Open Access

Drug resistance mechanisms and progress in the treatment of EGFR‑mutated lung adenocarcinoma (Review)

  • Authors:
    • Ruizhu Sun
    • Zhansheng Hou
    • Yankui Zhang
    • Bo Jiang
  • View Affiliations

  • Published online on: September 26, 2022     https://doi.org/10.3892/ol.2022.13528
  • Article Number: 408
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

According to global cancer data, lung cancer was the leading cause of cancer‑related death in 2020. With the diversification of treatment strategies, the survival outcomes of patients with advanced lung cancer have improved significantly, but the 5‑year overall survival rate remains <20%. Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR‑TKIs) are the preferred treatment for lung adenocarcinoma patients with EGFR‑sensitive mutations; however, acquired drug resistance is inevitable. Osimertinib (a third‑generation EGFR inhibitor) is the most commonly used drug for cancers with a secondary T790M mutation. Unfortunately, acquired drug resistance against third‑generation drugs still emerges. The C797s mutation is the primary acquired resistance mechanism against Osimertinib. Research on fourth‑generation EGFR‑TKI drugs with a C797s mutation is currently at various experimental stages, and no drug has been approved for clinical use. In addition to the resistance mechanisms described above, HER2 amplification, MET amplification, PIK3A mutation, KRAS mutation, BRAF mutation, transformation to small cell lung cancer, transformation to lung squamous cell carcinoma, and EMT have been reported as mechanisms of acquired drug resistance to first‑, second‑ and third‑generation EGFR‑TKIs. These mechanisms are noted in a relatively high proportion of tumors, but treatment options are limited. In recent years, immunotherapy has made progress in the treatment of several cancers, including advanced EGFR‑mutated non‑small cell lung cancer (NSCLC). Due to the relatively high frequency of EGFR mutation in patients with lung adenocarcinoma in China, an increased number of patients develop EGFR‑TKI resistance, and subsequent treatment options are critical. This article reviews the mechanisms of drug resistance to different EGFR‑TKIs and treatment progression, providing ideas for the follow‑up treatment for EGFR‑resistant patients.

Introduction

According to global cancer data statistics, lung cancer was the leading cause of cancer-related death in 2020 (1). Approximately 30% of cancer-related deaths in China are related to lung cancer, which remains the most common cancer type (2). Clinical statistics show that non-small cell lung cancer (NSCLC) accounts for ~85% of lung cancer cases, and lung adenocarcinoma is the most common type of NSCLC (1). Recently, with the introduction of molecular-targeted drugs and immune checkpoint inhibitors, the survival outcomes of patients with advanced lung cancer have improved greatly, but the 5-year overall survival rate of patients with lung adenocarcinoma remains less than 20% (35).

According to statistics, the incidence of epidermal growth factor receptor (EGFR) mutations in Caucasians is ~20% (6), whereas the rate is 44–50% among Asian nonsmoking NSCLC patients (7,8). The higher frequency of EGFR mutations appears to be beneficial for Asian lung adenocarcinoma patients. EGFR tyrosine kinase inhibitors (TKIs) are currently the first-line treatment for lung adenocarcinoma patients with EGFR-sensitive mutations (4,914); unfortunately, most patients develop acquired drug resistance after 10–14 months of EGFR-TKI treatment (14,15). The mechanisms of acquisition of drug resistance to first- and second-generation EGFR-TKIs are complex, and the most important mechanism of acquired drug resistance is the secondary T790M mutation, accounting for 50–60% of all cases (16). The third-generation EGFR inhibitor Osimertinib is the most common drug used for the treatment of patients with this mutation (17). However, acquired drug resistance still emerges against third-generation drugs typically 8–10 months after receiving Osimertinib (17,18). The C797s mutation is the primary mechanism of acquired drug resistance (19). Research on fourth-generation EGFR-TKI drugs for the treatment of tumors with a C797s mutation is currently at various experimental stages, although no drug has been approved for clinical use.

Due to the relatively high frequency of EGFR mutations in patients with lung adenocarcinoma in China, an increasing number of patients develop EGFR-TKI resistance, and subsequent treatment options are critical. This article reviews the mechanisms of drug resistance and treatment progress after EGFR-TKI resistance.

Drug resistance mechanisms and progress in the use of first- and second-generation EGFR-TKIs

The mechanisms of acquired drug resistance against first- and second-generation EGFR-TKIs are complex and can be divided into three categories: Changes in EGFR, activation of alternative bypass or downstream pathways, and changes in the phenotype (Fig. 1).

Changes in EGFR
T790M mutation

A secondary T790M mutation is the most important mechanism of acquired drug resistance against first-generation EGFR-TKIs. The crystal structure of the ATP binding pocket is altered due to this mutation, inhibiting the binding of TKIs and ATP. Thus, downstream signal transduction cannot be inhibited by TKIs, and these drugs do not subsequently restrict tumor growth (20,21). The mechanism of action of the first-generation EGFR-TKI differs from that of the second-generation EGFR-TKI; the second-generation EGFR-TKI irreversibly binds to the ErbB receptor, resulting in a more potent effect than the first-generation drugs (22), but the mechanism of drug resistance is similar (2325). In the ARCHER1050 study, dacomitinib had overall survival (OS) benefits relative to gefitinib in the Chinese population (median overall survival (mOS) duration was 32.5 months vs. 24.9 months, P=0.0097) (26). The LUX-Lung7 trial compared the efficacy of afatinib and gefitinib for the treatment of NSCLC patients with EGFR mutations. The results showed that the progression-free survival (PFS) duration of the afatinib group was longer than that of the gefitinib group (11.0 vs. 10.9 months; P=0.017) (11). These results suggest that compared with first-generation EGFR-TKIs, the effects of second-generation EGFR-TKIs are longer in the context of T790M.

Osimertinib is the most widely used third-generation EGFR-TKI and it can effectively and selectively inhibit tumors with EGFR-sensitive and T790M drug-resistant mutations (27), exhibiting a significant effect in NSCLC patients with brain metastases (28,29). Almonertinib (30,31) and furmonertinib (32,33) have also been approved in China, and several other third-generation EGFR-TKI inhibitors are in different stages of research and development (Table I). Lazertinib achieved a 57% overall response rate (ORR) in the T790M (+) population in a phase 2 clinical trial (34). The drug exhibited a potent beneficial effect on brain lesions, and the intracranial disease control rate in the entire population was 90.6% (35). In January 2021, the Korean Food and Drug Administration (MFDS) approved the listing of lazertinib for the treatment of patients with locally advanced or metastatic NSCLC positive for EGFR T790M mutations who previously received EGFR-TKI treatment (36). The third-generation EGFR-TKIs olmutinib (3739) and nazartinib (40) are also approved in South Korea.

Table I.

Research and development of third-generation EGFR inhibitors in China.

Table I.

Research and development of third-generation EGFR inhibitors in China.

NameManufacturerIndicationsDevelopment phase
MEK162Betta PharmaceuticalsAdvanced NSCLC with a T790M mutation after EGFR resistance Declared/listed
Previously untreated NSCLC patients with locally advanced or metastatic EGFR sensitive mutationsPhase 2/3 clinical trial
AC0010 (Avitinib)Acea BiosciencesAdvanced NSCLC with a T790M mutation after EGFR resistance Declared/listed
NSCLC with EGFR mutationsPhase 3 clinical trial
BPI-7711Beta Pharma, Inc.Advanced NSCLC with a T790M mutation after EGFR resistance Declared/listed
NSCLC with EGFR mutationsPhase 3 clinical trial
ASK120067Suzhou Aosaikang Biomedical Co.Advanced NSCLC with a T790M mutation after EGFR resistancePhase 1/2 clinical trial
NSCLC with EGFR mutationsPhase 3 clinical trial
SH-1028Nanjing Sanhome Pharmaceutical Co.Advanced NSCLC with a T790M mutation after EGFR resistancePhase 2 clinical trial
NSCLC with EGFR mutationsPhase 3 clinical trial
FHND9041Nanjing Chuangren Pharmaceutical Technology CenterAdvanced NSCLC with a T790M mutation after EGFR resistancePhase 1/2 clinical trial
YZJ-0318Yangtze River Pharmaceutical (Group) Co.Advanced NSCLC with a T790M mutationPhase 1 clinical trial
MED-1007Jiangsu Maidu Pharmaceutical Co.Advanced NSCLC with a T790M mutation after EGFR resistancePhase 1 clinical trial
BEBT-109Bebtter Medicine TechnologyAdvanced NSCLC with a T790M mutation after EGFR resistancePhase 1 clinical trial
TY-9591Zhejiang Tongyuan KangAdvanced NSCLC with a T790M mutationPhase 1 clinical trial
TQB3456Chiatai TianqingAdvanced NSCLC with a T790M mutationPhase 1 clinical trial
LazertinibYuhan CorporationCombination of amivantamab for treatment of Osimertinib resistant NSCLCPhase 1 clinical trial
NSCLCPhase 3 clinical trial

[i] NSCLC, non-small cell lung cancer; EGFR, epidermal growth factor receptor.

Secondary mutations

Other rare secondary mutations, such as L747S (41), D761Y (42), and T854A (43), have also been reported to be associated with gefitinib or erlotinib resistance. Due to the low incidence of these mutations, there are few in vitro studies and case reports showing whether Osimertinib is effective against these rare mutations (4447).

Activation of alternative bypass or downstream pathways
Human EGFR2 gene (Her2) amplification

Activation of HER2, also known as ERBB2, triggers functional abnormalities in several downstream signaling pathways, such as the mitogen-activated protein kinase (MAPK), inosine phosphate 3-kinase (PI3K)/protein kinase B (AKT), protein kinase C (PKC), and signal transducer and transcriptional activator (STAT) pathways, resulting in uncontrolled cell proliferation (48,49). HER2 overexpression occurs in ~12% of NSCLC patients who are resistant to first- and second-generation EGFR-TKIs and usually do not co-exist with the T790M secondary mutation (50). Standard treatment for managing this drug resistance mechanism is currently not available, and there is insufficient evidence to show that existing anti-HER2 therapies are effective. The selective HER2 tyrosine kinase inhibitors poziotinib (51) and pyrotinib (52,53), and the antibody conjugate drugs T-DM1 (54) and trastuzumabe-deruxtecan (55) are potential treatment options.

MET amplification

MET is a proto-oncogene and one of the key driver genes in several types of cancer (56). The MET gene encodes c-Met [a hepatocyte growth factor (HGF) receptor], which is responsible for regulating important processes, such as cell differentiation, proliferation, migration, and apoptosis (57). Hepatocyte growth factor (HGF) binds to c-Met to phosphorylate tyrosine kinase residues in the catalytic domain; activates the downstream pathways modulated by PI3K, MAPK, and STAT3 signaling, and promotes cell transformation, cell invasion, cell proliferation, and cell cycle progression (57,58). MET amplification accounts for 2–4% of untreated NSCLC cases (59) and for 5–20% of patients with acquired drug resistance against first- and second-generation EGFR-TKIs (58,60,61). Lai et al (62) showed that an increased copy number of the MET gene is not equal to MET amplification; only MET amplification is a determinant of EGFR-TKI resistance in NSCLC patients.

Due to the crosstalk between MET and RTK (EGFR) signaling pathways (63), it has been proposed that the combination of MET-TKIs and EGFR-TKIs may be a solution for MET-driven EGFR-TKI resistance (64). After disease progression in the context of EGFR-TKI treatment, patients with MET amplification were treated with camatinib combined with gefitinib. The ORR was 29%, and the PFS was 5.5 months (65). Tepotinib combined with gefitinib prolonged the survival time compared with chemotherapy (mOS: 37.3 vs. 13.1 months) (66). Sequist et al (67) evaluated the efficacy of Osimertinib + savolitinib in two global expansion cohorts (part B and part D) of the Tatton study, and the results showed a higher response rate in subgroup B3 (previously untreated with third-generation EGFR-TKIs and positive for T790M) and subgroup D (previously untreated with third-generation EGFR-TKIs), with ORR values of 67 and 64% and median (m)PFS values of 11 and 9.1 months, respectively. Osimertinib combined with savolitinib is also a potential treatment approach (67). In addition to the above combination of treatments, telisotuzumab-vedotin combined with erlotinib (68), capmatinib combined with erlotinib (69), and emibetuzumab combined with erlotinib (70) also achieved certain benefits.

PIK3A mutations

PIK3A can induce the phosphorylation and subsequent activation of the downstream AKT signal transduction pathway and it plays a central role in regulating tumor cell growth, reproduction, migration, and apoptosis (71). The role of PIK3CA mutations in NSCLC remains contested. Some researchers consider PIK3CA mutations to be an independent risk factor for NSCLC patient survival (72), and the survival time of patients with EGFR and PIK3CA mutations treated with EGFR-TKIs was shown to be shorter than that of people with only EGFR mutations (73). However, it has also been shown that PIK3CA mutations have no significant effect on NSCLC patient survival times (74). PIK3CA mutations are a mechanism of acquired EGFR-TKI resistance in patients with EGFR-mutated lung cancer (75). The frequency of PIK3CA mutations after EGFR-TKI resistance is 2–3% (76). Preclinical studies have found that double targeting of MEK and PI3K can effectively control the proliferation of EGFR-TKI drug-resistant NSCLC cell lines (77). Alpelisib (a PI3K inhibitor) has been approved by the Food and Drug Administration (FDA) for the treatment of breast cancer (78), but it has not been applied for NSCLC after the development of resistance to TKIs.

KRAS mutations

KRAS mutations activate downstream pathways, such as the MAPK and PI3K pathways, driving the occurrence and development of tumors (79). The proportion of KRAS mutations after the development of EGFR-TKI resistance is ~1% (76). Tanaka et al (80) suggested that the mechanism underlying KRASG12C-acquired drug resistance to KRAS-TKI is related to the activation of the RAS-MAPK signals and the production of KRASY96D resistance genes. The FDA approved the KRASG12C inhibitor sotorasib in May 2021 to treat NSCLC patients with KRASG12C mutations after at least one previous systematic treatment (81).

