Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Aug 18.
Published in final edited form as: Eur J Med Chem. 2017 May 3;136:497–510. doi: 10.1016/j.ejmech.2017.04.079

Discovery of a Potent Dual ALK and EGFR T790M Inhibitor

Jaebong Jang a,c,1, Jung Beom Son g,1, Ciric To b,f, Magda Bahcall d, So Young Kim g, Seock Yong Kang g, Mierzhati Mushajiang d, Younho Lee g, Pasi A Jänne b,d,e,h, Hwan Geun Choi g,**, Nathanael S Gray a,c,*
PMCID: PMC5800871  NIHMSID: NIHMS878864  PMID: 28528303

Abstract

The mutational activations of anaplastic lymphoma kinase (ALK) and epidermal growth factor receptor (EGFR) are validated oncogenic events and the targets of approved drugs to treat non-small cell lung cancer (NSCLC). Here we report highly potent dual small molecule inhibitors of both ALK and EGFR, particularly the T790M mutant which confers resistance to first generation EGFR inhibitors. Dual ALK/EGFR inhibitors may provide an efficient approach to prevent resistance that arises as a consequence of clinically reported reciprocal activation mechanisms. Our lead compound 7c displayed remarkable inhibitory activities against both ALK and EGFR in enzymatic and cellular assays. We demonstrate that 7c is capable of recapitulating the signaling effects and antiproliferative activity of combined treatment with the approved ALK inhibitor ceritinib and T790M EGFR inhibitor osimertinib against patient-derived non-small cell lung cancer cell line, DFCI032 which harbors both EML4-ALK and activated EGFR.

Keywords: ALK, EGFR T790M, dual inhibitor, non-small cell lung cancer, rational drug design

Graphical Abstract

graphic file with name nihms878864u1.jpg

1. Introduction

Oncogenic mutations and translocations of receptor tyrosine kinases (RTKs) cause the development and progression of many cancers [1]. Small molecule drugs targeting dysregulated RTKs have distinct clinical benefits especially for genetically defined patient populations [2]. The anaplastic lymphoma kinase (ALK) and epidermal growth factor receptor (EGFR) have been well characterized as the leading oncogenic drivers of non-small cell lung cancer (NSCLC) [3]. In the past two decades, several wild-type and mutant-directed inhibitors of both ALK and EGFR have been developed. For example, the first-generation ALK inhibitor, crizotinib [4] and the first and second generation EGFR inhibitors, gefitinib [5], erlotinib [6] and afatinib [7] have been developed to treat NSCLC. Despite excellent response rates to these drugs, patients invariably relapse due to the emergence of drug resistant tumors. The second generation ALK inhibitors, ceritinib [8], alectinib [9] and brigatinib [10] and the third generation EGFR inhibitor, osimertinib [1113] potently overcome the secondary resistant mutations.

Despite extensive exploration of diverse chemotypes as inhibitors of ALK and EGFR, there have been no reports of small molecules that can act as dual inhibitors of both kinases. EGFR activation has been demonstrated as one of the compensatory resistance mechanisms of oncogenic ALK inhibition by small molecule inhibitors such as crizotinib, alectinib and ceritinib [1416]. Additionally, with improved ability to perform single-cell sequencing there have been reports of tumors that harbor oncogenic alleles of both ALK and mutant EGFR concurrently [1720].

Here we report the rational design, synthesis and biological evaluation of a dual inhibitor of ALK and EGFR, especially the T790M resistant mutant, that acts as a reversible ATP-competitive inhibitor with respect to ALK and as an irreversible, covalent inhibitor of the T790M mutant of EGFR.

