Targeted Therapy for NSCLC Patients with EGFR Mutations: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , , ,

Oncogenic mutations within the epidermal growth factor receptor (EGFR) kinase domain are well-established driver mutations in non–small cell lung cancer (NSCLC). Small-molecule tyrosine kinase inhibitors (TKIs) specifically targeting these mutations have improved treatment outcomes for patients with this subtype of NSCLC. Multiple targeted agents, including first-, second- and third-generation EGFR TKIs, have been approved or are under active investigation for patients with EGFR-mutant NSCLC. The first-generation TKIs, including erlotinib and gefitinib, are reversible inhibitors, binding both to mutant as well as wild-type (WT) EGFR. The second-generation TKIs, including afatinib and dacomitinib, are irreversible inhibitors that covalently bind to EGFR. The third-generation EGFR TKIs, including osimertinib (AZD9291), aumolertinib (HS-10296) and alflutinib (AST2818), are irreversible inhibitors that selectively bind to mutant EGFR and show greater efficacy than the first- and second-generation TKIs. Osimertinib has been approved by the FDA for both frontline and second-line treatment of NSCLC with EGFR-sensitizing mutations. Aumolertinib (HS-10296) and alflutinib (AST2818) have been approved for the treatment of EGFR-mutant NSCLC in China. Acquired resistance inevitably occurs, and a promising new generation of EGFR-targeting agents is under investigation.

  • non–small cell lung cancer
  • EGFR
  • structure
  • targeted therapy

1. Classical Mutations

Ex19Del and L858R are found in 71.9% of NSCLCs with an EGFR mutation. Ex19Del and L858R are the sole EGFR mutations in 34.3% and 28.5%, respectively, while co-occurrence of Ex19Del or L858R with uncommon EGFR mutations accounts for 6.0% and 3.1% of cases (Figure 1a). Ex19Del and L858R as classical mutations confer sensitivity to all first-, second- and third-generation EGFR TKIs [1][2][3][4][5][6][7] (Figure 2), and co-occurrence of an uncommon mutation does not impact TKI sensitivity [8][9]. In fact, patients with complex “classical” mutations (Ex19Del or L858R in complex with an uncommon EGFR mutation) might have more favorable clinical outcomes than patients with uncommon mutations alone [10][11]. The researchers have not found any classical + classical complex mutations in the analysis of GENIE dataset. The sensitivity of this class of complex mutations might be equivalent to the corresponding single mutation, as L858R + Ex19Del complex mutations display similar patient outcomes with EGFR TKI treatment compared to patients with single classical mutations alone [12].
Figure 1. The frequencies of EGFR mutations in NSCLC. (a) Data were acquired from GENIE dataset (GENIE Cohort v12.0-public, n = 153,834). Data were filtered to contain mutations from NSCLC (n = 22,050). The common resistance mutations T790M and C797S were filtered out. (b) Pie chart showing the frequency of different uncommon EGFR mutations in NSCLC. (c) Pie chart showing the frequency of different uncommon complex EGFR mutations in NSCLC.
Figure 2. The position of EGFR mutations and sensitivity to EGFR TKIs based on exon-grouped classification. (a) Exons 18–21 in the tyrosine kinase region where the relevant mutations are located are expanded, and a detailed list of EGFR mutations in these exons is shown in the boxes below. TM, transmembrane; JM, juxtamembrane; KD, kinase domain. (b) The sensitivity to EGFR TKIs based on exon-grouped classification. Gray, insensitive to EGFR TKIs. Green, sensitive to EGFR TKIs in which the light-to-strong color represents weak-to-strong sensitivity.
Structurally, Leu858 lies within the two-turn helix of the A loop and packs against the αC helix in the “Src-like inactive” conformation. Arginine has a much larger side chain than leucine, and substitution of Leu858 with arginine is not tolerated with the inactive conformation [2]. The positively charged Arg858 is surrounded by a cluster of negatively charged residues (Glu758, Asp855, and Asp837), which stabilize the αC helix and the KE salt bridge. Therefore, the substitution holds the kinase domain in the active conformation likely at the expense of locally disordered conformation, rather than “Src-like inactive” conformation [13]. As a result, L858R mutation promotes the ligand-independent dimerization of EGFR kinase [14]. Structure-based approaches classify L858R into classical-like mutations, as Arg858 is located at the N-terminal portion of the A loop in the C-lobe, which is far from the ATP-binding pocket [15]. A spectrum of rare L858R + mutations were also categorized into classical-like mutations [15]. This subgroup of mutations is sensitive to all first-, second- and third-generation EGFR TKIs (Figure 3).
Figure 3. The positions and structure-based classification of EGFR mutations. (a) A detailed list of EGFR mutations in each exon are shown in the below boxes. Different colors of each box represent different classes of mutations following structure-based classification. Ex20ins-L, exon 20 loop insertions; Ex20ins-NL, exon 20 near-loop insertions; Ex20ins-FL, exon 20 far-loop insertions; PACC, P-loop and αC-helix compressing mutations. (b) The sensitivity to EGFR TKIs of each group of mutations based on structure-based classification.
Ex19Del mutations shorten the length of β3-αC loop and suppress the locally disordered conformation [13], and therefore constrain the αC helix to the active conformation [3], with the KE salt bridge making the interaction between the kinase domain and ATP favorable [16]. There have been more than 20 Ex19Del variants identified in NSCLC [17]. The length of the β3–αC loop functions as a rheostat for kinase activity. Specifically, deletion of five amino acids (E746_A760) makes an optimal orientation of the αC helix for catalytic activity, while other deletions also result in structural perturbations of the αC helix that impair kinase activity compared to WT EGFR [3]. This conformational difference leads to variable ability to form ligand-independent dimerization and therefore differential sensitivity to EGFR TKIs [17][18]. Clinically, variations in Ex19Del are translated into differential clinical features and treatment outcomes [19]. Ex19Del mutations were classified into classical-like mutations as, structurally, the β3–αC loop in the N-lobe is far from the ATP-binding pocket [15]. A spectrum of Ex19Del + rare mutations were also categorized into classical-like mutations (Figure 3).
Ex19Del and L858R mutants have a lower competitive binding affinity for ATP than WT EGFR. First-generation EGFR TKIs bind more favorably to the kinase domain of mutants than WT, thereby inhibiting mutant kinase activities in a therapeutically useful manner [2]. The substitution of Thr790 to the bulky methionine increases the ATP affinity and stabilizes the active conformation of EGFR kinase domain, which sterically prevents the binding of first-generation TKIs, thereby imparting drug resistance [20]. Second-generation TKIs bind irreversibly to EGFR Cys797 at the margin of the ATP-binding cleft by covalent adduct formation, and were anticipated to overcome acquired resistance caused by T790M mutation [21]. However, the poor selectivity of second-generation TKIs for T790M over WT EGFR results in dose-limiting toxicities that limit clinical efficacy [6]. Osimertinib has a differential binding pose with Met790 from Thr790 of WT EGFR, thus yielding greater selectivity for T790M mutation over WT EGFR than afatinib [22]. Greater selectivity has translated into more favorable treatment outcomes, as either second-line or first-line therapy, than earlier-generation EGFR TKIs [23][24].

