
One-quarter of global cancer deaths are caused by lung cancer (Sharma
EGFR belongs to the ERBB gene family (Wee and Wang, 2017). ERBB family kinases consist of four types i) EGFR/ERBB1/HER1, ii) NEU/ERBB2/HER2, iii) ERBB3/HER3, and iv) ERBB4/HER4, and are activated by ligand binding to the receptor to form homo-dimer or hetero-dimers (Table 1) (Finigan
Table 1 ERBB family receptors and ligands
Receptor | Ligand |
---|---|
ERBB1/HER1/EGFR | EGF |
TGF-α | |
Amphiregulin | |
Betacellulin | |
Epiregulin | |
ERBB2/HER2 | No ligand |
ERBB3/HER3 | Neuregulin 1, 2 |
ERBB4/HER 4 | Betacellulin |
Heparin-binding EGF | |
Epiregulin | |
Neuregulin 1-4 |
EGFR is a glycoprotein with 170 kDa and is a member of the receptor tyrosine kinase. EGFR contains three domains: i) an extracellular ligand-binding domain, ii) a transmembrane domain, and iii) an intracellular domain that contains an ATP-binding active site with a tyrosine moiety (Roskoski, 2014). The extracellular domain comprises 621 amino acids and is subdivided into four regions (exons 1-16). EGFR activation begins with ligand binding, followed by homo- or hetero-dimerization, resulting in tyrosine phosphorylation which is located in the intracellular tyrosine kinase domain (Carpenter
EGF binds to EGFR and forms homo- or hetero-dimerization with other ERBB members, leading to EGFR receptor phosphorylation and the activation of downstream pathways (Ferguson
EGFR plays a significant role in stimulating lung cancer generation and propagation (Ferlay
EGFR L858R and Del19 mutations are the two major driving mutations, and account for somatic mutations in the kinase domain of the EGFR gene related to lung adenocarcinoma. A total of 35% of Asians and 15% of Caucasians suffering from lung adenocarcinoma have EGFR-related mutations (Kobayashi and Mitsudomi, 2016). According to the COSMIC database, nearly 590 types of epithermal growth factor receptor mutations have been reported to date. The majority of these genes are present in the first four exons (18-21) of the tyrosine kinase domain (Fig. 2) (Kosaka
After EGF ligand binding to the EGFR, the C-helix conformation changes from outward to inward. This conformational change activates the enzyme from an inactive form to an active conformation (Purba
During the early 1970s, lung cancer was considered as one unit arising from the lung. Later, chemotherapy combination studies of all patients with lung cancer, irrespective of subtypes such as SCLC or NSCLC, resulted in a minimum benefit to the patients, with the overall survival rate less than seven months (Pao and Chmielecki, 2010). During the early 2000s, a groundbreaking chemotherapy trial with platinum doublets studied in all patients with NSCLC revealed no particular benefit in the overall survival rate of the patients (Sandler
Research has confirmed that almost 85% of EGFR mutations in NSCLC consist of Del19 (deletions in exon 19) and L858R point mutations (within exon 21). Several small molecule inhibitors have been designed and evaluated for their inhibitory activity against EGFR mutant kinases. These kinase inhibitors belong to competitive inhibitors as they bind to the intracellular active region of the tyrosine kinase domain, where ATP binds to the kinase activation (Du and Lovly, 2018). Anilinoquinazoline-based derivatives, such as gefitinib and erlotinib (Fig. 3), are first-generation EGFR inhibitors and are reversible inhibitors. Gefitinib and erlotinib exhibit significant potency against driving mutations (Del19 and L858R) (Eck and Yun, 2010). The treatment of NSCLC patients with first-generation EGFR inhibitors led to a positive response rate of approximately 60%-80%. Although these small molecule inhibitors showed promising results, most of the patients who responded to this treatment faced drug resistance after nearly 10-12 months of treatment (Karachaliou
To overcome the T790M mutation arising from the treatment of first-generation inhibitors, several irreversible 4-anilinoquinazoline analogs have been developed.
The second-generation EGFR TKIs such as afatinib, dacomitinib, neratinib, canertinib, and pelitinib (Fig. 4) have electrophilic acrylamide to form covalent interactions with Cys797 at the ATP-binding site of EGFR (Yu and Riely, 2013). The anilinoquinazoline moiety forms a hinge binding within the ATP-binding domain, providing a potential interaction with the gatekeeper Met790 residue and shows nanomolar potency toward EGFR Del19/T790M and EGFR L858R/T790M. However, the clinical benefits were compromised due to poor selectivity toward wild-type EGFR. Second-generation EGFR TKIs suffer intolerable low maximum tolerable dose due to dose-limiting toxicity, resulting from targeting wild-type EGFR and the repulsion created by the gatekeeper mutation T790M (Lin
Third-generation irreversible EGFR inhibitors, namely osimertinib and lazertinib, are potent against mutated EGFR and display encouraging efficacy in NSCLC patients who are resistant to first- and second-generation inhibitors. They selectively target Del19 and L858R mutations, as well as the gatekeeper T790M-resistance mutation, by forming a covalent bond with the C797 residue, located in the ATP-binding site of mutant EGFR (Grabe
Lazertinib showed promising antitumor activity in patients with tumors that had activated EGFR mutations and T790M resistance mutations. Lazertinib displayed less efficacy against wild-type EGFR than osimertinib, indicating that lazertinib might have fewer off-target side effects than osimertinib. Moreover, in a murine brain metastasis model, lazertinib inhibited intracranial tumor growth more efficiently than osimertinib. Clinical data have shown that lazertinib might be more effective in treating lung cancer with brain metastasis (Lategahn
The C797S mutation in EGFR is one of the mechanisms of drug resistance to third-generation EGFR TKIs such as osimertinib and lazertinib. Recent study findings have shown that C797S/T790M is in
The C797S point mutation is one of the major problems for NSCLC patients who are treated with currently available EGFR TKIs. According to updated clinical studies, approximately 40% of patients have developed C797S mutations after treatment with third-generation EGFR TKIs. Globally, many pharmaceutical companies are focused on the development of small molecules to treat EGFR triple mutant NSCLC (Table 2).
