Next Article in Journal
Can Be miR-126-3p a Biomarker of Premature Aging? An Ex Vivo and In Vitro Study in Fabry Disease
Next Article in Special Issue
Ellagic Acid and Schisandrins: Natural Biaryl Polyphenols with Therapeutic Potential to Overcome Multidrug Resistance in Cancer
Previous Article in Journal
NF-κB in Cancer Immunity: Friend or Foe?
Previous Article in Special Issue
Inhibition of HER Receptors Reveals Distinct Mechanisms of Compensatory Upregulation of Other HER Family Members: Basis for Acquired Resistance and for Combination Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Line Models for Acquired Resistance to First-Line Osimertinib in Lung Cancers—Applications and Limitations

Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama 589-8511, Japan
*
Author to whom correspondence should be addressed.
Cells 2021, 10(2), 354; https://doi.org/10.3390/cells10020354
Submission received: 9 January 2021 / Revised: 3 February 2021 / Accepted: 5 February 2021 / Published: 9 February 2021
(This article belongs to the Special Issue Studying Drug Resistance Using Cancer Cell Lines)

Abstract

:
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are first-line drugs for lung cancers with activating EGFR mutations. Although first- and second-generation EGFR-TKIs were standard first-line treatments, acquired resistance (AR) to these drugs is almost inevitable. Cell line models have been widely used to explore the molecular mechanisms of AR to first- and second-generation EGFR-TKIs. Many research groups, including ours, have established AR cell lines that harbor the EGFR T790M secondary mutation, MET gene amplification, or epithelial–mesenchymal transition (EMT) features, which are all found in clinical specimens obtained from TKI-refractory lesions. Currently, many oncologists prescribe osimertinib, a third-generation EGFR-TKI that can overcome T790M-mediated resistance, as a first-line TKI. Although few clinical data are available about AR mechanisms that arise when osimertinib is used as a first-line therapy, many research groups have established cell lines with AR to osimertinib and have reported on their AR mechanisms. In this review, we summarize the findings on AR mechanisms against first-line osimertinib obtained from analyses of cell line models.

1. Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide. Intensive molecular analyses of lung cancers, especially in lung adenocarcinomas, have identified several mutually exclusive aberrations that occur in proto-oncogenes. Subsequently, clinical trials have shown that orally available kinase inhibitors are highly active against tumors that harbor these driver gene mutations. Therefore, based on the results of many clinical trials, current guidelines recommend the first-line use of kinase inhibitors when treating unresectable lung cancers with mutations in epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), ROS1, BRAF, neurotrophic receptor tyrosine kinase 1/2/3 (NTRK1/2/3), or MET [1,2,3,4,5,6].
EGFR mutations are the most common molecular aberration found in lung cancers. They are found in ~50% of lung cancers in East Asians and ~15% of those in Caucasians [7]. The standard of care for lung cancers with EGFR mutations is EGFR tyrosine kinase inhibitor (TKI) monotherapies. The so-called first-generation EGFR-TKIs (gefitinib and erlotinib) and a second-generation EGFR-TKI (afatinib) were used as first-line EGFR-TKIs until the approval of a third-generation EGFR-TKI (osimertinib) for first-line use.
Despite the dramatic responses of lung cancers with EGFR mutations to EGFR-TKIs, the emergence of acquired resistance (AR) is almost inevitable [8]. To understand and to overcome AR to EGFR-TKIs, researchers have explored resistance mechanisms by analyzing tumor specimens obtained from patients who developed AR to an EGFR-TKI [9,10,11] or by analyzing cell lines that acquired resistance to an EGFR-TKI in vitro by chronic exposure to the drug [10,11,12]. Both of these research paths have identified many AR mechanisms to first- and second-generation EGFR-TKIs, including T790M EGFR secondary mutation, as the most common mechanism, followed by MET gene amplification, ERBB2 gene amplification, small-cell lung cancer (SCLC) transformation, and the acquisition of epithelial to mesenchymal transition (EMT) features [13].
Osimertinib is a third-generation EGFR-TKI that was designed to overcome T790M-mediated resistance while sparing wild-type EGFR [14]. Osimertinib was originally approved as a second-line EGFR-TKI for patients with lung cancer with EGFR mutations that acquired resistance to first or second-generation EGFR-TKIs through the T790M secondary mutation. However, the FLAURA trial showed that first-line osimertinib is superior to first-generation EGFR-TKIs in both progression-free and overall survivals [15]. Currently, many oncologists use osimertinib as the first-line EGFR-TKI.
However, developing resistance to first-line osimertinib is also inevitable. In this review, we summarize published findings regarding AR mechanisms to first-line osimertinib, with a particular focus on resistance mechanisms that were identified using cell line models.

2. Identifying Mechanisms of Resistance to EGFR-TKIs through Cell Line Models

Cell line models are widely used in lung cancer research to evaluate drug resistance mechanisms [16,17,18,19,20,21]. Engelman et al. were the first who successfully identified a novel AR mechanism against EGFR-TKI—MET gene amplification—by using a cell line model in 2007. They established a gefitinib-resistant cell line model with MET gene amplification by exposing a gefitinib-sensitive lung cancer cell line (HCC827) with an activating EGFR mutation (exon 19 E746_A750 del) to increasing concentrations of gefitinib. More importantly, they also used clinical specimens to show that MET gene amplification was detected in 4 of 18 (22%) of the lung cancer specimens that had developed AR to gefitinib or erlotinib [10]. The authors also showed that EGFR-TKI resistance mediated by MET gene amplification can be overcome by the combination of a MET-TKI plus gefitinib.
The EGFR T790M secondary mutation is the most common mechanism of AR to first- or second-generation EGFR-TKIs found in clinical specimens [22]. PC9 cells (exon 19 E746_A750 del) are a lung cancer cell line that often acquires resistance to EGFR-TKIs through T790M secondary mutation [22]. Hata et al. reported that PC9 cells contain a pre-existing minor subclone with the T790M mutation. The T790M secondary mutation was also induced in single-cell PC9 clones that were established to eliminate the pre-existing minor clone with a T790M mutation [23]. Furthermore, T790M-mediated AR to first-generation EGFR-TKIs has been reported in other cell lines, such as HCC4006 (exon 19 del L747_A750 ins P) [24] and HCC827 cells [11], which reflects the very high frequency of this resistance mechanism.
As another AR mechanism against first-generation EGFR-TKIs, in 2011, we established and reported an AR cell line model derived from HCC4006 cells with EMT features [12]. EMT has also been reported as an AR mechanism against EGFR-TKIs in clinical specimens [25,26]. Although ERBB2 gene amplification and SCLC transformation have been reported as AR mechanisms against EGFR-TKIs in clinical settings, as far as we know, no cell line model has been developed with either ERBB2 gene amplification or SCLC transformation by using chronic exposure to EGFR-TKIs. Many groups have reported potential AR mechanisms against first- or second-generation EGFR-TKIs using cell line models, such as activating β-catenin [27] or the insulin-like growth factor receptor (IGF-1R) [28,29,30,31]. However, the roles of these molecules in AR against EGFR-TKIs in clinical settings are unclear.

3. Cell Line Models Used to Analyze Resistance Mechanisms to First-Line Osimertinib

3.1. Search Criteria for Published Studies

To identify published articles that analyzed AR mechanisms against first-line osimertinib using cell line models, we systematically searched PubMed for relevant studies as of December 1, 2020. Our search criteria included the following terms: “osimertinib,” “resistance,” “lung cancer,” and “cell line” or “cell lines.” We also manually scanned the reference lists of selected articles for additional eligible publications. We finally identified 29 relevant papers [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60]. We excluded 2 of these papers [47,49], as they used osimertinib-resistant cell lines that had been established in other studies included in our survey.
In the 27 remaining studies, 17 AR cell lines were established from PC9 cells, 16 from H1975 cells (L858R plus T790M, a cell line model with primary resistance to first- or second-generation EGFR-TKIs), 10 from HCC827 cells, and 2 from HCC4006 cells (Figure 1). Twenty-five studies used chronic exposure to osimertinib at increasing concentrations, one study [57] used chronic exposure to osimertinib at a consistent concentration, and one study [52] used two different exposures (increasing concentration and a consistent concentration). We classified the AR mechanisms into five categories: (a) aberration of EGFR itself, (b) activation of bypass signaling, (c) suppression of apoptosis, (d) EMT, and (e) other mechanisms.

