First- and second-generation EGFR tyrosine kinase inhibitors (TKI) are effective clinical therapies for patients with non–small cell lung cancer (NSCLC) harboring EGFR-activating mutations. However, almost all patients develop resistance to these drugs. The EGFR T790M mutation of EGFR is the most predominant mechanism for resistance. In addition, activation of AXL signaling is one of the suggested alternative bypassing pathways for resistance to EGFR-TKIs. Here, we report that naquotinib, a pyrazine carboxamide–based EGFR-TKI, inhibited EGFR with activating mutations, as well as T790M resistance mutation while sparing wild-type (WT) EGFR. In in vivo murine xenograft models using cell lines and a patient-derived xenograft model, naquotinib induced tumor regression of NSCLC with EGFR-activating mutations with or without T790M resistance mutation, whereas it did not significantly inhibit WT EGFR signaling in skin. Furthermore, naquotinib suppressed tumor recurrence during the treatment period of 90 days. In addition, unlike erlotinib and osimertinib, naquotinib inhibited the phosphorylation of AXL and showed antitumor activity against PC-9 cells overexpressing AXL in vitro and in vivo. Our findings suggest that naquotinib has therapeutic potential in patients with NSCLC with EGFR-activating mutations, T790M resistance mutation, and AXL overexpression.

EGFR mutations are detected in approximately 10% of non–small cell lung cancer (NSCLC) in Caucasian patients and approximately 40% of NSCLC in East Asian patients (1–3). EGFR mutations lead to constitutive activation of EGFR signaling, including the MAPK/ERK and PI3K/AKT pathways (4, 5) and oncogenic transformation, such as increased malignant cell survival, proliferation, invasion, metastatic spread, and tumor angiogenesis in NSCLC (6, 7). The most common EGFR mutations are deletion in exon 19 (del ex19) and leucine-to-arginine substitution at amino acid position 858 (L858R) in exon 21 mutations, which together account for approximately 90% of activating EGFR mutations in NSCLC (8, 9).

The first-generation reversible EGFR tyrosine kinase inhibitors (TKI), gefitinib and erlotinib, and second-generation covalent EGFR-TKIs, afatinib and dacomitinib, dramatically improve progression-free survival in patients with advanced NSCLC. However, almost all patients eventually develop resistance to these drugs after a median of 10–13 months (10–13). The EGFR T790M mutation is the most predominant mechanism for acquired resistance, detected in approximately 50%–60% of patients with clinical resistance to these EGFR-TKIs (14–17).

Several third-generation irreversible EGFR-TKIs, including WZ4002, osimertinib, rociletinib, and olmutinib, have been developed to target activating EGFR mutations, as well as T790M mutation while sparing wild-type (WT) EGFR because second-generation EGFR-TKIs cause dose-limiting epithelial toxicities due to their activity against WT EGFR and the normal physiologic role of this kinase in skin and gastrointestinal tissues (18–23). Although osimertinib has been approved for second-line treatment in patients with NSCLC with EGFR T790M mutation, this therapeutic option is limited.

Aside from the EGFR T790M resistance mutation, activation of alternative bypassing pathways, such as the MET, HER2, or AXL signaling pathways, has also been indicated as a resistance mechanism to EGFR-TKIs (15–17, 24–29). AXL overexpression and AXL-mediated resistance have been detected in many human cancers, including breast cancer, pancreatic cancer, prostate cancer, and NSCLC (28–33). Increased AXL expression in tumors often results from pharmacologic selective pressure to multiple chemotherapies and targeted therapies (34). Inhibition of activated EGFR may trigger the tyrosine kinase switch to transactivation of AXL and its downstream pathways to maintain tumor growth, given that they share the same downstream pathways such as the MAPK/ERK and PI3K/AKT pathways (29, 35, 36). AXL may mediate acquired resistance to TKIs in an epithelial-to-mesenchymal transition setting in EGFR-mutant NSCLC (29, 32, 33). These findings suggest that AXL is a potential therapeutic target in patients with NSCLC with acquired resistance to EGFR-TKIs.

Here, we describe the preclinical in vitro and in vivo characteristics of naquotinib, which has a selective inhibitory effect on EGFR-activating mutations and T790M resistance mutation over WT. In addition, we show the inhibitory effect of naquotinib on the AXL signaling pathway, an alternative bypassing pathway in the development of resistance to EGFR-TKIs.

Reagents

Naquotinib and erlotinib were prepared at Astellas Pharma Inc. as described in PCT Patent Application WO 2016/121777 and WO2001/34574, respectively. Afatinib and osimertinib were synthesized according to the PCT Patent Application WO 2002/50043 and WO 2013/014448, respectively.

Cell lines and cell culture

NCI-H1975, HCC827, NCI-H292, and NCI-H1666 were obtained from ATCC in 2012, 2012, 2008, and 2008, respectively. PC-9 was obtained from Immuno-Biological Laboratories in 2011. II-18, A431, and Ba/F3 were obtained from RIKEN BRC Cell Bank in 2013, 2013, and 2008, respectively. Cells were cultured in RPMI1640 medium supplemented with 10% heat-inactivated FBS at 37°C in 5% CO2 atmosphere. The cell lines used in this study were not authenticated in our laboratory but were purchased from the providers of authenticated cell lines and stored at early passages in a central cell bank at Astellas Pharma Inc., with Mycoplasma testing performed using PCR. The experiments were conducted using low-passage cultures of these stocks.