Table 2 Fourth-generation EGFR inhibitors
Compound | Structure | Clinical trails | Reference |
---|---|---|---|
BLU-945 | Undisclosed | Phase 1 | Schalm |
BBT-176 | Undisclosed | Phase 1 |
The appearance of the C797S point mutation in EGFR prevented clinical treatment with third-generation inhibitors that are effective against EGFR T790M mutation. The C797S mutation prohibited the formation of covalent bonds with the Michael acceptor of the third-generation inhibitors. Because of this drawback, there is an immediate requirement for the development of next-generation EGFR inhibitors targeting the triple mutant L858R/T790M/C797S and Del19/T790M/C797S (Lu
These two small molecules showed activity in biochemical assays against L858R/T790M and L858R/T790M/C797S mutations (Jia
Allosteric EGFR inhibitors successfully inhibited the L858R/T790M/C797S mutation in an
As a promising alternative to inhibition, targeted protein degradation (TPD) using small molecules has emerged as a novel therapeutic approach. The TPD mechanism depends on degrader molecules that bind the protein target of interest and recruit it to an E3 ubiquitin ligase. This contiguity with E3 ligase affects target polyubiquitination and subsequent proteasomal degradation (Lai and Crews, 2017).
DDC-01-163 (Fig. 7) was developed as an allosteric degrader selective for EGFR triple mutants. It successfully inhibited the proliferation of EGFR mutant Ba/F3 cells (L858R/T790M) with no activity against the wild-type. Furthermore, the mechanism of allosteric degrader activity is independent of cetuximab. This result represents a noteworthy advance over existing EGFR allosteric inhibitors (De Clercq
DDC-01-163 induced the selective degradation and inhibition of mutant EGFR cells in a dose-dependent manner and was especially inactive toward wild-type EGFR. The human cancer cell line, H1975, also displayed consistent results. These encouraging results suggest that the development of allosteric PROTACs for EGFR could assist a wide range of patients with EGFR L858R/T790M/C797S mutations (Jang
Gefitinib and erlotinib are first-generation reversible EGFR TKIs. They are quinazoline-based derivatives that act as ATP competitive inhibitors binding reversibly to the tyrosine kinase pocket of EGFR. Both drugs exhibited high inhibitory activity against wild-type and mutant EGFR. However, they ultimately develop resistance due to secondary mutations (Wang
Afatinib and dacomitinib are irreversible EGFR TKIs. Although their structures are similar to gefitinib or erlotinib with a quinazoline backbone, their unique acrylate side chain feature binds covalently to the C797 of EGFR, creating irreversible inhibition of the EGFR tyrosine kinase. Afatinib and dacomitinib demonstrated
Osimertinib is an irreversible third-generation EGFR TKI. The key core structure changed from quinazolin to pyrimidine. Its acrylate side chain was introduced toward the solvent region to form covalent bonds with C797. Osimertinib potently inhibits T790M-positive tumors and spares wild-type EGFR. Osimertinib effectively inhibited EGFR-sensitizing and T790M-resistance mutations. It has also been approved as a first-line treatment for patients with EGFR mutation-positive NSCLC. Treatment was limited by generating third mutation C797S, which causes resistance to osimertinib (Kishikawa
Table 3 Summary of EGFR inhibitors
EGFR | EGFR mutations | Inhibition mode | |||
---|---|---|---|---|---|
del_19 L858R | del_19/T790M L858R/T790M | del_19/T790M/C797S L858R/T790M/C797S | |||
Gefitinib (First Gen.) | Sensitive | Sensitive | Resistant | Resistant | Reversible |
Afatinib (Second Gen.) | Sensitive | Sensitive | Sensitive | Resistant | Irreversible |
Osimertinib (Third Gen.) | Resistant | Sensitive | Sensitive | Resistant | Irreversible |
(Fourth Gen.) | Resistant | Sensitive | Sensitive | Sensitive |
TKIs are standard treatment options for NSCLC with EGFR mutations (Zarogoulidis
NSCLC patients treated with first-generation EGFR TKIs have shown a dramatic response. Retrospective analyses of first-generation EGFR TKI trials have reported response rates of 77% in patients with sensitizing EGFR mutations (L858R and Del19). However, resistance was acquired by developing the secondary mutation T790M. The development of second-generation TKIs has shown encouraging results for secondary mutations. Conversely, inhibition of wild-type EGFR caused adverse events such as rash, acne, and diarrhea.
Irreversible third-generation EGFR TKIs have better efficacy and safety profiles by sparing wild-type EGFR than other inhibitors. However, the treatment was limited by the third mutation C797S. The mechanism of resistance to third-generation EGFR TKIs is more heterogeneous than that of other inhibitors. In many cases, the putative mechanism of resistance has not yet been identified.
In conclusion, treatment options for third-generation EGFR TKIs resistant are not available. Patients with EGFR triple mutations may receive subsequent therapies, such as immunotherapy and cytotoxic chemotherapy. Nonetheless, several areas of unmet medical needs remain to be addressed to provide treatment options for EGFR-related NSCLC patients.
The authors gratefully acknowledge the generous financial support provided by the Korea Research Institute of Chemical Technology (KK2131-30).
![]() |
![]() |