3.2. Mechanisms of Resistance to First-Line Osimertinib That Were Identified in Cell Line Models

3.2.1. Aberration of EGFR Itself—On-Target Resistance Mechanism

As described above, the EGFR T790M secondary mutation is the most common AR mechanism against first- and second-generation EGFR-TKIs, and many TKI-resistant cell line models acquired this secondary mutation. Some EGFR secondary mutations (C797X: 7%, L718Q + C797S: 1%, L718Q + ex20ins: 1%, S768I: 1%) have been reported as AR mechanisms against first-line osimertinib in clinical settings [61]. However, we could not find a cell line model with AR against first-line osimertinib based on an EGFR secondary mutation.

3.2.2. Activation of Bypass Signaling

The activation of other receptor tyrosine kinases is a common AR mechanism against EGFR-TKIs. Reportedly, in vitro analyses have implicated several receptor tyrosine kinases in AR mechanisms against first-line osimertinib. These receptor tyrosine kinases are able to activate downstream pathways related to cell proliferation and survival, instead of the inhibited EGFR. Molecules and pathways that may have roles in AR to first-line osimertinib are summarized in Figure 2.

MET Gene Amplification

Similar to the AR mechanisms against first- or second-generation EGFR-TKIs [10,11], MET gene amplification is reported to be a resistance mechanism against first-line osimertinib in studies that used HCC827 cells [33]. This is reasonable because HCC827 cells have a pre-existing minor clone with MET gene amplification [62], and this cell line has repeatedly acquired resistance to several EGFR-TKIs through MET gene amplification [10,11,22].
In analyses of clinical specimens, MET gene amplification is a common AR mechanism against first-line osimertinib. For example, AR mechanisms were explored using circulating tumor DNA analysis in patients who were enrolled in the phase-III FLAURA study, which compared the efficacy of first-line osimertinib to first-generation EGFR-TKIs (gefitinib or erlotinib). In this study, MET gene amplification was reportedly the most common AR mechanism (n = 14/91; 15%) in the osimertinib arm [61]. Another study reported MET gene amplification as the AR mechanism against first-line osimertinib in 66% (n = 6/9) of their patients [63]. Currently, several clinical trials that co-target MET are ongoing for EGFR mutated non-small cell lung cancer (NSCLC) patients who acquired resistance to osimertinib; these include savolitinib plus osimertinib (SAVANNAH) or tepotinib plus osimertinib (INSIGHT 2).

AXL Activation

Increased AXL expression was reported by independent research groups as a resistance mechanism against first-line osimertinib using cell line models. AXL is a receptor tyrosine kinase that binds to a ligand (Gas6) and potentiates survival signaling by activating the PI3K–Akt and RAS–RAF–MEK–ERK pathways [64]. Originally, AR mediated by AXL was reported in HCC827 cells that acquired resistance to the first-generation EGFR-TKI, erlotinib [65]. As a resistance mechanism against first-line osimertinib, cells that acquired resistance through the activation of AXL and MET were established from HCC827 cells; a combination of an AXL/MET dual inhibitor (CB469) plus osimertinib could overcome the resistance [52]. Additionally, another group reported establishing acquired resistant PC9, H1975, and HCC4006 cell lines with AXL activation. The authors also explored the mechanism of AXL activation in osimertinib-resistant cells; mechanically, STC2 enhanced the AXL promoter activity by increasing the phosphorylation of c-Jun, which is an indispensable transcription factor that transactivates AXL [41]. Osimertinib-resistant cells with AXL activation were also established from PC9, HCC827, HCC4006, and H1975 cells; these resistant cells were sensitive to the combination of osimertinib plus cabozantinib [39].
Although AXL-mediated resistant cell lines have been repeatedly established in vitro, to our knowledge, there are no reported clinical cases in which AXL activation developed after first-line osimertinib treatment failure. However, several studies have reported high AXL expression in tumor specimens obtained after first or second EGFR-TKI treatment failure [65,66,67]. Another study reported that the response rate for osimertinib is relatively low (66.7%) in patients whose lung cancer has an EGFR mutation and a high AXL expression (IHC score 3+) [68]. These results, together with repeated development of osimertinib-resistant, AXL-activated cell lines, imply that AXL activation could be an AR mechanism against first-line osimertinib. However, this should be confirmed clinically. In addition to the AXL inhibitors described above, many AXL inhibitors (e.g., BGB324, TP-0903, ONO-7475, or DS-1205b) have been investigated in preclinical studies; therefore, we anticipate the clinical application of these agents in the near future.

ERBB3 Activation

ERBB3 is one of four members of the human ERBB family. ERBB3 lacks intrinsic tyrosine kinase activity but contains six YXXM-consensus binding sites for the SH2 domains of the p85 regulatory subunit of PI3K [69]. It therefore potentiates survival by activating PI3K/Akt signaling [70]. Yonesaka et al. reported the upregulation of ERBB3 in PC9 cells as a mechanism of AR to osimertinib. The authors also reported that an anti-HER3 antibody drug (U3-1402) might serve as a novel therapy that can overcome AR mediated by ERBB3 activation [45]. A clinical trial of U3-1402 plus osimertinib for EGFR mutated NSCLC patients who acquired resistance to osimertinib has just been posted (NCT04676477). So far, we have found no clinical cases with ERBB3 activation after first-line osimertinib treatment failure.

IGF-1R Activation

IGF-1R also potentiates survival by activating PI3K–Akt signaling and RAS–RAF–MEK–ERK signaling [71]. The role of IGF-1R as an AR mechanism against first- or second-generation EGFR-TKIs has been reported previously [29,30,31]. Two independent research groups, using H1975 cells, both reported that IGF-1R activation is an AR mechanism against osimertinib, and that combining osimertinib with an IGF-1R inhibitor (linsitinib) can overcome AR to osimertinib through IGF-1R activation [51,57]. Among clinical cases, Manabe et al. reported that 4 of 6 (66.7%) patients eventually showed elevated IGF2 after first-line osimertinib treatment failure [51].