In vitro cell proliferation assays

Cell viability was assessed using the CellTiter-Glo Luminescent Cell Viability Assay (Promega). A detailed protocol is provided in the Supplementary Materials and Methods.

Immunoblotting analysis

Protein was extracted using Cell Lysis Buffer (Cell Signaling Technology) or RIPA Buffer (Thermo Fisher Scientific) supplemented with a Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific) and Protease Inhibitor Cocktail (Sigma-Aldrich or Nacalai Tesque). Protein concentrations of the lysates were determined using a BCA protein assay reagent kit or Pierce 660 nm Protein Assay Kit (Thermo Fisher Scientific). Equal amounts of total protein were resolved by SDS-PAGE and transferred to a polyvinylidene fluoride membrane. After blocking at room temperature with Blocking One (Nacalai Tesque), each membrane was incubated overnight at 4°C with the primary antibodies. After washing with TBS with Tween 20 (TBST), membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibody for 1 hour at room temperature. Proteins of interest were visualized by enhanced chemiluminescence using ECL-Prime (GE Healthcare) and detected using ImageQuant LAS 4000 (GE Healthcare). Antibodies for Western blotting are described in the Supplementary Materials and Methods.

Pharmacokinetics studies

Plasma and tumor concentrations of naquotinib were determined by LC-MS/MS (Shimadzu 20AD LC System 2, Shimadzu Corporation and Triple Quad 5500, AB Sciex LLC). Naquotinib and deuterated naquotinib, used as an internal standard (IS), were added to 25 μL of mouse plasma and tumor homogenate containing 0.025% 2,2-dichlorovinyl dimethyl phosphate (Wako Pure Chemical Industries, Ltd.) with 300 μL of 0.5 mol/L NaHCO3 solution (Kokusan Chemical Co., Ltd.) and 3 mL of tert-butyl methyl ether (Kanto Chemical Co., Inc.), and the mixture was shaken and centrifuged. The organic layer was collected and evaporated to dryness under a stream of nitrogen gas at about 40°C. The residue was dissolved in 400 μL of 50 mmol/L NH4HCO3 buffer (Kanto Chemical Co., Inc.)/acetonitrile (Kokusan Chemical Co., Ltd.; 45:55, v/v), and 3 μL of the resulting solution was injected into the LC-MS/MS. The analytic column was a CAPCELL PAK C18 MGII (3.0 mm inner diameter × 75 mm, particle size 3 μm, Shiseido Co., Ltd.) and the mobile phase comprised of 50 mmol/L NH4HCO3 buffer (45%) and acetonitrile (55%). The parent and product ions, m/z 563 and m/z 323 for naquotinib, and m/z 571 and m/z 323 for the IS, respectively, were monitored using positive multiple reaction monitoring. The IS method with peak area ratio was used to determine levels of naquotinib.

In vivo xenograft studies

All animal experimental procedures were approved by the Institutional Animal Care and Use Committee of Astellas Pharma Inc. Furthermore, Astellas Pharma Inc., Tsukuba Research Center has been awarded accreditation status by the Association for Assessment and Accreditation of Laboratory Animal Care International. HCC827, NCI-H1975, A431, PC-9vec, and PC-9AXL cells were inoculated subcutaneously into the flank of male CAnN.Cg-Foxn1nu/CrlCrlj mice. A xenograft study using LU1868 cells, a human NSCLC patient-derived xenograft (PDX) model with EGFR L858R/T790M, was conducted at Crown Bioscience Inc. LU1868 cells were inoculated subcutaneously into the flank of female BALB/c nude mice.

Mice were randomized and administered vehicle, naquotinib, erlotinib, or osimertinib at indicated doses (for details, see the figures for each experiment). Body weight and tumor diameter were measured twice a week, and tumor volume was determined by calculating the volume of an ellipsoid using the formula: length × width2 × 0.5. All values are expressed as mean ± SEM.

IHC of phosphorylated ERK in mouse skin

Tissue sections (4–5 μm thick) were deparaffinized and rehydrated. For antigen retrieval, sections were heated in 10 mmol/L sodium citrate buffer, pH 6.0 at 95°C for 10 minutes. Slides were washed with TBST three times and immersed in 3% H2O2 for 10 minutes at room temperature to quench endogenous peroxidases. Sections were treated with 5% goat serum used as a blocking agent for 1 hour at room temperature and then incubated with phosphorylated ERK (pERK) 1/2 rabbit mAb (Cell Signaling Technology) diluted with SignalStain Antibody Diluent (Cell Signaling Technology). Sections were washed with TBST three times for 5 minutes and treated with SignalStain Boost IHC Detection Reagent (HRP, Rabbit, Cell Signaling Technology) for 30 minutes at room temperature, before reacting with 3,3′-diaminobenzidine at room temperature. Sections were counterstained with hematoxylin.

Statistical analysis

Data were statistically analyzed using Dunnett multiple comparison test or Student t test with SAS Software (SAS Institute) or GraphPad Prism (GraphPad Software). The IC50 value of each experiment was calculated using Sigmoid-Emax model nonlinear regression analysis and was expressed as the geometric mean of three independent experiments.