BRAF mutations

BRAF mutations increase the activity of RAF kinase, activates downstream MEK, and regulates cell growth, proliferation, differentiation, migration, and apoptosis (82). BRAFV600E is the most common BRAF mutation, accounting for 36% of all BRAF mutations (83). BRAF mutations account for only 1% of patients with acquired drug resistance to TKIs (84). Dabrafenib combined with trametinib has been approved by the FDA for the treatment of metastatic NSCLC with BRAFV600E mutations (85).

Other rare mutations

The AXL-mediated Gas6/Axl signaling pathway is associated with tumor cell growth, metastasis, invasion, EMT, angiogenesis, drug resistance, immune regulation, and stem cell maintenance (86,87). In 2012, a study found that Axl expression was upregulated in patients with acquired drug resistance to EGFR-TKIs, and EGFR-TKI sensitivity was restored after blocking Axl (88). Thus, Axl is a promising therapeutic target for patients with acquired drug resistance. Small molecule inhibitors, monoclonal antibodies, and antibody-drug conjugates targeting Axl are currently under development (89). DS-1205 (an AXL inhibitor) combined with gefitinib (90) and BGB324 (an AXL inhibitor) combined with erlotinib (91) were evaluated, and preliminary results were promising.

PTEN negatively regulates the PTEN/PI3K/Akt signaling pathway and regulates cell growth, apoptosis, and migration (92). Studies have shown that patients with EGFR mutations with PTEN deletions have significantly shorter PFS durations than those without PTEN deletions (6 vs. 18 months) (93). According to Xun et al (92), the deletion of PTEN in lung cancer promotes the carcinogenic function of STMN1 (overexpression of which is related to tumor growth, metastasis, and poor survival) through the PI3K/AKT pathway. Other reported drug resistance mechanisms include loss of neurofibromin 1 activity (94), amplification of the CT10 homologous oncogene of v-crk avian sarcoma virus (95), a multistep mechanism involved in the insulin-like growth factor 1 receptor (IGF1R) pathway (96), and the fibroblast growth factor (FGF) 2/FGF receptor 1 (FGFR1) autocrine growth pathway (97). As these drug resistance mutations are rare, no drugs targeting them have been approved.

EGFR compound mutations and co-mutations

Compound mutations indicate the presence of more than one EGFR mutation, either common or uncommon, within the same tumor. Attili et al (98) found high heterogeneity in the incidence of compound mutations (4–26% of total EGFR mutant cases), with the variance possibly due to the different testing methods adopted, and the specific mutations considered. In various combinations, compound EGFR mutations containing either exon 21 p. L858R or exon 19 deletions were common (99). The response rate of those tumors with compound mutations to EGFR-TKIs compared with those with single mutations is contested. Rossi et al (100) found a longer mOS in the compound mutation group than in the single rare mutation group (33.6 vs. 12 months; P=0.473), whereas Jiang et al (101) concluded that patients in the single mutation group exhibited a longer mOS than those in the co-mutation group (ORR: 64.6% vs. 27.4%, P<0.001). More prospective randomized clinical trials (RCTs) are required to reconcile these differences.

A co-mutation is defined as the coexistence of an EGFR mutation along with one or more other gene mutations. The co-mutation incidence rate was 66.0% in the retrospective study of Jiang (101). Co-mutations, including TP53 (102,103), HER family genes (104), KRAS, MET, and ROS1, are typically considered to be associated with poor prognosis (105,106).

Histological transformations
Transformation to SCLC

Among the patients who did not maintain a response to EGFR-TKI treatment, 3–14% had tumors that showed morphological transformation to SCLC (61,107). Although the tumors that transformed into SCLC had persistent EGFR activation, immunohistochemical analysis showed that EGFR expression decreased sharply (108). EGFR-TKI-resistant lung adenocarcinoma and SCLC share a common clonal origin. Significant inactivation of Rb and TP53 (a common mutation of classical SCLC) was found in patients with SCLC after the development of drug resistance (108110). In addition, PIK3CA (111) mutations and TERT amplification (112) were also observed. The specific mechanisms involved in this transition and TKI resistance have not been determined. In addition to the above mutations, other studies have suggested that the transformation may be related to EMT (113,114). A retrospective analysis of this mechanism of drug resistance showed that the etoposide/cisplatin regimen is currently the most effective treatment (115). In this retrospective study, patients treated with anlotinib also achieved an ORR of 66.7% and an mPFS duration of 6.2 months. Another small-sample study reported longer PFS durations were obtained with bevacizumab or other TKIs combined with chemotherapy (116).

Transformation to lung squamous cell carcinoma (SCC)

In recent years, several cases of transformation of EGFR-mutated NSCLC to SCC have been reported (117121), and some reports indicate the association between the T790M mutation and SCC transformation (122,123). As this morphological transformation is rare, the mechanism is unclear, although it has been shown that it may be related to changes in the PI3K/AKT/mTOR pathway during EGFR-TKI therapy (124). For patients with drug-resistant lung SCC, the prognosis is usually poor, and the mOS is only ~3.5 months (120). It is difficult to choose follow-up treatments due to the low incidence; Liao et al (121) reported the case of a patient who received almonertinib for 6 months after detection of the SCC phenotype. At the time of writing the study, the patient was continuing almonertinib monotherapy and the disease was stable.

EMT

EMT is a process in which epithelial cells lose polarity and adhesion to gain increased migratory ability, and in the process exhibit a mesenchymal phenotype characterized by decreased E-cadherin and increased vimentin expression as well as stem cell-like features (125,126). EMT is considered one of the possible mechanisms of acquired drug resistance to EGFR-TKIs (127). Increased expression of Aurora kinase A (AURKA) can induce EMT and contribute to the occurrence of acquired EGFR-TKI resistance (128). Nilsson et al (129) found that activation of the YAP and FOXM1 axes serves as a driver of EMT-related EGFR-TKI resistance. It has also been confirmed that reversing EMT can restore sensitivity to EGFR-TKI drugs (116). The AURKA inhibitor alisertib can restore the sensitivity of drug-resistant cells to EGFR-TKIs and partially reverse the EMT process (130). It has also been found that Bruton's tyrosine kinase (BTK) mediates dryness and EMT characteristics, and the BTK inhibitor acalabrutinib can enhance the effect of gefitinib and Osimertinib in TKI-resistant NSCLC cells (131).

Treatment of T790M-negative tumors after the development of resistance to first- and second-generation TKIs

Regarding the aforementioned drug resistance mechanisms, although researchers have performed extensive treatment-related research, no drugs specifically developed for acquired drug resistance mechanisms have been approved and marketed given the low incidence of these causative mutations. For the first- and second-generation TKI drug-resistant T790M-negative population, platinum-containing dual-drug chemotherapy is currently recommended, but its benefits are limited (132). Other treatment options are being explored and are summarized below.

Immunotherapy

The relationships between EGFR mutations, EGFR-TKIs, and immunotherapy efficacy are contested. A meta-analysis of large RCTs showed that patients with EGFR mutations showed no significant benefit from immunotherapy (133,134). In people with PD-L1 expression levels <50%, the use of EGFR-TKI inhibitors resulted in a better PFS rate and ORR (135). However, studies have also shown that for patients with EGFR mutations, the proportion of patients with PD-L1 expression levels ≥50% increased after EGFR-TKI treatment, and the mPFS resulting from subsequent treatment with PD-1 antibodies was longer than that of patients with low PD-L1 expression (7.1 vs. 1.7 months; P=0.0033) (136). These results indicate that EGFR-TKI drugs appear to have a positive effect on the tumor microenvironment (TME).

According to the IMpower150 study, a subgroup analysis of patients following EGFR-TKI failure showed that OS benefits were obtained after addition of atrizumab; this is the only study that has confirmed OS benefits from immunotherapy after the development of EGFR-TKI resistance (137). Another ongoing phase II study also showed that the addition of atezolizumab to the bevacizumab regimen improved the disease control rate (DCR) and PFS outcome (138). In the single-arm II phase study by Lam et al (139), a 9.4-month PFS duration was obtained using a quadruple combination of atezolizumab, bevacizumab, carboplatin, and pemetrexed. A total of 42.5% of the patients were resistant to first- and second-generation EGFR-TKIs. The incidence of treatment-related adverse events was 37.5% (15/40), which is within the range of controllable adverse events. Thus, this combination appears to be a feasible treatment.

CT18 is the first prospective immunotherapy study in patients with lung adenocarcinoma with EGFR mutations. The results showed good clinical benefits (ORR=50%, mPFS=7 months, OS=23.5 months) in T790M-negative patients with acquired drug resistance after treatment with toripalimab combined with carboplatin and pemetrexed (140). A phase III RCT (TREASURE) is underway, evaluating toripalimab plus chemotherapy as second-line treatment in patients with EGFR-mutant-advanced NSCLC who were previously treated with EGFR-TKIs; patients with failed first-line EGFR-TKIs and those who did not harbor T790M mutation were enrolled. (141). ORIENT-31 was a randomized, double-blind, multicenter, phase III RCT that evaluated the efficacy and safety of the combination of sintilimab and bevacizumab for treating EGFR-mutated SCLC after EGFR-TKI treatment. The study found that patients in the quadruple drug group had prolonged mPFS (6.9 vs. 4.3 months) and median duration of efficacy (8.3 vs. 7.0 months) outcomes compared to those in the chemotherapy group (142).

A phase II RCT conducted by Hayashi et al (143) compared nivolumab (NIVO) with carboplatin + pemetrexed for treating EGFR-TKI-resistant patients; NIVO did not exhibit an advantage over chemotherapy (mPFS 1.7 months vs. 5.6 months, mOS 20.7 vs. 19.9 months). Therefore, more prospective trials are needed to verify the feasibility of immunotherapy in patients with EGFR resistance.

Treatment regimens containing pemetrexed

Pemetrexed is an anti-folic acid drug that can interfere with folic acid metabolism, resulting in aberrant DNA synthesis in tumor cells (144). A cancer registration cohort analysis from Taiwan showed that pemetrexed may be suitable as a first choice for chemotherapy in patients undergoing chemotherapy after progression with EGFR-TKI treatment (145). A 2018 meta-analysis showed that second-line drugs combined pemetrexed chemotherapy resulted in a longer PFS and OS duration than therapy with pemetrexed (146). In a phase II study, researchers compared cisplatin plus pemetrexed against pemetrexed alone in patients with drug resistance and found no significant difference in PFS and OS outcomes between the two groups. The efficacy of pemetrexed in NSCLC patients with disease progression after first-line EGFR-TKI treatment was not improved by adding cisplatin (147).

Antiangiogenic drugs

Preclinical studies have shown that vascular endothelial growth factor (VEGF) and EGFR share a common downstream signaling pathway and acquired EGFR resistance is associated with increased VEGF levels (148). In vivo and in vitro studies have confirmed that anlotinib (a small molecular multitarget tyrosine kinase inhibitor) can overcome acquired resistance to EGFR-TKIs through modulation of the FGFR1 signaling pathway (149,150). Phase II clinical trials have shown that the use of bevacizumab combined with afatinib resulted in an ORR of 22% and a PFS of 7.1 months in T790M-negative patients who developed drug resistance (151). A patient with EGFRL858R and KRASG12D mutations administered a combination of bevacizumab, camrelizumab, and pemetrexed after developing EGFR-TKI resistance achieved a benefit lasting ~17 months (152). A retrospective analysis in China revealed that the longer the duration of the previous EGFR-TKI treatment had been, the longer the PFS duration was when patients received follow-up immunotherapy combined with chemotherapy and antiangiogenic drugs (153). Due to the limitations of the above studies, additional prospective studies are needed to confirm the efficacy of antiangiogenic drugs combined with targeted therapy or immunotherapy in the future.

Resistance mechanisms and treatment progress with Osimertinib (third-generation EGFR-TKIs)

At present, Osimertinib is the only third-generation EGFR-TKI preparation that is widely used and that has been studied relatively extensively. Drug resistance mechanisms associated with first-line use of Osimertinib are similar to those associated with second-line therapy, but the proportion of patients developing resistance differs (Fig. 2). At present, the reported mechanisms of drug resistance can be divided into EGFR-dependent and EGFR-independent mechanisms (154,155). The mechanisms of EGFR-dependent drug resistance include EGFR mutations, amplification, deletion, and ligand overexpression as well as tertiary EGFR mutations, whereas EGFR-independent resistance mechanisms include activation of abnormal accessory pathways, activation of downstream pathways, and histological/phenotypic transformation (156).

EGFR-dependent drug mechanisms
c797s mutation and treatment progress

The Aura3 study revealed that 49% of the patients had T790M loss, and 14% had EGFRC797S mutations, the most common mutations acquired after the development of Osimertinib resistance (154). EGFRC797S mutations include cis (98%) and trans mutations (2%). T790M and C797S mutations that occur simultaneously in the same allele are referred to as cis mutations, and mutations that occur in different alleles are referred to as trans mutations (157,158).

Recent studies have shown that a third-generation TKI combined with a first-generation EGFR-TKI can change the expression profile of drug resistance genes in lung adenocarcinoma patients with EGFR activation mutations, and T790M and trans-C797S triple mutations (158). Brigatinib combined with cetuximab is an effective treatment strategy for these lung adenocarcinoma patients with EGFR activation mutations and T790M and cis-C797S triple mutations (158,159). Chang et al (160) reported the case of a patient with lung adenocarcinoma with triple mutations (L858R, T790M, and cis-G796s/cis-C797s). After treatment failure with brigatinib combined with cetuximab, the patient responded to the combination of brigatinib, Osimertinib, and bevacizumab. Other reported treatments include Osimertinib combined with anlotinib (161), chemotherapy combined with antiangiogenic agents (162), and apatinib combined with afatinib (163). At present, fourth-generation EGFR-TKIs targeting drug-resistant T790M mutations are under development, although no drug has been approved. Fourth-generation EGFR-TKIs in clinical trials and currently undergoing research and development and are described in subsequent sections.