ERK or AKT Pathway Reactivation

The activation of signaling downstream of EGFR, including the PI3K–AKT, RAS–RAF–MEK–ERK, and JAK–STAT pathways, is a common AR mechanism against EGFR-TKIs. For example, we and other groups have reported that PTEN downregulation can activate the PI3K–AKT pathway as a resistance mechanism against first-generation EGFR-TKIs [72,73,74]. As AR mechanisms against first-line osimertinib, Src-mediated activation of AKT is reported as a mechanism of resistance in H1975 cells [35]. Another group reported that increased ACK1, which phosphorylates AKT and activates the AKT pathway [75], is an AR mechanism against osimertinib in PC9 and HCC827 cells, and that osimertinib plus an ACK1 inhibitor ([R]-9b) might overcome osimertinib resistance [48]. In addition, many groups have suggested that the RAS–RAF–MEK–ERK pathway is involved in the acquisition of resistance to first-line osimertinib in vitro.
Monica et al. established a PC9 cell line with AR to osimertinib and found that the resistant cells had acquired a BRAF G469A mutation. The authors also found that BRAF G469A, which is a class II mutation, maintained the constitutive activation of the ERK pathway and that combining selumetinib or trametinib with osimertinib can overcome resistance from the BRAF G469A mutation [43].
The BRAF mutation is a rare driver mutation in lung adenocarcinoma (1~2%) that is mutually exclusive with EGFR mutations [1,76,77,78]. V600E is the most common BRAF mutation. It is present in about half of NSCLCs with BRAF-activating mutations. The acquisition of a BRAF V600E mutation has been reported after the failure of first-line osimertinib treatment [79,80,81,82]. Additionally, BRAF activation through BRAF gene fusion has also been reported as a resistance mechanism against osimertinib in clinical specimens [83].
The activation of RAS family members has also been reported in cell line models as an AR mechanism against first-line osimertinib. Nukaga et al. reported that PC9 cells that acquired resistance to osimertinib developed a KRAS G13D mutation [35]. Another group reported that the upregulation of KRAS expression was a mechanism of AR to osimertinib in PC9 cells [32]. In a study in which osimertinib-resistant cells with ERK reactivation were established from PC9 cells, no specific mechanism of ERK reactivation was detected, but the HRAS G13R mutation partially contributed to ERK reactivation in these cells [37]. Aberration in another RAS gene, NRAS, may also contribute to AR to first-line osimertinib. Eberlein et al. reported that upregulation of NRAS RNA expression, and NRAS mutations (NRAS E63K, NRAS G12V, NRAS G12R) are mechanisms of AR to osimertinib in PC9 cells, and that the combination of osimertinib plus an MEK inhibitor (selumetinib) can overcome the osimertinib resistance [32].
Many groups have reported finding KRAS mutations (G12C, G12D, G12S), BRAF mutations (D594N, V600E), BRAF fusions, PIK3CA mutations (E545K, E542K, R88Q, N345K, E418K), and PTEN loss in specimens from patients who developed AR against second-line osimertinib [61,84,85,86,87,88,89,90,91]. Furthermore, a recent study reported that PIK3CA mutations (E453K, E545K, H1047R; 7%), BRAF V600E mutations (3%), and KRAS mutations (G12D/C, A146T; 3%) were detected in specimens from patients who developed AR to first-line osimertinib [61]. Therefore, ERK reactivation through several mechanisms may be an important and common AR mechanism against both first- and second-line osimertinib therapy.

3.2.3. Suppression of Apoptotic Response

Apoptotic response is the final step when EGFR-mutated lung cancer cells are killed by an EGFR-TKI. Therefore, dysregulation of apoptosis might be reasonably supposed as a cause of inherent or acquired resistance to EGFR-TKIs. For example, the polymorphism of BCL-2-like 11 (BIM), a pro-apoptotic member of the BCL2 family, is reportedly sufficient to confer intrinsic resistance to first-generation EGFR-TKIs in lung cancers [92]. Although we could not find a paper that described the role of BIM polymorphism in first-line osimertinib efficacy, a recent study suggested that BIM polymorphism is correlated with worse outcomes in patients who received osimertinib after first- or second-generation EGFR-TKI failure due to a T790M secondary mutation [93].
As an AR mechanism against first-line osimertinib in vitro, the upregulation of MCL-1 (another anti-apoptotic protein) and downregulation of BIM was reported as the mechanism of AR to osimertinib in experiments that used PC9 and HCC827 cells. The authors showed that MEK inhibitors (GSK1120212 or PD-0325901) combined with osimertinib reduced MCL-1 expression and could overcome the resistance [34]. The same group reported in a later study that honokiol (a natural product isolated from the bark, seed cones, and leaves of trees belonging to the genus Magnolia) plus osimertinib enhanced MCL-1 reduction by suppressing ERK-dependent MCL-1 phosphorylation [47], and that the histone deacetylase inhibitor (LBH589) enhanced the effects of osimertinib in the resistant cells by decreasing levels of p-ERK and increasing BIM levels [49]. Another group reported that in experiments using PC9 and H1975 cells, BIM downregulation was the mechanism of AR to osimertinib. The authors showed that combining aspirin with osimertinib could overcome this resistance by promoting BIM-dependent apoptosis [50]. In addition, significant upregulation of the BCL-2 protein was reported in osimertinib-resistant H1975 cells, compared to their H1975 parental cells. The authors of this study showed that adding a BCL-2 inhibitor (ABT263 or ABT199) to osimertinib could overcome the resistance mediated by BCL-2 overexpression [56].

3.2.4. EMT

EMT is a process by which epithelial cells lose their cell polarity and cell–cell adhesion and acquire mesenchymal phenotypes that are associated with increased migratory and invasive properties, as well as drug resistance. We were one of the first groups to report the involvement of EMT in the acquisition of resistance to EGFR-TKIs in 2011 [12]. Several later studies indicated that the HCC4006 and H1975 cell lines often acquire resistance to EGFR-TKIs through EMT [94]. The overexpression of the ankyrin repeat domain-1 (ANKRD1), which is associated with the EMT process and anti-apoptosis, was identified in afatinib- and osimertinib-resistant cells established from PC9 and HCC827 cells. The authors also observed that imatinib could inhibit ANKRD1 expression, resulting in the restoration of sensitivity to afatinib and osimertinib in EGFR-TKI-resistant cells [36]. Another group established osimertinib-resistant cell lines from PC9 and HCC827 cells and reported that increased TGFβ1 induces high integrin β3 expression, which in turn induces the cells to revert to a mesenchymal phenotype. The authors also reported that combining osimertinib with a TGFβR1 inhibitor (SB-431542) could overcome the osimertinib resistance mediated through this pathway [40]. Other studies that reported the involvement of EMT in acquiring resistance to first-line osimertinib used H1975 cells [13,46,53,54,58,59,60], which were destined to develop EMT as an acquired resistance mechanism as described in our previous study [94]. Among mechanisms of EMT induction, increased ZEB1 expression [58], or decreased microRNA-200c in combination with increased ZEB1 expression [46] have been reported. The latter study also reported that GSK-3 inhibitor (LY2090314) may circumvent EMT-associated resistance to osimertinib [46]. Other important inducers of EMT include TGFβ2 and NF-kB signaling. An NF-kB pathway inhibitor (BAY 11-7082) reportedly caused cell death in resistant cells with EMT features [59]. Other groups have reported the respective roles of the YAP–FOXM1 axis [54] and the Src pathway [13] as central regulators of EMT-associated osimertinib resistance.
Several studies have reported evidence of EMT in tumor specimens obtained from patients who developed AR to first-generation EGFR-TKIs [25,58]. We therefore expect further reports of EMT-mediated AR to first-line osimertinib in the near future.

3.2.5. Other Mechanisms

Many studies have reported AR mechanisms against first-line osimertinib that cannot be classified into the above four categories. We summarize these mechanisms below.
Increased inositol-requiring enzyme-1α (IRE1α) expression was reported as an AR mechanism against osimertinib in HCC827 cell line model [38]. IRE1α affects endoplasmic reticulum (ER)-based stress signals [95,96]. As the ER regulates protein folding, under stress conditions, the accumulation of immature proteins provokes the ER stress response and boosts autophagy, resulting in autophagic cell death [95]. This process is mainly regulated via the IRE1α–JNK and PERK–eIF2α–ATF4 pathways [96]. The authors reported that osimertinib sensitivity was restored through the knockdown of IRE1α or treatment with an IRE1α inhibitor (STF-083010) [38]. The potential role of enhanced autophagy in AR against first-line osimertinib has been reported by other groups that used H1975 or PC9 models [42,53]. One study suggested that combining osimertinib with an autophagy inhibitor (CQ) could improve osimertinib cytotoxicity [42]. Li et al. reported that autophagy is a common mechanism in second-line osimertinib resistance in clinical specimens [42].
Codony-Servat et al. reported that the activation of multiple oncogenes (AKT, STAT3, YAP, AXL, IGF-1R, MET) and the upregulation of BCL-2 constitute a mechanism of AR to osimertinib in PC9 cells, and that the combination of osimertinib plus an Hsp90 inhibitor (luminespib) can overcome osimertinib resistance mediated by multiple pathways [44].
Increased CDK4 expression and phosphorylation of Rb (the down-stream molecule of CDK4/6) were reported in cells with AR to osimertinib established from H1975 cells. In this resistant model, the combination of CDK4/6 inhibitor (palbociclib) plus osimertinib overcame the resistance [55]. The CDK4- and CDK6-mono-phosphorylated Rb proteins and the phosphorylated Rb protein partially relieved Rb-mediated suppression of the family of E2F transcription factors [97]. These processes promote cell progression from the G1 to the S phase [97]. Notably, CDK4/6 gene amplification was detected in 5% of clinical specimens obtained from patients who progressed after first-line osimertinib treatment [61].

