Next Article in Journal
Intramyocardial Inflammation after COVID-19 Vaccination: An Endomyocardial Biopsy-Proven Case Series
Next Article in Special Issue
Carborane-Containing Hydroxamate MMP Ligands for the Treatment of Tumors Using Boron Neutron Capture Therapy (BNCT): Efficacy without Tumor Cell Entry
Previous Article in Journal
Novel Drugs with High Efficacy against Tumor Angiogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It?

1
Dipartimento di Oncologia Medica, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
2
Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
3
Dipartimento di Radioterapia, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 6936; https://doi.org/10.3390/ijms23136936
Submission received: 11 May 2022 / Revised: 20 June 2022 / Accepted: 21 June 2022 / Published: 22 June 2022
(This article belongs to the Special Issue Molecular Targets of Anticancer Therapy)

Abstract

:
Osimertinib is currently the preferred first-line therapy in patients with non-small cell lung cancer (NSCLC) with common epidermal growth factor receptor (EGFR) mutation and the standard second-line therapy in T790M-positive patients in progression to previous EGFR tyrosine kinase inhibitor. Osimertinib is a highly effective treatment that shows a high response rate and long-lasting disease control. However, a resistance to the treatment inevitably develops among patients. Understanding the secondary mechanisms of resistance and the possible therapeutic options available is crucial to define the best management of patients in progression to osimertinib. We provide a comprehensive review of the emerging molecular resistance mechanism in EGFR-mutated NSCLC pre-treated with osimertinib and its future treatment applications.

1. Introduction

To date, the treatment of non-small-cell lung cancer (NSCLC) is strictly related to molecular profiling, which has allowed the development of target therapies that have changed the natural history of this disease in recent decades. The Epidermal Growth Factor Receptor (EGFR)-activating mutations are the most common molecular aberrations in NSCLC, being present in 15% of the Caucasian population and up to 50% of the Asian population with advanced NSCLC [1,2]. Exon 19 deletion or L858R point mutation in exon 21, also known as “common mutation”, account for about 90% of these mutations [2]. EGFR tyrosine kinase inhibitors (TKIs) have been demonstrated to be superior over classical chemotherapy in this population and represent the standard of care for EGFR-mutated NSCLC [3,4,5]. The secondary mutation of T790M is the most commonly acquired resistance mechanism to first- and second-generation EGFR TKIs and it develops in over half of patients [6,7,8]. T790M is a gatekeeper mutation that increases the affinity for ATP in the ATP-binding domain of EGFR. As a result, it reduces the potency of first- and second-generation EGFR TKIs that have an ATP-competitive mechanism of action [9]. To overcome this resistance mechanism, third-generation TKI have been developed. Osimertinib is the most used third-generation TKI designed to target both EGFR-sensitising mutations and T790M in a selective way [10]. Originally, osimertinib was approved only in EGFR T790M-positive mutation NSCLC patients progressing following EGFR TKI therapy [8]. Later, the FLAURA study demonstrated a significant improvement in progression-free survival (PFS) and overall survival (OS) compared to treatment with first-generation EGFR TKIs [11]. Additionally, an improved tolerability profile (due to high selectivity against mutant vs. wild-type EGFR) and substantial central nervous system activity made osimertinib a widely adopted treatment even in the first-line setting, regardless of whether T790M is present [11,12].
Unfortunately, patients inescapably develop secondary resistance, which constitutes a critical challenge due to the scarcity of post-osimertinib pharmacological options available. The resistance mechanisms to a third-generation TKI are complex and not fully understood, with some differences depending on whether osimertinib is the first- or second-line treatment [13,14]. Understanding the resistance mechanisms to osimertinib and the possible treatment options available is essential in choosing the subsequent therapy strategy, largely because of upfront setting anticipation of osimertinib. The purpose of this review is to give a usable overview of our current knowledge about the emerging resistance mechanisms to osimertinib in patients with EGFR-mutated NSCLC and relevant therapeutic options.

2. Acquired Resistance to Osimertinib

The acquired resistance mechanisms to osimertinib may be either dependent (-“on target”) or independent (-“off-target”) of the EGFR. In the first case, tumour cell proliferation remains directly linked to EGFR signalling. In off-target resistance, other parallel molecular pathways circumvent EGFR signalling. The resistance mechanisms to osimertinib appear to be analogous in the first- and second-line settings [15]. Nevertheless, the off-target mechanism of resistance may be more relevant in first-line osimertinib than in later-line treatment, in which tumour cells have previously displayed dependence on EGFR through T790M mutation [16].
The plasma analysis of circulating tumour DNA (ctDNA) by NGS in the FLAURA and AURA3 studies in patients with progressive disease during osimertinib therapy provided us the majority of available data [13,14].
Table 1 summarises the ongoing clinical trial in pretreated EGFR-mutant metastatic NSCLC.
Figure 1 summarises the acquired resistance mechanism to osimertinib in first- and second-line treatment.

2.1. EGFR On-Target Alterations

2.1.1. T790M Loss

In 50–60% of patients treated with first- or second-generation TKI, the somatic mutation of resistance p.Thr790Met (T790M) develops, resulting from a gatekeeper mutation in exon20 of EGFR [17]. T790M causes steric hindering of the binding to their connected ATP-binding site on EGFR of an ATP-competitive kinase inhibitor (first- and second-generation TKI), but irreversible inhibitors (third-generation TKIs) overcome this resistance simply through covalent binding [9]. More precisely, osimertinib irreversibly and covalently binds the cysteine-797 residue in the ATP binding pocket of EGFR, regardless the hindering of T790M. Furthermore, since osimertinib creates an irreversible link with the ATP pocket of the EGFR it is able to overcome the increased affinity of ATP determined by the T790M mutation [10]. Data from the AURA3 trial highlight that about 50% of patients who received second-line osimertinib (and 100% of whom developed a dependent EGFR tertiary mutation) at the time of progression preserved the T790M mutation [13]. In the remaining cases, resistance to osimertinib commonly showed the loss of T790M and was frequently associated with the development of KRAS mutation, gene fusions, histological transformation and other rarer mechanisms [13,18]. Interestingly, earlier resistance and poorer survival was associated with T790M loss [18,19]. As expected, at progression to front-line osimertinib treatment no evidence of T790M mutation occurred [14] and, given the anticipation of osimertinib in the first-line setting, the incidence of T790M is likely to become less and less relevant.

2.1.2. C797x

In EGFRm/T790M patients, second-line treatment with osimertinib can lead to the emergence of the so-called EGFR “triple mutant”. The most common tertiary EGFR mutation is the point mutation C797 in exon 20 and it accounts for 15–26% of cases of resistance to second-line osimertinib treatment [11,13,20]. The most commonly found substituted amino acid is serine (C797S), and glycine (C797G) has been anecdotally reported [20,21]. Indeed, the bond to the residue C797 in the ATP pocket is how osimertinib exceeds the T790M resistance [22,23]. Moreover, it is the second most recurrent resistance mechanism (7%) after MET amplification in first-line osimertinib [14]. Case reports have described that in the absence of T790M mutation, cancer cells harbouring C797S mutation maintain first- and second-generation EGFR TKI sensitivity [24]. The same is true in cases where C797S mutation coexists in a different allele with T790M (trans)—if the C797S mutation is in the same allele (cis), as in most T790M mutated cases, sensitivity to first and second-generation TKI is lost [22,25]. Two case reports reported patients with EGFR C797S mutation located in trans with T790M who experienced an initial response with the association of a first-generation TKIs and osimertinib [25,26]. Additionally, in vitro and in vivo activity of fourth-generation EGFR TKIs, alone or in combination with osimertinib, have been demonstrated. These new generation of EGFR TKIs such as EAI045, JBJ-04-125-02 and BLU-945, overcome both T790M and C797S mutations. However, they still have not been assessed in a clinical trial [27,28,29]. Moreover, the addition of the ALK inhibitor brigatinib to a fourth-generation EGFR TKI has revealed in vivo activity in triple-mutant EGFR/T790M/C797S [30]. Interestingly, amivantamab (a bispecific anti-EGFR and anti-MET inhibitor) showed response in patients with coexisting C797S mutation and MET amplification [31] and brigatinib plus cetuximab may be an efficacious therapy option in patients with T790M/cisC797S mutations resistant to osimertinib [32]. BDTX-1535, an orally available, highly potent, selective, irreversible inhibitor of allosteric EGFR alterations (NCT05256290), BLU-945, a selective EGFR inhibitor, as monotherapy or in combination with osimertinib [NCT04862780], BLU-701 in monotherapy or in combination with bot osimertinib or platinum-based chemotherapy (NCT05153408) are currently in testing in phase I/II clinical trials (Table 1).

2.1.3. Other EGFR Tertiary Mutations

EXON 18. Mutation in L718Q/V residue interacts directly with osimertinib in the EGFR kinase domain of the ATP binding site [33]. Of note, NSCLC with EGFR L858R/T790M/L718Q/V mutation are resistant to all EGFR-TKIs but L858R/L718Q/V (commonly found in patients who develop resistance to osimertinib [34]) seems to retain sensibility to afatinib [35]. Furthermore, the rare G724S mutation has been outlined as a resistance mechanism to second-line osimertinib [36]. In a subgroup of EGFR T790M negative but G724S mutated and osimertinib-resistant patients, Fassunke et al. demonstrated in vitro that afatinib reduces tumour growth of G724S driven cells [37].
EXON 20. G796R/D and L792 mutations were anecdotally reported in NSCLC treated with osimertinib and sterically hindered it, but drug sensitiveness against these newly on-target resistance mechanisms mandates additional investigations [36]. In vitro, double mutant T790M/M766Q are resistant to osimertinib but sensitive to neratinib and poziotinib (dual inhibitors of the human epidermal growth factor receptor 2 (HER2) and EGFR kinase) [38].