Other gene mutations and treatment progress

In a study where next-generation sequencing (NGS) analysis was performed on 93 samples obtained after the development of Osimertinib resistance, EGFRG796/C797, L792, and L718/G719 mutations were found in 24.7, 10.8, and 9.7% of cases, respectively (164). G724 mutations were also reported in some studies (165,166). At present, a drug that targets the aforementioned mutated genes is not available. In vitro studies have confirmed that L792 is still sensitive to gefitinib (167) and that tumors with the L718Q mutations remain sensitive to icotinib (168). In a patient with the EGFRG724S/19del mutation after second-line Osimertinib resistance, PFS was achieved after using afatinib for 3.8 months (169).

EGFR-independent mechanisms of drug resistance and treatment progress

EGFR-independent mechanisms of drug resistance primarily include activation of abnormal accessory and downstream pathways and histological/phenotypic transformation. Most of the mechanisms of Osimertinib drug resistance are the same as those of first- and second-generation TKIs.

MET amplification

Leonetti et al (155) demonstrated that the incidence of MET amplification after Osimertinib first-line treatment resistance was 7–15%, and that of MET amplification after second-line treatment resistance was 5–50%. In the B1 expansion cohort of the TATTON study (previously treated with third-generation EGFR-TKIs), a 5.5-month PFS duration was obtained with Osimertinib combined with sevotinib (67), and other therapeutic developments have been described. Zhang et al (170) found that MET amplification weakened the response of lung tumors to immunotherapy by inhibiting the STING signaling pathway, and that MET inhibitors combined with immune checkpoint inhibitors overcame this drug resistance; however, this information needs to be confirmed by further prospective studies. Amivantamab (JNJ-61186372) is a bispecific antibody against EGFR and MET that has been approved for the treatment of patients with EGFR exon 20 insertion mutations (171). Amivantamab can inhibit both the phosphorylation of EGFR and MET and the activation of downstream signals and has potent antibody-dependent cell-mediated cytotoxic effects (171,172). Amivantamab is inhibited by double targeting, which showed an inhibitory effect on several types of mutations secondary to EGFR-TKI resistance (C797S mutations, MET amplification, previous resistance to Osimertinib) (173).

HER2 amplification and PIK3CA mutation

HER2 amplification is one of the mechanisms of Osimertinib resistance (174). The AURA3 study showed that HER2 amplification was detected in 5% (4 out of 73) of second-line Osimertinib-resistant patients (157). In the FLAURA study, HER2 amplification occurred in 2% of first-line Osimertinib-resistant patients (175). PIK3CA mutations occur in patients with Osimertinib resistance, and T790M mutations may be retained or lost. Moreover, PIK3CA mutations showed different incidences in different studies (176,177).

Changes in other bypass pathways

Abnormal FGFR expression can lead to the activation of the FGFR cancer-related signaling pathway effectors (PI3K/AKT, STAT, and MAPK) and affect cell proliferation, survival, metabolism, and migration as well as the cell cycle (178). In vitro studies found that hypoxia can lead to acquired resistance to EGFR-TKIs by increasing the expression of FGFR1 (179). The combined use of EGFR-TKIs and FGFR1 inhibitors (BGJ398) may represent a potential therapeutic strategy for the management of NSCLC (179,180). Upregulation of IGF1R is one of the mechanisms of drug resistance to EGFR-TKIs, including Osimertinib (181183). In cells resistant to Osimertinib that exhibit low levels of AXL expression, short-term IGF-1R inhibition combined with Osimertinib can eradicate tumors and prevent regrowth (184).

Histology/bypass transformation

Histological transformation from lung adenocarcinoma to SCLC, SCC and EMT was also observed in patients with Osimertinib resistance (177,185). Platinum-containing dual-drug chemotherapy is still recommended for these patients.

Treatments available after the development of Osimertinib resistance

Fourth-generation EGFR-TKI inhibition

Drug resistance to third-generation targeted drugs (Osimertinib) is a dilemma faced by several lung cancer patients who receive targeted therapy. To date, the FDA has not approved targeted therapy for progression after treatment with Osimertinib. Thus, the research and development of fourth-generation EGFR-TKI drugs have become a focus recently, and several drugs have shown good results in clinical trials.

EAI045 is the first fourth-generation EGFR-TKI drug. EAI045 combined with cetuximab significantly reduced the tumor size in mice carrying L858R/T790M/C797S mutations, but no obvious effect was observed with single-agent use (186). To improve the activity of EAI045 and the ability to use the drug as a single agent, To et al (187) modified EAI045 and obtained a new allosteric inhibitor, JBJ-04-12502, which exhibited higher efficacy, lower toxicity, and efficacy against EGFR mutations compared with the parent compound. JBJ-04-12502 inhibits the triple-drug resistance mechanism of patients with L858R/T790M/C797S mutations, the double mutation of EGFR-T70M, and the L858R drug resistance mutation. The therapeutic effect of JBJ-04-12502 in combination with Osimertinib is more potent, although it is still in the research and development stage (188). CH7233163 is a fourth-generation EGFR-TKI inhibitor developed by Roche Chugai Pharmaceuticals for patients with a Del19 mutation. After the application of CH7233163 in Del19/L858R/T790M, L858R/T790M mutant, and Del19 mice, a substantial reduction in tumor volume was observed (189). The prospect of CH7233163 appears to be more promising than that of JBJ-04-12502.

Both BLU-945 and BLU-701 are fourth-generation EGFR-TKI inhibitors developed by Blueprint Medicines. Both can resist EGFR activation mutations (del19, 21L858R) as well as T790M and C797S drug resistance mutation activity (190,191). BLU-945 combined with Osimertinib or gefitinib provided a more significant tumor elimination effect in n NSCLC mouse model (192). BLU-701 also exhibited intracranial antitumor activity, and both BLU-701 alone and in combination with BLU-945 showed strong antitumor activity (193).

TQB3804 is a fourth-generation oral EGFR-targeted drug developed by the Zhengda Tianqing Pharmaceutical Group. It not only solves Osimertinib resistance caused by d746750 (19del)/T790M/C797S and L858R/T790M/C797S, but is also effective against the d746-750/T790M and L858R/T790M double mutations associated with resistance to first- and second-generation TKIs (194). Correlative clinical trials (NCT04128085 and NCT04180150) are currently underway (195).

BBT-176 is an innovative EGFR-TKI developed by Bridge Biotherapeutics in Korea. BBT-176 showed strong anticancer activity in xenotransplantation animal models carrying triple mutations Del19/T790M/C797S and L858R/T790M/C797S (196). Moreover, BBT-176 in combination with the anti-EGFR antibody cetuximab showed significantly enhanced activity (197).

The EGFR and MET bispecific antibody amivantamab is also classified as a fourth-generation EGFR-TKI. This drug is effective against the EGFR exon 20 insertion mutation (primary drug resistance mutation) (198), C797S mutation, and MET amplification after the acquisition of Osimertinib resistance. Amivantamab combined with Lazertinib effectively overcomes Osimertinib resistance. In 45 patients with Osimertinib resistance, the disease control rate reached 60% with a median follow-up period of 4 months (199). All the above drugs, except for amivantamab, which has been approved for the treatment of the NSCLC EGFR exon 20 insertion mutation, remain in different stages of research and development or clinical trials. Thus, it will be several years before these drugs are available for clinical use.

U3-1402

U3-1402 is an antibody-drug conjugate (ADC) developed by Daiichi Co., Ltd., which consists of patritumab acting on HER3 antibodies and the cytotoxic drug DX-8951 (Exitecan, a topoisomerase inhibitor). U3-1402 is effective against different drug resistance mechanisms against EGFR-TKIs, as demonstrated in phase I trial results published in 2019 (200). Another phase I study included 57 patients with EGFR-TKI resistance. The DCR was 68% in 44 patients who had previously received Osimertinib and platinum-containing chemotherapy. The mPFS reached 8.2 months (201). ADC drugs have considerable potential for solving the problem of resistance to Osimertinib.

Osimertinib rechallenge

Soo et al (202) showed no benefit regarding PFS in patients with T790M-positive NSCLC when treated with Osimertinib combined with bevacizumab compared with Osimertinib monotherapy. However, in a small-sample retrospective study, after the development of Osimertinib resistance, Osimertinib combined with bevacizumab showed certain benefits. The study compared the efficacy and safety of Osimertinib combined with bevacizumab against chemotherapy combined with bevacizumab in patients with Osimertinib resistance. The mPFS duration of the two groups was 7.0 vs. 4.9 months, and the mOS was 12.6 vs. 7.1 months, respectively; the difference was statistically significant (203).

The COMPEL study was a randomized, double-blind phase III clinical study that evaluated the efficacy and safety of chemotherapy plus Osimertinib or chemotherapy plus placebo in advanced NSCLC patients with progressive EGFR mutations after first-line treatment with Osimertinib. The study is currently underway and will be published in September 2024 (204).

Immunotherapy

The ORIENT-31 study included patients who were T790M-negative after first- and second-generation EGFR-TKI treatment and patients who received third-generation EGFR-TKI treatment. The results showed that the PFS duration was significantly prolonged in patients treated with sintilimab combined with bevacizumab and chemotherapy compared with that of patients treated with chemotherapy alone (142). This study was the first to confirm that PD-1 inhibitors combined with antivascular drugs and chemotherapy significantly improved PFS outcomes in EGFR-mutant non-squamous NSCLC patients with progression after EGFR-TKI treatment, providing options for the follow-up treatment of drug-resistant patients after targeted treatment.

In a single-arm phase II study of patients administered a quadruple combination of atezolizumab, bevacizumab, carboplatin, and pemetrexed, the PFS duration was 9.4 months; 57.5% of these patients had been treated with Osimertinib (139). The IMpower150 study is currently the only randomized prospective phase III clinical trial that demonstrated OS benefits in NSCLC patients in an EGFR-sensitive mutation subgroup (137), showing that the addition of atezolizumab to the standard therapy of bevacizumab and chemotherapy represents a novel treatment option.

Other treatment options

According to the subgroup analysis of the ALTER0303 study (205), patients with EGFR mutations exhibited PFS and OS benefits following treatment with anlotinib. Zhou et al (161) also reported on the case of a patient with a cisEGFRT790M-C797S mutation after Osimertinib resistance who was treated with amlotinib combined with Osimertinib and achieved partial remission that persisted for 9 months. The use of afatinib combined with bevacizumab has also been reported; it improved the patient's symptoms and was continued as the treatment for 12 months (206).

Summary and future perspectives

The 21st century is the era of targeted cancer treatment, and several promising options for lung adenocarcinoma patients are available. Although novel treatments provide survival benefits to varying degrees, the problem of drug resistance inevitably leads to disease progression. It has been demonstrated that tumors become increasingly molecularly heterogeneous following targeted therapy (5,207). There is a large body of literature implicating intratumoral heterogeneity as a major driver of drug resistance (208,209). NGS and single-cell RNA sequencing (scRNA-seq) are used to study the genetic and molecular characteristics of tumor development at various stages (210), revealing the heterogeneity of tumor cells and monitoring the progress of tumor development.

Maynard et al (211) performed scRNA-seq of metastatic lung adenocarcinoma using 49 clinical biopsies obtained from 30 patients before and during targeted therapy and found that the components of the TME differ at the stages of TKI naïve, residual disease (RD), and progression. A more inflammatory phenotype was observed in RD following targeted therapy that was characterized by T cell infiltration and decreased infiltration of immunosuppressive macrophages (211). In addition, various immunosuppressive cell states characterize progressive disease. Therefore, researchers have proposed that if deployed at the appropriate time, treatments that target a specific cell state or prevent further adaptation may help improve patient survival by constraining continued tumor evolution toward complete drug resistance (211).

In recent years, modified T-cell therapy, particularly those that use chimeric antigen receptor (CAR)-T cells, has attracted growing interest in various solid tumors with the clinical success of chimeric antigen receptor CAR T-cell therapy in hematological malignancies (212,213). The CAR T strategy aims to isolate T cells from the peripheral blood of patients or other donors and genetically engineer T cells with CAR structures to equip them with the capability of recognizing specific antigens on the tumor cell surface. After infusion back into patients, these ‘super’ T cells recognize and eliminate the cancer cells that express specific target antigens (214). The major difference between CAR T cells and tumor-specific T cells is that the former cells are not limited by the major histocompatibility complex (215,216). It is critical to identify targeted tumor-associated antigens (TAAs). Ideal TAAs are highly and selectively expressed in solid tumors, but weakly expressed or absent in normal tissues (217).

The lung adenocarcinoma-associated TAAs currently being investigated in clinical trials on CAR-T cells include mesothelin (MSLN), mucin 1 (MUC1), carcinoembryonic antigen (CEA) EGFR, PD-L1, prostate stem cell antigen (PSCA), disialoganglioside GD2 (GD2), and c-Met (218222).

For EGFR-mutated LUAD, EGFR is definitely an optimum TAA. A phase I clinical trial of EGFR-targeting CAR T-cell therapy to treat patients with EGFR-positive relapsed/refractory NSCLC achieved initial success (NCT01869166). The results showed that none of the patients exhibited significant toxic side effects after anti-EGFR CAR-T-cell therapy, 2 patients achieved partial remission, and 5 patients had stable disease for 2–8 months (223). This result provides preliminary evidence that EGFR-targeting CAR T therapy is safe and feasible in certain cases of relapsed/refractory NSCLC. Currently, there are two ongoing phase I clinical trials in patients with lung cancer on C-X-C chemokine receptor type 5 modified EGFR-targeted CAR-T cells (NCT05060796 and NCT04153799).