3.3. Correlation of Resistance Mechanisms Identified in Cell-Line Models and Clinical Specimens

As described above, the EGFR T790M secondary mutation [13,26,98] is the most common AR mechanism against first- and second-generation EGFR-TKIs at ~57%. Many studies have reported cell lines that develop resistance through T790M mutations after chronic exposure to first- or second-generation EGFR-TKIs [23]. However, as far as we know, no cell line has been reported to have developed AR to first-line osimertinib through EGFR secondary mutations. This is consistent with the lower frequency (~10%) of EGFR secondary mutations in clinical specimens obtained after first-line osimertinib failure.
That being said, many studies have reported the activation of a bypass pathway in cell lines that acquired resistance to first-line osimertinib. Some of them, such as MET gene amplification and KRAS/BRAF mutations, have been identified in clinical specimens.
Analyses of clinical specimens have reported higher frequency of SCLC transformation as a resistance mechanism to osimertinib. Interestingly, however, no cell line model with AR has shown evidence of SCLC transformation, which suggests a limitation of cell line models.

4. Summary

In this review, we summarized numerous mechanisms of AR to first-line osimertinib exposure in cell line models. We described that some of the resistance mechanisms that were found in cell line models were also identified in clinical specimens obtained from patients who developed AR to EGFR-TKIs, including those who acquired resistance to first-line osimertinib. Lessons from the experiments of acquired resistance mechanisms to first- or second-generation EGFR-TKIs have suggested that cell line models are useful tools to find novel mechanisms of resistance. Therefore, we hope that the catalog of resistance mechanisms to first-line osimertinib identified in cell line models summarized in this review helps researchers to identify acquired resistance mechanisms to first-line osimertinib in clinical specimens in the near future.

Author Contributions

Conceptualization, K.S.; methodology, S.O.; validation, K.S.; formal analysis, S.O. and K.S.; resources, S.O. and K.S.; data curation, S.O.; writing—original draft preparation, S.O. and K.S.; writing—review and editing, K.S. and T.M.; supervision, K.S.; funding acquisition, S.O., K.S. and T.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (20K17764 to S.O., 18K07336 to K.S. and 20H03773 to T.M.) and partially supported by a research grant from Boehringer Ingelheim to K.S. and T.M.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We thank Marla Brunker for editing a draft of this manuscript.

Conflicts of Interest

K.S. reports personal fees from AstraZeneca, grants and personal fees from Boehrinnger-Ingelheim related to the submitted work, and grants from Rain Therapeutics, outside the submitted work. T.M. reports grants and personal fees from AstraZeneca, grants and personal fees from Boehrinnger-Ingelheim related to the submitted work, grants and personal fees from Pfizer, personal fees from MSD, grants and personal fees from Chugai, grants and personal fees from Ono, personal fees from Eli-Lilly, grants and personal fees from Daiiichi-Sankyo, personal fees from ThermoFisher, personal fees from Guardant, grants and personal fees from Taiho, personal fees from Amgen, and personal fees from Novartis, outside of the submitted work. Other authors report no conflict of interest.