2.2. EGFR Off-Target Alteration

2.2.1. MET Amplification

MET amplification is one of the most common mechanisms of acquired resistance to osimertinib, with a prevalence of 19% and 15% of patients, respectively, receiving second- and first-line therapy [13,14,18]. MET amplification bypasses EGFR by causing persistent activation of downstream signalling paths mediated by phospho-inositide 3-kinase (PIK3CA), mitogen-activated protein kinase (MAPK), and signal transducer and activator of transcription (STAT) [39]. At present, MET amplification is generally defined as the presence of a MET gene copy number of ≥5 or a MET/CEP7 ratio of ≥2 [18]. So far, there is an absence of agreement on the definition of MET amplification detected by next-generation sequencing (NGS) in liquid biopsy, and NGS or fluorescence in situ hybridisation (FISH) to detect MET amplification should be carried out on all biopsies performed to define osimertinib resistance. NGS allows the parallel identification of single-nucleotide variants, rearrangements, deletions, insertions, copy number variations, and the definition of distinct thresholds [21,40]. However, not all NGS-based assays control for CEP7; consequently, a detected increase in copy number may actually be a polysomy instead of a proper MET amplification. [41]. Therefore, FISH is advised if NGS does not expressly assess for gene copy number gain [42]. MET amplification may emerge with or without T790M loss in the second-line osimertinib setting. In 7% of cases, it co-occurs with the tertiary mutation EGFR C797S [13] and is also potentially associated with CDK6 and BRAF amplification [43]. Preclinical evidence shows the osimertinib resistance in EGFR-mutated cell lines with MET amplification could be overcome by the concomitant use of MET inhibitors with afatinib [44]. Case reports suggested that combining crizotinib (a TKI with dual anti ALK and MET activity) with osimertinib or erlotinib might get over MET-mediated resistance [45,46,47]. Various combinations of EGFR and MET TKI are currently under investigation. Recently, data have been published from an interim analysis of the phase study Ib TATTON, which investigates the combination osimertinib-savolitinib, a MET TKI, in patients with MET amplification defined as MET gene copy number ≥ 5 or MET:CEPT7 ≥ 2. An objective response rate (ORR) of 30% and a PFS of 5.4 months were obtained in third-generation EGFR TKI pretreated patients, reaching 64–67% and 9–11 months, respectively, in third-generation EGFR TKI-naïve patients based on the different cohorts analyzed [48]. Phase II trials with this combination are currently ongoing (NCT03778229 (SAVANNAH), NCT03944772 (ORCHARD)). Two phase I/II studies tested the combination of the first-generation gefitinib with a MET inhibitor. The association of capmatinib with gefitinib obtained an ORR of 27% and an increased ORR of 47% was seen in patients with MET gene copy number ≥ 6 [49]. Similarly, the combination of tepotinib with gefitinib resulted in longer PFS and OS in compared to chemotherapy in the INSIGHT trial [50]. Initial results from the CHRYSALIS phase I trial, still ongoing, showed an ORR of 36% and a mPFS of 4.9 months in the osimertinib-resistant cohort was described with amivantamab combined with lazertinib (third-generation EGFR TKI, brain-penetrant) [51]. At the recent ASCO 2022 Annual meeting, updated results of pretreated cohort has been presented. At a median follow up of about 8 months, an ORR of 36% was confirmed in patients pretreated with first- or second-line osimertinib, with a clinical benefit rate of 58% and a median duration of response (mDoR) not reached. Moreover, in patients heavily pretreated with at least osimertinib plus a platinum-based chemotherapy, an ORR of 29% with a mDoR of 8.6 months was described. A manageable safety profile was confirmed [52].
Telisotuzumab vedotin is a MET-directed antibody–drug conjugate (ADC) that is in testing in a currently ongoing phase 1/1b study NCT02099058 in monotherapy or combination with osimertinib, erlotinib or nivolumab in patient harboring cMET overexpression after prior osimertinib therapy. In the interim analysis, the encouraging results of an ORR of 58% observed for the combination of telisotuzumab vedotin with osimertinib and an acceptable safety profile were presented at the ASCO 2022 annual meeting [53].
Other ongoing clinical trials targeting MET amplification are listed in Table 1.

2.2.2. HER2 Amplification

Another off-target mechanism that bypasses EGFR through the activation of downstream PI3K–Akt and MAPK/pathways is the overexpression of ErbB2, a tyrosine kinase receptor encoded by the HER2 gene. In patients who developed resistance to second-line and first-line osimertinib, HER2 amplification was detected in 5% and 2% of cases, respectively. Interestingly, HER2 amplification is mutually exclusive with T790M [13,14]. HER2 amplification resistance was sensitive to osimertinib plus the ADC anti-HER2 trastuzumab-emtansine (TDM1) in preclinical models. Recently, Li et al. demonstrated activity of TDM1 even in patients with EGFR-mutated HER2 amplification NSCLC who experienced disease progression on previous EGFR TKI [54]. Moreover, they described that ADC switching from TDM1 to trastuzumab deruxtecan (T-DXd), holding a distinct cytotoxic payload, achieves durable responses in a NSCLC patient that developed resistance to T-DM1 [54]. The TRAEMOS phase I/II trial is investigating the osimertinib–TDM1 combination in patients who progressed to an EGFR TKI gaining HER2 amplification (NCT03784599).

2.2.3. RAS/MAPK Pathway Mutations

KRAS mutation or amplification and NRAS, MEK1 and BRAF mutation have all been reported as acquired resistance to osimertinib [39]. In the FLAURA trial, variable mutations of NRAS (e.g., E63K mutation) and KRAS (G12S, G13D, Q61R, Q61K, G12D mutations) were discovered in 3% and 1% of patients who progressed on first-line and second-line treatment, respectively [14,43]. BRAF V600E mutation was found in about 3% of cases, with or without T790M, both in first- and second-line osimertinib [13,14,18]. Furthermore, BRAF V600E mutation coexisting with MET amplification as resistance mechanisms to first-line osimertinib therapy was reported [55]. BRAF V600E-mutated cell lines after osimertinib treatment showed sensitivity to combining a BRAF inhibitor (encorafenib) and osimertinib [56]. Likewise, Xie et al. proved that osimertinib combined with vemurafenib (a BRAF inhibitor) effectively overcame BRAF V600E-mediated osimertinib resistance [57]. Furthermore, both in vitro and in vivo, salumetinib (a MEK inhibitor) combined with osimertinib has been proven to overcome TKI resistance caused by NRAS mutations, although further evidence of this combination is required [58]. The association of dabrafenib and trametinib in BRAF V600E mutated NSCLC, including EGFR-mutated patients who progressed to an EGFR TKI is currently under investigation (NCT04452877).

2.2.4. PI3K Pathway Mutations

Activation of PI3K, either via PIK3CA mutations (E454K, E542K, R88Q, N345K, E418K) or PTEN deletion, is involved in 4–11% of patients who progressed to osimertinib [18]. Because of PIK3CA has a role in several oncogenic pathways in NSCLC, in contrast to other oncogenic driver mutations which are generally mutually exclusive, PIK3CA mutation is frequently contextual to other oncogenic gene mutations [59]. In patients with associated PIK3CA and EGFR mutations treated with EGFR TKI monotherapy no significant dissimilarities in clinical results were recorded [59]. To our knowledge, no targeted therapy against PIK3CA mutation has demonstrated clinical benefit thus far.

2.2.5. Oncogenic Fusions: FGFR3, RET, NTRK

Chromosomal rearrangements involving driver oncogenes, namely the oncogenic fusions, have been identified mainly in second-line osimertinib resistance (4–7%) [13,18]. Oncogenic fusions include but are not limited to ALK (SBTBN1-ALK, only in first-line osimertinib resistance, PLEKHA7-ALK), BRAF (AGK-BRAF, PCBP2-BRAF, ESYT2-BRAF, BAIAP2L1-BRAF), FGFR (FGFR3-TACC3), NTRK (NTRK-TMP3), RET (RET-ERC1, CCD&-RET, NCOA4-RET) and ROS1 (GOPC-ROS1) [13,14,18,60]. Zeng et al. reported that in a GOPC-ROS1 rearranged patients, a combination of crizotinib and osimertinib was proven effective and well tolerated [60]. Piotrowska et al. published the experience of two patients with acquired CCDC6-RET fusion who had rapid responses to the combination of osimertinib–RET inhibitor (BLU-667) [61]. Moreover, in a series of 12 patients and in a case report with RET fusions as mechanism of osimertinib resistance the combination of osimertinib and the RET inhibitor selpercatinib was feasible and achieved radiological response [62,63]. One patient in progression to osimertinib who developed PLEKHA7-ALK fusion obtained a durable response with the addition of alectinib (an ALK TKI) to osimertinib. The combination therapy targeting EGFR and the acquired fusion achieved clinical benefits in numerous patients [64]. Another brief report showed that in two patients with ALK-AML4 fusion, osimertinib association with both crizotinib and alectinib obtained disease control [65]. Although the data come from case reports or small series, they are encouraging to think about in the development of target therapy combinations even in this setting.

2.2.6. Cell Cycle Alterations

Plasma analysis of the studies AURA3 and FLAURA found that 10% of resistance mutations in first-line osimertinib and 12% in second-line treatment are represented by alteration of cell cycle-related genes. These include amplification or mutations in cyclin D1/2 and E1 genes, cyclin-dependent kinase (CDK) 4/6 and CDK inhibitor 2A genes [13,14]. The combination of CDK4/6 inhibitor palbociclib and osimertinib overcame the acquired resistance of osimertinib in cell lines [66]. Analogously, La Monica et al. provided preclinical evidence for employing abemaciclib (monotherapy or in addition to osimertinib) to overcome resistance in patients progressing to first-line osimertinib. They also suggested the combination of osimertinib and abemaciclib as a potential approach to prevent or delay resistance to osimertinib in first-line therapy [67]. Ongoing clinical trials are listed in Table 1.