Although CAR-T-cell therapies have achieved great success in hematological malignancies, the study of lung cancer is still in the early exploration stage. Numerous clinical trials have progressed slowly and have achieved very limited efficacy, and several challenges and hurdles remain, such as on-target/off-tumor toxicity, CAR-T cell trafficking and infiltration into the tumor, TME heterogeneity, immune suppression, and cytokine release syndrome (224226). Recently, Vasic et al (227) found that allogeneic double-negative CAR-T cells inhibit tumor growth with no off-tumor toxicity in either a lung cancer xenograft model or B-cell acute lymphoblastic leukemia (B-ALL) was observed. Therefore, double-negative CAR-T cells may serve as a patient-accessible form of CAR-T cell therapy.

Due to China's large population and relatively high EGFR mutation rate, the identification of the best treatment after the development of EGFR-TKI resistance has become an urgent problem. EGFR-TKIs have been continuously developed and are currently in their fourth generation of iteration, and this process is accompanied by the continuous optimization of pharmacological mechanisms, the emergence of novel drug resistance mechanisms, and the development of solutions to these new drug resistance mechanisms. Although it may take considerably more research to conquer cancer, significant levels of drug research and development remain ongoing.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Wu Jieping Medical Foundation of China (grant no. 320.6750.2021-22-8).

Availability of data and materials

Not applicable.

Authors' contributions

RS conceived the article, performed the literature search and drafted the manuscript. ZH and YZ contributed their knowledge on this topic and were involved in planning the structure of this review. BJ made critical modifications to the content within the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Cao M, Li H, Sun D and Chen W: Cancer burden of major cancers in China: A need for sustainable actions. Cancer Commun (Lond). 40:205–210. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Camidge DR, Doebele RC and Kerr KM: Comparing and contrasting predictive biomarkers for immunotherapy and targeted therapy of NSCLC. Nat Rev Clin Oncol. 16:341–355. 2019. View Article : Google Scholar

4 

Lee CK, Davies L, Wu YL, Mitsudomi T, Inoue A, Rosell R, Zhou C, Nakagawa K, Thongprasert S, Fukuoka M, et al: Gefitinib or erlotinib vs chemotherapy for EGFR mutation-positive lung cancer: Individual patient data meta-analysis of overall survival. J Natl Cancer Inst. 109:2017. View Article : Google Scholar

5 

Rotow J and Bivona TG: Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer. 17:637–658. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Kobayashi Y and Mitsudomi T: Not all epidermal growth factor receptor mutations in lung cancer are created equal: Perspectives for individualized treatment strategy. Cancer Sci. 107:1179–1186. 2016. View Article : Google Scholar

7 

Shi Y, Au JS, Thongprasert S, Srinivasan S, Tsai CM, Khoa MT, Heeroma K, Itoh Y, Cornelio G and Yang PC: A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER). J Thorac Oncol. 9:154–162. 2014. View Article : Google Scholar

8 

Shi Y, Li J, Zhang S, Wang M, Yang S, Li N, Wu G, Liu W, Liao G, Cai K, et al: Molecular Epidemiology of EGFR mutations in asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology-Mainland China Subset analysis of the PIONEER study. PLoS One. 10:e01435152015. View Article : Google Scholar : PubMed/NCBI

9 

Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, Sunpaweravong P, Han B, Margono B, Ichinose Y, et al: Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 361:947–957. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Wu YL, Zhou C, Liam CK, Wu G, Liu X, Zhong Z, Lu S, Cheng Y, Han B, Chen L, et al: First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: Analyses from the phase III, randomized, open-label, ENSURE study. Ann Oncol. 26:1883–1889. 2015. View Article : Google Scholar

11 

Schuler M, Paz-Ares L, Sequist LV, Hirsh V, Lee KH, Wu YL, Lu S, Zhou C, Feng J, Ellis SH, et al: First-line afatinib for advanced EGFRm+ NSCLC: Analysis of long-term responders in the LUX-Lung 3, 6, and 7 trials. Lung Cancer. 133:10–19. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Wu YL, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, Tsuji F, Linke R, Rosell R, Corral J, et al: Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): A randomised, open-label, phase 3 trial. Lancet Oncol. 18:1454–1466. 2017. View Article : Google Scholar

13 

Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, 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. 382. pp. 41–50. 2020, View Article : Google Scholar : PubMed/NCBI

14 

Shi YK, Wang L, Han BH, Li W, Yu P, Liu YP, Ding CM, Song X, Ma ZY, Ren XL, et al: First-line icotinib versus cisplatin/pemetrexed plus pemetrexed maintenance therapy for patients with advanced EGFR mutation-positive lung adenocarcinoma (CONVINCE): A phase 3, open-label, randomized study. Ann Oncol. 28:2443–2450. 2017. View Article : Google Scholar

15 

Rebuzzi SE, Alfieri R, La Monica S, Minari R, Petronini PG and Tiseo M: Combination of EGFR-TKIs and chemotherapy in advanced EGFR mutated NSCLC: Review of the literature and future perspectives. Crit Rev Oncol Hematol. 146:1028202020. View Article : Google Scholar

16 

Huang L and Fu L: Mechanisms of resistance to EGFR tyrosine kinase inhibitors. Acta Pharm Sin B. 5:390–401. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, Shepherd FA, He Y, Akamatsu H, Theelen WS, et al: Osimertinib or platinum-pemetrexed in EGFR T790M-Positive lung cancer. N Engl J Med. 376:629–640. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Thress KS, Paweletz CP, Felip E, Cho BC, 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. 21:560–562. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Wang S, Tsui ST, Liu C, Song Y and Liu D: EGFR C797S mutation mediates resistance to third-generation inhibitors in T790M-positive non-small cell lung cancer. J Hematol Oncol. 9:592016. View Article : Google Scholar

20 

Lim SM, Syn NL, Cho BC and Soo RA: Acquired resistance to EGFR targeted therapy in non-small cell lung cancer: Mechanisms and therapeutic strategies. Cancer Treat Rev. 65:1–10. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Nagano T, Tachihara M and Nishimura Y: Mechanism of resistance to epidermal growth factor receptor-tyrosine kinase inhibitors and a potential treatment strategy. Cells. 7:2122018. View Article : Google Scholar

22 

Park K, Tan EH, O'Byrne K, Zhang L, Boyer M, Mok T, Hirsh V, Yang JC, Lee KH, Lu S, et al: Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): A phase 2B, open-label, randomised controlled trial. Lancet Oncol. 17:577–589. 2016. View Article : Google Scholar

23 

Jänne PA, Ou SI, Kim DW, Oxnard GR, Martins R, Kris MG, Dunphy F, Nishio M, O'Connell J, Paweletz C, et al: Dacomitinib as first-line treatment in patients with clinically or molecularly selected advanced non-small-cell lung cancer: A multicentre, open-label, phase 2 trial. Lancet Oncol. 15:1433–1441. 2014. View Article : Google Scholar

24 

Wu SG, Liu YN, Tsai MF, Chang YL, Yu CJ, Yang PC, Yang JC, Wen YF and Shih JY: The mechanism of acquired resistance to irreversible EGFR tyrosine kinase inhibitor-afatinib in lung adenocarcinoma patients. Oncotarget. 7:12404–12413. 2016. View Article : Google Scholar

25 

Cabanero M, Sangha R, Sheffield BS, Sukhai M, Pakkal M, Kamel-Reid S, Karsan A, Ionescu D, Juergens RA, Butts C and Tsao MS: Management of EGFR-mutated non-small-cell lung cancer: Practical implications from a clinical and pathology perspective. Curr Oncol. 24:111–119. 2017. View Article : Google Scholar

26 

Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, Chawla A, Rosell R, Corral J, Migliorino MR, et al: Updated overall survival in a randomized study comparing dacomitinib with gefitinib as first-line treatment in patients with advanced non-small-cell lung cancer and EGFR-Activating mutations. Drugs. 81:257–266. 2021. View Article : Google Scholar : PubMed/NCBI

27 

Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, Dechaphunkul A, Imamura F, Nogami N, Kurata T, et al: Osimertinib in untreated EGFR-Mutated advanced non-small-cell lung cancer. N Engl J Med. 378:113–125. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Wu YL, Ahn MJ, Garassino MC, Han JY, Katakami N, Kim HR, Hodge R, Kaur P, Brown AP, Ghiorghiu D, et al: CNS efficacy of osimertinib in patients with T790M-Positive advanced non-small-cell lung cancer: Data from a Randomized phase III Trial (AURA3). J Clin Oncol. 36:2702–2709. 2018. View Article : Google Scholar

29 

Reungwetwattana T, Nakagawa K, Cho BC, Cobo M, Cho EK, Bertolini A, Bohnet S, Zhou C, Lee KH, Nogami N, et al: CNS response to osimertinib versus standard epidermal growth factor receptor tyrosine kinase inhibitors in patients with untreated EGFR-Mutated advanced non-small-cell lung cancer. J Clin Oncol. Aug 28–2018.(Epub ahead of print). View Article : Google Scholar

30 

Lu S, Wang Q, Zhang G, Dong X, Yang CT, Song Y, Chang GC, Lu Y, Pan H, Chiu CH, et al: Efficacy of aumolertinib (HS-10296) in patients with advanced EGFR T790M+ NSCLC: Updated post-national medical products administration approval results from the APOLLO registrational trial. J Thorac Oncol. 17:411–422. 2022. View Article : Google Scholar

31 

Lu S, Wang Q, Zhang G, Dong X, Yang C, Song Y, Chang GC, LU Y, Pan H, Chiu CH, et al: 1208P Final results of APOLLO study: Overall survival (OS) of aumolertinib in patients with pretreated EGFR T790M-positive locally advanced or metastatic non-small cell lung cancer (NSCLC). Ann Oncol. 32:S9622021. View Article : Google Scholar

32 

Shi Y, Hu X, Zhang S, Lv D, Wu L, Yu Q, Zhang Y, Liu L, Wang X, Cheng Y, et al: Efficacy, safety, and genetic analysis of furmonertinib (AST2818) in patients with EGFR T790M mutated non-small-cell lung cancer: A phase 2b, multicentre, single-arm, open-label study. Lancet Respir Med. 9:829–839. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Deeks ED: Furmonertinib: First approval. Drugs. 81:1775–1780. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Ahn MJ, Han JY, Kim SW, Ki Hyeong Lee5, Kim DW, Lee YG, Cho EK, Lee GW, Lee JS, Kim JH, et al: Lazertinib, a 3rd generation EGFR-TKI, in patients with EGFR-TKI resistant NSCLC: Updated results of phase I/II Study. Abstract #9037. May 31-June 4. 2019.

35 

Kim SW, Ahn MJ, Han JY, Lee KH, Cho EK, Lee YG, Kim DW, Kim JH, Lee JS, Lee GW, et al: Intracranial anti-tumor activity of lazertinib in patients with advanced NSCLC who progressed after prior EGFR TKI therapy: Data from a phase I/II study. Am Soc Clin Oncol. 38:95712020. View Article : Google Scholar

36 

Dhillon S: Lazertinib: First approval. Drugs. 81:1107–1113. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Kim ES: Olmutinib: First global approval. Drugs. 76:1153–1157. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Kim DW, Lee DH, Han JY, Lee J, Cho BC, Kang JH, Lee KH, Cho EK, Kim JS, Min YJ, et al: Safety, tolerability, and anti-tumor activity of olmutinib in non-small cell lung cancer with T790M mutation: A single arm, open label, phase 1/2 trial. Lung Cancer. 135:66–72. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Park K, Jӓnne PA, Kim DW, Han JY, Wu MF, Lee JS, Kang JH, Lee DH, Cho BC, Yu CJ, et al: Olmutinib in T790M-positive non-small cell lung cancer after failure of first-line epidermal growth factor receptor-tyrosine kinase inhibitor therapy: A global, phase 2 study. Cancer. 127:1407–1416. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Tan DS, Leighl NB, Riely GJ, Yang JC, Sequist LV, Wolf J, Seto T, Felip E, Aix SP, Jonnaert M, et al: Safety and efficacy of nazartinib (EGF816) in adults with EGFR-mutant non-small-cell lung carcinoma: A multicentre, open-label, phase 1 study. Lancet Respir Med. 8:561–572. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Costa DB, Schumer ST, Tenen DG and Kobayashi S: Differential responses to erlotinib in epidermal growth factor receptor (EGFR)-mutated lung cancers with acquired resistance to gefitinib carrying the L747S or T790M secondary mutations. J Clin Oncol. 26:1182–1184; author reply 1184–1186. 2008. View Article : Google Scholar

42 

Balak MN, Gong Y, Riely GJ, Somwar R, Li AR, Zakowski MF, Chiang A, Yang G, Ouerfelli O, Kris MG, et al: Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clin Cancer Res. 12:6494–6501. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Bean J, Riely GJ, Balak M, Marks JL, Ladanyi M, Miller VA and Pao W: Acquired resistance to epidermal growth factor receptor kinase inhibitors associated with a novel T854A mutation in a patient with EGFR-mutant lung adenocarcinoma. Clin Cancer Res. 14:7519–7525. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Grolleau E, Haddad V, Boissière L, Falchero L and Arpin D: Clinical efficacy of osimertinib in a patient presenting a double EGFR L747S and G719C mutation. J Thorac Oncol. 14:e151–e153. 2019. View Article : Google Scholar

45 

Chiba M, Togashi Y, Bannno E, Kobayashi Y, Nakamura Y, Hayashi H, Terashima M, De Velasco MA, Sakai K, Fujita Y, et al: Efficacy of irreversible EGFR-TKIs for the uncommon secondary resistant EGFR mutations L747S, D761Y, and T854A. BMC Cancer. 17:2812017. View Article : Google Scholar : PubMed/NCBI