References

  1. Hirsch, F.R.; Suda, K.; Wiens, J.; Bunn, P.A., Jr. New and emerging targeted treatments in advanced non-small-cell lung cancer. Lancet 2016, 388, 1012–1024. [Google Scholar] [CrossRef]
  2. Hida, T.; Nokihara, H.; Kondo, M.; Kim, Y.H.; Azuma, K.; Seto, T.; Takiguchi, Y.; Nishio, M.; Yoshioka, H.; Imamura, F.; et al. Alectinib versus crizotinib in patients with ALK-positive non-small-cell lung cancer (J-ALEX): An open-label, randomised phase 3 trial. Lancet 2017, 390, 29–39. [Google Scholar] [CrossRef]
  3. Wu, Y.L.; Yang, J.C.; Kim, D.W.; Lu, S.; Zhou, J.; Seto, T.; Yang, J.J.; Yamamoto, N.; Ahn, M.J.; Takahashi, T.; et al. Phase II Study of Crizotinib in East Asian Patients with ROS1-Positive Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 1405–1411. [Google Scholar] [CrossRef]
  4. Marcus, L.; Donoghue, M.; Aungst, S.; Myers, C.E.; Helms, W.S.; Shen, G.; Zhao, H.; Stephens, O.; Keegan, P.; Pazdur, R. FDA Approval Summary: Entrectinib for the treatment of NTRK-gene fusion solid tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  5. Planchard, D.; Besse, B.; Groen, H.J.M.; Souquet, P.J.; Quoix, E.; Baik, C.S.; Barlesi, F.; Kim, T.M.; Mazieres, J.; Novello, S.; et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: An open-label, multicentre phase 2 trial. Lancet Oncol. 2016, 17, 984–993. [Google Scholar] [CrossRef] [Green Version]
  6. Fujino, T.; Suda, K.; Mitsudomi, T. Emerging MET tyrosine kinase inhibitors for the treatment of non-small cell lung cancer. Exp. Opin. Emerg. Drugs 2020, 25, 229–249. [Google Scholar] [CrossRef]
  7. Suda, K.; Tomizawa, K.; Mitsudomi, T. Biological and clinical significance of KRAS mutations in lung cancer: An oncogenic driver that contrasts with EGFR mutation. Cancer Metast. Rev. 2010, 29, 49–60. [Google Scholar] [CrossRef]
  8. Suda, K.; Bunn, P.A., Jr.; Rivard, C.J.; Mitsudomi, T.; Hirsch, F.R. Primary Double-Strike Therapy for Cancers to Overcome EGFR Kinase Inhibitor Resistance: Proposal from the Bench. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2017, 12, 27–35. [Google Scholar] [CrossRef] [Green Version]
  9. Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Jänne, 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. New Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
  10. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  11. Suda, K.; Murakami, I.; Katayama, T.; Tomizawa, K.; Osada, H.; Sekido, Y.; Maehara, Y.; Yatabe, Y.; Mitsudomi, T. Reciprocal and complementary role of MET amplification and EGFR T790M mutation in acquired resistance to kinase inhibitors in lung cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 5489–5498. [Google Scholar] [CrossRef] [Green Version]
  12. Suda, K.; Tomizawa, K.; Fujii, M.; Murakami, H.; Osada, H.; Maehara, Y.; Yatabe, Y.; Sekido, Y.; Mitsudomi, T. Epithelial to mesenchymal transition in an epidermal growth factor receptor-mutant lung cancer cell line with acquired resistance to erlotinib. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2011, 6, 1152–1161. [Google Scholar] [CrossRef] [Green Version]
  13. Nagano, T.; Tachihara, M.; Nishimura, Y. Mechanism of Resistance to Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitors and a Potential Treatment Strategy. Cells 2018, 7, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cross, D.A.; Ashton, S.E.; Ghiorghiu, S.; Eberlein, C.; Nebhan, C.A.; Spitzler, P.J.; Orme, J.P.; Finlay, M.R.; 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] [PubMed] [Green Version]
  15. 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]
  16. Wang, H.J.; Yang, H.P.; Zhou, X.D.; Dai, X.T.; Chen, Y.F.; Xiong, W. CA916798 regulates multidrug resistance of lung cancer cells. Asian Pac. J. Cancer Prevent. 2011, 12, 3403–3408. [Google Scholar]
  17. Wang, H.J.; Yang, Z.X.; Dai, X.T.; Chen, Y.F.; Yang, H.P.; Zhou, X.D. Bisdemethoxycurcumin sensitizes cisplatin-resistant lung cancer cells to chemotherapy by inhibition of CA916798 and PI3K/AKT signaling. Apoptosis Int. J. Program. Cell Death 2017, 22, 1157–1168. [Google Scholar] [CrossRef]
  18. Qi, Z.; Wang, Y.; Zhou, X. CA916798 gene participates in cisplatin resistance of human lung adenocarcinoma A549 cells through PI3K/AKT/mTOR pathway. Nan Fang Yi Ke da Xue Xue Bao J. South. Med. Univ. 2012, 32, 1290–1293. [Google Scholar]
  19. Hanke, N.T.; Imler, E.; Marron, M.T.; Seligmann, B.E.; Garland, L.L.; Baker, A.F. Characterization of carfilzomib-resistant non-small cell lung cancer cell lines. J. Cancer Res. Clin. Oncol. 2018, 144, 1317–1327. [Google Scholar] [CrossRef]
  20. El-Awady, R.A.; Hersi, F.; Al-Tunaiji, H.; Saleh, E.M.; Abdel-Wahab, A.H.; Al Homssi, A.; Suhail, M.; El-Serafi, A.; Al-Tel, T. Epigenetics and miRNA as predictive markers and targets for lung cancer chemotherapy. Cancer Biol. Ther. 2015, 16, 1056–1070. [Google Scholar] [CrossRef] [Green Version]
  21. Cirigliano, S.M.; Díaz Bessone, M.I.; Berardi, D.E.; Flumian, C.; Bal de Kier Joffé, E.D.; Perea, S.E.; Farina, H.G.; Todaro, L.B.; Urtreger, A.J. The synthetic peptide CIGB-300 modulates CK2-dependent signaling pathways affecting the survival and chemoresistance of non-small cell lung cancer cell lines. Cancer Cell Int. 2017, 17, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Suda, K.; Mizuuchi, H.; Maehara, Y.; Mitsudomi, T. Acquired resistance mechanisms to tyrosine kinase inhibitors in lung cancer with activating epidermal growth factor receptor mutation--diversity, ductility, and destiny. Cancer Metastasis Rev. 2012, 31, 807–814. [Google Scholar] [CrossRef] [PubMed]
  23. Hata, A.N.; Niederst, M.J.; Archibald, H.L.; Gomez-Caraballo, M.; Siddiqui, F.M.; Mulvey, H.E.; Maruvka, Y.E.; Ji, F.; Bhang, H.E.; Krishnamurthy Radhakrishna, V.; et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat. Med. 2016, 22, 262–269. [Google Scholar] [CrossRef] [PubMed]
  24. Sesumi, Y.; Suda, K.; Mizuuchi, H.; Kobayashi, Y.; Sato, K.; Chiba, M.; Shimoji, M.; Tomizawa, K.; Takemoto, T.; Mitsudomi, T. Effect of dasatinib on EMT-mediated-mechanism of resistance against EGFR inhibitors in lung cancer cells. Lung Cancer 2017, 104, 85–90. [Google Scholar] [CrossRef] [PubMed]
  25. Uramoto, H.; Iwata, T.; Onitsuka, T.; Shimokawa, H.; Hanagiri, T.; Oyama, T. Epithelial-mesenchymal transition in EGFR-TKI acquired resistant lung adenocarcinoma. Anticancer Res. 2010, 30, 2513–2517. [Google Scholar] [PubMed]
  26. 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]
  27. Fang, X.; Gu, P.; Zhou, C.; Liang, A.; Ren, S.; Liu, F.; Zeng, Y.; Wu, Y.; Zhao, Y.; Huang, B.; et al. β-Catenin overexpression is associated with gefitinib resistance in non-small cell lung cancer cells. Pulmonary Pharmacol. Ther. 2014, 28, 41–48. [Google Scholar] [CrossRef]
  28. Suda, K.; Mizuuchi, H.; Sato, K.; Takemoto, T.; Iwasaki, T.; Mitsudomi, T. The insulin-like growth factor 1 receptor causes acquired resistance to erlotinib in lung cancer cells with the wild-type epidermal growth factor receptor. Int. J. Cancer 2014, 135, 1002–1006. [Google Scholar] [CrossRef]
  29. Guix, M.; Faber, A.C.; Wang, S.E.; Olivares, M.G.; Song, Y.; Qu, S.; Rinehart, C.; Seidel, B.; Yee, D.; Arteaga, C.L.; et al. Acquired resistance to EGFR tyrosine kinase inhibitors in cancer cells is mediated by loss of IGF-binding proteins. J. Clin. Investig. 2008, 118, 2609–2619. [Google Scholar] [CrossRef]