2.2.7. Other Mechanisms

Anexelekto (AXL) belongs to the receptor tyrosine kinase family, implicated in cell proliferation, survival and migration. Upregulated AXL interacting with EGFR and HER3 induces intrinsic and acquired osimertinib resistance [68]. In cell lines, the combination of osimertinib and cabozantinib has been reported to overcome osimertinib resistance [68]. Moreover, several authors have suggested the preclinical efficiency of using AXL inhibitors combined with osimertinib on cell lines resistant to osimertinib, making it an attractive pharmacological target [69,70,71].
Aberrant activation of the insulin-like growth factor 1 receptor (IGFR1R) has been suggested as one non-genetic cause of third-generation EGFR TKI resistance in T790M mutated NSCLC [72]. Adding an IGFR1 inhibitor to osimertinib might efficaciously overcome the acquired resistance to osimertinib elicited by IGF1R activation [73].
Patritumumab deruxtecan is an ADC directed against HER3 (ErBB3), another often-overexpressed receptor in EGFR-mutated NSCLC. HER3 alterations do not directly mediate resistance to EGFR-TKIs, but HER3 activates oncogenic signalling pathways, including PI3K and MAPK. Nevertheless, patritumab deruxtecan, in a phase I clinical trials in osimertinib-resistant patients, achieved a response rate of 39%, irrespective of the underlying resistance mechanism. HER3-directed ADC might provide a future agnostic treatment alternative for the TKI resistance mechanism of EGFR [74]. A prospective clinical trial is currently ongoing, testing patrimumab deruxtecan in combination with osimertinib in patients progressing to first-line osimertinib (NCT04676477).

2.2.8. Histological Transformation

Unlike mutational gene status, a study of tissue samples/re-biopsy is required to assess the existence of histologic transformation as an acquired resistance mechanism to osimertinib. The histologic conversion from EGFR-mutated NSCLC into small cell lung cancer (SCLC) has been recorded in 14% of patients progressing to first-line osimertinib and in 4–15% of patients experiencing disease progression in the second-line setting [6,18,61]. Notably, the risk of SCLC transformation has been significantly associated with the contemporary presence of RB1 and TP53 mutations, while no SCLC cases were recorded in wild-type patients [75,76]. Therefore, for lack of other resistance mechanisms, a liquid biopsy positive for RB1 or TP53 alterations may imply that a tissue re-biopsy should be considered to search for SCLC transformation. Likewise, in about 15% of patients receiving both first- and second-line osimertinib, squamous cell transformation is described [77]. Both in SCLC and squamous transformation, the original EGFR-mutation is retained [75,78]. Currently, for EGFR-mutant NSCLC with transformed histology, which generally has a worse prognosis as a consequence of intrinsic resistance mechanisms, there are no target therapies or therapeutic strategies validated [75,76]. Histology-driven chemotherapy would remain the standard of care in this patient subgroup even if traditional systemic chemotherapy yielded limited efficacy, notwithstanding some evidence of efficacy of platinum-etoposide chemotherapy—but no immune checkpoint inhibitors (ICIs)—in SCLC-transformed NSCLC [75,76,79]. Of note, the association of a PARP inhibitor (niraparib) and anti PDL1 durvalumab is currently under investigation in SCLC-transformed EGFR-mutated NSCLC (NCT04538378).
Resistance to osimertinib has also been described in epithelial-to-mesenchymal transition (EMT) and over-expression of its transcription factor TWIST-1 by NSCLC cells. The assumption of a mesenchymal phenotype confers migratory capacity to the cells through the loss of the expression of cadherin in favor of vimentin [80]. In the preclinical setting, TWIST-1 inhibitors are under investigation [81].

3. New First-Line Combinations Aming to Prevent the Onset of Resistance

Because in up to 40–50% of cases, there are currently no known detectable changes and as osimertinib is the first-line choice TKI and chemotherapy is the second standard line, the researchers are studying the possibility of preventing the emergence of resistance to third-generation EGFR TKI by combining, in various ways, TKI, chemotherapy, and immunotherapy in the front-line setting. Ongoing clinical trials in this setting are listed in Table 2.

3.1. Chemotherapy

Clinical data support the superiority in terms of PFS, of the combination of a first-generation TKI and chemotherapy versus EGFR TKI monotherapy [82,83,84]. Moreover, the osimertinib–chemotherapy combination had a good safety profile and demonstrated promising control of central nervous system (CNS) disease in a group of patients who progressed systemically to at least two lines of therapy, including an EGFR TKI [85]. The ongoing phase III trial FLAURA2 compares first-line osimertinib plus a platinum-pemetrexed based chemotherapy with osimertinib alone in EGFR-mutated NSCLC. The first results published encouragingly demonstrated manageable safety and tolerability of this combination [86]. Additionally, a study designed to prevent SCLC transformation is combining platinum–etoposide chemotherapy plus osimertinib in the first-line setting for EGFR-mutated NSCLC with concurrent RB1 and TP53 mutations (NCT03567642).

3.2. VEGF Inhibitors

Another association of interest is between EGFR TKI and an antiangiogenic agent. The rationale is that VEGF signalling is regulated by EGFR expression and shares common downstream pathways; conversely, an EGFR independent VEGF up-regulation is supposed to promote resistance to EGFR inhibition. [87]. Even if the exact mechanism is currently not well understood, there is preclinical evidence that VEGF/VEGF receptor inhibition boosts EGFR TKI activity [88]. Various trials demonstrate a significant PFS benefit, but not translated into OS advantage, with first-generation EGFR TKI and the anti-VEGF monoclonal antibody bevacizumab or the monoclonal antibody targeting VEGF receptor 2 ramucirumab [89,90,91]. To date, the association in the second-line of bevacizumab and osimertinib in T790M patients failed to show prolongation of PFS vs. osimertinib alone [92], but other clinical trials are ongoing in the first-line setting (Table 1). Nishio et al. recently reported data from the RELAY+ phase III trial in which ramucirumab plus gefitinib achieved a positive 1-year PFS rate with a manageable safety profile in Asian patients in a first-line setting [93]. Data from a concluded phaseI/II study of combination osimertinib and bevacizumab in first-line EGFR mutant NSCLC are awaited (NCT02803203).

3.3. First-Generation TKI

In the absence of T790M mutation development, the on-target EGFR resistance mechanism to osimertinib seems to maintain sensitivity to first- and second-generation EGFR TKI. Consequently, combining a previous generation of TKI with osimertinib could potentially prevent on-target resistance [21]. Concurrent osimertinib plus gefitinib in the first-line setting was safe and obtained an objective response rate consistent with previously reported first-line osimertinib. However, survival outcomes and acquired resistance mechanism results are still awaited [94].

3.4. Alternative Pathways Inhibitors

To avoid the emergence of off-target resistance mechanism various strategies trying to co-target EGFR and alternative pathways are under investigation in several clinical trials (Table 1). Among these are awaited with particular interest the result of the amivantamab–lazertinib combination. Besides the CHRYSALIS phase I study mentioned above, two phase III clinical trials are currently ongoing. The MARIPOSA study (NCT04487080) compares the efficacy and safety of amivantamab–lazertinib combination therapy versus single-agent osimertinib and the AMIGO-1 trial evaluates the combination of amivantamab–lazertinib with platinum-pemetrexed based chemotherapy in treatment naïve EGFR-mutated NSCLC (NCT05299125).

3.5. Immunotherapy

EGFR-mutant NSCLC patients have historically been excluded from most first-line trials with immune checkpoint inhibitors. Previous evidence suggested that PD-L1 expression does not predict benefit in EGFR-mutant NSCLC [95]. Data from Impower130 showed that the addition of anti-PDL1 atezolizumab to chemotherapy does not improve survival in mutated EGFR patients, while from the Impower150 study comes the suggestion that there may be a certain synergy of the anti-VEGF and-PDL1 inhibitors since the addition of bevacizumab to atezolizumab and chemotherapy gave a survival benefit in this population [96,97]. More recently, the final exploratory analysis of Impower150 reported OS benefits for the atezolizumab–bevacizumab–carboplatin–paclitaxel combination against bevacizumab–carboplatin–paclitaxel therapy in patients with sensitising EGFR mutations and with liver metastases. Although exploratory, and therefore to be interpreted with caution, these data support a possible use of this combination at the forefront, even if there is no comparison with the standard current therapy with EGFR TKI [98].
No data are currently available in patients with EGFR-mutated NSCLC in progression after osimertinib. To elucidate this, the phase III trial KEYNOTE 789 is currently testing the efficacy and safety of pembrolizumab in addition to platinum–pemetrexed-based chemotherapy specifically in EGFR-mutated NSCLC in progression after an EGFR TKI, osimertinib included (NCT03515837). The combination of osimertinib with the anti-PDL1 durvalumab investigated in the TATTON study was burdened with high rates of immune-mediated adverse events and, in particular, interstitial lung disease, leading to premature termination of enrollment in this study and in the CAURAL phase III study (NCT02454933) [99]. A meta-analysis of adverse events in combination EGFR TKI and ICIs in advanced EGFR-mutant NSCLC confirmed that the joint incidences of gastrointestinal grade 3 skin and adverse events and ILD were significantly higher in combination therapy than in osimertinib monotherapy, limiting future clinical development of this association [100].

