Advanced Non-small Cell Lung Cancer Patients in Belgium: Results from the REVEAL Study. Drugs Real World Outcomes 2021, 8, 141–152. [Google Scholar] [CrossRef]
  141. Gray, J.E.; Thakrar, B.; Sun, P.; Maclachlan, S.; Chehab, N.; Potter, D. Treatment (tx) patterns in patients (pts) with lung cancer starting 1st or 2nd generation (1G/2G) EGFR-TKI: A US insurance claims database analysis. Ann. Oncol. 2018, 29, 156–157. [Google Scholar] [CrossRef]
  142. Ballard, P.; Yates, J.W.; Yang, Z.; Kim, D.W.; Yang, J.C.; Cantarini, M.; Pickup, K.; Jordan, A.; Hickey, M.; Grist, M.; et al. Preclinical Comparison of Osimertinib with Other EGFR-TKIs in EGFR-Mutant NSCLC Brain Metastases Models, and Early Evidence of Clinical Brain Metastases Activity. Clin. Cancer Res. 2016, 22, 5130–5140. [Google Scholar] [CrossRef] [PubMed]
  143. US Food and Drug Administration. FDA Broadens Afatinib Indication to Previously Untreated, Metastatic NSCLC with Other Non-Resistant EGFR Mutations. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-broadens-afatinib-indication-previously-untreated-metastatic-nsclc-other-non-resistant-egfr#:~:text=FDA%20initially%20approved%20afatinib%20in,progressing%20after%20platinum%2Dbased%20chemotherapy (accessed on 1 September 2022).
Figure 1. Timeline depicting the publication of landmark trials on anti-EGFR TKIs, approval by the FDA and EMA, and publication of data pertaining novel therapies; EMA, European Medicines Agency; FDA, Food and Drugs Administration; TKIs, tyrosine kinase inhibitors. Created with BioRender.com, accessed on 8 January 2023.
Figure 1. Timeline depicting the publication of landmark trials on anti-EGFR TKIs, approval by the FDA and EMA, and publication of data pertaining novel therapies; EMA, European Medicines Agency; FDA, Food and Drugs Administration; TKIs, tyrosine kinase inhibitors. Created with BioRender.com, accessed on 8 January 2023.
Cancers 15 00629 g001
Table 1. Landmark Clinical Trials of EGFR TKIs as First-Line Therapy for Advanced NSCLC.
Table 1. Landmark Clinical Trials of EGFR TKIs as First-Line Therapy for Advanced NSCLC.
Drug(s)Trial
(NCT #)
PhasePopulation
Characteristics
Treatment Regimen
/Cohorts
OutcomesSide EffectsRate of
Discontinuation from
Toxicity
FDA Approval Date
ErlotinibEURTAC
(NCT00446225)
IIIN = 173
Stages IIIB/IV
Adenocarcinoma only
-Race: White (99%)
-EGFR Mutations: ex19del and L858R
-Asymptomatic brain metastases were
allowed
Cohort A:
Erlotinib 150 mg daily
Cohort B:
Platinum-based chemotherapy
ORR: 63.6% (A) vs. 17.8% (B)
mPFS: 9.7 months (A) vs. 5.2 months (B) p < 0.0001
mOS: 19.3 months (A) vs. 19.5 months (B) p = 0.87
Overall grade ≥3 AE: 45% (A) vs. 67% (B)13% (A) vs. 23% (B)14 May 2013
GefitinibIPASS
(NCT00322452)
IIIN = 1217
Stage IIIB/IV
Adenocarcinoma only
-Race: Asian (98%)
-EGFR Mutations: ex19del, L858R, T790M, other
-Excluded patients with untreated brain metastases
Cohort A:
Gefitinib 250 mg daily
Cohort B:
Platinum-based chemotherapy
ORR: 84.8% (A) vs. 43.2% (B)