  30. Cortot, A.B.; Repellin, C.E.; Shimamura, T.; Capelletti, M.; Zejnullahu, K.; Ercan, D.; Christensen, J.G.; Wong, K.K.; Gray, N.S.; Jänne, P.A. Resistance to irreversible EGF receptor tyrosine kinase inhibitors through a multistep mechanism involving the IGF1R pathway. Cancer Res. 2013, 73, 834–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Vazquez-Martin, A.; Cufí, S.; Oliveras-Ferraros, C.; Torres-Garcia, V.Z.; Corominas-Faja, B.; Cuyàs, E.; Bonavia, R.; Visa, J.; Martin-Castillo, B.; Barrajón-Catalán, E.; et al. IGF-1R/epithelial-to-mesenchymal transition (EMT) crosstalk suppresses the erlotinib-sensitizing effect of EGFR exon 19 deletion mutations. Sci. Rep. 2013, 3, 2560. [Google Scholar] [CrossRef] [PubMed]
  32. 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]
  33. Shi, P.; Oh, Y.T.; Zhang, G.; Yao, W.; Yue, P.; Li, Y.; Kanteti, R.; Riehm, J.; Salgia, R.; Owonikoko, T.K.; et al. Met gene amplification and protein hyperactivation is a mechanism of resistance to both first and third generation EGFR inhibitors in lung cancer treatment. Cancer Lett. 2016, 380, 494–504. [Google Scholar] [CrossRef] [PubMed]
  34. Shi, P.; Oh, Y.T.; Deng, L.; Zhang, G.; Qian, G.; Zhang, S.; Ren, H.; Wu, G.; Legendre, B., Jr.; Anderson, E.; et al. Overcoming Acquired Resistance to AZD9291, A Third-Generation EGFR Inhibitor, through Modulation of MEK/ERK-Dependent Bim and Mcl-1 Degradation. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 6567–6579. [Google Scholar] [CrossRef] [Green Version]
  35. Nukaga, S.; Yasuda, H.; Tsuchihara, K.; Hamamoto, J.; Masuzawa, K.; Kawada, I.; Naoki, K.; Matsumoto, S.; Mimaki, S.; Ikemura, S.; et al. Amplification of EGFR Wild-Type Alleles in Non-Small Cell Lung Cancer Cells Confers Acquired Resistance to Mutation-Selective EGFR Tyrosine Kinase Inhibitors. Cancer Res. 2017, 77, 2078–2089. [Google Scholar] [CrossRef] [Green Version]
  36. Takahashi, A.; Seike, M.; Chiba, M.; Takahashi, S.; Nakamichi, S.; Matsumoto, M.; Takeuchi, S.; Minegishi, Y.; Noro, R.; Kunugi, S.; et al. Ankyrin Repeat Domain 1 Overexpression is Associated with Common Resistance to Afatinib and Osimertinib in EGFR-mutant Lung Cancer. Sci. Rep. 2018, 8, 14896. [Google Scholar] [CrossRef]
  37. Ku, B.M.; Choi, M.K.; Sun, J.M.; Lee, S.H.; Ahn, J.S.; Park, K.; Ahn, M.J. Acquired resistance to AZD9291 as an upfront treatment is dependent on ERK signaling in a preclinical model. PLoS ONE 2018, 13, e0194730. [Google Scholar] [CrossRef]
  38. Tang, Z.H.; Su, M.X.; Guo, X.; Jiang, X.M.; Jia, L.; Chen, X.; Lu, J.J. Increased Expression of IRE1α Associates with the Resistant Mechanism of Osimertinib (AZD9291)-resistant non-small Cell Lung Cancer HCC827/OSIR Cells. Anti-Cancer Agents Med. Chem. 2018, 18, 550–555. [Google Scholar] [CrossRef]
  39. 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] [PubMed] [Green Version]
  40. Wang, C.; Wang, T.; Lv, D.; Li, L.; Yue, J.; Chen, H.Z.; Xu, L. Acquired Resistance to EGFR TKIs Mediated by TGFβ1/Integrin β3 Signaling in EGFR-Mutant Lung Cancer. Mol. Cancer Ther. 2019, 18, 2357–2367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Liu, Y.N.; Tsai, M.F.; Wu, S.G.; Chang, T.H.; Tsai, T.H.; Gow, C.H.; Chang, Y.L.; Shih, J.Y. Acquired resistance to EGFR tyrosine kinase inhibitors is mediated by the reactivation of STC2/JUN/AXL signaling in lung cancer. Int. J. Cancer 2019, 145, 1609–1624. [Google Scholar] [CrossRef]
  42. Li, L.; Wang, Y.; Jiao, L.; Lin, C.; Lu, C.; Zhang, K.; Hu, C.; Ye, J.; Zhang, D.; Wu, H.; et al. Protective autophagy decreases osimertinib cytotoxicity through regulation of stem cell-like properties in lung cancer. Cancer Lett. 2019, 452, 191–202. [Google Scholar] [CrossRef] [PubMed]
  43. La Monica, S.; Minari, R.; Cretella, D.; Bonelli, M.; Fumarola, C.; Cavazzoni, A.; Galetti, M.; Digiacomo, G.; Riccardi, F.; Petronini, P.G.; et al. Acquired BRAF G469A Mutation as a Resistance Mechanism to First-Line Osimertinib Treatment in NSCLC Cell Lines Harboring an EGFR Exon 19 Deletion. Targ. Oncol. 2019, 14, 619–626. [Google Scholar] [CrossRef] [PubMed]
  44. Codony-Servat, J.; Viteri, S.; Codony-Servat, C.; Ito, M.; Bracht, J.W.P.; Berenguer, J.; Chaib, I.; Molina-Vila, M.A.; Karachaliou, N.; Rosell, R. Hsp90 inhibitors enhance the antitumoral effect of osimertinib in parental and osimertinib-resistant non-small cell lung cancer cell lines. Transl. Lung Cancer Res. 2019, 8, 340–351. [Google Scholar] [CrossRef]
  45. Yonesaka, K.; Takegawa, N.; Watanabe, S.; Haratani, K.; Kawakami, H.; Sakai, K.; Chiba, Y.; Maeda, N.; Kagari, T.; Hirotani, K.; et al. An HER3-targeting antibody-drug conjugate incorporating a DNA topoisomerase I inhibitor U3-1402 conquers EGFR tyrosine kinase inhibitor-resistant NSCLC. Oncogene 2019, 38, 1398–1409. [Google Scholar] [CrossRef]
  46. Fukuda, K.; Takeuchi, S.; Arai, S.; Kita, K.; Tanimoto, A.; Nishiyama, A.; Yano, S. Glycogen synthase kinase-3 inhibition overcomes epithelial-mesenchymal transition-associated resistance to osimertinib in EGFR-mutant lung cancer. Cancer Sci. 2020, 111, 2374–2384. [Google Scholar] [CrossRef]
  47. Zang, H.; Qian, G.; Arbiser, J.; Owonikoko, T.K.; Ramalingam, S.S.; Fan, S.; Sun, S.Y. Overcoming acquired resistance of EGFR-mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib, with the natural product honokiol. Mol. Oncol. 2020, 14, 882–895. [Google Scholar] [CrossRef] [Green Version]
  48. Gu, J.; Qian, L.; Zhang, G.; Mahajan, N.P.; Owonikoko, T.K.; Ramalingam, S.S.; Sun, S.Y. Inhibition of ACK1 delays and overcomes acquired resistance of EGFR mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib. Lung Cancer 2020, 150, 26–35. [Google Scholar] [CrossRef] [PubMed]
  49. Zang, H.; Qian, G.; Zong, D.; Fan, S.; Owonikoko, T.K.; Ramalingam, S.S.; Sun, S.Y. Overcoming acquired resistance of epidermal growth factor receptor-mutant non-small cell lung cancer cells to osimertinib by combining osimertinib with the histone deacetylase inhibitor panobinostat (LBH589). Cancer 2020, 126, 2024–2033. [Google Scholar] [CrossRef]
  50. Han, R.; Hao, S.; Lu, C.; Zhang, C.; Lin, C.; Li, L.; Wang, Y.; Hu, C.; He, Y. Aspirin sensitizes osimertinib-resistant NSCLC cells in vitro and in vivo via Bim-dependent apoptosis induction. Mol. Oncol. 2020, 14, 1152–1169. [Google Scholar] [CrossRef]
  51. 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]
  52. 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] [PubMed]
  53. Verusingam, N.D.; Chen, Y.C.; Lin, H.F.; Liu, C.Y.; Lee, M.C.; Lu, K.H.; Cheong, S.K.; Ong, A.H.; Chiou, S.H.; Wang, M.L. Generation of Osimertinib-Resistant Cells from EGFR L858R/T790M Mutant NSCLC Cell Line. J. Chin. Med. Assoc. 2020. [Google Scholar] [CrossRef] [PubMed]
  54. Nilsson, M.B.; Sun, H.; Robichaux, J.; Pfeifer, M.; McDermott, U.; Travers, J.; Diao, L.; Xi, Y.; Tong, P.; Shen, L.; et al. A YAP/FOXM1 axis mediates EMT-associated EGFR inhibitor resistance and increased expression of spindle assembly checkpoint components. Sci. Transl. Med. 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  55. 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). Thoracic Cancer 2020, 11, 2389–2397. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, Z.; Gao, W. Synergistic effects of Bcl-2 inhibitors with AZD9291 on overcoming the acquired resistance of AZD9291 in H1975 cells. Arch. Toxicol. 2020, 94, 3125–3136. [Google Scholar] [CrossRef]