46 

Zhu Y, Tang J, Li X, Qin T and Wei Y: Durable response to osimertinib in a Chinese patient with metastatic lung adenocarcinoma harboring a rare EGFR L858R/D761Y compound mutation. Onco Targets Ther. 13:10447–10451. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Zhang L, Yang X, Ming Z, Shi J, Lv X, Li W, Yuan B, Chen Y, Liu B, Qin K, et al: Molecular characteristics of the uncommon EGFR Exon 21 T854A Mutation and response to osimertinib in patients with non-small cell lung cancer. Clin Lung Cancer. 23:311–319. 2022. View Article : Google Scholar : PubMed/NCBI

48 

Patil T, Mushtaq R, Marsh S, Azelby C, Pujara M, Davies KD, Aisner DL, Purcell WT, Schenk EL, Pacheco JM, et al: Clinicopathologic characteristics, treatment outcomes, and acquired resistance patterns of atypical EGFR mutations and HER2 alterations in stage IV non-small-cell lung cancer. Clin Lung Cancer. 21:e191–e204. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Riudavets M, Sullivan I, Abdayem P and Planchard D: Targeting HER2 in non-small-cell lung cancer (NSCLC): A glimpse of hope? An updated review on therapeutic strategies in NSCLC harbouring HER2 alterations. ESMO Open. 6:1002602021. View Article : Google Scholar : PubMed/NCBI

50 

Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E, Ohashi K, Janjigian YY, Spitzler PJ, Melnick MA, et al: HER2 amplification: A potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov. 2:922–933. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Elamin YY, Robichaux JP, Carter BW, Altan M, Gibbons DL, Fossella FV, Lam VK, Patel AB, Negrao MV, Le X, et al: Poziotinib for patients With HER2 Exon 20 mutant non-small-cell lung cancer: Results from a phase II Trial. J Clin Oncol. 40:702–709. 2022. View Article : Google Scholar

52 

Song Z, Lv D, Chen SQ, Huang J, Li Y, Ying S, Wu X, Hua F, Wang W, Xu C, et al: Pyrotinib in patients with HER2-Amplified advanced non-small cell lung cancer: A prospective, multicenter, single-arm trial. Clin Cancer Res. 28:461–467. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Zhou C, Li X, Wang Q, Gao G, Zhang Y, Chen J, Shu Y, Hu Y, Fan Y, Fang J, et al: Pyrotinib in HER2-Mutant advanced lung adenocarcinoma after platinum-based chemotherapy: A multicenter, open-label, single-arm, phase II Study. J Clin Oncol. 38:2753–2761. 2020. View Article : Google Scholar

54 

Li BT, Shen R, Buonocore D, Olah ZT, Ni A, Ginsberg MS, Ulaner GA, Offin M, Feldman D, Hembrough T, et al: Ado-Trastuzumab emtansine for patients with HER2-Mutant lung cancers: Results from a phase II basket trial. J Clin Oncol. 36:2532–2537. 2018. View Article : Google Scholar

55 

Li BT, Smit EF, Goto Y, Nakagawa K, Udagawa H, Mazières J, Nagasaka M, Bazhenova L, Saltos AN, Felip E, et al: Trastuzumab deruxtecan in HER2-Mutant non-small-cell lung cancer. N Engl J Med. 386:241–251. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Koch JP, Aebersold DM, Zimmer Y and Medová M: MET targeting: Time for a rematch. Oncogene. 39:2845–2862. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Pasquini G and Giaccone G: C-MET inhibitors for advanced non-small cell lung cancer. Expert Opin Investig Drugs. 27:363–375. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al: MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 316:1039–1043. 2007. View Article : Google Scholar : PubMed/NCBI

59 

Bean J, Brennan C, Shih JY, Riely G, Viale A, Wang L, Chitale D, Motoi N, Szoke J, Broderick S, et al: MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci USA. 104:20932–20937. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, Bergethon K, Shaw AT, Gettinger S, Cosper AK, et al: Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 3:75ra262011. View Article : Google Scholar : PubMed/NCBI

61 

Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, Kris MG, Miller VA, Ladanyi M and Riely GJ: 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. 19:2240–2247. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Lai GG, Lim TH, Lim J, Liew PJ, Kwang XL, Nahar R, Aung ZW, Takano A, Lee YY, Lau DP, et al: Clonal MET amplification as a determinant of tyrosine kinase inhibitor resistance in epidermal growth factor receptor-mutant non-small-cell lung cancer. J Clin Oncol. 37:876–884. 2019. View Article : Google Scholar

63 

Dulak AM, Gubish CT, Stabile LP, Henry C and Siegfried JM: HGF-independent potentiation of EGFR action by c-Met. Oncogene. 30:3625–3635. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Dong Y, Xu J, Sun B, Wang J and Wang Z: MET-Targeted therapies and clinical outcomes: A systematic literature review. Mol Diagn Ther. 26:203–227. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Wu YL, Zhang L, Kim DW, Liu X, Lee DH, Yang JC, Ahn MJ, Vansteenkiste JF, Su WC, 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. 36:3101–3109. 2018. View Article : Google Scholar

66 

Wu YL, Cheng Y, Zhou J, Lu S, Zhang Y, Zhao J, Kim DW, Soo RA, Kim SW, 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. 8:1132–1143. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Sequist LV, Han JY, Ahn MJ, Cho BC, Yu H, Kim SW, Yang JC, Lee JS, Su WC, 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. 21:373–386. 2020. View Article : Google Scholar

68 

Camidge D, Barlesi F, Goldman J, Morgensztern D, Heist R, Vokes E, Spira A, Angevin E, Su W, Hong D, Strickler J, Motwani M, Sun Z, et al: MA14. 03 EGFR M+ Subgroup of Phase 1b study of telisotuzumab vedotin (Teliso-V) plus erlotinib in c-Met+ non-small cell lung cancer. J Thor Oncol. 14:S305–S306. 2019. View Article : Google Scholar

69 

McCoach CE, Yu A, Gandara DR, Riess JW, Vang DP, Li T, Lara PN, Gubens M, Lara F, Mack PC, et al: Phase I/II study of capmatinib plus erlotinib in patients with MET-positive non-small-cell lung cancer. JCO Precis Oncol. 1:PO.20.00279. 2021.

70 

Camidge DR, Moran T, Demedts I, Grosch H, Mileham K, Molina J, Juan-Vidal O, Bepler G, Goldman JW, Park K, et al: A Randomized, open-label phase II study evaluating emibetuzumab plus erlotinib and emibetuzumab monotherapy in MET immunohistochemistry positive NSCLC patients with acquired resistance to erlotinib. Clin Lung Cancer. 23:300–310. 2022. View Article : Google Scholar : PubMed/NCBI

71 

Alqahtani A, Ayesh HSK and Halawani H: PIK3CA gene mutations in solid malignancies: Association with clinicopathological parameters and prognosis. Cancers (Basel). 12:932019. View Article : Google Scholar

72 

Wang Y, Wang Y, Li J, Li J and Che G: Clinical significance of PIK3CA gene in non-small-cell lung cancer: A systematic review and meta-analysis. Biomed Res Int. 2020:36082412020.

73 

Qiu X, Wang Y, Liu F, Peng L, Fang C, Qian X, Zhang X, Wang Q, Xiao Z, Chen R, et al: Survival and prognosis analyses of concurrent PIK3CA mutations in EGFR mutant non-small cell lung cancer treated with EGFR tyrosine kinase inhibitors. Am J Cancer Res. 11:3189–3200. 2021.PubMed/NCBI

74 

Song Z, Yu X and Zhang Y: Mutation and prognostic analyses of PIK3CA in patients with completely resected lung adenocarcinoma. Cancer Med. 5:2694–2700. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Engelman JA: Targeting PI3K signalling in cancer: Opportunities, challenges and limitations. Nat Rev Cancer. 9:550–562. 2009. View Article : Google Scholar : PubMed/NCBI

76 

Westover D, Zugazagoitia J, Cho BC, Lovly CM and Paz-Ares L: Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol. 29 (Suppl 1):i10–i19. 2018. View Article : Google Scholar

77 

Qu GP, Shi M, Wang D, Wu JH, Wang P, Gong ML and Zhang ZJ: Dual targeting of MEK and PI3K effectively controls the proliferation of human EGFR-TKI resistant non-small cell lung carcinoma cell lines with different genetic backgrounds. BMC Pulm Med. 21:2082021. View Article : Google Scholar : PubMed/NCBI

78 

Markham AJD: Alpelisib: First global approval. Drugs. 79:1249–1253. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, Italiano A, Schuler M, Borghaei H, Barlesi F, et al: Sotorasib for lung cancers with KRAS p.G12C Mutation. N Engl J Med. 384:2371–2381. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, Michel AG, Syed MU, Fella KA, Sakhi M, et al: Clinical acquired resistance to KRASG12C inhibition through a Novel KRAS Switch-II pocket mutation and polyclonal alterations converging on RAS-MAPK Reactivation. Cancer Discov. 11:1913–1922. 2021. View Article : Google Scholar : PubMed/NCBI

81 

Zhang SS; Nagasaka MJLCT and Therapy, : Spotlight on Sotorasib (AMG 510) for KRASG12C positive non-small cell lung cancer. Lung Cancer (Auckl). 12:115–122. 2021.PubMed/NCBI

82 

Pratilas CA, Hanrahan AJ, Halilovic E, Persaud Y, Soh J, Chitale D, Shigematsu H, Yamamoto H, Sawai A, Janakiraman M, et al: Genetic predictors of MEK dependence in non-small cell lung cancer. Cancer Res. 68:9375–9383. 2008. View Article : Google Scholar : PubMed/NCBI

83 

Ai X, Li Y, Chen R, Gu D and Mao Y: P59. 07 mutation profile of BRAF in Chinese non-small cell lung cancer patients. J Thorac Oncol. 16:S11492021. View Article : Google Scholar

84 

Ohashi K, Sequist LV, Arcila ME, Moran T, Chmielecki J, Lin YL, Pan Y, Wang L, de Stanchina E, Shien K, et al: Lung cancers with acquired resistance to EGFR inhibitors occasionally harbor BRAF gene mutations but lack mutations in KRAS, NRAS, or MEK1. Proc Natl Acad Sci USA. 109:E2127–E2133. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Odogwu L, Mathieu L, Blumenthal G, Larkins E, Goldberg KB, Griffin N, Bijwaard K, Lee EY, Philip R, Jiang X, et al: FDA approval summary: Dabrafenib and trametinib for the treatment of metastatic non-small cell lung cancers harboring BRAF V600E mutations. Oncologist. 23:740–745. 2018. View Article : Google Scholar

86 

Zhu C, Wei Y and Wei X: AXL receptor tyrosine kinase as a promising anti-cancer approach: Functions, molecular mechanisms and clinical applications. Mol Cancer. 18:1532019. View Article : Google Scholar : PubMed/NCBI

87 

Goyette MA and Côté JF: AXL receptor tyrosine kinase as a promising therapeutic target directing multiple aspects of cancer progression and metastasis. Cancers (Basel). 14:4662022. View Article : Google Scholar : PubMed/NCBI

88 

Zhang Z, Lee JC, Lin L, Olivas V, Au V, LaFramboise T, Abdel-Rahman M, Wang X, Levine AD, Rho JK, et al: Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet. 44:852–860. 2012. View Article : Google Scholar

89 

Sang YB, Kim JH, Kim CG, Hong MH, Kim HR, Cho BC and Lim SM: The Development of AXL inhibitors in lung cancer: Recent progress and challenges. Front Oncol. 12:8112472022. View Article : Google Scholar

90 

Nishio M, Okamoto I, Murakami H, Horinouchi H, Toyozawa R, Takeda M, Uno M, Crawford N, Jimbo T, Ishigami M, et al: 570P A first-in-human phase I study of the AXL inhibitor DS-1205c in combination with gefitinib in subjects with EGFR-mutant NSCLC. Ann Oncol. 31:S4882020. View Article : Google Scholar

91 

Byers LA, Gold KA and Peguero JA: Ph I/II study of oral selective AXL inhibitor bemcentinib (BGB324) in combination with erlotinib in patients with advanced EGFRm NSCLC: End of trial update. Wolters Kluwer Health; 2021, View Article : Google Scholar

92 

Xun G, Hu W and Li B: PTEN loss promotes oncogenic function of STMN1 via PI3K/AKT pathway in lung cancer. Sci Rep. 11:143182021. View Article : Google Scholar : PubMed/NCBI

93 

Ferrara MG, Martini M, D'Argento E, Forcella C, Vita E, Di Noia V, Sperduti I, Bilotta M, Ribelli M, Damiano P, et al: PTEN loss as a predictor of tumor heterogeneity and poor prognosis in patients with EGFR-mutant advanced non-small-cell lung cancer receiving tyrosine kinase inhibitors. Clin Lung Cancer. 22:351–360. 2021. View Article : Google Scholar : PubMed/NCBI

94 

Redig AJ, Capelletti M, Dahlberg SE, Sholl LM, Mach S, Fontes C, Shi Y, Chalasani P and Jänne PA: Clinical and molecular characteristics of NF1-mutant lung cancer. Clin Cancer Res. 22:3148–3156. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Cheung HW, Du J, Boehm JS, He F, Weir BA, Wang X, Butaney M, Sequist LV, Luo B, Engelman JA, et al: Amplification of CRKL induces transformation and epidermal growth factor receptor inhibitor resistance in human non-small cell lung cancers. Cancer Discov. 1:608–625. 2011. View Article : Google Scholar : PubMed/NCBI

96 

Cortot AB, Repellin CE, Shimamura T, Capelletti M, Zejnullahu K, Ercan D, Christensen JG, Wong KK, Gray NS and Jänne PA: Resistance to irreversible EGF receptor tyrosine kinase inhibitors through a multistep mechanism involving the IGF1R pathway. Cancer Res. 73:834–843. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Ware KE, Hinz TK, Kleczko E, Singleton KR, Marek LA, Helfrich BA, Cummings CT, Graham DK, Astling D, Tan AC and Heasley LE: A mechanism of resistance to gefitinib mediated by cellular reprogramming and the acquisition of an FGF2-FGFR1 autocrine growth loop. Oncogenesis. 2:e392013. View Article : Google Scholar : PubMed/NCBI

98 

Attili I, Passaro A, Pisapia P, Malapelle U and de Marinis F: Uncommon EGFR compound mutations in non-small cell lung cancer (NSCLC): A systematic review of available evidence. Curr Oncol. 29:255–266. 2022. View Article : Google Scholar

99 

Hayashi T, Kohsaka S, Takamochi K, Hara K, Kishikawa S, Sano K, Takahashi F, Suehara Y, Saito T, Takahashi K, et al: Clinicopathological characteristics of lung adenocarcinoma with compound EGFR mutations. Hum Pathol. 103:42–51. 2020. View Article : Google Scholar

100 

Rossi S, Damiano P, Toschi L, Finocchiaro G, Giordano L, Marinello A, Bria E, D'Argento E and Santoro A: Uncommon single and compound EGFR mutations: Clinical outcomes of a heterogeneous subgroup of NSCLC. Curr Probl Cancer. 46:1007872022. View Article : Google Scholar : PubMed/NCBI

101 

Jiang D, Fu Y, Zhou X, Li Y, Cui Y, Hong L, Jin H, Shi K, Huang F, Zhang X, et al: The prognosis of EGFR complex mutation or co-mutation with tyrosine kinase inhibitor treatment in non-small cell lung cancer. Am Soc Clin Oncol. 40:e210862022. View Article : Google Scholar

102 

Wang R, Pan S and Song X: Research Advances of EGFR-TP53 Co-mutation in advanced non-small cell lung cancer. Zhongguo Fei Ai Za Zhi. 25:174–182. 2022.(In Chinese).