mPFS: not reported; HR, 0.48 (95% CI, 0.34 to 0.67) for those with EGFR mutation
mOS: 18.8 months (A) vs. 17.4 months (B) p = 0.109
Overall grade ≥3 AE: not reported *6.9% (A) vs. 13.6% (B)13 July 2015
NEJ-002
(N/A–Japan)
IIIN = 230
Stage IIIB/IV
NSCLC
-Race: Asian/Japanese (100%)
-EGFR Mutations: ex19del, L858R, and other (6.1%)
-Asymptomatic brain metastases were allowed
Cohort A:
Gefitinib 250 mg daily
Cohort B:
Platinum-based chemotherapy
ORR: 73.7% (A) vs. 30.7% (B)
mPFS: 10.8 months (A) vs. 5.4 months (B) p < 0.001
mOS: 27.7 months (A) vs. 26.6 months (B) p = 0.483
Overall grade ≥3 AE: 41.2% (A) vs. 71.7% (B)Not reported
WJTOG3405
(N/A–Japan)
IIIN = 118
Stage IIIB/IV
NSCLC
-Race: Asian/Japanese (100%)
-EGFR Mutations: ex19del and L858R
-Asymptomatic brain metastases were allowed
Cohort A:
Gefitinib 250 mg daily
Cohort B:
Platinum-based chemotherapy
ORR: 62.1% (A) vs. 32.2% (B)
mPFS: 9.2 months (A) vs. 6.3 months (B) p < 0.0001
mOS: 34.9 months (A) vs. 37.3 months (B) p = 0.2070
Overall grade ≥3 AE: not reported *Not Reported
IFUM
(NCT01203917)
IVN = 106
Stage IIIA/B/IV
NSCLC
-Race: White (100%)
-EGFR Mutations: ex19del, L858R. T790M, S768I
-Inclusion of brain metastatic disease not mentioned
Single-Arm:
Gefitinib 250 mg daily
ORR: 69.8%
mPFS: 9.7 months
mOS: 19.2 months
Overall grade ≥3 AE: 15%7.5%
AfatinibLUX-Lung 3
(NCT00949650)
IIIN = 345
Stage IIIB/IV
Adenocarcinoma only
-Race: White (26.5%), Asian (71.7%), Other (1.7%)
-EGFR Mutations: ex19del, L858R, Other (10.3%)
-Asymptomatic stable brain metastases were allowed
Cohort A:
Afatinib 40 mg daily
Cohort B:
Platinum-based chemotherapy
ORR: 56% (A) vs. 23% (B)
mPFS: 11.1 months (A) vs. 6.9 months (B) p = 0.0004
mOS: 28.2 months (A) vs. 28.2 months (B) p = 0.39
mOS ex19del: 33.3 months (A) vs. 21.1 months (B) p = 0.0015
mOS L858R: 27.6 months (A) vs. 40.3 months (B) p = 0.29
Overall grade ≥3 AE: 49% (A) vs. 48% (B)8% (A) vs. 12% (B)Approval for EGFR exon 19 deletions or exon 21 (L858R):
23 July 2013
Expansion of indication to all non-resistant EGFR mutations:
12 January 2018
LUX-Lung 6
(NCT01121393)
IIIN = 364
Stage IIIB/IV
Adenocarcinoma only
-Race: Asian 100%
-EGFR Mutations: ex19del, L858R, other (11%)
-Asymptomatic, stable brain metastases were allowed
Cohort A:
Afatinib 40 mg daily
Cohort B:
Platinum-based chemotherapy
ORR: 66.9% (A) vs. 23% (B)
mPFS: 13.7 months (A) vs. 5.6 months (B) p < 0.0001
mOS: 23.1 months (A) vs. 23.5 months (B) p = 0.61
mOS ex19del: 31.4 months (A) vs. 18.4 months (B) p = 0.023
mOS L858R: 19.6 months (A) vs. 24.3 months (B) p = 0.34
Overall grade ≥3 AE: 36% (A) vs. 60.2% (B)5.9% (A) vs. 39.8% (B)
LUX-Lung 7IIbN = 319
Stage IIIB/IV
Adenocarcinoma only
-Race: Asian (59%), White (30%), Black (1%), not available (11%)
-EGFR Mutations: ex19del and L858R
-Active brain metastases (symptomatic or requiring treatment) excluded
Cohort A:
Afatinib 40 mg daily with escalation to 50 mg daily if well tolerated after 4 weeks
Cohort B:
Gefitinib 250 mg daily
ORR: 70% (A) vs. 56% (B)