2. Uncommon EGFR Mutations

2.1. Exon 18 Mutations

EGFR exon 18 mutations include E709_T710insX (e.g., exon 18 deletion, Ex18Del), E709X and G719X (Figure 1b). From the analysis of the GENIE dataset, either E709X or E709-T710insX as a single EGFR mutation accounts for around 0.68% of all EGFR mutations and 1.7% of rare EGFR mutations in NSCLC (data not shown). Around 96% of E709X co-occurs with another EGFR mutation, such as G719X or L861X. G719X mutations as independent EGFR mutations, including G719A, G719C, G719S, G719D and G719V, and account for 4.0% of rare EGFR mutations and 1.6% of all EGFR mutations in NSCLC. G719X mutations also co-occur with other EGFR mutations, which represent 8.4% of rare EGFR mutations and 3.2% of all EGFR mutations in NSCLC (Figure 1c).
Preclinical studies show that exon 18 mutations including E709K, E709_T710insD and G719A are sensitive to first-, second- and third-generation EGFR TKIs, but show less sensitivity than Ex19Del [25] (Figure 2). Exon18 mutations tend to be more sensitive to second-generation TKIs than first- and third-generation EGFR TKIs [8][17][18]. E709K and G719A tend to be more sensitive to EGFR TKIs than E709-T710insD [8][17]. Clinically, patients with G719X and Ex18Del respond poorly to first-generation TKIs [25], and patients with G719X also respond poorly to the third-generation TKI osimertinib [19]. Few clinical data about second- and third-generation TKIs exist for E709X as a single EGFR mutation.
Both Glu709 and Gly719 are located in the N-lobe. Glu709 is located on the N-terminal end of the β1 strand, which is immediately followed by the P loop. Gly719 is the first glycine in the evolutionarily conserved “GXGXXG” motif of the P loop. Substitution of Gly719 to non-glycine residues decreases the flexibility of the P loop and attenuates the hydrophobic interactions that constrain the αC-helix in the inactive conformation [2]. Therefore, G719X mutations destabilize the inactive conformation and are well accommodated in the active conformation [2][13]. This conformation increases the propensity for dimerization and subsequent receptor activation. Exon 18 mutations, including E709X, G719X and Ex18Del mutations, were categorized as PACC mutations [15] (Figure 3). Structurally, PACC mutations change the flexibility of the P loop and destabilize an osimertinib-binding pattern. By contrast, second-generation TKIs do not interact with the P loop and favor the binding pose. Therefore, PACC mutations are more sensitive to the second-generation TKIs than other-generation TKIs [15]. Clinically, patients with G719X mutations are more responsive to second-generation EGFR TKIs than first- and third-generation TKIs [21][26][27]. Similarly, patients who develop resistance to osimertinib via a mutation in Gly724, the third glycine in the “GXGXXG” motif, often respond to second-generation EGFR TKIs [28][29][30][31], but not first-generation TKIs [32]. The variable sensitivity of different exon 18 mutations observed in previous studies might be explained by the differential flexibility of the P loop induced by mutations.

2.2. Exon 19 Mutations

The researchers found 13 cases with L747P mutation in the GENIE dataset. The researchers also found 5 cases with L747X complex mutations, including L747F + L861Q (1 case), L747S + G719C (2 cases) and L747A + I744M (2 cases). The researchers have not found exon 19 insertions in the GENIE dataset, while this subset of mutations has been reported to account for approximately 1% of EGFR-mutant NSCLC [9]. Available clinical data indicate that L747P/S mutations are resistant to first- and third-generation EGFR TKIs [33][34] and confer sensitivity to second-generation EGFR TKIs [35][36]. Consistent with this evidence, L747X mutations were included in PACC mutations, as substitution of Leu747 to proline or serine might alter the rigidity of the β3–αC loop, which stabilizes the αC-helix and KE salt bridge interactions in the active conformation [37].