103 

Wang F, Zhao N, Gao G, Deng HB, Wang ZH, Deng LL, Yang Y and Lu C: Prognostic value of TP53 co-mutation status combined with EGFR mutation in patients with lung adenocarcinoma. J Cancer Res Clin Oncol. 146:2851–2859. 2020. View Article : Google Scholar

104 

Cheng Y, Ma L, Liu Y, Zhu J, Xin Y, Liu X, Wang Y, Zhang T, Yang C, Wang S, et al: Comprehensive characterization and clinical impact of concomitant genomic alterations in EGFR-mutant NSCLCs treated with EGFR kinase inhibitors. Lung Cancer. 145:63–70. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Zhang Y, Li S, Lyu Z, Cai J, Zheng N, Li Y, Xu T and Zeng H: The co-mutation of EGFR and tumor-related genes leads to a worse prognosis and a higher level of tumor mutational burden in Chinese non-small cell lung cancer patients. J Thorac Dis. 14:185–193. 2022. View Article : Google Scholar : PubMed/NCBI

106 

Li HS, Liu CM and Wang Y: Limited role of KRAS mutation in guiding immunotherapy in advanced non-small-cell lung cancer. Future Oncol. 18:2433–2443. 2022. View Article : Google Scholar

107 

Marcoux N, Gettinger SN, O'Kane G, Arbour KC, Neal JW, Husain H, Evans TL, Brahmer JR, Muzikansky A, Bonomi PD, et al: EGFR-Mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: Clinical outcomes. J Clin Oncol. 37:278–285. 2019. View Article : Google Scholar

108 

Niederst MJ, Sequist LV, Poirier JT, Mermel CH, Lockerman EL, Garcia AR, Katayama R, Costa C, Ross KN, Moran T, et al: RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer. Nat Commun. 6:63772015. View Article : Google Scholar : PubMed/NCBI

109 

Lee JK, Lee J, Kim S, Kim S, Youk J, Park S, An Y, Keam B, Kim DW, Heo DS, et al: Clonal history and genetic predictors of transformation into small-cell carcinomas from lung adenocarcinomas. J Clin Oncol. 35:3065–3074. 2017. View Article : Google Scholar

110 

Offin M, Chan JM, Tenet M, Rizvi HA, Shen R, Riely GJ, 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. 14:1784–1793. 2019. View Article : Google Scholar

111 

Mambetsariev I, Arvanitis L, Fricke J, Pharaon R, Baroz AR, Afkhami M, Koczywas M, Massarelli E and Salgia R: Small cell lung cancer transformation following treatment in EGFR-Mutated non-small cell lung cancer. J Clin Med. 11:14292022. View Article : Google Scholar : PubMed/NCBI

112 

Mc Leer A, Foll M, Brevet M, Antoine M, Novello S, Mondet J, Cadranel J, Girard N, Giaj Levra M, Demontrond P, et al: Detection of acquired TERT amplification in addition to predisposing p53 and Rb pathways alterations in EGFR-mutant lung adenocarcinomas transformed into small-cell lung cancers. Lung Cancer. 167:98–106. 2022. View Article : Google Scholar : PubMed/NCBI

113 

Song KA, Niederst MJ, Lochmann TL, Hata AN, Kitai H, Ham J, Floros KV, Hicks MA, Hu H, Mulvey HE, et al: Epithelial-to-Mesenchymal transition antagonizes response to targeted therapies in lung cancer by suppressing BIM. Clin Cancer Res. 24:197–208. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Shaurova T, Zhang L, Goodrich DW and Hershberger PA: Understanding lineage plasticity as a path to targeted therapy failure in EGFR-Mutant non-small cell lung cancer. Front Genet. 11:2812020. View Article : Google Scholar

115 

Wang W, Xu C, Chen H, Jia J, Wang L, Feng H, Wang H, Song Z, Yang N and Zhang Y: Genomic alterations and clinical outcomes in patients with lung adenocarcinoma with transformation to small cell lung cancer after treatment with EGFR tyrosine kinase inhibitors: A multicenter retrospective study. Lung Cancer. 155:20–27. 2021. View Article : Google Scholar : PubMed/NCBI

116 

Zhang C, Zhang S, Yao Y, Huang J, Peng K, Gao Q, Chen H, Xu C, Zhang X, Wu Y, Yang J, et al: MA12. 08 Chemotherapy plus EGFR TKIs or bevacizumab versus chemotherapy alone in SCLC-transformed EGFR-mutant lung adenocarcinoma. J Thor Oncol. 16:S178–S179. 2021. View Article : Google Scholar

117 

Kuiper JL, Ronden MI, Becker A, Heideman DA, van Hengel P, Ylstra B, Thunnissen E and Smit EF: Transformation to a squamous cell carcinoma phenotype of an EGFR-mutated NSCLC patient after treatment with an EGFR-tyrosine kinase inhibitor. J Clin Pathol. 68:320–321. 2015. View Article : Google Scholar

118 

Levin PA, Mayer M, Hoskin S, Sailors J, Oliver DH and Gerber DE: Histologic transformation from adenocarcinoma to squamous cell carcinoma as a mechanism of resistance to EGFR inhibition. J Thorac Oncol. 10:e86–e88. 2015. View Article : Google Scholar

119 

Longo L, Mengoli MC, Bertolini F, Bettelli S, Manfredini S and Rossi G: Synchronous occurrence of squamous-cell carcinoma ‘transformation’ and EGFR exon 20 S768I mutation as a novel mechanism of resistance in EGFR-mutated lung adenocarcinoma. Lung Cancer. 103:24–26. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Roca E, Pozzari M, Vermi W, Tovazzi V, Baggi A, Amoroso V, Nonnis D, Intagliata S and Berruti A: Outcome of EGFR-mutated adenocarcinoma NSCLC patients with changed phenotype to squamous cell carcinoma after tyrosine kinase inhibitors: A pooled analysis with an additional case. Lung Cancer. 127:12–18. 2019. View Article : Google Scholar : PubMed/NCBI

121 

Liao J, Li Y, Liu C, Long Q and Wang J: Case report: EGFR-Positive early-stage lung adenocarcinoma transforming to squamous cell carcinoma after TKI treatment. Front Oncol. 11:6968812021. View Article : Google Scholar

122 

Jukna A, Montanari G, Mengoli MC, Cavazza A, Covi M, Barbieri F, Bertolini F and Rossi G: Squamous Cell Carcinoma ‘Transformation’ concurrent with secondary T790M mutation in resistant EGFR-Mutated Adenocarcinomas. J Thorac Oncol. 11:e49–e51. 2016. View Article : Google Scholar

123 

Bugano DDG, Kalhor N, Zhang J, Neskey M and William WN Jr: Squamous-cell transformation in a patient with lung adenocarcinoma receiving erlotinib: Co-occurrence with T790M mutation. Cancer Treat Comm. 4:34–36. 2015. View Article : Google Scholar

124 

Park S, Shim JH, Lee B, Cho I, Park WY, Kim Y, Lee SH, Choi Y, Han J, Ahn JS, et al: Paired genomic analysis of squamous cell carcinoma transformed from EGFR-mutated lung adenocarcinoma. Lung Cancer. 134:7–15. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Weng CH, Chen LY, Lin YC, Shih JY, Lin YC, Tseng RY, Chiu AC, Yeh YH, Liu C, Lin YT, et al: Epithelial-mesenchymal transition (EMT) beyond EGFR mutations per se is a common mechanism for acquired resistance to EGFR TKI. Oncogene. 38:455–468. 2019. View Article : Google Scholar : PubMed/NCBI

126 

Brabletz S, Schuhwerk H, Brabletz T and Stemmler MP: Dynamic EMT: A multi-tool for tumor progression. EMBO J. 40:e1086472021. View Article : Google Scholar : PubMed/NCBI

127 

Zhu X, Chen L, Liu L and Niu X: EMT-Mediated Acquired EGFR-TKI resistance in NSCLC: Mechanisms and strategies. Front Oncol. 9:10442019. View Article : Google Scholar

128 

Miralaei N, Majd A, Ghaedi K, Peymani M and Safaei M: Integrated pan-cancer of AURKA expression and drug sensitivity analysis reveals increased expression of AURKA is responsible for drug resistance. Cancer Med. 10:6428–6441. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Nilsson MB, 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. 12:eaaz45892020. View Article : Google Scholar : PubMed/NCBI

130 

Wang CY, Lee MH, Kao YR, Hsiao SH, Hong SY and Wu CW: Alisertib inhibits migration and invasion of EGFR-TKI resistant cells by partially reversing the epithelial-mesenchymal transition. Biochim Biophys Acta Mol Cell Res. 1868:1190162021. View Article : Google Scholar : PubMed/NCBI

131 

Yeh CT, Chen TT, Satriyo PB, Wang CH, Wu ATH, Chao TY, Lee KY, Hsiao M, Wang LS and Kuo KT: Bruton's tyrosine kinase (BTK) mediates resistance to EGFR inhibition in non-small-cell lung carcinoma. Oncogenesis. 10:562021. View Article : Google Scholar : PubMed/NCBI

132 

Liao BC, Griesing S and Yang JC: Second-line treatment of EGFR T790M-negative non-small cell lung cancer patients. Ther Adv Med Oncol. Nov 25–2019.(Epub ahead of print). View Article : Google Scholar

133 

Lee CK, Man J, Lord S, Links M, Gebski V, Mok T and Yang JC: Checkpoint Inhibitors in Metastatic EGFR-Mutated non-small cell lung cancer-A meta-analysis. J Thorac Oncol. 12:403–407. 2017. View Article : Google Scholar

134 

Lee CK, Man J, Lord S, Cooper W, Links M, Gebski V, Herbst RS, Gralla RJ, Mok T and Yang JC: Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: A systematic review and meta-analysis. JAMA Oncol. 4:210–216. 2018. View Article : Google Scholar

135 

Yang CY, Liao WY, Ho CC, Chen KY, Tsai TH, Hsu CL, Su KY, Chang YL, Wu CT, Hsu CC, et al: Association between programmed death-ligand 1 expression, immune microenvironments, and clinical outcomes in epidermal growth factor receptor mutant lung adenocarcinoma patients treated with tyrosine kinase inhibitors. Eur J Cancer. 124:110–122. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Isomoto K, Haratani K, Hayashi H, Shimizu S, Tomida S, Niwa T, Yokoyama T, Fukuda Y, Chiba Y, Kato R, et al: Impact of EGFR-TKI treatment on the tumor immune microenvironment in EGFR mutation-positive non-small cell lung cancer. Clin Cancer Res. 26:2037–2046. 2020. View Article : Google Scholar : PubMed/NCBI

137 

Reck M, Mok TS, Nishio M, Jotte RM, 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. 7:387–401. 2019. View Article : Google Scholar : PubMed/NCBI

138 

Wu SG, Ho CC, Yang JC, Lia BC, Yang CY, Lin YT, Yu CJ, Liao WY and Shih JY: 12P A phase II study of atezolizumab in combination with bevacizumab, carboplatin or cisplatin, and pemetrexed for EGFR-mutant metastatic NSCLC patients after failure of EGFR TKIs. Ann Oncol. 33:S33–S34. 2022. View Article : Google Scholar

139 

Lam TC, Tsang KC, Choi HC, Lee VH, Lam KO, Chiang CL, So TH, Chan WW, Nyaw SF, Lim F, et al: Combination atezolizumab, bevacizumab, pemetrexed and carboplatin for metastatic EGFR mutated NSCLC after TKI failure. Lung Cancer. 159:18–26. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Ren S, Zhang J, Zhao Y, Zhou J, Fan Y, Shu Y, Liu X, Zhang H, He J, Gao G, et al: A multi-center phase II study of toripalimab with chemotherapy in patients with EGFR mutant advanced NSCLC patients resistant to EGFR TKIs: Efficacy and biomarker analysis. Am Soc Clin Oncol. 6:3552020.