2. Results and discussion

2.1 Rational drug design

Our study commenced by recognizing a structural similarity that exists between several of the known ALK inhibitors and EGFR inhibitors. The ALK inhibitors, ceritinib [21], TAE684 [22] and brigatinib [23] share a 2,4-disubstituted pyrimidine scaffold with the third generation EGFR inhibitors, osimertinib [1113], WZ4002 [24] and rociletinib [25, 26]. Despite sharing the 2,4-disubstituted pyrimidine core, both ALK and EGFR inhibitors have distinct substituents that are critical to the recognition of their respective targets. In particular, the T790M EGFR inhibitors use a meta-acrylamide to enable the formation of a covalent bond with Cys797 located at the edge of the EGFR ATP binding pocket. The ALK inhibitors exploit an ortho-substituted isopropylsulfone (TAE684/ceritinib) or a dimethylphosphine oxide (brigatinib) to form a hydrogen bond with the catalytic Lys1150, which is critical to the binding affinity. Based on this structural analysis, we decided to combine both the acrylamide and ortho-substituted hydrogen bond acceptors with the 2,4-disubstituted pyrimidine scaffold with the aim of generating a dual ALK/EGFR inhibitor. (Figure 1) The ortho-alkoxy group at the C2-aniline substituent is a well-known functional group which is required to impart kinome-wide selectivity [21, 22]. For example, the methoxy group of WZ4002 led to improved selectivity for EGFR relative to its non-substituted analog, WZ3146 [24]. We also selected the methoxy group instead of a bulkier alkoxy group in our ALK/EGFR dual inhibitor because the isopropoxy substituent of ceritinib causes a loss of binding affinity for EGFR (Figure 3).

Figure 1.

Figure 1

Rational design of dual ALK and EGFR inhibitor.

Figure 3.

Figure 3

Figure 3

Figure 3

Kinome-wide selectivity profiles of (A) TAE684, (B) ceritinib and (C) compound 7c at a 1 μM concentration. Wild type and mutants ALK and EGFR are indicated by blue spots.

2.2 Structure-Activity Relationship

The potencies of newly synthesized compounds were first assessed biochemically using a Z′-Lyte enzymatic assay for wild type ALK and an ATP-depletion (ADP-Glo) enzymatic assay for wild type EGFR, EGFR T790M and EGFR T790M/L858R. Compounds were subsequently evaluated using a proliferation assay employing Ba/F3 cells [27] transformed by either EML4-ALK, wild type EGFR or mutant EGFRs such as L858R, exon19del, L858R/T70M or exon19del/T790M. Furthermore, we utilized parental Ba/F3 cells grown in the presence of interlukin-3 (IL-3) to assess whether the compounds exhibited any general cytotoxicity.

First, we benchmarked our enzymatic and cellular ALK and EGFR assays using the ALK inhibitor, ceritinib and the EGFR inhibitors WZ4002 and osimertinib which share the 2,4-disubstituted pyrimidine core structure (Table 1). As expected, ceritinib exhibited excellent biochemical potency against wild type ALK with IC50 value of 20 nM and exhibited weak potency against wild type EGFR with IC50 value of > 2 μM. Interestingly, ceritinib also displayed potent inhibition of T790M and T790M/L858R EGFR mutants using the ADP-Glo assays, although less potent than the IC50 values measured for WZ4002 and osimertinib. This result was reproduced by a Z′-LYTE enzymatic assay (Invitrogen, Table S1). The observed biochemical potency of ceritinib for T790M EGFR was not observed in the Ba/F3 cellular assay (Table 2), highlighting the importance of tracking both the biochemical and cellular potencies. The two EGFR inhibitors displayed excellent potencies against all forms of EGFRs, especially T790M containing mutants (IC50 = 5 ~ 7 nM), but displayed moderate to low potencies against wild type ALK (IC50 = 990 nM and 177 nM). These known drugs also showed similar trends in their cell growth inhibitory activities in the proliferation assays of Ba/F3 cells transformed with EML4-ALK, wild type EGFR and four different EGFR mutants (Table 2).

Table 1.

Enzymatic Inhibitory Activities against ALK and EGFR.

Compd. No. R1 R2 ALK (IC50, nM) EGFR (IC50, nM)

wt wt T790M T790M/L858R
Ceritinib 20 2156 28 19
WZ4002 990 120 5 4
Osimertinib 177 114 5 7
7a H graphic file with name nihms878864t1.jpg 103 2480 26 86
7b F graphic file with name nihms878864t1.jpg 17 46 2 2
7c Cl graphic file with name nihms878864t1.jpg 18 151 2 4
7d I graphic file with name nihms878864t1.jpg 91 865 9 33
7e H graphic file with name nihms878864t2.jpg 695 149 5 35
7f F graphic file with name nihms878864t2.jpg 123 106 2 5
7g Cl graphic file with name nihms878864t2.jpg 40 65 2 12
7h I graphic file with name nihms878864t2.jpg 41 54 2 8
7i H graphic file with name nihms878864t3.jpg 31 307 4 12
7j F graphic file with name nihms878864t3.jpg 15 172 2 3
7k Cl graphic file with name nihms878864t3.jpg 17 204 2 4
7l I graphic file with name nihms878864t3.jpg 38 244 3 5

Table 2.