2.3. Exon 20 Mutations

EGFR exon 20 insertion (Ex20ins) mutations contain a broad spectrum of small insertions of 1–7 amino acids that occur within the C-terminal end of the αC-helix and the immediately following loop [38]. After classical mutations, Ex20ins mutations are the next most prevalent EGFR mutation in NSCLC [39]. From the GENIE dataset, the researchers found more than 60 different forms of Ex20ins mutations, accounting for 16% of rare EGFR mutations and 6.2% of all EGFR mutations in NSCLC (Figure 1). The researchers also found other forms of exon 20 mutations, including S768I (3 cases), V774L (1 case) and Q787L (1 case) (Figure 2). Exon 20 mutations also co-occur with other EGFR mutations (Figure 1c).
Variable TKI sensitivity has been found in different types of Ex20ins mutations [40][41]. Most patients harboring Ex20ins mutations are insensitive to most of the FDA-approved EGFR TKIs [42][43][44][45]. However, a specific subset of insertions within the C-terminal portion of the αC-helix, for example, A763_Y764insFQEA, are sensitive to first-generation EGFR TKIs [46]. Multiple reports have demonstrated A763_Y764insFQEA is also responsive to second- and third-generation EGFR TKIs with comparable sensitivity to classical mutations [44][47][48]. For the rest of the Ex20ins mutations, most of them do not respond to first- and third-generation TKIs [49], but are sensitive to second-generation TKIs [50]. Exceptionally, the three insertion mutations (D770_N771insG, D770>GY, and N771_P772insN) are at least partially responsive to first-generation EGFR TKIs [49].
Structural variations in Ex20ins mutations lead to the heterogeneity of TKI sensitivity. The insertions form a wedge that “pushes” the αC-helix and hold the αC in the active conformation [46][51], leading to constitutive activation of the receptor [46][51]. Sequence and conformational variation in the αC–β4 loop may function as a rheostat that regulates the kinase activity via modulating the αC-helix conformation [51]. Exceptionally, the insertion A763_764insFQEA that occurs within the αC-helix differs from insertions within the αC–β4 loop, and may constitutively activate the receptor most similarly to classical mutations, rather than other Ex20ins mutations. As might be expected, A763_764insFQEA is sensitive to first-, second- and third-generation EGFR TKIs [44][47][48], and is classified as a classical-like mutation. Other Ex20ins mutations are categorized as a unique class called Ex20-L mutations [15]. Ex20ins-L are sensitive only to select second-generation TKIs such as poziotinib [52], and Ex20ins-active TKIs such as CLN-081 [53] and mobocertinib [54]. Ex20ins-L mutations could be further subdivided into Ex20ins near αC-β4 loop (Ex20ins-NL) and far αC–β4 loop insertions (Ex20ins-FL). Ex20ins-NL is more sensitive to second-generation and Ex20ins-active TKIs than Ex20ins-FL (Figure 3).
Patients with single S768I mutation have variable responses to first-generation EGFR TKIs [45][55][56][57]. Patients whose cancers harbor theS768I mutation responded to afatinib [42], leading to FDA approval of this drug for this subset of patients. Notably, S768I usually co-occurs with additional EGFR mutations (7/8 cases in this study), making the response of the single S768I mutation to afatinib ambiguous. Co-occurrence of S768I with additional EGFR mutations might impact TKI sensitivity [58][59]. One patient with S768I+V769L, for example, was resistant to afatinib [60]. S768I-complex mutations are sensitive to the third-generation EGFR TKI osimertinib [61][62], though little data exists on single-mutant S768I sensitivity. S768I and its complex mutations are classified as PACC mutations. Ser768 is located at the αC–β4 loop. Substitution of Ser768 to isoline improves the hydrophobic interactions between the αC helix and the adjacent β9 strand, which might enthalpically stabilize the “αC-in” active conformation [13]. As such, S768I and its complex mutations are classified as PACC mutations (Figure 3).
Finally, little clinical or structural data exist for V774L and Q787L, though V774M and its complex mutations are classified as PACC mutations.

2.4. Exon 21 Mutations

Apart from L858R mutation, the researchers found another exon 21 mutation L861X, including L861Q and L861R, in 2.3% of all EGFR mutations and 5.7% of rare EGFR mutations in the GENIE lung cancer dataset (Figure 1b). L861X mutations were also found to co-occur with additional EGFR mutations (Figure 1c). Preclinical and clinical studies show that L861Q mutation has an intermediate sensitivity to first-generation EGFR TKIs comparable to S768I and G719X mutations [26][63][64] (Figure 2). Patients with L861Q mutation are responsive to the second-generation EGFR TKI afatinib [42], which led to FDA approval of afatinib for this subtype [1]. In preclinical studies, L861Q mutations are sensitive to the third-generation EGFR TKI osimertinib [63], and a phase II trial reported patients with L861Q having partial responses to osimertinib [61]. Structurally, substitution of Leu861 to Gln allows the formation of new hydrogen bonds near the C-terminal of the αC helix that might enthalpically stabilize the “αC-in” active conformation [13]. L861X mutation and its complex mutations have been classified as PACC mutations (Figure 3).

2.5. EGFR Kinase Domain Duplication

In-frame, tandem duplication of EGFR exons 18–25, encoding EGFR kinase domain duplication (EGFR-KDD), has been identified in patients with NSCLC [65], with cases of a duplication of exons 14–26, 17–25 and 18–26 also reported in NSCLC [66]. EGFR-KDD occurs in 0.2–0.24% of all EGFR-mutant NSCLC patients [66][67][68]. Current evidence suggests that EGFR-KDD is sensitive to multiple EGFR TKIs [65][66]. Case studies show that NSCLC patients with EGFR-KDD can have durable partial response to either first-line [66] or second-line treatment with gefitinib [69], though at least one patient with EGFR-KDD did to respond to first-line gefitinib [66]. Case studies also show that NSCLC patients with EGFR-KDD are at least partially responsive to afatinib treatment [65][70], and the third-generation TKI osimertinib has been found to be effective in patients with EGFR-KDD [71][72].
Structure–function studies indicate that EGFR-KDD can form ligand-independent intramolecular asymmetric dimers in which one kinase domain functions as “activator” (donor) and the other functions as “receiver” (acceptor), and ligand-dependent intermolecular asymmetric dimers and higher-order oligomer. The linker between the two kinase domains provides additional enthalpic stabilization to promote activation. Preclinical studies show that the inhibition of EGFR-KDD activity can be achieved by the inhibition of intramolecular kinase activity (afatinib) and intermolecular kinase activity (cetuximab) [73]. Compared to gefitinib, structural studies predict osimertinib to bind more favorably with, and thus better inhibit, EGFR-KDD [74]. More structure–function studies are required to understand the detailed conformations when EGFR-KDD binds to the available TKIs, so that the researchers can classify this type of mutations based on the structure-based grouping.