In vitro activity of naquotinib on EGFR mutations and cancer cell lines

Naquotinib is a pyrazine carboxamide–based compound with a reactive acrylamide moiety. It is structurally different from other third-generation EGFR-TKIs such as osimertinib, rociletinib, and olmutinib, which have pyrimidine-based chemical structures (refs. 37–39; Supplementary Fig. S1). In vitro biochemical enzymatic assays revealed that naquotinib inhibited EGFR del ex19, L858R, del ex19/T790M, and L858R/T790M with IC50 values of 5.5, 4.6, 0.26, and 0.41 nmol/L, respectively, and an IC50 value of 13 nmol/L against WT EGFR (Supplementary Table S1). In vitro cell proliferation assays in human cancer cell lines harboring mutant EGFR or WT EGFR revealed that naquotinib inhibited the growth of NCI-H1975 (L858R/T790M), HCC827 (del ex19), PC-9 (del ex19), II-18 (L858R), A431 (WT), NCI-H292 (WT), and NCI-H1666 (WT) cells with IC50 values of 26, 7.3, 6.9, 43, 600, 260, and 230 nmol/L, respectively (Table 1). We further evaluated the inhibitory effect of naquotinib on EGFR and its downstream signals, ERK and AKT, by Western blotting. Naquotinib dose-dependently suppressed the phosphorylation of EGFR, ERK, and AKT in HCC827 and NCI-H1975 cells at concentrations that showed antiproliferative effects in these cell lines. In contrast, the inhibitory effect of naquotinib on these molecules in A431 cells was weaker (Fig. 1A). In addition, we compared the inhibitory effect of naquotinib on EGFR activating mutations and T790M resistance mutation under the same genetic background using Ba/F3 transfectants. Naquotinib showed comparable inhibitory effects on the growth of Ba/F3 cells expressing EGFR del ex19, L858R, del ex19/T790M, and L858R/T790M with IC50 values of 4.1, 9.4, 2.1, and 4.2 nmol/L, respectively (Supplementary Table S2).

Table 1.

Inhibitory effect of naquotinib on the proliferation of cancer cell lines expressing mutant or WT EGFR

IC50 (nmol/L)NCI-H1975HCC827PC-9IL18A431NCI-H292NCI-H1666
EGFR(L858R/T790M)(del ex19)(del ex19)(L858R)(WT)(WT)(WT)
Naquotinib 26 7.3 6.9 43 600 260 230 
Erlotinib >1,000 9.8 6.6 81 530 270 48 
Osimertinib 28 4.3 2.5 21 340 160 110 
IC50 (nmol/L)NCI-H1975HCC827PC-9IL18A431NCI-H292NCI-H1666
EGFR(L858R/T790M)(del ex19)(del ex19)(L858R)(WT)(WT)(WT)
Naquotinib 26 7.3 6.9 43 600 260 230 
Erlotinib >1,000 9.8 6.6 81 530 270 48 
Osimertinib 28 4.3 2.5 21 340 160 110 

NOTE: The IC50 value of each experiment was calculated using Sigmoid-Emax model nonlinear regression analysis and was expressed as the geometric mean of three independent experiments.

Figure 1.

Inhibitory effect of naquotinib against EGFR and its downstream signaling molecules in cell lines. A, Inhibitory effect of naquotinib on the phosphorylation of EGFR and its downstream molecules ERK and AKT was evaluated in NCI-H1975 (L858R/T790M), HCC827 (del ex19), and A431 (WT) cells. Cells were treated with naquotinib for 4 hours and examined for the expression and phosphorylation of these molecules by Western blotting. B, NCI-H1975 cells were exposed to 300 nmol/L naquotinib for 1 hour, washed with D-PBS(−), and cultured for 0, 4, 8, and 24 hours after removal of the compound. Phosphorylated EGFR (pEGFR) and EGFR protein levels were detected by Western blotting.

Figure 1.

Inhibitory effect of naquotinib against EGFR and its downstream signaling molecules in cell lines. A, Inhibitory effect of naquotinib on the phosphorylation of EGFR and its downstream molecules ERK and AKT was evaluated in NCI-H1975 (L858R/T790M), HCC827 (del ex19), and A431 (WT) cells. Cells were treated with naquotinib for 4 hours and examined for the expression and phosphorylation of these molecules by Western blotting. B, NCI-H1975 cells were exposed to 300 nmol/L naquotinib for 1 hour, washed with D-PBS(−), and cultured for 0, 4, 8, and 24 hours after removal of the compound. Phosphorylated EGFR (pEGFR) and EGFR protein levels were detected by Western blotting.

Close modal

Naquotinib covalently binds to the Cys-797 residue of EGFR L858R/T790M via its acrylamide moiety (ref. 42; Supplementary Fig. S2). We therefore investigated whether the covalent binding of naquotinib to mutant EGFR results in the prolonged inhibition of EGFR phosphorylation in NCI-H1975 cells by Western blotting. Naquotinib continuously inhibited the phosphorylation of EGFR for at least 24 hours after removal of the compound (Fig. 1B).

In vivo antitumor activity of naquotinib in EGFR-mutant tumor models

The antitumor activity of naquotinib was evaluated in murine xenograft models using HCC827, NCI-H1975, and A431 cells and in a PDX model using LU1868 cells. In HCC827 and NCI-H1975 xenograft models, once-daily oral administration of naquotinib inhibited tumor growth with tumor regression at 10, 30, and 100 mg/kg (Fig. 2A and B) without affecting body weight (Supplementary Fig. S3). In contrast, naquotinib did not significantly inhibit tumor growth at 10 and 30 mg/kg in an A431 xenograft model, but inhibited tumor growth at 100 mg/kg (Fig. 2C). These results indicate that naquotinib is selective for mutant EGFR in cell line xenograft models. In the PDX model using LU1868 (L858R/T790M) cells, erlotinib at 50 mg/kg did not inhibit tumor growth, which is in agreement with the resistance shown in clinical settings. In contrast, naquotinib significantly inhibited tumor growth at 10, 30, and 100 mg/kg, which is comparable with its effects in cell line xenograft models (Fig. 2B and D).