mPFS: 11.0 months (A) vs. 10.9 months (B), HR 0.73 p < 0.017
mOS: 27.9 months (A) vs. 24.9 months (B) p = 0.258
Overall grade ≥3 AE: 57% (A) vs. 52% (B)6% (A) vs. 6% (B)
DacomitinibARCHER 1050
(NCT01774721)
IIIN = 452
Stage IIIB/IV
NSCLC
-Race: Asian (75%), Black (<1%), White (25%)
-EGFR Mutations: ex19del, L858R
-Brain or leptomeningeal metastases excluded
Cohort A:
Dacomitinib 45 mg daily
Cohort B:
Gefitinib 250 mg daily
ORR: 75% (A) vs. 72% (B)
mPFS: 14.7 months (A) vs. 9.2 months (B) p < 0.0001
mOS: 34.1 months (A) vs. 26.8 months (B) p = 0.438
Overall grade ≥3 AE: 63% (A) vs. 41% (B)10% (A) vs. 7% (B)27 September 2018
OsimertinibFLAURA
(NCT02296125)
IIIN = 556
Stage IIIB/IV
NSCLC
-Race: Asian (62%), White (36%), Other (1%)
-EGFR Mutations: ex19del, L858R
-Asymptomatic, stable brain metastases were allowed
Cohort A:
Osimertinib 80 mg daily
Cohort B:
Gefitinib 250 mg daily or
Erlotinib 150 mg daily
ORR: 80% (A) vs. 76% (B)
mPFS: 18.9 months (A) vs. 10.2 months (B) p < 0.001
mOS: 38.6 months (A) vs. 31.8 months (B) p = 0.046
Overall grade ≥3 AE: 42% (A) vs. 47% (B)15% (A) vs. 18% (B)18 April 2018
Erlotinib + ramucirumabRELAY
(NCT02411448)
IIIN = 449
Stage IV
NSCLC
-Race: Asian (77%), White (22.3%), Other (1%)
-EGFR Mutations: ex19del, L858R
-Brain or leptomeningeal metastases excluded
Cohort A:
Erlotinib 150 mg daily
+ ramucirumab 10 mg/kg once every 2 weeks
Cohort B:
Erlotinib 150 mg daily
+ placebo once every 2 weeks
ORR: 76% (A) vs. 75% (B)
mPFS: 19.4 months (A) vs. 12.4 month
p ≤ 0.0001
mOS: Not available
Overall grade ≥3 AE: 72% (A) vs. 54% (B)13% (A) vs. 11% (B)29 May 2020
* Only individual grade ≥3 AEs are reported; Abbreviations: AEs, adverse events; FDA, Food and Drug Administration; mPFS, median progression-free survival; mOS, median overall survival; ORR, overall response rate; NSCLC, non-small cell lung cancer; TKI, tyrosine kinase inhibitor.
Table 2. Mechanisms of Resistance to EGFR-TKI.
Table 2. Mechanisms of Resistance to EGFR-TKI.
ClassificationSub-ClassificationExamples
PrimaryCoexisting Activating Mutations/fusionsUncommon EGFR Mutations: EGFR Exon 20 insertions or duplications, de novo EGFR T790M
Other: MET amplifications, ALK fusions/EML4-ALK fusions
Heterogeneity in TKI ResponseCellular apoptotic machinery heterogeneity/Baseline BIM protein expression differences
Secondary/
Acquired
EGFR-DependentEGFR T790M (“gatekeeper” mutation)
Non-T790M EGFR Mutations: D761Y, S768I, V769L, C797X, L792X, G719A, G769X, L718Q, or G724S
EGFR-IndependentBypass Mechanisms:
(A) Genetic alterations: MET exon 14 skipping mutation, ERBB2/HER2 mutations/amplification, HER3 upregulation, RET or FGFR3 fusions, PIK3CA/BRAF/KRAS mutations
(B) Immune escape: PD-L1 upregulation
Histologic Transformation:
(A) Small Cell Lung Cancer
(B) EMT
Abbreviations: EMT: Epithelial-to-mesenchymal transformation; PD-L1: Programmed Death-Ligand 1; TKI, tyrosine kinase inhibitor.
Table 3. Strategies to Overcome TKI Resistance.
Table 3. Strategies to Overcome TKI Resistance.