2.6. Uncommon Complex Mutations

The researchers found uncommon complex mutations (rare + rare) occur in 36.5% of rare EGFR mutations and 6.7% of all EGFR mutations in the GENIE lung cancer dataset (Figure 1C). Of them, G719X complex mutations are the most common subtype of mutations and account for almost a half of all uncommon complex mutations (Figure 1C), with G719X + S768I most common one (Table 1). E709X complex mutations are the second most common of this subtype of mutations and account for 14.6% of all uncommon complex mutations (Figure 1C). Interestingly, all E709X mutations co-occur with G719X mutations (Table 1). The researchers also found Ex20ins, L861X, L858X and S768I complex mutations (Figure 1C, Table 2).
Table 1. Complex mutations identified in exon 18 from GENIE lung cancer dataset.
Table 2. Complex mutations identified in exon 20 and 21 from GENIE lung cancer dataset.
Uncommon EGFR complex mutations encompass a wide range of patient responses to EGFR TKIs. Structure-based approaches classify the majority of uncommon EGFR complex mutations as PACC mutations, including E709X + G719X, G719X + rare and S768I + rare, like their single mutation partners. These complex mutations tend to yield more favorable patient outcomes in response to TKIs than the single rare mutations alone [43][64][75][76]. Interestingly, the sensitivity to EGFR TKIs is likely influenced by the specific co-occurring partner mutation. For example, patients with G719X + S768I mutations have dramatically different clinical outcomes compared to patients with G719X+L861Q mutations [64]. Single S768I mutation might not be sensitive to erlotinib, but co-occurrence with sensitizing EGFR mutations will confer sensitivity to the single S768I mutation [58].