Figure 2.

In vivo antitumor efficacy of naquotinib in subcutaneous xenograft models. A, HCC827 (del ex19) xenograft model (n = 5). B, NCI-H1975 (L858R/T790M) xenograft model (n = 5). C, A431 (WT) xenograft model (n = 5). D, LU1868 (L858R/T790M) PDX model (n = 10). E, Chronic daily oral dosing of naquotinib in the NCI-H1975 xenograft model (n = 5). Mice were treated with vehicle, naquotinib, or erlotinib at the indicated doses for 14 days (A–D) or 90 days (E). Each datapoint represents the mean ± SEM. *, P < 0.05; **, P < 0.01 compared with the control group on day 14 (Dunnett test; A–D).

Figure 2.

In vivo antitumor efficacy of naquotinib in subcutaneous xenograft models. A, HCC827 (del ex19) xenograft model (n = 5). B, NCI-H1975 (L858R/T790M) xenograft model (n = 5). C, A431 (WT) xenograft model (n = 5). D, LU1868 (L858R/T790M) PDX model (n = 10). E, Chronic daily oral dosing of naquotinib in the NCI-H1975 xenograft model (n = 5). Mice were treated with vehicle, naquotinib, or erlotinib at the indicated doses for 14 days (A–D) or 90 days (E). Each datapoint represents the mean ± SEM. *, P < 0.05; **, P < 0.01 compared with the control group on day 14 (Dunnett test; A–D).

Close modal

As described above, robust tumor regression was observed after 14 days of treatment with naquotinib in several xenograft models with EGFR mutations. We next investigated the duration of the antitumor efficacy of naquotinib in a mutant EGFR model. In the NCI-H1975 xenograft model, while naquotinib at 10 mg/kg inhibited tumor growth for around 30 days, tumor regrowth was observed despite treatment. Naquotinib at 30 and 100 mg/kg induced tumor regression and the antitumor effect was sustained during the treatment period of 90 days with no sign of recurrence (Fig. 2E).

Naquotinib showed almost dose-proportional pharmacokinetics in both plasma and tumors after a single oral dose of the compound in the NCI-H1975 xenograft model. Interestingly, naquotinib showed a higher concentration and a longer elimination half-life in tumors than plasma (Fig. 3A and B). Evaluation of the phosphorylation of EGFR in tumors under the same conditions confirmed that the inhibitory effect of naquotinib on EGFR phosphorylation in tumors also showed dose proportionality, and revealed that inhibition continued for at least 24 hours after dosing at 100 mg/kg (Fig. 3C). These data may suggest that, in addition to the irreversible covalent binding property of naquotinib, the retention of naquotinib in tumors also contributes to its prolonged EGFR inhibition in tumors.

Figure 3.

Pharmacokinetics and pharmacodynamics of naquotinib in the NCI-H1975 xenograft model. After single oral administration, naquotinib concentration was determined in plasma (A) and tumors (B) by LC-MS/MS. Each datapoint represents the mean ± SD (n = 3). C, Inhibitory effect of naquotinib on the phosphorylation of EGFR in tumors was evaluated by ELISA. Signals for phosphorylated EGFR (pEGFR) were normalized to those for EGFR. The percentage of pEGFR in each group relative to the control (nontreated) group was calculated. Each datapoint represents the mean ± SEM (n = 3).

Figure 3.

Pharmacokinetics and pharmacodynamics of naquotinib in the NCI-H1975 xenograft model. After single oral administration, naquotinib concentration was determined in plasma (A) and tumors (B) by LC-MS/MS. Each datapoint represents the mean ± SD (n = 3). C, Inhibitory effect of naquotinib on the phosphorylation of EGFR in tumors was evaluated by ELISA. Signals for phosphorylated EGFR (pEGFR) were normalized to those for EGFR. The percentage of pEGFR in each group relative to the control (nontreated) group was calculated. Each datapoint represents the mean ± SEM (n = 3).

Close modal

Naquotinib spared inhibition of WT EGFR signaling in skin

To investigate the inhibitory effect of naquotinib, erlotinib, and afatinib on WT EGFR in skin, we conducted IHC staining for pERK in nude mice. Repeated dosing with erlotinib at 100 mg/kg and afatinib at 25 mg/kg eliminated the pERK signal in skin compared with nontreated controls. In contrast, naquotinib did not change the staining pattern of pERK even at 100 mg/kg (Fig. 4).

Figure 4.

IHC staining of phosphorylated ERK, a downstream molecule of EGFR. Nude mice were treated with EGFR-TKIs by oral administration over a 24-hour period. Three hours after the second treatment, the mice were sacrificed, and skin segments were dissected and fixed in 10% PBS-buffered formalin for 24 hours. The sections were subjected to IHC for phosphorylated ERK. A, Nontreated. B, Naquotinib 100 mg/kg. C, Afatinib 25 mg/kg. D, Erlotinib 100 mg/kg. Images were taken at a magnification of 80 × A (a) or 400 × B (b).