4. Conclusions

The optimal subsequent treatment after the progression to osimertinib must be tailored according to sites of progression and resistance mechanisms. In patients with limited progression sites, either in the brain or other organs, data support the beyond-progression osimertinib treatment with definitive local therapy, such as stereotactic radiotherapy or surgery of sites of oligo-progression [15,101] and other ongoing clinical trials such as NRG LU002 and SARON are ongoing with the same rationale (NCT03137771, NCT02417662).
In patients requiring a change in systemic therapy, although the standard of treatment is still platinum-based chemotherapy, strategies targeting specific resistance mechanisms are showing promising results, and they should be encouraging access to clinical trials with specific agents targeted at resistance alterations [102].
To date, for assessing osimertinib-resistance mechanisms, the gold standard remains a tissue re-biopsy, which allows histological evaluation, NGS, and an RNA-based fusion panel analysis. If tissue biopsy is not feasible, the ctDNA analysis (liquid biopsy) can be assessed, but it must be taken into account that not all cancers have a ctDNA shedding detectable [18], that histological transformation cannot be detected in liquid biopsy samples [77], and that oncogenic fusions and acquired gene amplifications are not always surely detected by available ctDNA testing. The tissue biopsy should be reconsidered if the ctDNA does not identify the basal EGFR mutation or resistance mechanisms. More comprehensive data are awaited from the ELIOS study (NCT03239340). Plasma genotyping and paired tumour biopsy from patients treated with first-line osimertinib will be analyzed by NGS to assess resistance mechanism.
In a first-line osimertinib-relapsed setting, the molecular-driven designed phase II ORCHARD platform trial is testing different agents in combination with osimertinib, according to the identified TKI resistance mechanism. They include, but are not limited to, savolitinib, gefitinib, necitumumab or others in the case of MET alteration, C797X mutation, EGFR amplification or no biomarker, respectively (NCT03944772). Considering the tropism of the oncogene disease addicted to the diffusion of the brain, the results of the phase 3 study COMPEL (NCT04765059) will be very interesting, evaluating the continuation of osimertinib or placebo with platinum-based chemotherapy in patients with EGFR-mutated metastatic NSCLC who responded to first-line osimertinib therapy and subsequently experienced radiological, extracranial disease progression, with stratification based on the presence or absence of specific endpoint encephalic metastases for CNS outcomes. Regardless of the identification of a biomarker, the combination of osimertinib with necitumumab, a highly selective monoclonal antibody against EGFR, was shown to be active and safe in patients with advanced EGFR-mutated NSCLC pre-treated with osimertinib in the first-line setting in a phase I study recently presented [103]. Although further studies are warranted, this could be another approach to be explored.
To conclude, after the failure of osimertinib treatment, access to clinical trials, subject to an extensive evaluation of the genomic profile, should be granted to all patients, not only to allow a potential treatment tailored on the basis of the resistance mechanism identified but also to delve deeper into the knowledge of the resistance mechanisms themselves.

Author Contributions

Conceptualization E.B.; resources, E.B.; writing—original draft preparation, E.B.; writing—review and editing, E.B., A.D.C., E.D.C., B.S.; visualization, E.B., A.D.C., E.D.C., B.S., A.R., K.F., A.B., M.S.; supervision, E.B., A.B., M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