Antiproliferation Activities against EML4-ALK Ba/F3 and EGFR Ba/F3 Cells.

Compd. No. R1 R2 EML4-ALK (EC50, nM) EGFR (EC50, nM) Parental (EC50, nM)

wt L858R Del T790M/L858R T790M/Del
Ceritinib 26 > 3000 3000 2500 2500 2660 > 5000
WZ4002 6258 989 26 68 31 27 > 5000
Osimertinib 1748 66 6 18 20 16 > 5000
7a H graphic file with name nihms878864t1.jpg 2182 827 78 195 183 158 > 5000
7b F graphic file with name nihms878864t1.jpg 20 25 2 5 5 5 > 5000
7c Cl graphic file with name nihms878864t1.jpg 7 32 4 6 6 5 > 5000
7d I graphic file with name nihms878864t1.jpg 87 240 19 47 22 20 > 5000
7e H graphic file with name nihms878864t2.jpg > 5000 988 236 680 830 400 > 5000
7f F graphic file with name nihms878864t2.jpg 2506 178 68 157 162 74 > 5000
7g Cl graphic file with name nihms878864t2.jpg 961 68 21 44 45 30 > 5000
7h I graphic file with name nihms878864t2.jpg 2357 96 23 84 31 32 > 5000
7i H graphic file with name nihms878864t3.jpg 217 93 19 21 22 19 > 5000
7j F graphic file with name nihms878864t3.jpg 39 31 8 6 6 5 4661
7k Cl graphic file with name nihms878864t3.jpg 8 69 4 6 6 5 2904
7l I graphic file with name nihms878864t3.jpg 21 262 19 24 19 17 2270

We next investigated which combination of functional groups are critical to achieving potent inhibition of ALK and EGFR, including the aforementioned ortho-hydrogen bond acceptor and the meta-acrylamide functional groups. We first synthesized and evaluated 7a containing the ortho-isopropylsulfone at C4-aniline substituent and the meta-acrylamide at C2-aniline substituent of the pyrimidine core. 7a displayed moderate enzyme inhibitory activities against wild type ALK, T790M EGFR and L858R/T790M EGFR with IC50 values of 103 nM, 26 nM and 86 nM respectively and it showed weak potency against wild type EGFR (Table 1). Consistent with the biochemical activities, 7a showed moderate antiproliferative potencies against mutant EGFR transformed Ba/F3 cells [EC50 = 78 nM (L858R), 195 nM (exon19del), 183 nM (T790M/L858R), 158 nM (exon19del/T790M)] while exhibiting weak proliferative inhibition of wild type EGFR (EC50 = 827 nM) as well as no meaningful inhibition against parental Ba/F3 cells (EC50 ≥ 5000 nM). However, contrary to the biochemical assay, compound 7a displayed poor antiproliferative activity against EML4-ALK transformed Ba/F3 cells which encouraged us to explore further the structure-activity relationships.

Previously, the C5-substituents of pyrimidine-based kinase inhibitors have been demonstrated as a key functional group for improving potency and kinome selectivity via the interaction with the gatekeeper residue [23, 28, 29]. Ceritinib, TAE684, brigatinib, and WZ4002 all possess a chlorine substituent at C5-pyrimidine which is required to achieve potent inhibition of ALK or EGFR. For WZ4002 an interaction of the C5-chlorine with the thioether of M790 was observed in the crystal structure and was critical to achieving selectivity for T790M over wild type EGFR [24, 30]. Similar to the known ALK and EGFR inhibitors, introduction of diverse halogens such as fluorine, chlorine and iodine at C5 of the pyrimidine core led to remarkable increases in inhibitory activities against both ALK and EGFRs (Tables 1 and 2). Especially compound 7c containing a chlorine substituent, exhibited more than 300-fold improved anti-proliferative activity against EML4-ALK transformed Ba/F3 cells. Moreover, inhibition of wild type EGFR and mutant EGFRs in enzymatic and cellular assays were improved by 15 ~ 30-fold, with maintenance of selectivity between wild type EGFR and mutant EGFRs. The fluorine derivative (7b) was roughly equipotent and had similar selectivity to 7c but the iodine derivative (7d) showed slightly lower potencies against both ALK and EGFR while retaining selectivity for wild type and mutant EGFRs.