141 

Jiang T, Wang P, Zhang J, Zhao Y, Zhou J, Fan Y, Shu Y, Liu X, Zhang H, He J, et al: Toripalimab plus chemotherapy as second-line treatment in previously EGFR-TKI treated patients with EGFR-mutant-advanced NSCLC: A multicenter phase-II trial. Signal Transduct Target Ther. 6:3552021. View Article : Google Scholar : PubMed/NCBI

142 

Lu S, Wu L, Jian H, Cheng Y, Wang Q, Fang J, Wang Z, Hu Y, Sun M, Han L, et al: VP9-2021: ORIENT-31: Phase III study of sintilimab with or without IBI305 plus chemotherapy in patients with EGFR mutated nonsquamous NSCLC who progressed after EGFR-TKI therapy. Ann Oncol. 33:112–113. 2022. View Article : Google Scholar

143 

Hayashi H, Sugawara S, Fukuda Y, Fujimoto D, Miura S, Ota K, Ozawa Y, Hara S, Tanizaki J, Azuma K, et al: A randomized phase II study comparing nivolumab with carboplatin-pemetrexed for EGFR-mutated NSCLC with resistance to EGFR tyrosine kinase inhibitors (WJOG8515L). Clin Cancer Res. 28:893–902. 2022. View Article : Google Scholar : PubMed/NCBI

144 

de Rouw N, Piet B, Derijks HJ, van den Heuvel MM and Ter Heine R: Mechanisms, management and prevention of pemetrexed-related toxicity. Drug Saf. 44:1271–1281. 2021. View Article : Google Scholar : PubMed/NCBI

145 

Liang SK, Keng LT, Chang CH, Wen YF, Lee MR, Yang CY, Wang JY, Ko JC, Shih JY and Yu CJ: Treatment options of first-line tyrosine kinase inhibitors and subsequent systemic chemotherapy agents for advanced EGFR mutant lung adenocarcinoma patients: Implications from Taiwan cancer registry cohort. Front Oncol. 10:5903562021. View Article : Google Scholar

146 

Li Z, Guo H, Lu Y, Hu J, Luo H and Gu W: Chemotherapy with or without pemetrexed as second-line regimens for advanced non-small-cell lung cancer patients who have progressed after first-line EGFR TKIs: A systematic review and meta-analysis. Onco Targets Ther. 11:3697–3703. 2018. View Article : Google Scholar : PubMed/NCBI

147 

Yoo KH, Lee SJ, Cho J, Lee KH, Park KU, Kim KH, Cho EK, Choi YH, Kim HR, Kim HG, et al: A randomized, open-label, Phase II study comparing pemetrexed plus cisplatin followed by maintenance pemetrexed versus pemetrexed alone in patients with epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer after failure of first-line EGFR tyrosine kinase inhibitor: KCSG-LU12-13. Cancer Res Treat. 51:718–726. 2019. View Article : Google Scholar

148 

Le X, Nilsson M, Goldman J, Reck M, Nakagawa K, Kato T, Ares LP, Frimodt-Moller B, Wolff K, Visseren-Grul C, et al: Dual EGFR-VEGF Pathway inhibition: A promising strategy for patients with EGFR-Mutant NSCLC. J Thorac Oncol. 16:205–215. 2021. View Article : Google Scholar

149 

Lian Z, Du W, Zhang Y, Fu Y, Liu T, Wang A, Cai T, Zhu J, Zeng Y, Liu Z and Huang JA: Anlotinib can overcome acquired resistance to EGFR-TKIs via FGFR1 signaling in non-small cell lung cancer without harboring EGFR T790M mutation. Thorac Cancer. 11:1934–1943. 2020. View Article : Google Scholar : PubMed/NCBI

150 

Zhang C, Cao H, Cui Y, Jin S, Gao W, Huang C and Guo R: Concurrent use of anlotinib overcomes acquired resistance to EGFR-TKI in patients with advanced EGFR-mutant non-small cell lung cancer. Thorac Cancer. 12:2574–2584. 2021. View Article : Google Scholar : PubMed/NCBI

151 

Hata A, Katakami N, Kaji R, Yokoyama T, Kaneda T, Tamiya M, Inoue T, Kimura H, Yano Y, Tamura D, et al: Afatinib plus bevacizumab combination after acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant non-small cell lung cancer: Multicenter, single-arm, phase 2 trial (ABC Study). Cancer. 124:3830–3838. 2018. View Article : Google Scholar : PubMed/NCBI

152 

Yang R, Wang D, Li X, Mao K, Wang J, Li P, Shi X, Zhang S and Wang Y: An advanced non-small cell lung cancer patient with EGFR and KRAS mutations, and PD-L1 positive, benefited from immunotherapy: A case report. Ann Transl Med. 10:3812022. View Article : Google Scholar : PubMed/NCBI

153 

Bai M, Wang W, Gao X, Wu L, Jin P, Wu H, Yu J and Meng X: Efficacy of immune checkpoint inhibitors in patients with EGFR Mutated NSCLC and potential risk factors associated with prognosis: A single institution experience. Front Immunol. 13:8324192022. View Article : Google Scholar

154 

Mu Y, Hao X, Xing P, Hu X, Wang Y, Li T, Zhang J, Xu Z and Li J: Acquired resistance to osimertinib in patients with non-small-cell lung cancer: Mechanisms and clinical outcomes. J Cancer Res Clin Oncol. 146:2427–2433. 2020. View Article : Google Scholar

155 

Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E and Tiseo M: Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer. 121:725–737. 2019. View Article : Google Scholar : PubMed/NCBI

156 

He J, Huang Z, Han L, Gong Y and Xie C: Mechanisms and management of 3rd-generation EGFR-TKI resistance in advanced non-small cell lung cancer (Review). Int J Oncol. 59:902021. View Article : Google Scholar

157 

Papadimitrakopoulou V, Wu YL, Han JY, Ahn MJ, Ramalingam SS, John T, Okamoto I, Yang JC, Bulusu K, Laus G, et al: Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Annal Oncol. 29:viii7412018. View Article : Google Scholar

158 

Piotrowska Z, Nagy R, Fairclough S, Lanman R, Marcoux N, Gettinger S, Owonikoko T, Ramalingam S and Sequist L: Characterizing the genomic landscape of EGFR C797S in lung cancer using ctDNA next-generation sequencing. J Thorac Oncol. 12:S17672017. View Article : Google Scholar

159 

Wang X, Zhou L, Yin JC, Wu X, Shao YW and Gao B: Lung adenocarcinoma harboring EGFR 19del/C797S/T790M triple mutations responds to brigatinib and Anti-EGFR antibody combination therapy. J Thorac Oncol. 14:e85–e88. 2019. View Article : Google Scholar

160 

Chang Y, Liu S, Jiang Y, Hua L and Wen L: Effective treatment of pulmonary adenocarcinoma harboring triple EGFR mutations of L858R, T790M, cis-G796s/cis-C797s by osimertinib, brigatinib, and bevacizumab combination therapy: A case report. Respir Med Case Rep. 36:1015822022.PubMed/NCBI

161 

Zhou R, Song L, Zhang W, Shao L and Li X and Li X: Combination of osimertinib and anlotinib may overcome the resistance mediated by in cis EGFR T790M-C797S in NSCLC: A case report. Onco Targets Ther. 14:2847–2851. 2021. View Article : Google Scholar : PubMed/NCBI

162 

Yang Y, Xu H, Ma L, Yang L, Yang G, Zhang S, Ai X, Zhang S and Wang Y: Possibility of brigatinib-based therapy, or chemotherapy plus anti-angiogenic treatment after resistance of osimertinib harboring EGFR T790M-cis-C797S mutations in lung adenocarcinoma patients. Cancer Med. 10:8328–8337. 2021. View Article : Google Scholar : PubMed/NCBI

163 

Zhao Y, Chen Y, Huang H, Li X, Shao L and Ding H: Significant benefits of afatinib and apatinib in a refractory advanced NSCLC patient resistant to osimertinib: A case report. OncoTargets Ther. 14:3063–3067. 2021. View Article : Google Scholar

164 

Yang Z, Yang N, Ou Q, Xiang Y, Jiang T, Wu X, Bao H, Tong X, Wang X, Shao YW, et al: Investigating novel resistance mechanisms to third-generation EGFR tyrosine kinase inhibitor osimertinib in non-small cell lung cancer patients. Clin Cancer Res. 24:3097–3107. 2018. View Article : Google Scholar : PubMed/NCBI

165 

Zhang Y, He B, Zhou D, Li M and 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. 12:51–56. 2018. View Article : Google Scholar

166 

Schoenfeld AJ, Chan JM, Kubota D, Sato H, Rizvi H, Daneshbod Y, Chang JC, Paik PK, Offin M, Arcila ME, 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. 26:2654–2663. 2020. View Article : Google Scholar : PubMed/NCBI

167 

Fairclough SR, Kiedrowski LA, Lin JJ, Zelichov O, Tarcic G, Stinchcombe TE, Odegaard JI, Lanman RB, Shaw AT and Nagy RJ: Identification of osimertinib-resistant EGFR L792 mutations by cfDNA sequencing: oncogenic activity assessment and prevalence in large cfDNA cohort. Exp Hematol Oncol. 8:242019. View Article : Google Scholar

168 

Ma L, Chen R, Wang F, Ma LL, Yuan MM, Chen RR and Liu J: EGFR L718Q mutation occurs without T790M mutation in a lung adenocarcinoma patient with acquired resistance to osimertinib. Ann Transl Med. 7:2072019. View Article : Google Scholar : PubMed/NCBI

169 

Fang W, Huang Y, Gan J, Zheng Q and Zhang L: Emergence of EGFR G724S after progression on osimertinib responded to afatinib monotherapy. J Thorac Oncol. 15:e36–e37. 2020. View Article : Google Scholar

170 

Zhang Y, Yang Q, Zeng X, Wang M, Dong S, Yang B, Tu X, Wei T, Xie W, Zhang C, et al: MET amplification attenuates lung tumor response to immunotherapy by inhibiting STING. Cancer Discov. 11:2726–2737. 2021. View Article : Google Scholar : PubMed/NCBI

171 

Syed YY: Amivantamab: First approval. Drugs. 81:1349–1353. 2021. View Article : Google Scholar : PubMed/NCBI

172 

Amivantamab OK'd for EGFR-Mutant NSCLC, . Cancer Discov. 11:16042021. View Article : Google Scholar

173 

Neijssen J, Cardoso RM, Chevalier KM, Wiegman L, Valerius T, Anderson GM, Moores SL, Schuurman J, Parren PW, Strohl WR and Chiu ML: Discovery of amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR and MET. J Biol Chem. 296:1006412021. View Article : Google Scholar : PubMed/NCBI

174 

Planchard D, Loriot Y, André F, Gobert A, Auger N, Lacroix L and Soria JC: EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann Oncol. 26:2073–2078. 2015. View Article : Google Scholar

175 

Ramalingam S, Cheng Y, Zhou C, Ohe Y, Imamura F, Cho BC, Lin M, Majem M, Shah R, Rukazenkov Y, et al: Mechanisms of acquired resistance to first-line osimertinib: preliminary data from the phase III FLAURA study. OncologyPro. 29:viii7402018.

176 

Oxnard GR, Hu Y, Mileham KF, Husain H, Costa DB, Tracy P, Feeney N, Sholl LM, Dahlberg SE, Redig AJ, et al: Assessment of resistance mechanisms and clinical implications in patients With EGFR T790M-Positive lung cancer and acquired resistance to osimertinib. JAMA Oncol. 4:1527–1534. 2018. View Article : Google Scholar

177 

Qu F, Zhou Y and Yu WJA-CD: A review of research progress on mechanisms and overcoming strategies of acquired osimertinib resistance. Anticancer Drugs. 33:e76–e83. 2022. View Article : Google Scholar : PubMed/NCBI

178 

Beenken A and Mohammadi M: The FGF family: Biology, pathophysiology and therapy. Nat Rev Drug Discov. 8:235–253. 2009. View Article : Google Scholar : PubMed/NCBI

179 

Lu Y, Liu Y, Oeck S, Zhang GJ, Schramm A and Glazer PM: Hypoxia induces resistance to EGFR inhibitors in lung cancer cells via upregulation of FGFR1 and the MAPK pathway. Cancer Res. 80:4655–4667. 2020. View Article : Google Scholar : PubMed/NCBI

180 

Quintanal-Villalonga A, Molina-Pinelo S, Cirauqui C, Ojeda-Márquez L, Marrugal Á, Suarez R, Conde E, Ponce-Aix S, Enguita AB, Carnero A, et al: FGFR1 Cooperates with EGFR in lung cancer oncogenesis, and their combined inhibition shows improved efficacy. J Thorac Oncol. 14:641–655. 2019. View Article : Google Scholar

181 

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. 11:140–149. 2020. View Article : Google Scholar : PubMed/NCBI

182 

Zhao Y, Wang H and He C: Drug resistance of targeted therapy for advanced non-small cell lung cancer harbored EGFR mutation: From mechanism analysis to clinical strategy. J Cancer Res Clin Oncol. 147:3653–3664. 2021. View Article : Google Scholar

183 

Makimoto G, Ninomiya K, Kubo T, Sunami R, Kato Y, Ichihara E, Ohashi K, Rai K, Hotta K, Tabata M, et al: A novel osimertinib-resistant human lung adenocarcinoma cell line harbouring mutant EGFR and activated IGF1R. Jpn J Clin Oncol. 51:956–965. 2021. View Article : Google Scholar

184 

Wang R, Yamada T, Kita K, Taniguchi H, Arai S, Fukuda K, Terashima M, Ishimura A, Nishiyama A, Tanimoto A, et al: Transient IGF-1R inhibition combined with osimertinib eradicates AXL-low expressing EGFR mutated lung cancer. Nat Commun. 11:46072020. View Article : Google Scholar : PubMed/NCBI