This work was supported by the Italian Ministry of Health (Ricerca Corrente); no grant provided.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Midha, A.; Dearden, S.; McCormack, R. EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: A systematic review and global map by ethnicity (mutMapII). Am. J. Cancer Res. 2015, 5, 2892–2911. [Google Scholar] [PubMed]
  2. Graham, R.P.; Treece, A.L.; Lindeman, N.I.; Vasalos, P.; Shan, M.; Jennings, L.J.; Rimm, D.L. Worldwide Frequency of Commonly Detected EGFR Mutations. Arch. Pathol. Lab. Med. 2018, 142, 163–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. 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] [PubMed]
  4. 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]
  5. Sequist, L.V.; Yang, J.C.-H.; Yamamoto, N.; Obyrne, 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]
  6. 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]
  7. 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] [Green Version]
  8. 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]
  9. 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] [Green Version]
  10. Cross, D.A.; Ashton, S.E.; Ghiorghiu, S.; Eberlein, C.; Nebhan, C.A.; Spitzler, P.J.; Orme, J.P.; Finlay, M.R.V.; Ward, R.A.; Mellor, M.J.; et al. AZD9291, an Irreversible EGFR TKI, Overcomes T790M-Mediated Resistance to EGFR Inhibitors in Lung Cancer. Cancer Discov. 2014, 4, 1046–1061. [Google Scholar] [CrossRef] [Green Version]
  11. Ramalingam, S.S.; Vansteenkiste, J.; Planchard, D.; Cho, B.C.; Gray, J.E.; Ohe, Y.; Zhou, C.; Reungwetwattana, T.; Cheng, Y.; Chewaskulyong, B.; et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2020, 382, 41–50. [Google Scholar] [CrossRef] [PubMed]
  12. Ballard, P.; Yates, J.W.T.; Yang, Z.; Kim, D.-W.; Yang, J.C.-H.; 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] [Green Version]
  13. Papadimitrakopoulou, V.A.; Wu, Y.L.; Han, J.Y.; Ahn, M.J.; Ramalingam, S.S.; John, T.; Okamoto, I.; Yang, J.H.; Bulusu, K.C.; Laus, G.J.A.O.O.; et al. Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Ann. Oncol. 2018, 29, viii741. [Google Scholar] [CrossRef]
  14. Ramalingam, S.S.; Cheng, Y.; Zhou, C.; Ohe, Y.; Imamura, F.; Cho, B.C.; Lin, M.C.; Majem, M.; Shah, R.; Rukazenkov, Y.; et al. Mechanisms of acquired resistance to first-line osimertinib: Preliminary data from the phase III FLAURA study. Ann. Oncol. 2018, 29, viii740. [Google Scholar] [CrossRef]
  15. 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]
  16. Schoenfeld, A.J.; Yu, H.A. The Evolving Landscape of Resistance to Osimertinib. J. Thorac. Oncol. 2020, 15, 18–21. [Google Scholar] [CrossRef]
  17. Kobayashi, S.; Jänne, P.A.; Meyerson, M.; Eck, M.J. EGFR Mutation and Resistance of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
  18. Oxnard, G.R.; Hu, Y.; Mileham, K.F.; Husain, H.; Costa, D.B.; Tracy, P.; Feeney, N.; Sholl, L.M.; Dahlberg, S.E.; Redig, A.J.; et al. Assessment of Resistance Mechanisms and Clinical Implications in Patients with EGFR T790M–Positive Lung Cancer and Acquired Resistance to Osimertinib. JAMA Oncol. 2018, 4, 1527–1534. [Google Scholar] [CrossRef] [Green Version]
  19. Lin, C.-C.; Shih, J.-Y.; Yu, C.-J.; Ho, C.-C.; Liao, W.-Y.; Lee, J.-H.; Tsai, T.-H.; Su, K.-Y.; Hsieh, M.-S.; Chang, Y.-L.; et al. Outcomes in patients with non-small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: A genomic study. Lancet Respir. Med. 2018, 6, 107–116. [Google Scholar] [CrossRef]
  20. DE Carlo, E.; Schiappacassi, M.; Pelizzari, G.; Baresic, T.; DEL Conte, A.; Stanzione, B.; DA Ros, V.; Doliana, R.; Baldassarre, G.; Bearz, A. Acquired EGFR C797G Mutation Detected by Liquid Biopsy as Resistance Mechanism after Treatment with Osimertinib: A Case Report. Vivo 2021, 35, 2941–2945. [Google Scholar] [CrossRef]
  21. Passaro, A.; Jänne, P.A.; Mok, T.; Peters, S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat. Cancer 2021, 2, 377–391. [Google Scholar] [CrossRef] [PubMed]
  22. 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] [PubMed] [Green Version]
  23. Bersanelli, M.; Minari, R.; Bordi, P.; Gnetti, L.; Bozzetti, C.; Squadrilli, A.; Lagrasta, C.A.M.; Bottarelli, L.; Osipova, G.; Capelletto, E.; et al. L718Q Mutation as New Mechanism of Acquired Resistance to AZD9291 in EGFR -Mutated NSCLC. J. Thorac. Oncol. 2016, 11, e121–e123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Rangachari, D.; To, C.; Shpilsky, J.E.; VanderLaan, P.A.; Kobayashi, S.S.; Mushajiang, M.; Lau, C.J.; Paweletz, C.P.; Oxnard, G.R.; Jänne, P.A.; et al. EGFR-Mutated Lung Cancers Resistant to Osimertinib through EGFR-C797S Respond to 1st Generation Reversible EGFR Inhibitors but Eventually Acquire EGFR-T790M/C797S in Preclinical Models and Clinical Samples. J. Thorac. Oncol. 2020, 14, 1995–2002. [Google Scholar] [CrossRef] [PubMed]
  25. Arulananda, S.; Do, H.; Musafer, A.; Mitchell, P.; Dobrovic, A.; John, T. Combination Osimertinib and Gefitinib in C797S and T790M EGFR-Mutated Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 1728–1732. [Google Scholar] [CrossRef] [Green Version]
  26. Wang, Z.; Yang, J.-J.; Huang, J.; Ye, J.-Y.; Zhang, X.-C.; Tu, H.-Y.; Han-Zhang, H.; Wu, Y.-L. Lung Adenocarcinoma Harboring EGFR T790M and In Trans C797S Responds to Combination Therapy of First- and Third-Generation EGFR TKIs and Shifts Allelic Configuration at Resistance. J. Thorac. Oncol. 2017, 12, 1723–1727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Schalm, S.; Dineen, T.; Lim, S.; Park, C.-W.; Hsieh, J.; Woessner, R.; Zhang, Z.; Wilson, K.; Eno, M.; Wilson, D.; et al. 1296P BLU-945, a highly potent and selective 4th generation EGFR TKI for the treatment of EGFR T790M/C797S resistant NSCLC. Ann. Oncol. 2020, 31, S839. [Google Scholar] [CrossRef]
  28. To, C.; Jang, J.; Chen, T.; Park, E.; Mushajiang, M.; De Clercq, D.J.; Xu, M.; Wang, S.; Cameron, M.D.; Heppner, D.E.; et al. Single and Dual Targeting of Mutant EGFR with an Allosteric Inhibitor. Cancer Discov. 2019, 9, 926–943. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, S.; Song, Y.; Liu, D. EAI045: The fourth-generation EGFR inhibitor overcoming T790M and C797S resistance. Cancer Lett. 2016, 385, 51–54. [Google Scholar] [CrossRef]
  30. Maity, S.; Pai, K.S.R.; Nayak, Y. Advances in targeting EGFR allosteric site as anti-NSCLC therapy to overcome the drug resistance. Pharmacol. Rep. 2020, 72, 799–813. [Google Scholar] [CrossRef]
  31. Haura, E.B.; Cho, B.C.; Lee, J.S.; Han, J.Y.; Lee, K.H.; Sanborn, R.E.; Govindan, R.; Cho, E.K.; Kim, S.W.; Reckamp, K.L. JNJ-61186372 (JNJ-372), an EGFR-CMet Bispecific Antibody, in EGFR-Driven Advanced Non-Small Cell Lung Cancer (NSCLC). J. Clin. Oncol. 2021, 37, 9009. [Google Scholar] [CrossRef]
  32. Wang, Y.; Yang, N.; Zhang, Y.; Li, L.; Han, R.; Zhu, M.; Feng, M.; Chen, H.; Lizaso, A.; Qin, T.; et al. Effective Treatment of Lung Adenocarcinoma Harboring EGFR-Activating Mutation, T790M, and cis-C797S Triple Mutations by Brigatinib and Cetuximab Combination Therapy. J. Thorac. Oncol. 2020, 15, 1369–1375. [Google Scholar] [CrossRef] [PubMed]
  33. Dong, R.-F.; Zhu, M.-L.; Liu, M.-M.; Xu, Y.-T.; Yuan, L.-L.; Bian, J.; Xia, Y.-Z.; Kong, L.-Y. EGFR mutation mediates resistance to EGFR tyrosine kinase inhibitors in NSCLC: From molecular mechanisms to clinical research. Pharmacol. Res. 2021, 167, 105583. [Google Scholar] [CrossRef]
  34. Ma, L.; Chen, R.; Wang, F.; Ma, L.-L.; Yuan, M.-M.; Chen, R.-R.; Liu, J. EGFR L718Q Mutation Occurs without T790M Mutation in a Lung Adenocarcinoma Patient with Acquired Resistance to Osimertinib. Ann. Transl. Med. 2019, 7, 207. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, J.; Jin, B.; Su, H.; Qu, X.; Liu, Y. Afatinib helped overcome subsequent resistance to osimertinib in a patient with NSCLC having leptomeningeal metastasis baring acquired EGFR L718Q mutation: A case report. BMC Cancer 2019, 19, 702. [Google Scholar] [CrossRef]
  36. Zhang, Y.; He, B.; Zhou, D.; Li, M.; Hu, C. Newly emergent acquired EGFR exon 18 G724S mutation after resistance of a T790M specific EGFR inhibitor osimertinib in non-small-cell lung cancer: A case report. OncoTargets Ther. 2018, 12, 51–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Fassunke, J.; Müller, F.; Keul, M.; Michels, S.; Dammert, M.A.; Schmitt, A.; Plenker, D.; Lategahn, J.; Heydt, C.; Brägelmann, J.; et al. Overcoming EGFRG724S-mediated osimertinib resistance through unique binding characteristics of second-generation EGFR inhibitors. Nat. Commun. 2018, 9, 4655. [Google Scholar] [CrossRef]
  38. Castellano, G.M.; Aisner, J.; Burley, S.K.; Vallat, B.; Yu, H.A.; Pine, S.R.; Ganesan, S. A Novel Acquired Exon 20 EGFR M766Q Mutation in Lung Adenocarcinoma Mediates Osimertinib Resistance but is Sensitive to Neratinib and Poziotinib. J. Thorac. Oncol. 2019, 14, 1982–1988. [Google Scholar] [CrossRef]
  39. Chabon, J.J.; Simmons, A.D.; Lovejoy, A.F.; Esfahani, M.S.; Newman, A.M.; Haringsma, H.J.; Kurtz, D.M.; Stehr, H.; Scherer, F.; Karlovich, C.A.; et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat. Commun. 2016, 7, 11815. [Google Scholar] [CrossRef]
  40. Wang, Y.; Li, L.; Han, R.; Jiao, L.; Zheng, J.; He, Y. Clinical analysis by next-generation sequencing for NSCLC patients with MET amplification resistant to osimertinib. Lung Cancer 2018, 118, 105–110. [Google Scholar] [CrossRef] [Green Version]