We next directed our attention to evaluate the combination of various ortho-hydrogen bond acceptors such as dimethylsulfonamide and dimethylphosphine oxide at C4-aniline with C5-halogen substituents (Table 1 and 2, 7e – l). The dimethylphosphine oxide analogs (7e – 7h) were slightly less potent against ALK than the isopropylsulfone analogs (7a – 7d) but, equipotent against wild type EGFR and mutant EGFRs in enzymatic assays and retained the potencies against mutant EGFR transformed Ba/F3 cells. However, these analogs totally lost their growth inhibitory activity against EML4-ALK transformed Ba/F3 cells. In contrast to the dimethylphosphine oxide, introduction of the dimethylsulfonamide group led to increases in inhibitory activities of both ALK and EGFRs in enzymatic and cellular assays (7a and 7i) and when combined with the halogen substituents at C5 of the pyrimidine as in 7j – 7l, biochemical and cellular potencies were similar to those of the isopropylsulfone analogs (7b – 7d). However, these analogs showed a slight cytotoxicity against parental Ba/F3 cells (EC50 = 2.3 ~ 4.7 μM) which led us to choose compound 7c as a lead compound.

2.3 Characterization of compound 7c

We next performed detailed characterization of 7c including a molecular modeling study, kinome selectivity profile, effects on ALK and EGFR-dependent signaling pathways, antiproliferative activity on patient-derived NSCLC and pharmacokinetic profile in mouse.

To better rationalize the structural-basis for the observed dual inhibition of ALK and EGFR, we performed a molecular modeling based on the co-crystal structures of ALK and ceritinib (PDB 4MKC), wild type EGFR and osimertinib (PDB 4ZAU) and T790M EGFR and WZ4002 (PDB 3IKA) (Figure 2). 7c is predicted to bind to the ATP binding site similarly to ceritinib and WZ4002 making identical hydrogen bonds to the hinge residues Met1199 and Met793. We carefully investigated the binding modes of two distinct functional groups such as the meta-acrylamide in ALK and the ortho-isopropylsulfone in EGFR. The residue corresponding to the Cys797 in EGFR, which forms the covalent bond with the meta-acrylamide (Table S2), is the Asp1203 in ALK. We did not find an additional interaction between the meta-acrylamide of 7c and the Asp1203, but it is well accommodated by this residue at the edge of the ATP binding pocket. On the other hand, ALK and EGFR have the same residues around the region of the binding pocket occupied by the ortho-isopropylsulfone, except for the DFG-1 residue (Gly1269 in ALK and Thr854 in EGFR), which creates similar binding pockets. As a result, the ortho-isopropylsulfone is positioned in proximity to the catalytic Lys745 in wild type EGFR and T790M mutant EGFR, which forms an additional hydrogen bond as observed in the ceritinib-ALK co-crystal structure.

Figure 2.

Figure 2

Molecular docking models of compound 7c bound to (A) wild type ALK, (B) wild type EGFR and (C) T790M EGFR. Hydrogen bonds are indicated by dashed lines.