Figure 4.

IHC staining of phosphorylated ERK, a downstream molecule of EGFR. Nude mice were treated with EGFR-TKIs by oral administration over a 24-hour period. Three hours after the second treatment, the mice were sacrificed, and skin segments were dissected and fixed in 10% PBS-buffered formalin for 24 hours. The sections were subjected to IHC for phosphorylated ERK. A, Nontreated. B, Naquotinib 100 mg/kg. C, Afatinib 25 mg/kg. D, Erlotinib 100 mg/kg. Images were taken at a magnification of 80 × A (a) or 400 × B (b).

Close modal

Effect of naquotinib in an AXL overexpression model

In vitro biochemical enzymatic assays revealed that naquotinib inhibited AXL with IC50 values of 6.2 nmol/L at 50 μmol/L ATP and 37 nmol/L at 1 mmol/L ATP (Supplementary Table S3). These results indicate that naquotinib inhibited AXL kinase activity in an ATP-competitive manner. To evaluate the inhibitory effect of naquotinib on AXL overexpression, we constructed a retrovirus-based expression vector to upregulate AXL expression in PC-9 cells (PC-9AXL) and a vector control (PC-9vec), and investigated the pharmacologic effects of EGFR-TKIs on these cells. In the cell proliferation assays, PC-9AXL cells showed resistance to erlotinib and osimertinib, whereas PC-9vec cells did not; neither compound showed cell growth inhibition in PC-9AXL cells at up to 1 μmol/L, although both inhibited cell growth in PC-9vec cells. In contrast, naquotinib showed a comparable inhibitory effect on PC-9AXL and PC-9vec cells with IC50 values of 18 and 9.6 nmol/L, respectively (Fig. 5A and B). In Western blotting, all tested compounds inhibited the phosphorylation of EGFR in PC-9AXL and PC-9vec cells, but only naquotinib inhibited the phosphorylation of AXL, AKT, and ERK in PC-9AXL cells (Fig. 5C and D). In xenograft models, osimertinib showed partial tumor growth inhibition in PC-9AXL cells and tumor growth inhibition with tumor regression in PC-9vec cells. In contrast, naquotinib showed complete tumor growth inhibition in PC-9AXL cells and tumor growth inhibition with tumor regression in PC-9vec cells (Fig. 5E and F).

Figure 5.

Inhibitory effect of naquotinib in AXL-expressing PC-9 cells (PC-9AXL). Inhibitory effect of naquotinib on proliferation was evaluated in PC-9AXL (A) or PC-9vec (B) cells. Cell viability was assessed after treatment with naquotinib, osimertinib, or erlotinib for 3 days. Inhibitory effect on the phosphorylation of AXL and EGFR and its downstream molecules ERK and AKT was evaluated in PC-9AXL (C) or PC-9vec (D) cells. Cells were treated with naquotinib, osimertinib, or erlotinib for 24 hours and examined for the expression and phosphorylation of these molecules by Western blotting. The in vivo antitumor efficacy of naquotinib was evaluated in the PC-9AXL xenograft model (n = 5; E) or PC-9vec xenograft model (n = 5; F). Mice were treated with vehicle, naquotinib, osimertinib, or erlotinib at the indicated doses for 14 days. Each datapoint represents the mean ± SEM (**, P < 0.01 compared with the control group on day 14; Student t test).

Figure 5.

Inhibitory effect of naquotinib in AXL-expressing PC-9 cells (PC-9AXL). Inhibitory effect of naquotinib on proliferation was evaluated in PC-9AXL (A) or PC-9vec (B) cells. Cell viability was assessed after treatment with naquotinib, osimertinib, or erlotinib for 3 days. Inhibitory effect on the phosphorylation of AXL and EGFR and its downstream molecules ERK and AKT was evaluated in PC-9AXL (C) or PC-9vec (D) cells. Cells were treated with naquotinib, osimertinib, or erlotinib for 24 hours and examined for the expression and phosphorylation of these molecules by Western blotting. The in vivo antitumor efficacy of naquotinib was evaluated in the PC-9AXL xenograft model (n = 5; E) or PC-9vec xenograft model (n = 5; F). Mice were treated with vehicle, naquotinib, osimertinib, or erlotinib at the indicated doses for 14 days. Each datapoint represents the mean ± SEM (**, P < 0.01 compared with the control group on day 14; Student t test).