  41. Coleman, N.; Hong, L.; Zhang, J.; Heymach, J.; Hong, D.; Le, X. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open 2021, 6, 100319. [Google Scholar] [CrossRef] [PubMed]
  42. Piper-Vallillo, A.J.; Sequist, L.V.; Piotrowska, Z. Emerging Treatment Paradigms for EGFR-Mutant Lung Cancers Progressing on Osimertinib: A Review. J. Clin. Oncol. 2020, 38, 2926–2936. [Google Scholar] [CrossRef] [PubMed]
  43. 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] [PubMed]
  44. Martinez-Marti, A.; Felip, E.; Matito, J.; Mereu, E.; Navarro, A.; Cedrés, S.; Pardo, N.; de Castro, A.M.; Remon, J.; Miquel, J.; et al. DualMET and ERBB inhibition overcomes intratumor plasticity in osimertinib-resistant-advanced non-small-cell lung cancer (NSCLC). Ann. Oncol. 2017, 28, 2451–2457. [Google Scholar] [CrossRef]
  45. Deng, L.; Kiedrowski, L.A.; Ravera, E.; Cheng, H.; Halmos, B. Response to Dual Crizotinib and Osimertinib Treatment in a Lung Cancer Patient with MET Amplification Detected by Liquid Biopsy Who Acquired Secondary Resistance to EGFR Tyrosine Kinase Inhibition. J. Thorac. Oncol. 2018, 13, e169–e172. [Google Scholar] [CrossRef] [Green Version]
  46. Zhu, V.W.; Schrock, A.B.; Ali, S.M.; Ou, S.-H.I. Differential response to a combination of full-dose osimertinib and crizotinib in a patient with EGFR-mutant non-small cell lung cancer and emergent MET amplification. Lung Cancer Targets Ther. 2019, 10, 21–26. [Google Scholar] [CrossRef] [Green Version]
  47. Giroux-Leprieur, E.; Dumenil, C.; Chinet, T. Combination of Crizotinib and Osimertinib or Erlotinib Might Overcome MET-Mediated Resistance to EGFR Tyrosine Kinase Inhibitor in EGFR-Mutated Adenocarcinoma. J. Thorac. Oncol. 2018, 13, e232–e234. [Google Scholar] [CrossRef] [Green Version]
  48. Sequist, L.V.; Han, J.-Y.; Ahn, M.-J.; Cho, B.C.; Yu, H.; Kim, S.-W.; Yang, J.C.-H.; Lee, J.S.; Su, W.-C.; Kowalski, D.; et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: Interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef]
  49. Wu, Y.-L.; Zhang, L.; Kim, D.-W.; Liu, X.; Lee, D.H.; Yang, J.C.-H.; Ahn, M.-J.; Vansteenkiste, J.F.; Su, W.-C.; Felip, E.; et al. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib after Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients with EGFR-Mutated, MET Factor–Dysregulated Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3101–3109. [Google Scholar] [CrossRef]
  50. Wu, Y.-L.; Cheng, Y.; Zhou, J.; Lu, S.; Zhang, Y.; Zhao, J.; Kim, D.-W.; Soo, R.A.; Kim, S.-W.; Pan, H.; et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): An open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir. Med. 2020, 8, 1132–1143. [Google Scholar] [CrossRef]
  51. Bauml, J.; Cho, B.C.; Park, K.; Lee, K.H.; Cho, E.K.; Kim, D.W.; Kim, S.W.; Haura, E.B.; Sabari, J.K.; Sanborn, R.E. Amivantamab in Combination with Lazertinib for the Treatment of Osimertinib-Relapsed, Chemotherapy-Naïve EGFR Mutant (EGFRm) Non-Small Cell Lung Cancer (NSCLC) and Potential Biomarkers for Response. J. Clin. Oncol. 2021, 39, 9006. [Google Scholar] [CrossRef]
  52. 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]
  53. Goldman, J.W.; Horinouchi, H.; Cho, B.C.; Tomasini, P.; Dunbar, M.; Hoffman, D.; Parikh, A.; Blot, V.; Camidge, D.R. Phase 1/1b study of telisotuzumab vedotin (Teliso-V) + osimertinib (Osi), after failure on prior Osi, in patients with advanced, c-Met overexpressing, EGFR-mutated non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2022, 40, 9013. [Google Scholar] [CrossRef]
  54. Li, B.T.; Michelini, F.; Misale, S.; Cocco, E.; Baldino, L.; Cai, Y.; Shifman, S.; Tu, H.-Y.; Myers, M.L.; Xu, C.; et al. HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers. Cancer Discov. 2020, 10, 674–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Minari, R.; Bordi, P.; La Monica, S.; Squadrilli, A.; Leonetti, A.; Bottarelli, L.; Azzoni, C.; Lagrasta, C.A.M.; Gnetti, L.; Campanini, N.; et al. Concurrent Acquired BRAF V600E Mutation and MET Amplification as Resistance Mechanism of First-Line Osimertinib Treatment in a Patient with EGFR-Mutated NSCLC. J. Thoracic Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2018, 13, e89–e91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ho, C.-C.; Liao, W.-Y.; Lin, C.-A.; Shih, J.-Y.; Yu, C.-J.; Yang, J.C.-H. Acquired BRAF V600E Mutation as Resistant Mechanism after Treatment with Osimertinib. J. Thoracic Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2017, 12, 567–572. [Google Scholar] [CrossRef] [Green Version]
  57. Xie, Z.; Gu, Y.; Xie, X.; Lin, X.; Ouyang, M.; Qin, Y.; Zhang, J.; Lizaso, A.; Chen, S.; Zhou, C. Lung Adenocarcinoma Harboring Concomitant EGFR Mutations and BRAF V600E Responds to a Combination of Osimertinib and Vemurafenib to Overcome Osimertinib Resistance. Clin. Lung Cancer 2020, 22, e390–e394. [Google Scholar] [CrossRef]
  58. Eberlein, C.A.; Stetson, D.; Markovets, A.A.; Al-Kadhimi, K.J.; Lai, Z.; Fisher, P.R.; Meador, C.B.; Spitzler, P.; Ichihara, E.; Ross, S.J.; et al. Acquired Resistance to the Mutant-Selective EGFR Inhibitor AZD9291 Is Associated with Increased Dependence on RAS Signaling in Preclinical Models. Cancer Res. 2015, 75, 2489–2500. [Google Scholar] [CrossRef] [Green Version]
  59. Eng, J.; Woo, K.M.; Sima, C.S.; Plodkowski, A.; Hellmann, M.D.; Chaft, J.; Kris, M.; Arcila, M.E.; Ladanyi, M.; Drilon, A. Impact of Concurrent PIK3CA Mutations on Response to EGFR Tyrosine Kinase Inhibition in EGFR-Mutant Lung Cancers and on Prognosis in Oncogene-Driven Lung Adenocarcinomas. J. Thorac. Oncol. 2015, 10, 1713–1719. [Google Scholar] [CrossRef] [Green Version]
  60. Zeng, L.; Yang, N.; Zhang, Y. GOPC-ROS1 Rearrangement as an Acquired Resistance Mechanism to Osimertinib and Responding to Crizotinib Combined Treatments in Lung Adenocarcinoma. J. Thorac. Oncol. 2018, 13, e114–e116. [Google Scholar] [CrossRef] [Green Version]
  61. Piotrowska, Z.; Isozaki, H.; Lennerz, J.K.; Gainor, J.F.; Lennes, I.T.; Zhu, V.W.; Marcoux, N.; Banwait, M.K.; Digumarthy, S.R.; Su, W.; et al. Landscape of Acquired Resistance to Osimertinib in EGFR-Mutant NSCLC and Clinical Validation of Combined EGFR and RET Inhibition with Osimertinib and BLU-667 for Acquired RET Fusion. Cancer Discov. 2018, 8, 1529–1539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Rotow, J.; Patel, J.; Hanley, M.; Yu, H.; Goldman, J.; Nechustan, H.; Scheffler, M.; Awad, M.; Clifford, S.; Santucci, A.; et al. FP14.07 Combination Osimertinib plus Selpercatinib for EGFR-mutant Non-Small Cell Lung Cancer (NSCLC) with Acquired RET fusions. J. Thorac. Oncol. 2021, 16, S230. [Google Scholar] [CrossRef]
  63. Freydman, J.; Henshaw, L.; Patel, J.V.; Smith, C.E.; Everett, P.C. Combination EGFR and RET Inhibition in Acquired Resistance to Osimertinib in EGFR-Mutant NSCLC. Ann. Pharmacother. 2021, 56, 503–504. [Google Scholar] [CrossRef] [PubMed]
  64. Schrock, A.B.; Zhu, V.W.; Hsieh, W.-S.; Madison, R.; Creelan, B.; Silberberg, J.; Costin, D.; Bharne, A.; Bonta, I.; Bosemani, T.; et al. Receptor Tyrosine Kinase Fusions and BRAF Kinase Fusions are Rare but Actionable Resistance Mechanisms to EGFR Tyrosine Kinase Inhibitors. J. Thorac. Oncol. 2018, 13, 1312–1323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Offin, M.; Somwar, R.; Rekhtman, N.; Benayed, R.; Chang, J.C.; Plodkowski, A.; Lui, A.J.; Eng, J.; Rosenblum, M.; Li, B.T.; et al. Acquired ALK and RET Gene Fusions as Mechanisms of Resistance to Osimertinib in EGFR-Mutant Lung Cancers. JCO Precis. Oncol. 2018, 2, 1–12. [Google Scholar] [CrossRef]
  66. Qin, Q.; Li, X.; Liang, X.; Zeng, L.; Wang, J.; Sun, L.; Zhong, D. CDK4/6 inhibitor palbociclib overcomes acquired resistance to third-generation EGFR inhibitor osimertinib in non-small cell lung cancer (NSCLC). Thorac. Cancer 2020, 11, 2389–2397. [Google Scholar] [CrossRef]
  67. La Monica, S.; Fumarola, C.; Cretella, D.; Bonelli, M.; Minari, R.; Cavazzoni, A.; Digiacomo, G.; Galetti, M.; Volta, F.; Mancini, M.; et al. Efficacy of the CDK4/6 Dual Inhibitor Abemaciclib in EGFR-Mutated NSCLC Cell Lines with Different Resistance Mechanisms to Osimertinib. Cancers 2020, 13, 6. [Google Scholar] [CrossRef]
  68. Namba, K.; Shien, K.; Takahashi, Y.; Torigoe, H.; Sato, H.; Yoshioka, T.; Takeda, T.; Kurihara, E.; Ogoshi, Y.; Yamamoto, H.; et al. Activation of AXL as a Preclinical Acquired Resistance Mechanism Against Osimertinib Treatment in EGFR-Mutant Non–Small Cell Lung Cancer Cells. Mol. Cancer Res. 2019, 17, 499–507. [Google Scholar] [CrossRef] [Green Version]
  69. Okura, N.; Nishioka, N.; Yamada, T.; Taniguchi, H.; Tanimura, K.; Katayama, Y.; Yoshimura, A.; Watanabe, S.; Kikuchi, T.; Shiotsu, S.; et al. ONO-7475, a Novel AXL Inhibitor, Suppresses the Adaptive Resistance to Initial EGFR-TKI Treatment in EGFR-Mutated Non–Small Cell Lung Cancer. Clin. Cancer Res. 2020, 26, 2244–2256. [Google Scholar] [CrossRef]
  70. Yang, Y.-M.; Jang, Y.; Lee, S.H.; Kang, B.; Lim, S.M. AXL/MET dual inhibitor, CB469, has activity in non-small cell lung cancer with acquired resistance to EGFR TKI with AXL or MET activation. Lung Cancer 2020, 146, 70–77. [Google Scholar] [CrossRef]
  71. Sang, Y.B.; Kim, J.-H.; Kim, C.-G.; Hong, M.H.; Kim, H.R.; Cho, B.C.; Lim, S.M. The Development of AXL Inhibitors in Lung Cancer: Recent Progress and Challenges. Front. Oncol. 2022, 12, 811247. [Google Scholar] [CrossRef] [PubMed]