185 

Lin CC, Shih JY, Yu CJ, Ho CC, Liao WY, Lee JH, Tsai TH, Su KY, Hsieh MS, Chang YL, et al: Outcomes in patients with non-small-cell lung cancer and acquired Thr790Met mutation treated with osimertinib: A genomic study. Lancet Respir Med. 6:107–116. 2018. View Article : Google Scholar : PubMed/NCBI

186 

Jia Y, Yun CH, Park E, Ercan D, Manuia M, Juarez J, Xu C, Rhee K, Chen T, Zhang H, et al: Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors. Nature. 534:129–132. 2016. View Article : Google Scholar : PubMed/NCBI

187 

To C, Jang J, Chen T, Park E, Mushajiang M, De Clercq DJH, Xu M, Wang S, Cameron MD, Heppner DE, et al: Single and dual targeting of mutant EGFR with an allosteric inhibitor. Cancer Discov. 9:926–943. 2019. View Article : Google Scholar : PubMed/NCBI

188 

Tripathi SK and Biswal BK: Allosteric mutant-selective fourth-generation EGFR inhibitors as an efficient combination therapeutic in the treatment of non-small cell lung carcinoma. Drug Discov Today. 26:1466–1472. 2021. View Article : Google Scholar : PubMed/NCBI

189 

Kashima K, Kawauchi H, Tanimura H, Tachibana Y, Chiba T, Torizawa T and Sakamoto H: CH7233163 overcomes osimertinib-resistant EGFR-Del19/T790M/C797S Mutation. Mol Cancer Ther. 19:2288–2297. 2020. View Article : Google Scholar : PubMed/NCBI

190 

Schalm S, Dineen T, Lim S, Park CW, 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. 31:S8392020. View Article : Google Scholar

191 

Conti C, Campbell J, Woessner R, Guo J, Timsit Y, Iliou M, Wardwell S, Davis A, Chicklas S, Hsieh J, et al: BLU-701 is a highly potent, brain-penetrant and WT-sparing next-generation EGFR TKI for the treatment of sensitizing (ex19del, L858R) and C797S resistance mutations in metastatic NSCLC. Cancer Res. 81 (Suppl 13):12622021. View Article : Google Scholar

192 

Lim SM, Park CW, Zhang Z, Woessner R, Dineen T, Stevison F, Hsieh J, Eno M, Wilson D, Campbell J, et al: BLU-945, a fourth-generation, potent and highly selective epidermal growth factor receptor tyrosine kinase inhibitor with intracranial activity, demonstrates robust in vivo anti-tumor activity in models of osimertinib-resistant non-small cell lung cancer. Cancer Res. 81 (Suppl 13):14672021. View Article : Google Scholar

193 

Tavera L, Zhang Z, Wardwell S, Job E, McGinn K, Chen M, Iliou M, Albayya F, Campbell J, Eno M, et al: BLU-701 tumour suppression and intracranial activity as a single agent and in combination with BLU-945 in models of non-small cell lung cancer (NSCLC) driven by EGFR mutations. Mol Cell Biol. 165:S372022.

194 

Liu X, Zhang X, Yang L, Chen S, Tian X, Dong T, Ding CZ, Hu L, Wu L, Zhao L, Mao J, et al: Preclinical evaluation of TQB3804, a potent EGFR C797S inhibitor. Cancer Res. 79 (Suppl 13):13202019. View Article : Google Scholar

195 

Huang J and Wang H: Targeted therapy and mechanism of drug resistance in non-small cell lung cancer with epidermal growth factor receptor gene mutation. Zhongguo Fei Ai Za Zhi. 25:183–192. 2022.(In Chinese).

196 

Lim S, Kim DW, Jung JE, Lee G, Ryou JH, Kang SU, Lee YH, Shin HJ, Yum SY and Yim Ε: A Phase 1/2, open-label study of BBT-176, a triple mutation targeting EGFR TKI, in patients with NSCLC who progressed after prior EGFR TKI therapy. Ann Oncol. 32:S949–S1039. 2021. View Article : Google Scholar

197 

Lim S, Kim D and Jung J: A phase I/II, open-label study of BBT-176, a triple mutation targeting EGFR TKI, in patients with NSCLC who progressed after prior EGFR TKI therapy. Ann Oncol. 32:S1035(Suppl 5):2021.

198 

Park K, Haura EB, Leighl NB, Mitchell P, Shu CA, Girard N, Viteri S, Han JY, Kim SW, Lee CK, et al: Amivantamab in EGFR exon 20 insertion-mutated non-small-cell lung cancer progressing on platinum chemotherapy: Initial results from the CHRYSALIS phase I study. J Clin Oncol. 39:3391–3402. 2021. View Article : Google Scholar

199 

Cho B, Lee K, Cho E, Kim DW, Lee JS, Han JY, Kim SW, Spira A, Haura EB, Sabari JK, et al: 1258O Amivantamab (JNJ-61186372), an EGFR-MET bispecific antibody, in combination with lazertinib, a 3rd-generation tyrosine kinase inhibitor (TKI), in advanced EGFR NSCLC. Ann Oncol. 31:S813(Suppl 4):2020. View Article : Google Scholar

200 

Yu H, Johnson M, Steuer C, Vigliotti M, Chen S, Kamai Y, Yu C and Jänne P: Preliminary phase 1 results of U3-1402-A novel HER3-targeted antibody-drug conjugate-in EGFR TKI-resistant, EGFR-mutant NSCLC. Mol Cell Biol. 14:S336–S337. 2019.

201 

Jänne PA, Baik C, Su WC, Johnson ML, Hayashi H, Nishio M, Kim DW, Koczywas M, Gold KA, Steuer CE, et al: Efficacy and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated non-small cell lung cancer. Cancer Discov. 12:74–89. 2022. View Article : Google Scholar

202 

Soo RA, Han JY, Dafni U, Cho BC, Yeo CM, Nadal E, Carcereny E, de Castro J, Sala MA, Bernabé R, et al: A randomised phase II study of osimertinib and bevacizumab versus osimertinib alone as second-line targeted treatment in advanced NSCLC with confirmed EGFR and acquired T790M mutations: The European Thoracic Oncology Platform (ETOP 10–16) BOOSTER trial. Ann Oncol. 33:181–192. 2022. View Article : Google Scholar

203 

Cui Q, Hu Y, Cui Q, Wu D, Mao Y, Ma D and Liu H: Osimertinib rechallenge with bevacizumab vs. chemotherapy plus bevacizumab in EGFR-Mutant NSCLC patients with osimertinib resistance. Front Pharmacol. 12:7467072022. View Article : Google Scholar

204 

Sequist L, Peled N, Tufman A, Servidio L, Li J, Taylor R and Zhao J: COMPEL: Chemotherapy with/without osimertinib in patients with EGFRm advanced NSCLC and progression on first-line osimertinib. J Thor Oncol. 16:S11012021. View Article : Google Scholar

205 

Han B, Li K, Wang Q, Zhang L, Shi J, Wang Z, Cheng Y, He J, Shi Y, Zhao Y, et al: Effect of anlotinib as a third-line or further treatment on overall survival of patients with advanced non-small cell lung cancer: The ALTER 0303 phase 3 Randomized clinical trial. JAMA Oncol. 4:1569–1575. 2018. View Article : Google Scholar

206 

Tamiya M, Kunimasa K, Nishino K, Matsumoto S, Kawachi H, Kuno K, Inoue T, Kuhara H, Imamura F, Goto K and Kumagai T: Successful treatment of an osimertinib-resistant lung adenocarcinoma with an exon 18 EGFR mutation (G719S) with afatinib plus bevacizumab. Invest New Drugs. 39:232–236. 2021. View Article : Google Scholar : PubMed/NCBI

207 

Blakely CM, Watkins TBK, Wu W, Gini B, Chabon JJ, McCoach CE, McGranahan N, Wilson GA, Birkbak NJ, Olivas VR, et al: Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat Genet. 49:1693–1704. 2017. View Article : Google Scholar

208 

Dagogo-Jack I and Shaw AT: Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 15:81–94. 2018. View Article : Google Scholar

209 

Assaraf YG, Brozovic A, Gonçalves AC, Jurkovicova D, Linē A, Machuqueiro M, Saponara S, Sarmento-Ribeiro AB, Xavier CPR and Vasconcelos MH: The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist Updat. 46:1006452019. View Article : Google Scholar

210 

Zhang Y, Wang D, Peng M, Tang L, Ouyang J, Xiong F, Guo C, Tang Y, Zhou Y, Liao Q, et al: Single-cell RNA sequencing in cancer research. J Exp Clin Cancer Res. 40:812021. View Article : Google Scholar : PubMed/NCBI

211 

Maynard A, McCoach CE, Rotow JK, Harris L, Haderk F, Kerr DL, Yu EA, Schenk EL, Tan W, Zee A, et al: Therapy-Induced evolution of human lung cancer revealed by single-cell RNA sequencing. Cell. 182:1232–1251.e22. 2020. View Article : Google Scholar

212 

Kim DW and Cho JY: Recent advances in allogeneic CAR-T cells. Biomolecules. 10:2632020. View Article : Google Scholar

213 

Patel AJ, Richter A, Drayson MT and Middleton GW: The role of B lymphocytes in the immuno-biology of non-small-cell lung cancer. Cancer Immunol Immunother. 69:325–342. 2020. View Article : Google Scholar : PubMed/NCBI

214 

Hung LVM, Ngo HT and Van Pham P: Clinical trials with cytokine-induced killer cells and CAR-T cell transplantation for non-small cell lung cancer treatment. Adv Exp Med Biol. 1292:113–130. 2020. View Article : Google Scholar

215 

Johnson LA and June CH: Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 27:38–58. 2017. View Article : Google Scholar : PubMed/NCBI

216 

Xu J, Zhang Q, Tian K, Wang H, Yin H and Zheng J: Current status and future prospects of the strategy of combining CAR-T with PD-1 blockade for antitumor therapy (Review). Mol Med Rep. 17:2083–2088. 2018.PubMed/NCBI

217 

Kandra P, Nandigama R, Eul B, Huber M, Kobold S, Seeger W, Grimminger F and Savai R: Utility and drawbacks of chimeric antigen receptor T Cell (CAR-T) therapy in lung cancer. Front Immunol. 13:9035622022. View Article : Google Scholar

218 

Xu C, Ju D and Zhang X: Chimeric antigen receptor T-cell therapy: Challenges and opportunities in lung cancer. Antib Ther. 5:73–83. 2022.PubMed/NCBI

219 

Yang P, Qiao Y, Meng M and Zhou Q: Cancer/Testis antigens as biomarker and target for the diagnosis, prognosis, and therapy of lung cancer. Front Oncol. 12:8641592022. View Article : Google Scholar

220 

Yeku O, Li X and Brentjens RJ: Adoptive T-Cell therapy for solid tumors. Am Soc Clin Oncol Educ Book. 37:193–204. 2017. View Article : Google Scholar : PubMed/NCBI

221 

Reppel L, Tsahouridis O, Akulian J, Davis IJ, Lee H, Fucà G, Weiss J, Dotti G, Pecot CV and Savoldo B: Targeting disialoganglioside GD2 with chimeric antigen receptor-redirected T cells in lung cancer. J Immunother Cancer. 10:e0038972022. View Article : Google Scholar : PubMed/NCBI

222 

Min J, Long C, Zhang L, Duan J, Fan H, Chu F and Li Z: c-Met specific CAR-T cells as a targeted therapy for non-small cell lung cancer cell A549. Bioengineered. 13:9216–9232. 2022. View Article : Google Scholar : PubMed/NCBI

223 

Feng K, Guo Y, Dai H, Wang Y, Li X, Jia H and Han W: Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci. 59:468–479. 2016. View Article : Google Scholar

224 

Xiao BF, Zhang JT, Zhu YG, Cui XR, Lu ZM, Yu BT and Wu N: Chimeric antigen receptor T-Cell therapy in lung cancer: Potential and challenges. Front Immunol. 12:7827752021. View Article : Google Scholar

225 

Qu J, Mei Q, Chen L and Zhou J: Chimeric antigen receptor (CAR)-T-cell therapy in non-small-cell lung cancer (NSCLC): Current status and future Aperspectives. Cancer Immunol Immunother. 70:619–631. 2021. View Article : Google Scholar : PubMed/NCBI

226 

Chen L, Chen F, Li J, Pu Y, Yang C, Wang Y, Lei Y and Huang Y: CAR-T cell therapy for lung cancer: Potential and perspective. Thorac Cancer. 13:889–899. 2022. View Article : Google Scholar : PubMed/NCBI

227 

Vasic D, Lee JB, Leung Y, Khatri I, Na Y, Abate-Daga D and Zhang L: Allogeneic double-negative CAR-T cells inhibit tumor growth without off-tumor toxicities. Sci Immunol. 7:eabl36422022. View Article : Google Scholar

Related Articles

Journal Cover

November-2022
Volume 24 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun R, Hou Z, Zhang Y and Jiang B: Drug resistance mechanisms and progress in the treatment of EGFR‑mutated lung adenocarcinoma (Review). Oncol Lett 24: 408, 2022
APA
Sun, R., Hou, Z., Zhang, Y., & Jiang, B. (2022). Drug resistance mechanisms and progress in the treatment of EGFR‑mutated lung adenocarcinoma (Review). Oncology Letters, 24, 408. https://doi.org/10.3892/ol.2022.13528
MLA
Sun, R., Hou, Z., Zhang, Y., Jiang, B."Drug resistance mechanisms and progress in the treatment of EGFR‑mutated lung adenocarcinoma (Review)". Oncology Letters 24.5 (2022): 408.
Chicago
Sun, R., Hou, Z., Zhang, Y., Jiang, B."Drug resistance mechanisms and progress in the treatment of EGFR‑mutated lung adenocarcinoma (Review)". Oncology Letters 24, no. 5 (2022): 408. https://doi.org/10.3892/ol.2022.13528