Close modal

In this study, we characterized the efficacy of naquotinib, a pyrazine carboxamide–based compound, as a third-generation EGFR-TKI. In in vitro experiments, naquotinib inhibited cell growth and EGFR signaling in NSCLC cell lines harboring mutant EGFR with or without T790M resistance mutation (Table 1). In xenograft models, naquotinib showed robust inhibition of tumor growth in NSCLC cell lines with EGFR mutations (Fig. 2A and B) and in a PDX model, which is thought to maintain intratumoral heterogeneity, indicating clinical efficacy in more than just monoclonal cell lines (Fig. 2D). Our findings also suggest that naquotinib shows mutant selectivity and spares WT EGFR. Naquotinib inhibited cell growth and EGFR signaling in NSCLC cell lines harboring mutant EGFR with or without T790M more potently than those harboring WT EGFR in vitro (Fig. 1A; Table 1; Supplementary Table S2). While we did not examine Ba/F3 cells overexpressing WT EGFR, our collaborator previously demonstrated that naquotinib showed very low activity against these cells (40). IHC evaluation of EGFR signaling molecules in murine skin showed that erlotinib and afatinib inhibited the phosphorylation of ERK, which corresponds to the prevalence of severe skin rashes in clinical settings (11, 22). Naquotinib showed comparable mutant selectivity, sparing WT EGFR, to that of erlotinib in vitro (Table 1), and showed some inhibition of tumor growth in the A431 xenograft model (Fig. 2C). In contrast, naquotinib did not inhibit the phosphorylation of ERK in skin at 100 mg/kg (Fig. 4), the same dose that induced tumor regression in xenograft models with EGFR mutations (Fig. 2A, B, and D). Pharmacokinetic studies in the NCI-H1975 xenograft model showed that naquotinib had a higher concentration and a longer half-life in tumors than in plasma (Fig. 3A and B). This distribution may explain why naquotinib did not inhibit EGFR downstream signaling in skin. Considering that second-generation EGFR-TKIs are limited because of toxicity that likely arises from WT EGFR inhibition, the WT-sparing selectivity of naquotinib suggests that it may exert antitumor activity in patients with NSCLC harboring EGFR T790M resistance mutation without such limitations. This is in agreement with recent reports from naquotinib phase I–II clinical trials, in which skin toxicities were rarely observed (41).

We also examined the inhibitory effect of naquotinib on AXL overexpression, an alternative bypassing pathway involved in acquired resistance. We generated PC-9AXL cells stably expressing AXL. Studies have demonstrated that erlotinib, gefitinib, afatinib, rociletinib, and osimertinib do not inhibit any AXL kinase activity (37, 38, 42, 43). Consistent with these reports, PC-9AXL cells were resistant to erlotinib and osimertinib. In contrast, naquotinib inhibited AXL kinase activity in an ATP-competitive manner (Supplementary Table S3). Furthermore, naquotinib inhibited the phosphorylation of AXL, ERK, and AKT, and decreased cell proliferation more potently than erlotinib and osimertinib. In addition, in xenograft models, naquotinib showed complete tumor growth inhibition both in PC-9AXL and PC-9vec cells, whereas osimertinib showed only partial growth inhibition in PC-9AXL cells (Fig. 5). Furthermore, naquotinib inhibited the phosphorylation of AXL in PC-9AXL cells more potently than the phosphorylation of EGFR in A431 cells (Figs. 1A and 5C), and decreased the cell proliferation of PC-9AXL cells more potently than WT EGFR–expressing cells in vitro (Fig. 5A and Table 1). In xenograft models, naquotinib showed complete tumor growth inhibition in PC-9AXL cells and partial tumor growth inhibition in A431 (WT) cells (Figs. 2C and 5E). Interestingly, AXL expression has not only been observed in tumors from EGFR-TKI–resistant patients but also in those from a certain rare subgroup of patients with treatment-naïve NSCLC (28, 29, 44, 45). In addition, patients with NSCLC with EGFR L858R+T790M mutations and high AXL mRNA expression show resistance to osimertinib (46). Further investigation, such as in drug exposure–derived resistant cells with high AXL expression, is expected to reveal how AXL activation may cooperate with other genomic or genetic alterations to induce acquired resistance to EGFR-TKIs.

In conclusion, naquotinib, a mutant-selective irreversible EGFR inhibitor, shows antitumor activity against NSCLC tumors that harbor EGFR-activating mutations, as well as EGFR T790M resistance mutation and that overexpress AXL.

No potential conflicts of interest were disclosed.

Conception and design: H. Sakagami, N. Kaneko, S. Konagai, Y. Yamanaka, M. Mori, M. Takeuchi, H. Koshio, M. Hirano, S. Kuromitsu

Development of methodology: H. Sakagami, S. Konagai, M. Yuri, Y. Yamanaka

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Tanaka, N. Kaneko, S. Konagai, H. Yamamoto, T. Matsuya, M. Yuri, Y. Yamanaka, H. Koshio

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Tanaka, H. Sakagami, N. Kaneko, S. Konagai, M. Yuri, Y. Yamanaka, M. Mori, H. Koshio, M. Hirano

Writing, review, and/or revision of the manuscript: H. Tanaka, H. Sakagami, N. Kaneko, S. Konagai, T. Matsuya, M. Yuri, Y. Yamanaka, M. Mori, H. Koshio, M. Hirano, S. Kuromitsu