This entry is adapted from the peer-reviewed paper 10.3390/biom13020210

References

  1. Mok, T.S.; Wu, Y.L.; Thongprasert, S.; Yang, C.H.; Chu, D.T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957.
  2. Yun, C.H.; Boggon, T.J.; Li, Y.; Woo, M.S.; Greulich, H.; Meyerson, M.; Eck, M.J. Structures of lung cancer-derived EGFR mutants and inhibitor complexes: Mechanism of activation and insights into differential inhibitor sensitivity. Cancer Cell 2007, 11, 217–227.
  3. Foster, S.A.; Whalen, D.M.; Ozen, A.; Wongchenko, M.J.; Yin, J.; Yen, I.; Schaefer, G.; Mayfield, J.D.; Chmielecki, J.; Stephens, P.J.; et al. Activation Mechanism of Oncogenic Deletion Mutations in BRAF, EGFR, and HER2. Cancer Cell 2016, 29, 477–493.
  4. Lu, S.; Wang, Q.; Zhang, G.; Dong, X.; Yang, C.T.; Song, Y.; Chang, G.C.; Lu, Y.; Pan, H.; Chiu, C.H.; 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. 2022, 17, 411–422.
  5. Shi, Y.; Zhang, S.; Hu, X.; Feng, J.; Ma, Z.; Zhou, J.; Yang, N.; Wu, L.; Liao, W.; Zhong, D.; et al. Safety, Clinical Activity, and Pharmacokinetics of Alflutinib (AST2818) in Patients with Advanced NSCLC With EGFR T790M Mutation. J. Thorac. Oncol. 2020, 15, 1015–1026.
  6. Zhou, W.; Ercan, D.; Chen, L.; Yun, C.H.; Li, D.; Capelletti, M.; Cortot, A.B.; Chirieac, L.; Iacob, R.E.; Padera, R.; et al. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature 2009, 462, 1070–1074.
  7. Schuler, M.; Yang, J.C.; Park, K.; Kim, J.H.; Bennouna, J.; Chen, Y.M.; Chouaid, C.; De Marinis, F.; Feng, J.F.; Grossi, F.; et al. Afatinib beyond progression in patients with non-small-cell lung cancer following chemotherapy, erlotinib/gefitinib and afatinib: Phase III randomized LUX-Lung 5 trial. Ann. Oncol. 2016, 27, 417–423.
  8. Bazhenova, L.; Minchom, A.; Viteri, S.; Bauml, J.M.; Ou, S.I.; Gadgeel, S.M.; Trigo, J.M.; Backenroth, D.; Li, T.; Londhe, A.; et al. Comparative clinical outcomes for patients with advanced NSCLC harboring EGFR exon 20 insertion mutations and common EGFR mutations. Lung Cancer 2021, 162, 154–161.
  9. He, M.; Capelletti, M.; Nafa, K.; Yun, C.H.; Arcila, M.E.; Miller, V.A.; Ginsberg, M.S.; Zhao, B.; Kris, M.G.; Eck, M.J.; et al. EGFR exon 19 insertions: A new family of sensitizing EGFR mutations in lung adenocarcinoma. Clin. Cancer Res. 2012, 18, 1790–1797.
  10. Baek, J.H.; Sun, J.M.; Min, Y.J.; Cho, E.K.; Cho, B.C.; Kim, J.H.; Ahn, M.J.; Park, K. Efficacy of EGFR tyrosine kinase inhibitors in patients with EGFR-mutated non-small cell lung cancer except both exon 19 deletion and exon 21 L858R: A retrospective analysis in Korea. Lung Cancer 2015, 87, 148–154.
  11. Kohsaka, S.; Nagano, M.; Ueno, T.; Suehara, Y.; Hayashi, T.; Shimada, N.; Takahashi, K.; Suzuki, K.; Takamochi, K.; Takahashi, F.; et al. A method of high-throughput functional evaluation of EGFR gene variants of unknown significance in cancer. Sci. Transl. Med. 2017, 9, eaan6566.
  12. Xu, J.; Jin, B.; Chu, T.; Dong, X.; Yang, H.; Zhang, Y.; Wu, D.; Lou, Y.; Zhang, X.; Wang, H.; et al. EGFR tyrosine kinase inhibitor (TKI) in patients with advanced non-small cell lung cancer (NSCLC) harboring uncommon EGFR mutations: A real-world study in China. Lung Cancer 2016, 96, 87–92.
  13. Shan, Y.; Eastwood, M.P.; Zhang, X.; Kim, E.T.; Arkhipov, A.; Dror, R.O.; Jumper, J.; Kuriyan, J.; Shaw, D.E. Oncogenic mutations counteract intrinsic disorder in the EGFR kinase and promote receptor dimerization. Cell 2012, 149, 860–870.
  14. Red Brewer, M.; Yun, C.H.; Lai, D.; Lemmon, M.A.; Eck, M.J.; Pao, W. Mechanism for activation of mutated epidermal growth factor receptors in lung cancer. Proc. Natl. Acad. Sci. USA 2013, 110, E3595–E3604.
  15. Robichaux, J.P.; Le, X.; Vijayan, R.S.K.; Hicks, J.K.; Heeke, S.; Elamin, Y.Y.; Lin, H.Y.; Udagawa, H.; Skoulidis, F.; Tran, H.; et al. Structure-based classification predicts drug response in EGFR-mutant NSCLC. Nature 2021, 597, 732–737.
  16. Tamirat, M.Z.; Koivu, M.; Elenius, K.; Johnson, M.S. Structural characterization of EGFR exon 19 deletion mutation using molecular dynamics simulation. PLoS ONE 2019, 14, e0222814.
  17. Brown, B.P.; Zhang, Y.K.; Kim, S.; Finneran, P.; Yan, Y.; Du, Z.; Kim, J.; Hartzler, A.L.; LeNoue-Newton, M.L.; Smith, A.W.; et al. Allele-specific activation, enzyme kinetics, and inhibitor sensitivities of EGFR exon 19 deletion mutations in lung cancer. Proc. Natl. Acad. Sci. USA 2022, 119, e2206588119.
  18. Truini, A.; Starrett, J.H.; Stewart, T.; Ashtekar, K.; Walther, Z.; Wurtz, A.; Lu, D.; Park, J.H.; DeVeaux, M.; Song, X.; et al. The EGFR Exon 19 Mutant L747-A750>P Exhibits Distinct Sensitivity to Tyrosine Kinase Inhibitors in Lung Adenocarcinoma. Clin. Cancer Res. 2019, 25, 6382–6391.
  19. Zhao, C.; Jiang, T.; Li, J.; Wang, Y.; Su, C.; Chen, X.; Ren, S.; Li, X.; Zhou, C. The impact of EGFR exon 19 deletion subtypes on clinical outcomes in non-small cell lung cancer. Transl. Lung Cancer Res. 2020, 9, 1149–1158.