StrategyDrugsSupporting Clinical TrialPopulationInterventionORRPFSOS
3rd Generation TKIsOsimertinibAURA 3Stages IIIB/IV Adenocarcinoma
EGFR T790M after failure to 1st or 2nd generation TKIs
N = 419
Cohort A: Osimertinib 80 mg daily
Cohort B:
Pemetrexed 500 mg/m2 with either carboplatin AUC 5 or cisplatin 75 mg/m2 every 3 weeks
71% (A) vs. 31% (B)
p < 0.001
10.1 months (A) vs. 4.4 months (B), HR 0.30 p < 0.00126.8 months (A) vs. 22.5 months (B)
p = 0.277
Chemo-Immunotherapy +/− anti-VEGF therapyAtezolizumab +/− BevacizumabIMpower150Stage IV non-squamous NSCLC. Those with EGFR mutations should have received and progressed or had unacceptable toxicities while on TKI
N = 124/1202 EGFR positive
Cohort A: ABCP:
Atezolizumab 1200 mg, bevacizumab 15 mg/Kg, carboplatin AUC 6, paclitaxel 200 mg/m2 every 3 weeks
Cohort B: ACP: Atezolizumab 1200 mg, carboplatin AUC 6, paclitaxel 200 mg/m2 every 3 weeks
Cohort C: BCP: Bevacizumab 15 mg/Kg, carboplatin AUC 6, paclitaxel 200 mg/m2 every 3 weeks
70.6% (A) vs. 35.6% (B) vs. 41.9% (C)10.2 months (A) vs. 6.9 months (C)
HR 0.61
CI 0.36–1.03
6.9 months (B) vs. 6.9 months (C),
HR 1.14
CI 0.73–1.78
26.1 months (A) vs. 20.3 months (C),
HR 0.91
CI 0.53–1.59
21.4 months (B) vs. 20.3 months (C),
HR 1.16
CI 0.71–1.89
IMpower130Stage IV non-squamous NSCLC. Those with EGFR mutations should have received and progressed or had unacceptable toxicities while on TKI
N = 44/724 with EGFR or ALK genomic aberrations
Cohort A: Atezolizumab 1200 mg, carboplatin AUC 6, and nab-paclitaxel 100 mg/m2 every 3 weeks
Cohort B:
Carboplatin AUC 6, and nab-paclitaxel 100 mg/m2 every 3 weeks
EGFR-cohort not reported7.0 months (A) vs. 6.0 months (B),
HR 0.75
CI 0.36–1.54
14.4 months (A) vs. 10.0 months (B),
HR 0.98
CI 0.41–2.31
Sintilimab +/− IBI305ORIENT-31EGFR-mutant non-squamous NSCLC who had progressed after EGFR TKI
N = 444
Cohort A: Sintilimab 200 mg, IBI305 15 mg/Kg, cisplatin 75 mg/m2, pemetrexed 500 mg/m2 every 3 weeks
Cohort B: Sintilimab 200 mg, placebo, cisplatin 75 mg/m2, pemetrexed 500 mg/m2 every 3 weeks
Cohort C: Placebo, cisplatin 75 mg/m2, pemetrexed 500 mg/m2 every 3 weeks
43.9% (A) vs. 33.1% (B) vs. 25.2% (C)6.9 months (A) vs. 4.3 months (C)
HR 0.464
p < 0.0001
5.6 months (B) vs. 4.3 months (C)
HR 0.726
p = 0.0584
NA
ImmunotherapyPembrolizumabKEYNOTE-001Advanced NSCLC
N = 78/550 with common EGFR mutations
Pembrolizumab 2 or 10 mg/Kg every 3 weeks, or 10 mg/Kg every 2 weeks7.7% (all), 20% PD-L1 ≥50%, 8.7% PD-L1 1–49%, 0% <1%NANA
DurvalumabATLANTICAdvanced NSCLC and disease progression after ≥2 systemic therapies
N = 111/444 with EGFR or ALK mutations
Durvalumab 10 mg/Kg every 2 weeks3.6% (PD-L1 <25%) vs. 12.2% PD-L1 ≥25%1.9 months (PD-L1 <25%) vs. 1.9 months (PD-L1 ≥25%)9.9 months (PD-L1 <25%) vs. 13.3 months (PD-L1 ≥25%)
c-MET Agents ORCHARD
(Experimental Module 1)
Metastatic NSCLC with EGFR and MET alterations after progression on first line osimertinib
N = 20
Osimertinib 80 mg daily with savolitinib 300 or 600 mg daily41%NANA
TATTONLocally advanced or metastatic NSCLC with EGFR mutation and MET amplification after progression on EGFR TKIs