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Tanaka, H. Sakagami

Study supervision: H. Sakagami, M. Mori, M. Takeuchi, M. Hirano, S. Kuromitsu

This research was funded by Astellas Pharma Inc. The authors are grateful to Masafumi Kudo, Ryuichi Takezawa, Yukihiro Takemoto, Nobuaki Shindo, and Tomohiro Eguchi for their helpful discussions and work, and thank Regina Switzer, Choice Healthcare Solutions, for logistical support during the development of this article, which was funded by the study sponsor.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Lynch
TJ
,
Bell
DW
,
Sordella
R
,
Gurubhagavatula
S
,
Okimoto
RA
,
Brannigan
BW
, et al
Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib
.
N Engl J Med
2004
;
350
:
2129
39
.
2.
Marchetti
A
,
Martella
C
,
Felicioni
L
,
Barassi
F
,
Salvatore
S
,
Chella
A
, et al
EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment
.
J Clin Oncol
2005
;
23
:
857
65
.
3.
Kosaka
T
,
Yatabe
Y
,
Endoh
H
,
Kuwano
H
,
Takahashi
T
,
Mitsudomi
T
. 
Mutations of the epidermal growth factor receptor gene in lung cancer: biological and clinical implications
.
Cancer Res
2004
;
64
:
8919
23
.
4.
Hsuan
JJ
,
Tan
SH
. 
Growth factor-dependent phosphoinositide signalling
.
Int J Biochem Cell Biol
1997
;
29
:
415
35
.
5.
Burgering
BM
,
Coffer
PJ
. 
Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction
.
Nature
1995
;
376
:
599
602
.
6.
Herbst
RS
,
Heymach
JV
,
Lippman
SM
. 
Lung cancer
.
N Engl J Med
2008
;
359
:
1367
80
.
7.
Wells
A
. 
EGF receptor
.
Int J Biochem Cell Biol
1999
;
31
:
637
43
.
8.
Rosell
R
,
Moran
T
,
Queralt
C
,
Porta
R
,
Cardenal
F
,
Camps
C
, et al
Screening for epidermal growth factor receptor mutations in lung cancer
.
N Engl J Med
2009
;
361
:
958
67
.
9.
Sharma
SV
,
Bell
DW
,
Settleman
J
,
Haber
DA
. 
Epidermal growth factor receptor mutations in lung cancer
.
Nat Rev Cancer
2007
;
7
:
169
81
.
10.
Mok
TS
,
Wu
YL
,
Thongprasert
S
,
Yang
CH
,
Chu
DT
,
Saijo
N
, et al
Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma
.
N Engl J Med
2009
;
361
:
947
57
.
11.
Cappuzzo
F
,
Ciuleanu
T
,
Stelmakh
L
,
Cicenas
S
,
Szczesna
A
,
Juhasz
E
, et al
Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: a multicentre, randomised, placebo-controlled phase 3 study
.
Lancet Oncol
2010
;
11
:
521
9
.
12.
Sequist
LV
,
Yang
JC
,
Yamamoto
N
,
O'Byrne
K
,
Hirsh
V
,
Mok
T
, et al
Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations
.
J Clin Oncol
2013
;
31
:
3327
34
.
13.
Janne
PA
,
Ou
SH
,
Kim
DW
,
Oxnard
GR
,
Martins
R
,
Kris
MG
, 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
2014
;
15
:
1433
41
.
14.
Kobayashi
S
,
Boggon
TJ
,
Dayaram
T
,
Janne
PA
,
Kocher
O
,
Meyerson
M
, et al
EGFR mutation and resistance of non-small-cell lung cancer to gefitinib
.
N Engl J Med
2005
;
352
:
786
92
.
15.
Blakely
CM
,
Bivona
TG
. 
Resiliency of lung cancers to EGFR inhibitor treatment unveiled, offering opportunities to divide and conquer EGFR inhibitor resistance
.
Cancer Discov
2012
;
2
:
872
5
.
16.
Sequist
LV
,
Waltman
BA
,
Dias-Santagata
D
,
Digumarthy
S
,
Turke
AB
,
Fidias
P
, et al
Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors
.
Sci Transl Med
2011
;
3
:
75ra26
.
17.
Yu
HA
,
Arcila
ME
,
Rekhtman
N
,
Sima
CS
,
Zakowski
MF
,
Pao
W
, et al
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
2013
;
19
:
2240
7
.
18.
Zhou
W
,
Ercan
D
,
Chen
L
,
Yun
CH
,
Li
D
,
Capelletti
M
, et al
Novel mutant-selective EGFR kinase inhibitors against EGFR T790M
.
Nature
2009
;
462
:
1070
4
.
19.
Mok
TS
,
Wu
YL
,
Ahn
MJ
,
Garassino
MC
,
Kim
HR
,
Ramalingam
SS
, et al
Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer
.
N Engl J Med
2017
;
376
:
629
40
.
20.
Sequist
LV
,
Soria
JC
,
Goldman
JW
,
Wakelee
HA
,
Gadgeel
SM
,
Varga
A
, et al
Rociletinib in EGFR-mutated non-small-cell lung cancer
.
N Engl J Med
2015
;
372
:
1700
9
.
21.
Park
K
,
Lee
JS
,
Han
JY
,
Lee
KH
,
Kim
JH
,
Cho
EK
, et al
Efficacy and safety of BI 1482694 (HM61713), an EGFR mutant-specific inhibitor, in T790M-positive NSCLC at the recommended phase II dose
.
J Thoracic Oncol
2016
;
11
:
S113
.
22.
Schuler
M
,
Yang
JC
,
Park
K
,
Kim
JH
,
Bennouna
J
,
Chen
YM
, 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
23
.
23.
Wu
YL
,
Cheng
Y
,
Zhou
X
,
Lee
KH
,
Nakagawa
K
,
Niho
S
, 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
2017
;
18
:
1454
66
.
24.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