  20. Yun, C.H.; Mengwasser, K.E.; Toms, A.V.; Woo, M.S.; Greulich, H.; Wong, K.K.; Meyerson, M.; Eck, M.J. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl. Acad. Sci. USA 2008, 105, 2070–2075.
  21. Yu, H.A.; Pao, W. Targeted therapies: Afatinib--new therapy option for EGFR-mutant lung cancer. Nat. Rev. Clin. Oncol. 2013, 10, 551–552.
  22. Yan, X.E.; Ayaz, P.; Zhu, S.J.; Zhao, P.; Liang, L.; Zhang, C.H.; Wu, Y.C.; Li, J.L.; Choi, H.G.; Huang, X.; et al. Structural Basis of AZD9291 Selectivity for EGFR T790M. J. Med. Chem. 2020, 63, 8502–8511.
  23. Soria, J.C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 113–125.
  24. Cho, B.C.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Sriuranpong, V.; Imamura, F.; Nogami, N.; Kurata, T.; Okamoto, I.; Zhou, C.; et al. Osimertinib versus Standard of Care EGFR TKI as First-Line Treatment in Patients with EGFRm Advanced NSCLC: FLAURA Asian Subset. J. Thorac. Oncol. 2019, 14, 99–106.
  25. Yasuda, H.; Ichihara, E.; Sakakibara-Konishi, J.; Zenke, Y.; Takeuchi, S.; Morise, M.; Hotta, K.; Sato, M.; Matsumoto, S.; Tanimoto, A.; et al. A phase I/II study of osimertinib in EGFR exon 20 insertion mutation-positive non-small cell lung cancer. Lung Cancer 2021, 162, 140–146.
  26. Kobayashi, Y.; Mitsudomi, T. Not all epidermal growth factor receptor mutations in lung cancer are created equal: Perspectives for individualized treatment strategy. Cancer Sci. 2016, 107, 1179–1186.
  27. Bar, J.; Peled, N.; Schokrpur, S.; Wolner, M.; Rotem, O.; Girard, N.; Aboubakar Nana, F.; Derijcke, S.; Kian, W.; Patel, S.; et al. UNcommon EGFR Mutations: International Case Series on Efficacy of Osimertinib in Real-Life Practice in First-LiNe Setting (UNICORN). J. Thorac. Oncol. 2022; in press.
  28. Kobayashi, Y.; Togashi, Y.; Yatabe, Y.; Mizuuchi, H.; Jangchul, P.; Kondo, C.; Shimoji, M.; Sato, K.; Suda, K.; Tomizawa, K.; et al. EGFR Exon 18 Mutations in Lung Cancer: Molecular Predictors of Augmented Sensitivity to Afatinib or Neratinib as Compared with First- or Third-Generation TKIs. Clin. Cancer Res. 2015, 21, 5305–5313.
  29. Minari, R.; Leonetti, A.; Gnetti, L.; Zielli, T.; Ventura, L.; Bottarelli, L.; Lagrasta, C.; La Monica, S.; Petronini, P.G.; Alfieri, R.; et al. Afatinib therapy in case of EGFR G724S emergence as resistance mechanism to osimertinib. Anticancer Drugs 2021, 32, 758–762.
  30. Li, Y.; Lin, Y.; Wu, J.; Ye, F. Meningeal metastasis patients with EGFR G724S who develop resistance to osimertinib benefit from the addition of afatinib. Transl. Lung Cancer Res. 2020, 9, 2188–2190.
  31. Fang, W.; Huang, Y.; Gan, J.; Zheng, Q.; Zhang, L. Emergence of EGFR G724S After Progression on Osimertinib Responded to Afatinib Monotherapy. J. Thorac. Oncol. 2020, 15, e36–e37.
  32. Wei, Y.; Jiang, B.; Liu, S.; Zhang, Z.; Fang, W.; Yang, Y.; Li, X.; Zhao, J.; Zhao, H. Afatinib as a Potential Therapeutic Option for Patients with NSCLC With EGFR G724S. JTO Clin. Res. Rep. 2021, 2, 100193.
  33. Wang, Y.T.; Ning, W.W.; Li, J.; Huang, J.A. Exon 19 L747P mutation presented as a primary resistance to EGFR-TKI: A case report. J. Thorac. Dis. 2016, 8, E542–E546.
  34. Huang, J.; Wang, Y.; Zhai, Y.; Wang, J. Non-small cell lung cancer harboring a rare EGFR L747P mutation showing intrinsic resistance to both gefitinib and osimertinib (AZD9291): A case report. Thorac. Cancer 2018, 9, 745–749.
  35. Li, J.; Zhu, L.; Stebbing, J.; Peng, L. Afatinib treatment in a lung adenocarcinoma patient harboring a rare EGFR L747P mutation. J. Cancer Res. Ther. 2022, 18, 1436–1439.
  36. Li, Y.; Guo, W.; Jiang, B.; Han, C.; Ye, F.; Wu, J. Case Report: Dacomitinib is effective in lung adenocarcinoma with rare EGFR mutation L747P and brain metastases. Front. Oncol. 2022, 12, 863771.
  37. Yoshizawa, T.; Uchibori, K.; Araki, M.; Matsumoto, S.; Ma, B.; Kanada, R.; Seto, Y.; Oh-Hara, T.; Koike, S.; Ariyasu, R.; et al. Microsecond-timescale MD simulation of EGFR minor mutation predicts the structural flexibility of EGFR kinase core that reflects EGFR inhibitor sensitivity. NPJ Precis. Oncol. 2021, 5, 32.
  38. Meador, C.B.; Sequist, L.V.; Piotrowska, Z. Targeting EGFR Exon 20 Insertions in Non-Small Cell Lung Cancer: Recent Advances and Clinical Updates. Cancer Discov. 2021, 11, 2145–2157.
  39. Friedlaender, A.; Subbiah, V.; Russo, A.; Banna, G.L.; Malapelle, U.; Rolfo, C.; Addeo, A. EGFR and HER2 exon 20 insertions in solid tumours: From biology to treatment. Nat. Rev. Clin. Oncol. 2022, 19, 51–69.
  40. Lin, Y.T.; Shih, J.Y. Not All EGFR Exon 20 Insertions Are Created Equal. JTO Clin. Res. Rep. 2020, 1, 100069.
  41. Shi, C.; Xing, R.; Li, M.; Feng, J.; Sun, R.; Wei, B.; Guo, Y.; Ma, J.; Wang, H. Real-world clinical treatment outcomes in Chinese non-small cell lung cancer with EGFR exon 20 insertion mutations. Front. Oncol. 2022, 12, 949304.