  57. 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. Thoracic Cancer 2020, 11, 140–149. [Google Scholar] [CrossRef] [Green Version]
  58. Weng, C.H.; Chen, L.Y.; Lin, Y.C.; Shih, J.Y.; Lin, Y.C.; Tseng, R.Y.; Chiu, A.C.; Yeh, Y.H.; Liu, C.; Lin, Y.T.; et al. Epithelial-mesenchymal transition (EMT) beyond EGFR mutations per se is a common mechanism for acquired resistance to EGFR TKI. Oncogene 2019, 38, 455–468. [Google Scholar] [CrossRef]
  59. Jiang, X.M.; Xu, Y.L.; Yuan, L.W.; Zhang, L.L.; Huang, M.Y.; Ye, Z.H.; Su, M.X.; Chen, X.P.; Zhu, H.; Ye, R.D.; et al. TGFβ2-mediated epithelial-mesenchymal transition and NF-κB pathway activation contribute to osimertinib resistance. Acta Pharmacol. Sinica 2020. [Google Scholar] [CrossRef]
  60. Ji, W.; Choi, Y.J.; Kang, M.H.; Sung, K.J.; Kim, D.H.; Jung, S.; Choi, C.M.; Lee, J.C.; Rho, J.K. Efficacy of the CDK7 Inhibitor on EMT-Associated Resistance to 3rd Generation EGFR-TKIs in Non-Small Cell Lung Cancer Cell Lines. Cells 2020, 9, 2596. [Google Scholar] [CrossRef]
  61. 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]
  62. Turke, A.B.; Zejnullahu, K.; Wu, Y.L.; Song, Y.; Dias-Santagata, D.; Lifshits, E.; Toschi, L.; Rogers, A.; Mok, T.; Sequist, L.; et al. Preexistence and clonal selection of MET amplification in EGFR mutant NSCLC. Cancer Cell 2010, 17, 77–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Roper, N.; Brown, A.L.; Wei, J.S.; Pack, S.; Trindade, C.; Kim, C.; Restifo, O.; Gao, S.; Sindiri, S.; Mehrabadi, F.; et al. Clonal Evolution and Heterogeneity of Osimertinib Acquired Resistance Mechanisms in EGFR Mutant Lung Cancer. Cell Rep. Med. 2020, 1. [Google Scholar] [CrossRef]
  64. Antony, J.; Huang, R.Y.-J. AXL-Driven EMT State as a Targetable Conduit in Cancer. J. Cancer Res. 2017, 77, 3725–3732. [Google Scholar] [CrossRef] [Green Version]
  65. Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; Abdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef]
  66. Ji, W.; Choi, C.M.; Rho, J.K.; Jang, S.J.; Park, Y.S.; Chun, S.M.; Kim, W.S.; Lee, J.S.; Kim, S.W.; Lee, D.H.; et al. Mechanisms of acquired resistance to EGFR-tyrosine kinase inhibitor in Korean patients with lung cancer. BMC Cancer 2013, 13, 606. [Google Scholar] [CrossRef] [Green Version]
  67. Nonagase, Y.; Takeda, M.; Azuma, K.; Hayashi, H.; Haratani, K.; Tanaka, K.; Yonesaka, K.; Ishii, H.; Hoshino, T.; Nakagawa, K. Tumor tissue and plasma levels of AXL and GAS6 before and after tyrosine kinase inhibitor treatment in EGFR-mutated non-small cell lung cancer. Thoracic Cancer 2019, 10, 1928–1935. [Google Scholar] [CrossRef] [Green Version]
  68. Taniguchi, H.; Yamada, T.; Wang, R.; Tanimura, K.; Adachi, Y.; Nishiyama, A.; Tanimoto, A.; Takeuchi, S.; Araujo, L.H.; Boroni, M.; et al. AXL confers intrinsic resistance to osimertinib and advances the emergence of tolerant cells. Nat. Commun. 2019, 10, 259. [Google Scholar] [CrossRef] [PubMed]
  69. Hsieh, A.C.; Moasser, M.M. Targeting HER proteins in cancer therapy and the role of the non-target HER3. Br. J. Cancer 2007, 97, 453–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Liang, J.; Slingerland, J.M. Multiple roles of the PI3K/PKB (Akt) pathway in cell cycle progression. Cell Cycle 2003, 2, 339–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. LeRoith, D.; Roberts, C.T., Jr. The insulin-like growth factor system and cancer. Cancer Lett. 2003, 195, 127–137. [Google Scholar] [CrossRef] [Green Version]
  72. Suda, K.; Tomizawa, K.; Osada, H.; Maehara, Y.; Yatabe, Y.; Sekido, Y.; Mitsudomi, T. Conversion from the “oncogene addiction” to “drug addiction” by intensive inhibition of the EGFR and MET in lung cancer with activating EGFR mutation. Lung Cancer 2012, 76, 292–299. [Google Scholar] [CrossRef]
  73. Shen, H.; Zhu, F.; Liu, J.; Xu, T.; Pei, D.; Wang, R.; Qian, Y.; Li, Q.; Wang, L.; Shi, Z.; et al. Alteration in Mir-21/PTEN expression modulates gefitinib resistance in non-small cell lung cancer. PLoS ONE 2014, 9, e103305. [Google Scholar] [CrossRef]
  74. Li, B.; Ren, S.; Li, X.; Wang, Y.; Garfield, D.; Zhou, S.; Chen, X.; Su, C.; Chen, M.; Kuang, P.; et al. MiR-21 overexpression is associated with acquired resistance of EGFR-TKI in non-small cell lung cancer. Lung Cancer 2014, 83, 146–153. [Google Scholar] [CrossRef]
  75. Xu, S.H.; Huang, J.Z.; Xu, M.L.; Yu, G.; Yin, X.F.; Chen, D.; Yan, G.R. ACK1 promotes gastric cancer epithelial-mesenchymal transition and metastasis through AKT-POU2F1-ECD signalling. J. Pathol. 2015, 236, 175–185. [Google Scholar] [CrossRef]
  76. Barlesi, F.; Mazieres, J.; Merlio, J.P.; Debieuvre, D.; Mosser, J.; Lena, H.; Ouafik, L.; Besse, B.; Rouquette, I.; Westeel, V.; et al. Routine molecular profiling of patients with advanced non-small-cell lung cancer: Results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT). Lancet 2016, 387, 1415–1426. [Google Scholar] [CrossRef]
  77. Kris, M.G.; Johnson, B.E.; Berry, L.D.; Kwiatkowski, D.J.; Iafrate, A.J.; Wistuba, I.I.; Varella-Garcia, M.; Franklin, W.A.; Aronson, S.L.; Su, P.F.; et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014, 311, 1998–2006. [Google Scholar] [CrossRef] [PubMed]
  78. Paik, P.K.; Arcila, M.E.; Fara, M.; Sima, C.S.; Miller, V.A.; Kris, M.G.; Ladanyi, M.; Riely, G.J. Clinical Characteristics of Patients with Lung Adenocarcinomas Harboring BRAF Mutations. J. Clin. Oncol. 2011, 29, 2046–2051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Ho, C.C.; Liao, W.Y.; Lin, C.A.; Shih, J.Y.; Yu, C.J.; Yang, J.C. Acquired BRAF V600E Mutation as Resistant Mechanism after Treatment with Osimertinib. J. Thoracic Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2017, 12, 567–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. 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]
  81. Huang, Y.; Gan, J.; Guo, K.; Deng, Y.; Fang, W. Acquired BRAF V600E Mutation Mediated Resistance to Osimertinib and Responded to Osimertinib, Dabrafenib, and Trametinib Combination Therapy. J. Thoracic Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2019, 14, e236–e237. [Google Scholar] [CrossRef]
  82. Meng, P.; Koopman, B.; Kok, K.; Ter Elst, A.; Schuuring, E.; van Kempen, L.C.; Timens, W.; Hiltermann, T.J.N.; Groen, H.J.M.; van den Berg, A.; et al. Combined osimertinib, dabrafenib and trametinib treatment for advanced non-small-cell lung cancer patients with an osimertinib-induced BRAF V600E mutation. Lung Cancer 2020, 146, 358–361. [Google Scholar] [CrossRef]
  83. Suda, K.; Mitsudomi, T.J.T.L.C.R. Emerging oncogenic fusions other than ALK, ROS1, RET, and NTRK in NSCLC and the role of fusions as resistance mechanisms to targeted therapy. Transl. Lung Cancer Res. 2020, 2020. [Google Scholar] [CrossRef]
  84. Papadimitrakopoulou, V.; Wu, Y.L.; Han, J.Y.; Ahn, M.J.; Ramalingam, S.; John, T.; Okamoto, I.; Yang, J.C.H.; Bulusu, K.; Laus, G.; et al. LBA51Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Ann. Oncol. 2018, 29. [Google Scholar] [CrossRef]