Cohort B1: Previously treated with 3rd generation TKI
N = 69
Cohort B2: No 3rd previous generation TKI T790M negative N = 51
Cohort B3: No 3rd previous generation TKI T790M positive N = 18
Cohort D: No previous 3rd generation TKI T790M negative N = 42
Cohort B: Osimertinib 80 mg daily with savolitinib 300 mg (if ≤55 Kg) or 600 mg daily
Cohort D: Osimertinib 80 mg daily with savolitinib 300 mg daily
All B: 48%
B1: 30%
B2: 65%
B3: 67%
D: 64%
All B: 7.6 months
B1: 5.4 months
B2: 9.0 months
B3: 11.0 months
D: 9.1 months
NA
CHRYSALISMetastatic or unresectable NSCLC with EGFR mutation who progressed on osimertinib and were chemotherapy naïve
N = 166 (121 cohort A, 45 cohort B)
Cohort A: Amivantamab 1050 mg (1400 mg for patients ≥80 kg) weekly
Cohort B: Amivantamab 1050 mg (1400 mg for patients ≥80 kg) weekly with lazertinib 240 mg daily
Cohort A: 19%
Cohort B: 36%
NA
Cohort A
median DOR: 5.9 months
Cohort B median DOR: 9.6 months
NA
CHRYSALIS 2Advanced or metastatic NSCLC with EGFR exon 19 deletion or L858R that progressed after osimertinib (1st or 2nd line) and
platinum-based
chemotherapy
N = 162
Amivantamab 1050 mg (1400 mg if ≥80 Kg) with lazertinib 240 mg daily36%NA
DOR: not reached
NA
ADC NCT03260491Locally advanced or metastatic EGFR-mutant NSCLC who fail prior TKI
N = 57
Prior osimertinib and platinum-based chemotherapy:
N = 44
HER3-DXd (pertuzumab deruxtecan) 5.6 mg/kg every 3 weeksAll: 39%
Prior osimertinib and platinum chemotherapy: 39%
All: 8.2 months
Prior osimertinib and platinum chemotherapy: 8.2 months
NA
TROPION-PanTumor 01Advanced or metastatic NSCLC with Actionable Mutations who failed TKI and
chemotherapy
N = 34
EGFR-mutant: N = 29, 65% after osimertinib
Datopotamab deruxtecan 4 mg/Kg, 6 mg/Kg, or 8 mg/Kg35%NA
Median DOR: 9.5 months
NA
Abbreviations: ADC, Antibody Drug Conjugate; AEs, adverse events; AUC, area under the curve; DOR, duration of response; NA, not available; NSCLC, non-small cell lung cancer; OS, overall survival; ORR, overall response rate; PFS, progression-free survival; TKI, tyrosine kinase inhibitor.
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

Marin-Acevedo, J.A.; Pellini, B.; Kimbrough, E.O.; Hicks, J.K.; Chiappori, A. Treatment Strategies for Non-Small Cell Lung Cancer with Common EGFR Mutations: A Review of the History of EGFR TKIs Approval and Emerging Data. Cancers 2023, 15, 629. https://doi.org/10.3390/cancers15030629

AMA Style

Marin-Acevedo JA, Pellini B, Kimbrough EO, Hicks JK, Chiappori A. Treatment Strategies for Non-Small Cell Lung Cancer with Common EGFR Mutations: A Review of the History of EGFR TKIs Approval and Emerging Data. Cancers. 2023; 15(3):629. https://doi.org/10.3390/cancers15030629

Chicago/Turabian Style

Marin-Acevedo, Julian A., Bruna Pellini, ErinMarie O. Kimbrough, J. Kevin Hicks, and Alberto Chiappori. 2023. "Treatment Strategies for Non-Small Cell Lung Cancer with Common EGFR Mutations: A Review of the History of EGFR TKIs Approval and Emerging Data" Cancers 15, no. 3: 629. https://doi.org/10.3390/cancers15030629

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