  72. Manabe, T.; Yasuda, H.; Terai, H.; Kagiwada, H.; Hamamoto, J.; Ebisudani, T.; Kobayashi, K.; Masuzawa, K.; Ikemura, S.; Kawada, I.; et al. IGF2 Autocrine-Mediated IGF1R Activation Is a Clinically Relevant Mechanism of Osimertinib Resistance in Lung Cancer. Mol. Cancer Res. 2020, 18, 549–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hayakawa, D.; Takahashi, F.; Mitsuishi, Y.; Tajima, K.; Hidayat, M.; Winardi, W.; Ihara, H.; Kanamori, K.; Matsumoto, N.; Asao, T.; et al. Activation of insulin-like growth factor-1 receptor confers acquired resistance to osimertinib in non-small cell lung cancer with EGFR T790M mutation. Thorac. Cancer 2019, 11, 140–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Jänne, 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. 2021, 12, 74–89. [Google Scholar] [CrossRef]
  75. 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]
  76. 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]
  77. Schoenfeld, A.J.; Chan, J.M.; Kubota, D.; Sato, H.; Rizvi, H.; Daneshbod, Y.; Chang, J.C.; Paik, P.K.; Offin, M.; Arcila, M.E.; et al. Tumor Analyses Reveal Squamous Transformation and Off-Target Alterations as Early Resistance Mechanisms to First-line Osimertinib in EGFR-Mutant Lung Cancer. Clin. Cancer Res. 2020, 26, 2654–2663. [Google Scholar] [CrossRef] [Green Version]
  78. Hakozaki, T.; Kitazono, M.; Takamori, M.; Kiriu, T. Combined Small and Squamous Transformation in EGFR-mutated Lung Adenocarcinoma. Intern. Med. 2020, 59, 1291–1294. [Google Scholar] [CrossRef] [Green Version]
  79. Roca, E.; Gurizzan, C.; Amoroso, V.; Vermi, W.; Ferrari, V.; Berruti, A. Outcome of patients with lung adenocarcinoma with transformation to small-cell lung cancer following tyrosine kinase inhibitors treatment: A systematic review and pooled analysis. Cancer Treat. Rev. 2017, 59, 117–122. [Google Scholar] [CrossRef]
  80. Qin, Q.; Li, X.; Liang, X.; Zeng, L.; Wang, J.; Sun, L.; Zhong, D. Targeting the EMT transcription factor Snail overcomes resistance to osimertinib in EGFR -mutant non-small cell lung cancer. Thorac. Cancer 2021, 12, 1708–1715. [Google Scholar] [CrossRef]
  81. Yochum, Z.A.; Cades, J.; Wang, H.; Chatterjee, S.; Simons, B.W.; O’Brien, J.P.; Khetarpal, S.K.; Lemtiri-Chlieh, G.; Myers, K.; Huang, E.H.-B.; et al. Targeting the EMT transcription factor TWIST1 overcomes resistance to EGFR inhibitors in EGFR-mutant non-small-cell lung cancer. Oncogene 2018, 38, 656–670. [Google Scholar] [CrossRef]
  82. Oizumi, S.; Sugawara, S.; Minato, K.; Harada, T.; Inoue, A.; Fujita, Y.; Maemondo, M.; Watanabe, S.; Ito, K.; Gemma, A.; et al. Updated survival outcomes of NEJ005/TCOG0902: A randomised phase II study of concurrent versus sequential alternating gefitinib and chemotherapy in previously untreated non-small cell lung cancer with sensitive EGFR mutations. ESMO Open 2018, 3, e000313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. 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]
  84. 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]
  85. 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]
  86. Planchard, D.; Feng, P.-H.; Karaseva, N.; Kim, S.-W.; Kim, T.; Lee, C.; Poltoratskiy, A.; Yanagitani, N.; Marshall, R.; Huang, X.; et al. Osimertinib plus platinum–pemetrexed in newly diagnosed epidermal growth factor receptor mutation-positive advanced/metastatic non-small-cell lung cancer: Safety run-in results from the FLAURA2 study. ESMO Open 2021, 6, 100271. [Google Scholar] [CrossRef]
  87. Herbst, R.S.; Johnson, D.H.; Mininberg, E.; Carbone, D.P.; Henderson, T.; Kim, E.S.; Jr, G.B.; Lee, J.J.; Liu, D.D.; Truong, M.T.; et al. Phase I/II Trial Evaluating the Anti-Vascular Endothelial Growth Factor Monoclonal Antibody Bevacizumab in Combination with the HER-1/Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Erlotinib for Patients with Recurrent Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2005, 23, 2544–2555. [Google Scholar] [CrossRef]
  88. Naumov, G.N.; Nilsson, M.B.; Cascone, T.; Briggs, A.; Straume, O.; Akslen, L.A.; Lifshits, E.; Byers, L.A.; Xu, L.; Wu, H.-K.; et al. Combined Vascular Endothelial Growth Factor Receptor and Epidermal Growth Factor Receptor (EGFR) Blockade Inhibits Tumor Growth in Xenograft Models of EGFR Inhibitor Resistance. Clin. Cancer Res. 2009, 15, 3484–3494. [Google Scholar] [CrossRef] [Green Version]
  89. Seto, T.; Kato, T.; Nishio, M.; Goto, K.; Atagi, S.; Hosomi, Y.; Yamamoto, N.; Hida, T.; Maemondo, M.; Nakagawa, K.; et al. Erlotinib alone or with bevacizumab as first-line therapy in patients with advanced non-squamous non-small-cell lung cancer harbouring EGFR mutations (JO25567): An open-label, randomised, multicentre, phase 2 study. Lancet Oncol. 2014, 15, 1236–1244. [Google Scholar] [CrossRef]
  90. Nakagawa, K.; Garon, E.B.; Seto, T.; Nishio, M.; Aix, S.P.; Paz-Ares, L.; Chiu, C.-H.; Park, K.; Novello, S.; Nadal, E.; et al. Ramucirumab plus erlotinib in patients with untreated, EGFR-mutated, advanced non-small-cell lung cancer (RELAY): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019, 20, 1655–1669. [Google Scholar] [CrossRef] [Green Version]
  91. Maemondo, M.; Fukuhara, T.; Saito, H.; Furuya, N.; Watanabe, K.; Sugawara, S.; Iwasawa, S.; Tsunezuka, Y.; Yamaguchi, O.; Okada, M. NEJ026: Final Overall Survival Analysis of Bevacizumab plus Erlotinib Treatment for NSCLC Patients Harboring Activating EGFR-Mutations. J. Clin. Oncol. 2020, 15, 9506. [Google Scholar] [CrossRef]
  92. Akamatsu, H.; Toi, Y.; Hayashi, H.; Fujimoto, D.; Tachihara, M.; Furuya, N.; Otani, S.; Shimizu, J.; Katakami, N.; Azuma, K. 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. JAMA Oncol. 2021, 7, 386–394. [Google Scholar] [CrossRef] [PubMed]
  93. Nishio, M.; Nishio, K.; Reck, M.; Garon, E.B.; Imamura, F.; Kawaguchi, T.; Yamaguchi, H.; Ikeda, S.; Hirano, K.; Visseren-Grul, C.; et al. RELAY+: Exploratory Study of Ramucirumab Plus Gefitinib in Untreated Patients with EGFR-Mutated Metastatic NSCLC. JTO Clin. Res. Rep. 2022, 3, 100303. [Google Scholar] [CrossRef] [PubMed]
  94. Rotow, J.K.; Costa, D.B.; Paweletz, C.P.; Awad, M.M.; Marcoux, P.; Rangachari, D.; Barbie, D.A.; Sands, J.; Cheng, M.L.; Johnson, B.E. Concurrent Osimertinib plus Gefitinib for First-Line Treatment of EGFR-Mutated Non-Small Cell Lung Cancer (NSCLC). J. Clin. Oncol. 2020, 38, 9507. [Google Scholar] [CrossRef]
  95. Jin, R.; Zhao, J.; Xia, L.; Li, Q.; Li, W.; Peng, L.; Xia, Y. Application of immune checkpoint inhibitors in EGFR-mutant non-small-cell lung cancer: From bed to bench. Ther. Adv. Med. Oncol. 2020, 12, 1758835920930333. [Google Scholar] [CrossRef] [PubMed]
  96. 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]
  97. Reck, M.; Mok, T.; Nishio, M.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): Key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir. Med. 2019, 7, 387–401. [Google Scholar] [CrossRef]
  98. Nogami, N.; Barlesi, F.; Socinski, M.A.; Reck, M.; Thomas, C.A.; Cappuzzo, F.; Mok, T.S.; 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. 2021, 17, 309–323. [Google Scholar] [CrossRef]
  99. Ahn, M.-J.; Cho, B.C.; Ou, X.; Walding, A.; Dymond, A.W.; Ren, S.; Cantarini, M.; Jänne, P.A. Osimertinib Plus Durvalumab in Patients with EGFR-Mutated, Advanced NSCLC: A Phase 1b, Open-Label, Multicenter Trial. J. Thorac. Oncol. 2022, 17, 718–723. [Google Scholar] [CrossRef]
  100. Chan, D.W.-K.; Choi, H.C.-W.; Lee, V.H.-F. Treatment-Related Adverse Events of Combination EGFR Tyrosine Kinase Inhibitor and Immune Checkpoint Inhibitor in EGFR-Mutant Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 2157. [Google Scholar] [CrossRef]
  101. Gomez, D.R.; Blumenschein, G.R., Jr.; Lee, J.J.; Hernández, M.; Ye, R.; Camidge, D.R.; Doebele, R.C.; Skoulidis, F.; Gaspar, L.E.; Gibbons, D.L.; et al. Local consolidative therapy versus maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer without progression after first-line systemic therapy: A multicentre, randomised, controlled, phase 2 study. Lancet Oncol. 2016, 17, 1672–1682. [Google Scholar] [CrossRef] [Green Version]
  102. Passaro, A.; Leighl, N.; Blackhall, F.; Popat, S.; Kerr, K.; Ahn, M.; Arcila, M.; Arrieta, O.; Planchard, D.; de Marinis, F.; et al. ESMO expert consensus statements on the management of EGFR mutant non-small-cell lung cancer. Ann. Oncol. 2022, 33, 466–487. [Google Scholar] [CrossRef] [PubMed]
  103. Riess, J.W.; Krailo, M.D.; Padda, S.K.; Groshen, S.G.; Wakelee, H.A.; Reckamp, K.L.; Koczywas, M.; Piotrowska, Z.; Steuer, C.E.; Kim, C.; et al. Osimertinib plus necitumumab in EGFR-mutant NSCLC: Final results from an ETCTN California Cancer Consortium phase I study. J. Clin. Oncol. 2022, 40, 9014. [Google Scholar] [CrossRef]
Figure 1. Acquired resistance mechanism to osimertinib in first- and second-line treatment. Amp—amplification; SCLC—small cell lung cancer; squamous CC—squamous cell carcinoma.
Figure 1. Acquired resistance mechanism to osimertinib in first- and second-line treatment. Amp—amplification; SCLC—small cell lung cancer; squamous CC—squamous cell carcinoma.
Ijms 23 06936 g001
Table 1. Selection of ongoing clinical trials in osimertinib-pretreated EGFR-mutated advanced NSCLC.
Table 1. Selection of ongoing clinical trials in osimertinib-pretreated EGFR-mutated advanced NSCLC.
NCT IdentifierPhaseDrug(S) ClassPopulationTreatment ArmsStatusPrimary Endpoint
NCT05256290ISelective 4th gen EGFR TKINSCLC with acquired resistance EGFR mutation (eg, C797S) in the absence of concurrent T790M.