  85. Ortiz-Cuaran, S.; Scheffler, M.; Plenker, D.; Dahmen, L.; Scheel, A.H.; Fernandez-Cuesta, L.; Meder, L.; Lovly, C.M.; Persigehl, T.; Merkelbach-Bruse, S.; et al. Heterogeneous Mechanisms of Primary and Acquired Resistance to Third-Generation EGFR Inhibitors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 4837–4847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Park, S.; Shim, J.H.; Lee, B.; Cho, I.; Park, W.Y.; Kim, Y.; Lee, S.H.; Choi, Y.; Han, J.; Ahn, J.S.; et al. Paired genomic analysis of squamous cell carcinoma transformed from EGFR-mutated lung adenocarcinoma. Lung Cancer 2019, 134, 7–15. [Google Scholar] [CrossRef] [PubMed]
  87. Oxnard, G.R.; Thress, K.; Paweletz, C.; Stetson, D.; Dougherty, B.; Lai, Z.; Markovets, A.; Felip, E.; Vivancos, A.; Kuang, Y. ORAL17.07 Mechanisms of Acquired Resistance to AZD9291 in EGFR T790M Positive Lung. J. Thorac. Oncol. 2015, 10, S207. [Google Scholar]
  88. 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]
  89. 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. Off. J. Am. Assoc. Cancer Res. 2018, 24, 6195–6203. [Google Scholar] [CrossRef] [Green Version]
  90. Shi, J.H.; Lou, Y.Y.; Zhou, K.L.; Pan, D.Q. Exploration of intermolecular interaction of calf thymus DNA with sulfosulfuron using multi-spectroscopic and molecular docking techniques. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2018, 204, 209–216. [Google Scholar] [CrossRef] [PubMed]
  91. Hong, M.H.; Kim, M.H.; Kim, S.Y.; Heo, S.G.; Kang, H.N.; Park, C.W.; Barrett, J.C.; Stetson, D.; Chmielecki, J.; Markovets, A.; et al. Molecular landscape of osimertinib resistance revealed by targeted panel sequencing and patient-derived cancer models in non-small cell lung cancer patients. Ann. Oncology 2018, 29, viii516. [Google Scholar] [CrossRef]
  92. Ng, K.P.; Hillmer, A.M.; Chuah, C.T.; Juan, W.C.; Ko, T.K.; Teo, A.S.; Ariyaratne, P.N.; Takahashi, N.; Sawada, K.; Fei, Y.; et al. A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer. Nat. Med. 2012, 18, 521–528. [Google Scholar] [CrossRef] [PubMed]
  93. Li, X.; Zhang, D.; Li, B.; Zou, B.; Wang, S.; Fan, B.; Li, W.; Yu, J.; Wang, L. Clinical implications of germline BCL2L11 deletion polymorphism in pretreated advanced NSCLC patients with osimertinib therapy. Lung Cancer 2020, 151, 39–43. [Google Scholar] [CrossRef] [PubMed]
  94. Suda, K.; Murakami, I.; Yu, H.; Kim, J.; Tan, A.C.; Mizuuchi, H.; Rozeboom, L.; Ellison, K.; Rivard, C.J.; Mitsudomi, T.; et al. CD44 Facilitates Epithelial-to-Mesenchymal Transition Phenotypic Change at Acquisition of Resistance to EGFR Kinase Inhibitors in Lung Cancer. Mol. Cancer Ther. 2018, 17, 2257–2265. [Google Scholar] [CrossRef] [Green Version]
  95. Ogata, M.; Hino, S.; Saito, A.; Morikawa, K.; Kondo, S.; Kanemoto, S.; Murakami, T.; Taniguchi, M.; Tanii, I.; Yoshinaga, K.; et al. Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol. Cell. Biol. 2006, 26, 9220–9231. [Google Scholar] [CrossRef] [Green Version]
  96. Moretti, L.; Yang, E.S.; Kim, K.W.; Lu, B. Autophagy signaling in cancer and its potential as novel target to improve anticancer therapy. Drug Resist. Updates Rev. Comment. Antimicrob. Anticancer Chemother. 2007, 10, 135–143. [Google Scholar] [CrossRef]
  97. Narasimha, A.M.; Kaulich, M.; Shapiro, G.S.; Choi, Y.J.; Sicinski, P.; Dowdy, S.F. Cyclin D activates the Rb tumor suppressor by mono-phosphorylation. eLife 2014, 3. [Google Scholar] [CrossRef]
  98. 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. Off. J. Am. Assoc. Cancer Res. 2013, 19, 2240–2247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Mechanisms of acquired resistance to first-line osimertinib that were identified in cell line models [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,48,50,51,52,53,54,55,56,57,58,59,60]. This graph is color-coded by cell line models and mechanisms of resistance to first-line osimertinib. EGFR, epidermal growth factor receptor; AXL, AXL receptor tyrosine kinase; ERBB3, human epidermal growth factor receptor-3; IGF1R, insulin-like growth factor-1 receptor; TGF, transforming growth factor; ACK1, activated Cdc42-associated kinase-1; CDK4/6, cyclin-dependent kinase; BCL-2, B-cell lymphoma-2; YAP, yes-associated protein; ANKRD1, ankyrin repeat domain-1; IRE1α, inositol-requiring enzyme-1α.
Figure 1. Mechanisms of acquired resistance to first-line osimertinib that were identified in cell line models [32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,48,50,51,52,53,54,55,56,57,58,59,60]. This graph is color-coded by cell line models and mechanisms of resistance to first-line osimertinib. EGFR, epidermal growth factor receptor; AXL, AXL receptor tyrosine kinase; ERBB3, human epidermal growth factor receptor-3; IGF1R, insulin-like growth factor-1 receptor; TGF, transforming growth factor; ACK1, activated Cdc42-associated kinase-1; CDK4/6, cyclin-dependent kinase; BCL-2, B-cell lymphoma-2; YAP, yes-associated protein; ANKRD1, ankyrin repeat domain-1; IRE1α, inositol-requiring enzyme-1α.
Cells 10 00354 g001
Figure 2. Scheme of the signal pathway and molecules that reportedly cause osimertinib resistance—AXL [39,41,52], HER3 [45], IGF1R [51,57], MET [33,52], TGFβ1 [40], TGFβ2 [59], BRAF [43], RAS [32,35], ERK [37], Src-AKT [35], ACK1 [48], CDK4/6 [55], BCL-2 [56], MCL-1 [34], BIM [34,50], YAP [54], ANKRD1 [36], microRNA-200c [46], ZEB1 [46,58], and IRE1α [38] (highlighted in red)—in cell line models treated with osimertinib. EGFR, epidermal growth factor receptor; AXL, AXL receptor tyrosine kinase; ERBB3, human epidermal growth factor receptor-3; IGF1R, insulin-like growth factor-1 receptor; MET, MET proto-oncogene, receptor tyrosine kinase; TGF, transforming growth factor; ACK1, activated Cdc42-associated kinase-1; CDK4/6, cyclin-dependent kinase; BCL-2, B-cell lymphoma-2; YAP, yes-associated protein; ANKRD1, ankyrin repeat domain-1; IRE1α, inositol-requiring enzyme-1α.
Figure 2. Scheme of the signal pathway and molecules that reportedly cause osimertinib resistance—AXL [39,41,52], HER3 [45], IGF1R [51,57], MET [33,52], TGFβ1 [40], TGFβ2 [59], BRAF [43], RAS [32,35], ERK [37], Src-AKT [35], ACK1 [48], CDK4/6 [55], BCL-2 [56], MCL-1 [34], BIM [34,50], YAP [54], ANKRD1 [36], microRNA-200c [46], ZEB1 [46,58], and IRE1α [38] (highlighted in red)—in cell line models treated with osimertinib. EGFR, epidermal growth factor receptor; AXL, AXL receptor tyrosine kinase; ERBB3, human epidermal growth factor receptor-3; IGF1R, insulin-like growth factor-1 receptor; MET, MET proto-oncogene, receptor tyrosine kinase; TGF, transforming growth factor; ACK1, activated Cdc42-associated kinase-1; CDK4/6, cyclin-dependent kinase; BCL-2, B-cell lymphoma-2; YAP, yes-associated protein; ANKRD1, ankyrin repeat domain-1; IRE1α, inositol-requiring enzyme-1α.
Cells 10 00354 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ohara, S.; Suda, K.; Mitsudomi, T. Cell Line Models for Acquired Resistance to First-Line Osimertinib in Lung Cancers—Applications and Limitations. Cells 2021, 10, 354. https://doi.org/10.3390/cells10020354

AMA Style

Ohara S, Suda K, Mitsudomi T. Cell Line Models for Acquired Resistance to First-Line Osimertinib in Lung Cancers—Applications and Limitations. Cells. 2021; 10(2):354. https://doi.org/10.3390/cells10020354

Chicago/Turabian Style

Ohara, Shuta, Kenichi Suda, and Tetsuya Mitsudomi. 2021. "Cell Line Models for Acquired Resistance to First-Line Osimertinib in Lung Cancers—Applications and Limitations" Cells 10, no. 2: 354. https://doi.org/10.3390/cells10020354

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