  42. Yang, J.C.; Sequist, L.V.; Geater, S.L.; Tsai, C.M.; Mok, T.S.; Schuler, M.; Yamamoto, N.; Yu, C.J.; Ou, S.H.; Zhou, C.; et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: A combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol. 2015, 16, 830–838.
  43. Beau-Faller, M.; Prim, N.; Ruppert, A.M.; Nanni-Metellus, I.; Lacave, R.; Lacroix, L.; Escande, F.; Lizard, S.; Pretet, J.L.; Rouquette, I.; et al. Rare EGFR exon 18 and exon 20 mutations in non-small-cell lung cancer on 10 117 patients: A multicentre observational study by the French ERMETIC-IFCT network. Ann. Oncol. 2014, 25, 126–131.
  44. Naidoo, J.; Sima, C.S.; Rodriguez, K.; Busby, N.; Nafa, K.; Ladanyi, M.; Riely, G.J.; Kris, M.G.; Arcila, M.E.; Yu, H.A. Epidermal growth factor receptor exon 20 insertions in advanced lung adenocarcinomas: Clinical outcomes and response to erlotinib. Cancer 2015, 121, 3212–3220.
  45. Janning, M.; Suptitz, J.; Albers-Leischner, C.; Delpy, P.; Tufman, A.; Velthaus-Rusik, J.L.; Reck, M.; Jung, A.; Kauffmann-Guerrero, D.; Bonzheim, I.; et al. Treatment outcome of atypical EGFR mutations in the German National Network Genomic Medicine Lung Cancer (nNGM). Ann. Oncol. 2022, 33, 602–615.
  46. Yasuda, H.; Park, E.; Yun, C.H.; Sng, N.J.; Lucena-Araujo, A.R.; Yeo, W.L.; Huberman, M.S.; Cohen, D.W.; Nakayama, S.; Ishioka, K.; et al. Structural, biochemical, and clinical characterization of epidermal growth factor receptor (EGFR) exon 20 insertion mutations in lung cancer. Sci. Transl. Med. 2013, 5, 216ra177.
  47. Vasconcelos, P.; Gergis, C.; Viray, H.; Varkaris, A.; Fujii, M.; Rangachari, D.; VanderLaan, P.A.; Kobayashi, I.S.; Kobayashi, S.S.; Costa, D.B. EGFR-A763_Y764insFQEA Is a Unique Exon 20 Insertion Mutation That Displays Sensitivity to Approved and In-Development Lung Cancer EGFR Tyrosine Kinase Inhibitors. JTO Clin. Res. Rep. 2020, 1, 100051.
  48. Voon, P.J.; Tsui, D.W.; Rosenfeld, N.; Chin, T.M. EGFR exon 20 insertion A763-Y764insFQEA and response to erlotinib--Letter. Mol. Cancer Ther. 2013, 12, 2614–2615.
  49. Qin, Y.; Jian, H.; Tong, X.; Wu, X.; Wang, F.; Shao, Y.W.; Zhao, X. Variability of EGFR exon 20 insertions in 24 468 Chinese lung cancer patients and their divergent responses to EGFR inhibitors. Mol. Oncol. 2020, 14, 1695–1704.
  50. Kobayashi, I.S.; Viray, H.; Rangachari, D.; Kobayashi, S.S.; Costa, D.B. EGFR-D770>GY and Other Rare EGFR Exon 20 Insertion Mutations with a G770 Equivalence Are Sensitive to Dacomitinib or Afatinib and Responsive to EGFR Exon 20 Insertion Mutant-Active Inhibitors in Preclinical Models and Clinical Scenarios. Cells 2021, 10, 3561.
  51. Ruan, Z.; Kannan, N. Altered conformational landscape and dimerization dependency underpins the activation of EGFR by alphaC-beta4 loop insertion mutations. Proc. Natl. Acad. Sci. USA 2018, 115, E8162–E8171.
  52. Elamin, Y.Y.; Robichaux, J.P.; Carter, B.W.; Altan, M.; Tran, H.; Gibbons, D.L.; Heeke, S.; Fossella, F.V.; Lam, V.K.; Le, X.; et al. Poziotinib for EGFR exon 20-mutant NSCLC: Clinical efficacy, resistance mechanisms, and impact of insertion location on drug sensitivity. Cancer Cell 2022, 40, 754–767.e6.
  53. Udagawa, H.; Hasako, S.; Ohashi, A.; Fujioka, R.; Hakozaki, Y.; Shibuya, M.; Abe, N.; Komori, T.; Haruma, T.; Terasaka, M.; et al. TAS6417/CLN-081 Is a Pan-Mutation-Selective EGFR Tyrosine Kinase Inhibitor with a Broad Spectrum of Preclinical Activity against Clinically Relevant EGFR Mutations. Mol. Cancer Res. 2019, 17, 2233–2243.
  54. Gonzalvez, F.; Vincent, S.; Baker, T.E.; Gould, A.E.; Li, S.; Wardwell, S.D.; Nadworny, S.; Ning, Y.; Zhang, S.; Huang, W.S.; et al. Mobocertinib (TAK-788): A Targeted Inhibitor of EGFR Exon 20 Insertion Mutants in Non-Small Cell Lung Cancer. Cancer Discov. 2021, 11, 1672–1687.
  55. Leal, J.L.; Alexander, M.; Itchins, M.; Wright, G.M.; Kao, S.; Hughes, B.G.M.; Pavlakis, N.; Clarke, S.; Gill, A.J.; Ainsworth, H.; et al. EGFR Exon 20 Insertion Mutations: Clinicopathological Characteristics and Treatment Outcomes in Advanced Non-Small Cell Lung Cancer. Clin. Lung Cancer 2021, 22, e859–e869.
  56. Popat, S.; Hsia, T.C.; Hung, J.Y.; Jung, H.A.; Shih, J.Y.; Park, C.K.; Lee, S.H.; Okamoto, T.; Ahn, H.K.; Lee, Y.C.; et al. Tyrosine Kinase Inhibitor Activity in Patients with NSCLC Harboring Uncommon EGFR Mutations: A Retrospective International Cohort Study (UpSwinG). Oncologist 2022, 27, 255–265.
  57. Hellmann, M.D.; Reva, B.; Yu, H.; Rusch, V.W.; Rizvi, N.A.; Kris, M.G.; Arcila, M.E. Clinical and in vivo evidence that EGFR S768I mutant lung adenocarcinomas are sensitive to erlotinib. J. Thorac. Oncol. 2014, 9, e73–e74.
  58. Leventakos, K.; Kipp, B.R.; Rumilla, K.M.; Winters, J.L.; Yi, E.S.; Mansfield, A.S. S768I Mutation in EGFR in Patients with Lung Cancer. J. Thorac. Oncol. 2016, 11, 1798–1801.