previous EGFR TKI: mandatory (osimertinib in first line)
BDTX-1535recruitingDLT
NCT04862780
(SYMPHONY)
I/IISelective 4th gen EGFR TKINSCLC harboring EGFR T790M and/or C797S mutation
previous EGFR TKI: mandatory at least 1 prior EGFR-targeted TKI with activity against the T790M mutation
BLU-945 as monotherapy and BLU-945 in combination with osimertinibrecruitingDLT
NCT05153408
(HARMONY)
I/IISelective 4th gen EGFR TKI
Chemotherapy
EGFRm NSCLC, EGFR C797X in part 2
previous EGFR TKI: mandatory at least 1 prior 3rd gen EGFR-targeted TKI (osimertinib)
BLU-701 as monotherapy or in combination with either osimertinib or platinum-based chemotherapyrecruitingMTD
safety
ORR
NCT02496663IAnti-EGFR mAb
3th gen EGFR TKI
EGFR NSCLC
previous EGFR TKI: mandatory (either)
Necitumumab + osimertinibactive, not recruitingMTD
safety
NCT03944772
(ORCHARD)
IIMET inhibitor
1st gen EGFR TKI
Anti-EGFR mAb
ALK TKI
RET inhibitor
MEK inhibitor
Chemotherapy
3th gen EGFR TKI
EGFRm NSCLC
previous EGFR TKI: mandatory osimertinib
Biomarker driven:
Osimertinib + savolitinib
Osimertinib + gefitinib
Osimertinib + necitumumab
Durvalumab + carboplatin + pemetrexed
Osimertinib + alectinib
Osimertinib + selpercatinib
Durvalumab + carbo/cis-platin + etoposide
Osimertinib + carbo/cis-platin + pemetrexed
Osimertinib + salumetinib
recruitingORR
NCT02609776
(CHRISALYS)
IAnti EGFR+ MET mAb
3rd gen EGFR TKI
Chemotherapy
EGFRm NSCLC naïve or pretreated with TKI
previous EGFR TKI: permitted
Amivantamab + Lazertinib + Carboplatin + PemetrexedrecruitingDLT, AE, ORR, DOR, clinical benefit rate
NCT04816214
(GEOMETRY-E)
IIIMET inhibitor
EGFR 3rd gen TKI
EGFR+, T790M-, MET amplification NSCLC
previous EGFR TKI: mandatory (either, osimertinib included)
Capmatinib + osimertinibrecruitingDLT
PFS
NCT03940703
(INSIGHT-2)
IIMET inhibitor
EGFR 3rd gen TKI
EGFR+, MET amplification NSCLC
previous EGFR TKI: mandatory (osimertinib)
Tepotinib + osimertinibrecruitingDLT
ORR
NCT03778229
(SAVANNAH)
IIMET TKI
3rd gen EGFR TKI
EGFRm+/MET+ NSCLC
previous EGFR TKI: mandatory (Osimertinib)
Osimertinib + savolitinibrecruitingORR
NCT05261399
(SAFFRON)
IIIMET inhibitor
3rd gen EGFR TKI
Chemotherapy
EGFR NSCLC MET-overexpressed and/or amplified
previous EGFR TKI: mandatory (Osimertinib)
Savolitinib + osimertinib vs. platinum-pemetrexed chemotherapynot yet recruitingPFS
NCT02099058IMET-directed ADC
3rd gen EGFR TKi
Anti-PD11st gen EGFR TKI
EGFR+/MET+ NSCLC
previous EGFR TKI: mandatory (either)
Telisotuzumab vedotin
Telisotuzumab vedotin + osimertinib Telisotuzumab vedotin + erlotinib
Telisotuzumab vedotin + nivolumab
recruitingsafety
RPTD
NCT04042701IHER3-directed ADC
anti PD1
HER2+ breast cancer and NSCLC
previous EGFR TKI: If EGFR+ NSCLC, mandatory (either Osimertinib included)
Trastuzumab deruxtecan + PembrolizumabrecruitingDLT, ORR
NCT03784599
(TRAEMOS)
IIHER2-directed ADCEGFR+ HER2+ NSCLC
previous EGFR TKI: mandatory (either). If 1st or 2nd gen TKI must be T790M negative
TDM1 + osimertinibrecruitingsafety, ORR
NCT05338970IIIHER3-directed ADCEGFR+ NSCLC
previous EGFR TKI: mandatory (third generation TKI)
Patritumab Deruxtecan
vs. platinum-pemetrexed based chemotherapy
recruitingPFS
NCT04619004
(HERTHENA—Lung01)
IIHER3-directed ADCEGFR+ NSCLC
previous EGFR TKI: mandatory (either) + 1 line of platinum based chemotherapy
Patritumab deruxtecanrecruitingORR
NCT04676477IHER3-directed ADCEGFR+ NSCLC
previous EGFR TKI: mandatory (osimertinib)
Patritumab deruxtecan + osimertinibrecruitingDLT, afety
NCT03260491IHER3-directed ADCEGFR+ NSCLC
previous EGFR TKI: mandatory (either, Osimertinib included)
U3-1402active, not ecruitingDLT, ORR
NCT04452877IIBRAF + MEK inhibitorsBRAF V600E NSCLC
previous EGFR TKI: mandatory (either, Osimertinib included)
Dabrafenib + trametinibrecruitingORR
NCT04545710IICDK4/6 inhibitor
3rd gen EGFR TKI
EGFR+ NSCLC
previous EGFR TKI: mandatory (osimertinib)
Abemaciclib + osimertinibrecruiiting6 months-PFS
NCT03455829I/IICDK4/6 inhibitor
3rd gen EGFR TKI
EGFR+ NSCLC
previous EGFR TKI: mandatory (osimertinib)
Lerociclib + Osimertinibactive, not recruitingDLT
safety
PFS
NCT02729298IAXL inhibitorSolid tumors including EGFR+ NSCLC
previous EGFR TKI: mandatory (either, osimertinib included)
TP-0903active, not recruitingDLT
NCT03891615IPARP inhibitorEGFR+ NSCLC
previous EGFR TKI: mandatory (osimertinib)
Niraparib + osimertinibrecruitingMTD
NCT04538378IIPARP inhibitor
Anti-PDL1
EGFR+ NSCLC transformed into SCLC in progression to platinum-based chemotherapy
previous EGFR TKI: mandatory (either, Osimertinib included)
Niraparib + durvalumabrecruitingbest overall response
NCT04484142
(TROPION-Lung05)
IIHER3-directed ADCEGFR, ALK, ROS1, NTRK, BRAF, MET exon 14 skipping, or RET positive NSCLC
previous EGFR TKI: mandatory (osimertinib included if T790M)
DS-1062aactive, not recruitingORR
NCT04765059
(COMPEL)
IIIChemotherapy
3rd gen EGFR TKI
EGFRm+NSCLC in extracranial disease progression
previous EGFR TKI: mandatory, (osimertinib)
Platinum/pemetrexed/osimertinib vs. platinum/pemetrexedrecruitingPFS
NCT04438902IIAnti-VEGFR TKI
3rd gen EGFR TKI
EGFRm+/T790M NSCLC with gradual progression on osimertinib
previous EGFR TKI: mandatory, (osimertinib)
Anlotinib + osimertinibrecruitingPFS
NCT04405674IIAnti-PD1
Chemotherapy
anti-VEGF mAb
EGFRm+ NSCLC
previous EGFR TKI: mandatory, (either, osimertinib if T790M mandatory)
Tislelizumab + carboplatin + nabpaclitaxel followed by tislelizumab + pemetrexed manteinance therapyrecruiting1 y-PFS rate
NCT02864251IIIAnti-PDL1
Anti-CTLA4
chemotherapy
EGFRm+/T790M- NSCLC
previous EGFR TKI: mandatory, (either, osimertinib included)
Nivolumab + platinum + pemetrexed vs. nivolumab + ipilimumab vs. platinum-pemetrexed chemotherapyactive, not recruitingPFS
Gen—generation; PFS—progression-free survival; mAb—monoclonal antibody; TKI—tyrosine kinase inhibitors; DLT—dose-limiting toxicity; MTD—maximum tolerated dose; ORR—objective response rate.
Table 2. Ongoing clinical trials in treatment naïve EGFR-mutated advanced NSCLC.
Table 2. Ongoing clinical trials in treatment naïve EGFR-mutated advanced NSCLC.
NCT IdentifierPhaseDrug(s) ClassPopulationTreatment ArmsStatusPrimary Endpoint
NCT05299125
(AMIGO-1)
IIAnti-EGFR+ MET mAb
3rd gen EGFR TKI
Chemotherapy
Advanced NSCLC with common EGFR sensitising mutationAmivantamab + Lazertinib + carboplatin + pemetrexedNot yet recruiting18 months PFS rate
NCT03865511
(MELROSE)
II3rd gen EGFR TKIAdvanced NSCLC with common EGFR sensitising mutationosimertinibrecruitingGenetic profile at disease progression in EGFRm+ compared to baseline
NCT04487080
MARIPOSA
IIIAnti-EGFR+ MET mAb
3rd gen EGFR TKI
Advanced NSCLC with common EGFR sensitising mutationAmivantamb + Lazertinib vs. osimertinibrecruitingPFS
NCT04248829
LASER301
III3rd gen EGFR TKI
1st gen EGFR TKI
Advanced NSCLC with common EGFR sensitising mutationLazertininb + gefitinibActive, not recruitingPFS
NCT04181060
(EA5182)
IIIAnti-VEGF mAb
3rd gen EGFR TKI
Advanced NSCLC with EGFR sensitising mutation (uncommon included)Bevacizumab + osimertinibrecruitingPFS
NCT03909334IIAnti-VEGFR2 mAb
3rd gen EGFR TKI
Advanced NSCLC with EGFR sensitising mutation (uncommon included)Osimertinib + ramucirumab vs. osimertinibrecruitingPFS
NCT04035486
FLAURA2
III3rd gen EGFR TKI
chemotherapy
Advanced NSCLC with EGFR sensitising mutationOsimertinib + platinum-pemetrexed chemotherapy vs. osimertinibActive, not recruitingPFS
NCT03567642I3rd gen EGFR TKI
chemotherapy
Advanced EGFR+ NSCLC with concurrent RB1 and TP53 AlterationsPlatinum-etoposide + osimertinibrecruitingMTD
NCT03392246IIMEK inhibitor
3rd gen EGFR TKI
Advanced NSCLC with EGFR sensitising mutationOsimertinib + salumetinibrecruitingBest objective response
NCT04695925III3rd gen EGFR TKI
chemotherapy
Advanced EGFR+ NSCLC with concurrent TP53 mutationOsimertinib vs. Osimertinib + carboplatin + pemetrexedNot yet recruitingPFS
NCT02971501IIAnti-VEGF mAb
3rd gen EGFR TKI
Advanced NSCLC with EGFR sensitising mutation with brain metastasisBevacizumab + osimertinibActive, not recruitingPFS
NCT02954523I/IIBCR/AbL inhibitor
3rd gen EGFR TKI
Advanced NSCLC with EGFR sensitising mutation (uncommon included)Dasatinib + OsimertinibActive, not recruitingSafety
NCT03122717I/II3rd gen EGFR TKI
1st gen EGFR TKI
Advanced NSCLC with EGFR sensitising mutationOsimertinib + gefitinibActive, not recruitingNumber of patients completing combination therapy for 6 × 28 day cycles
Gen—generation; PFS—progression-free survival; mAb—monoclonal antibody; TKI—tyrosine kinase inhibitors.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bertoli, E.; De Carlo, E.; Del Conte, A.; Stanzione, B.; Revelant, A.; Fassetta, K.; Spina, M.; Bearz, A. Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It? Int. J. Mol. Sci. 2022, 23, 6936. https://doi.org/10.3390/ijms23136936

AMA Style

Bertoli E, De Carlo E, Del Conte A, Stanzione B, Revelant A, Fassetta K, Spina M, Bearz A. Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It? International Journal of Molecular Sciences. 2022; 23(13):6936. https://doi.org/10.3390/ijms23136936

Chicago/Turabian Style

Bertoli, Elisa, Elisa De Carlo, Alessandro Del Conte, Brigida Stanzione, Alberto Revelant, Kelly Fassetta, Michele Spina, and Alessandra Bearz. 2022. "Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It?" International Journal of Molecular Sciences 23, no. 13: 6936. https://doi.org/10.3390/ijms23136936

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