,
Song
Y
,
Hyland
C
,
Park
JO
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
25.
Bean
J
,
Brennan
C
,
Shih
JY
,
Riely
G
,
Viale
A
,
Wang
L
, 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 U S A
2007
;
104
:
20932
7
.
26.
Takezawa
K
,
Pirazzoli
V
,
Arcila
ME
,
Nebhan
CA
,
Song
X
,
de Stanchina
E
, 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
2012
;
2
:
922
33
.
27.
Shieh
YS
,
Lai
CY
,
Kao
YR
,
Shiah
SG
,
Chu
YW
,
Lee
HS
, et al
Expression of axl in lung adenocarcinoma and correlation with tumor progression
.
Neoplasia
2005
;
7
:
1058
64
.
28.
Zhang
Z
,
Lee
JC
,
Lin
L
,
Olivas
V
,
Au
V
,
LaFramboise
T
, et al
Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer
.
Nat Genet
2012
;
44
:
852
60
.
29.
Zhang
G
,
Wang
M
,
Zhao
H
,
Cui
W
. 
Function of Axl receptor tyrosine kinase in non-small cell lung cancer
.
Oncol Lett
2018
;
15
:
2726
34
.
30.
Graham
DK
,
DeRyckere
D
,
Davies
KD
,
Earp
HS
. 
The TAM family: phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer
.
Nat Rev Cancer
2014
;
14
:
769
85
.
31.
Hong
CC
,
Lay
JD
,
Huang
JS
,
Cheng
AL
,
Tang
JL
,
Lin
MT
, et al
Receptor tyrosine kinase AXL is induced by chemotherapy drugs and overexpression of AXL confers drug resistance in acute myeloid leukemia
.
Cancer Lett
2008
;
268
:
314
24
.
32.
Gjerdrum
C
,
Tiron
C
,
Hoiby
T
,
Stefansson
I
,
Haugen
H
,
Sandal
T
, et al
Axl is an essential epithelial-to-mesenchymal transition-induced regulator of breast cancer metastasis and patient survival
.
Proc Natl Acad Sci U S A
2010
;
107
:
1124
9
.
33.
Koorstra
JB
,
Karikari
CA
,
Feldmann
G
,
Bisht
S
,
Rojas
PL
,
Offerhaus
GJ
, et al
The Axl receptor tyrosine kinase confers an adverse prognostic influence in pancreatic cancer and represents a new therapeutic target
.
Cancer Biol Ther
2009
;
8
:
618
26
.
34.
Scaltriti
M
,
Elkabets
M
,
Baselga
J
. 
Molecular pathways: AXL, a membrane receptor mediator of resistance to therapy
.
Clin Cancer Res
2016
;
22
:
1313
7
.
35.
Stommel
JM
,
Kimmelman
AC
,
Ying
H
,
Nabioullin
R
,
Ponugoti
AH
,
Wiedemeyer
R
, et al
Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies
.
Science
2007
;
318
:
287
90
.
36.
Mahadevan
D
,
Cooke
L
,
Riley
C
,
Swart
R
,
Simons
B
,
Della Croce
K
, et al
A novel tyrosine kinase switch is a mechanism of imatinib resistance in gastrointestinal stromal tumors
.
Oncogene
2007
;
26
:
3909
19
.
37.
Finlay
MR
,
Anderton
M
,
Ashton
S
,
Ballard
P
,
Bethel
PA
,
Box
MR
, et al
Discovery of a potent and selective EGFR inhibitor (AZD9291) of both sensitizing and T790M resistance mutations that spares the wild type form of the receptor
.
J Med Chem
2014
;
57
:
8249
67
.
38.
Walter
AO
,
Sjin
RT
,
Haringsma
HJ
,
Ohashi
K
,
Sun
J
,
Lee
K
, et al
Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC
.
Cancer Discov
2013
;
3
:
1404
15
.
39.
Kim
ES
. 
Olmutinib: first global approval
.
Drugs
2016
;
76
:
1153
7
.
40.
Hirano
T
,
Yasuda
H
,
Hamamoto
J
,
Nukaga
S
,
Masuzawa
K
,
Kawada
I
, et al
Pharmacological and structural characterizations of naquotinib, a novel third-generation EGFR tyrosine kinase inhibitor, in EGFR-mutated non-small cell lung cancer
.
Mol Cancer Ther
2018
;
17
:
740
50
.
41.
Yu
HA
,
Spira
A
,
Horn
L
,
Weiss
J
,
West
H
,
Giaccone
G
, et al
A phase I, dose escalation study of oral ASP8273 in patients with non-small cell lung cancers with epidermal growth factor receptor mutations
.
Clin Cancer Res
2017
;
23
:
7467
73
.
42.
Anastassiadis
T
,
Deacon
SW
,
Devarajan
K
,
Ma
H
,
Peterson
JR
. 
Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity
.
Nat Biotechnol
2011
;
29
:
1039
45
.
43.
Li
D
,
Ambrogio
L
,
Shimamura
T
,
Kubo
S
,
Takahashi
M
,
Chirieac
LR
, et al
BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models
.
Oncogene
2008
;
27
:
4702
11
.
44.
Kim
GW
,
Song
JS
,
Choi
CM
,
Rho
JK
,
Kim
SY
,
Jang
SJ
, et al
Multiple resistant factors in lung cancer with primary resistance to EGFR-TK inhibitors confer poor survival
.
Lung Cancer
2015
;
88
:
139
46
.
45.
Byers
LA
,
Diao
L
,
Wang
J
,
Saintigny
P
,
Girard
L
,
Peyton
M
, et al
An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance
.
Clin Cancer Res
2013
;
19
:
279
90
.
46.
Karachaliou
N
,
Chaib
I
,
Cardona
AF
,
Berenguer
J
,
Bracht
JWP
,
Yang
J
, et al
Common co-activation of AXL and CDCP1 in EGFR-mutation-positive non-smallcell lung cancer associated with poor prognosis
.
EBioMedicine
2018
;
29
:
112
27
.