  59. Kuiper, J.L.; Hashemi, S.M.; Thunnissen, E.; Snijders, P.J.; Grunberg, K.; Bloemena, E.; Sie, D.; Postmus, P.E.; Heideman, D.A.; Smit, E.F. Non-classic EGFR mutations in a cohort of Dutch EGFR-mutated NSCLC patients and outcomes following EGFR-TKI treatment. Br. J. Cancer 2016, 115, 1504–1512.
  60. Niogret, J.; Coudert, B.; Boidot, R. Primary Resistance to Afatinib in a Patient with Lung Adenocarcinoma Harboring Uncommon EGFR Mutations: S768I and V769L. J. Thorac. Oncol. 2018, 13, e113.
  61. Cho, J.H.; Lim, S.H.; An, H.J.; Kim, K.H.; Park, K.U.; Kang, E.J.; Choi, Y.H.; Ahn, M.S.; Lee, M.H.; Sun, J.M.; et al. Osimertinib for Patients with Non-Small-Cell Lung Cancer Harboring Uncommon EGFR Mutations: A Multicenter, Open-Label, Phase II Trial (KCSG-LU15-09). J. Clin. Oncol. 2020, 38, 488–495.
  62. Eide, I.J.Z.; Stensgaard, S.; Helland, A.; Ekman, S.; Mellemgaard, A.; Hansen, K.H.; Cicenas, S.; Koivunen, J.; Gronberg, B.H.; Sorensen, B.S.; et al. Osimertinib in non-small cell lung cancer with uncommon EGFR-mutations: A post-hoc subgroup analysis with pooled data from two phase II clinical trials. Transl. Lung Cancer Res. 2022, 11, 953–963.
  63. Banno, E.; Togashi, Y.; Nakamura, Y.; Chiba, M.; Kobayashi, Y.; Hayashi, H.; Terashima, M.; de Velasco, M.A.; Sakai, K.; Fujita, Y.; et al. Sensitivities to various epidermal growth factor receptor-tyrosine kinase inhibitors of uncommon epidermal growth factor receptor mutations L861Q and S768I: What is the optimal epidermal growth factor receptor-tyrosine kinase inhibitor? Cancer Sci. 2016, 107, 1134–1140.
  64. Chiu, C.H.; Yang, C.T.; Shih, J.Y.; Huang, M.S.; Su, W.C.; Lai, R.S.; Wang, C.C.; Hsiao, S.H.; Lin, Y.C.; Ho, C.L.; et al. Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Treatment Response in Advanced Lung Adenocarcinomas with G719X/L861Q/S768I Mutations. J. Thorac. Oncol. 2015, 10, 793–799.
  65. Gallant, J.N.; Sheehan, J.H.; Shaver, T.M.; Bailey, M.; Lipson, D.; Chandramohan, R.; Red Brewer, M.; York, S.J.; Kris, M.G.; Pietenpol, J.A.; et al. EGFR Kinase Domain Duplication (EGFR-KDD) Is a Novel Oncogenic Driver in Lung Cancer That Is Clinically Responsive to Afatinib. Cancer Discov. 2015, 5, 1155–1163.
  66. Wang, J.; Li, X.; Xue, X.; Ou, Q.; Wu, X.; Liang, Y.; Wang, X.; You, M.; Shao, Y.W.; Zhang, Z.; et al. Clinical outcomes of EGFR kinase domain duplication to targeted therapies in NSCLC. Int. J. Cancer 2019, 144, 2677–2682.
  67. Costa, D.B. Kinase inhibitor-responsive genotypes in EGFR mutated lung adenocarcinomas: Moving past common point mutations or indels into uncommon kinase domain duplications and rearrangements. Transl. Lung Cancer Res. 2016, 5, 331–337.
  68. Wu, D.; Xie, Y.; Jin, C.; Qiu, J.; Hou, T.; Du, H.; Chen, S.; Xiang, J.; Shi, X.; Liu, J. The landscape of kinase domain duplication in Chinese lung cancer patients. Ann. Transl. Med. 2020, 8, 1642.
  69. Baik, C.S.; Wu, D.; Smith, C.; Martins, R.G.; Pritchard, C.C. Durable Response to Tyrosine Kinase Inhibitor Therapy in a Lung Cancer Patient Harboring Epidermal Growth Factor Receptor Tandem Kinase Domain Duplication. J. Thorac. Oncol. 2015, 10, e97-99.
  70. Chen, D.; Li, X.L.; Wu, B.; Zheng, X.B.; Wang, W.X.; Chen, H.F.; Dong, Y.Y.; Xu, C.W.; Fang, M.Y. A Novel Oncogenic Driver in a Lung Adenocarcinoma Patient Harboring an EGFR-KDD and Response to Afatinib. Front. Oncol. 2020, 10, 867.
  71. Zhang, L.D.; Gao, H.; Qin, S.M.; Zeng, Q.; Chen, Q.F. Osimertinib is an effective epidermal growth factor receptor-tyrosine kinase inhibitor choice for lung cancer with epidermal growth factor receptor exon 18-25 kinase domain duplication: Report of two cases. Anticancer Drugs 2022, 33, e486–e490.
  72. Taek Kim, J.; Zhang, W.; Lopategui, J.; Vail, E.; Balmanoukian, A. Patient with Stage IV NSCLC and CNS Metastasis with EGFR Exon 18-25 Kinase Domain Duplication with Response to Osimertinib as a First-Line Therapy. JCO Precis. Oncol. 2021, 5, 88–92.
  73. Du, Z.; Brown, B.P.; Kim, S.; Ferguson, D.; Pavlick, D.C.; Jayakumaran, G.; Benayed, R.; Gallant, J.N.; Zhang, Y.K.; Yan, Y.; et al. Structure-function analysis of oncogenic EGFR Kinase Domain Duplication reveals insights into activation and a potential approach for therapeutic targeting. Nat. Commun. 2021, 12, 1382.
  74. Jin, R.; Li, J.; Jin, Z.; Lu, Y.; Shao, Y.W.; Li, W.; Zhao, G.; Xia, Y. Osimertinib confers potent binding affinity to EGFR kinase domain duplication. Int. J. Cancer 2019, 145, 2884–2885.
  75. Wu, J.Y.; Shih, J.Y. Effectiveness of tyrosine kinase inhibitors on uncommon E709X epidermal growth factor receptor mutations in non-small-cell lung cancer. OncoTargets Ther. 2016, 9, 6137–6145.
  76. Cheng, C.; Wang, R.; Li, Y.; Pan, Y.; Zhang, Y.; Li, H.; Zheng, D.; Zheng, S.; Shen, X.; Sun, Y.; et al. EGFR Exon 18 Mutations in East Asian Patients with Lung Adenocarcinomas: A Comprehensive Investigation of Prevalence, Clinicopathologic Characteristics and Prognosis. Sci. Rep. 2015, 5, 13959.
More
This entry is offline, you can click here to edit this entry!
Video Production Service