We assessed the kinase selectivity of 7c using the KINOMEscan technology [DiscoverX] [31, 32] against a diverse panel of 468 kinases at a concentration of 1 μM and compared it to the profiles obtained for both TAE684 (panel of 353 kinases) and ceritinib (Figure 3). Hydrogen bond acceptors such as sulfone, sulfonamide and phosphine oxide, are well known functional groups which interact with the conserved catalytic lysine. Installing these functional groups at ortho-position of C4-anilinopyrimidine leads to poor kinome selectivity. However, ceritinib, which has a bulky ortho-isopropoxy substituent at C2-anilinopyrimidine, achieved good kinase selectivity [S-score(1) = 0.02] even though it contains the ortho-isopropylsulfone at the C4-anilinopyrimidine, compared to TAE684 which has the ortho-methoxy group [S-score(1) = 0.37]. However, ceritinib lost affinity for EGFR proteins in KINOMEscan which encouraged us to retain the ortho-methoxy substituent of 2-anilinopyrimidine. Fortunately, 7c displayed superior overall kinome selectivity [S-score(1) = 0.07] relative to TAE684, and as expected it still bound to both wild type and mutants ALK and EGFR. The profiling revealed a number of potential off-targets such as ErbB2, ErbB4, BTK and BLK, which have a cysteine residue at the same position as the Cys797 of EGFR [24], and several non-receptor tyrosine kinases (Table S3).

We investigated the cell growth inhibitory activity of 7c against patient-derived lung cancer cell line, DFCI032, which harbors wild type EML4-ALK together with activated EGFR [33]. We also tested DFCI076 cell line, which harbors both activated EGFR and an L1152R mutation in the ALK kinase domain, which renders cells resistant to most ALK inhibitors [14, 34] (Figure 4A and 4B). As expected, ceritinib or osimertinib alone were not able to efficiently inhibit the proliferation of DFCI032 (EC50 = 4.44 μM and 4.73 μM, respectively). In contrast, our lead compound 7c exhibited remarkable inhibitory effect against DFCI032 cells with an EC50 value of 0.17 μM, which is 25-fold more potent than ceritinib or osimertinib alone and equipotent to a 1:1 combination treatment of ceritinib and osimertinib (EC50 = 0.19 μM). On the other hand, the 1:1 combination of ceritinib and osimertinib was not able to efficiently inhibit the proliferation of DFCI076 due to the loss of ALK inhibition. 7c was also less effective against DFCI076, but was 5-fold more potent than the 1:1 combination of ceritinib and osimertinib, which was presumably due to stronger ALK inhibition than ceritinib. This result strongly supports the ability of 7c to inhibit both ALK and EGFR in a patient-derived cellular context, in good agreement with the engineered Ba/F3 cellular assays.

Figure 4.

Figure 4

Figure 4

Profiles of compound 7c in patient-derived NSCLC. (A) Dose-dependent cell growth curves of ceritinib (orange), osimertinib (cyan), compound 7c (red) and the 1:1 combination of ceritinib and osimertinib (black) against DFCI032 and DCI076. (B) Cellular contexts of DFCI032 and DFCI076 and EC50 values. (C) Effects on phosphorylation of ALK, EGFR, Akt and ERK in DFCI032 cells.

To assess the capability of 7c to downregulate ALK and EGFR signaling pathways simultaneously in a cellular context, we performed Western blotting for phosphorylated ALK and EGFR as well as downstream signaling mediators such as Akt and ERK in DFCI032 cells (Figure 4C). Ceritinib and osimertinib suppressed phosphorylation of their primary target respectively but were not able to sufficiently reduce phosphorylation of Akt and ERK. On the other hand, the 1:1 combination of ceritinib and osimertinib effectively inhibited phosphorylation of both ALK and EGFR as well as Akt and ERK. Consistent with the cell growth inhibitory activity, 7c alone also showed dual inhibition of ALK and EGFR phosphorylation which led to better suppression of both PI3K/Akt and MAPK/ERK pathways than the 1:1 combination of ceritinib and osimertinib.

We profiled the in vitro metabolic stability of newly synthesized compounds using human, dog, rat and mouse liver microsomes and we evaluated their ability to inhibit cytochrome P450 (CYP) in vitro (Table 3). Generally, the dimethylphosphine oxide analogs showed superior microsomal stability and lower CYP enzyme inhibition than the corresponding isopropylsulfone analogs and the dimethylsulfonamide analogs which displayed 30 – 60 % inhibition of CYP 3A4 and approximately 20 % inhibition of other CYP enzymes. Our lead compound 7c was not stable in liver microsomes of all species whereas corresponding dimethylphosphine oxide analog 7g displayed good microsomal stability. This result was not surprising because the dimethylphosphine oxide analogs showed lower clog P values than the isopropylsulfone analogs and dimethylsulfonamide analogs.

Table 3.

Metabolic Stability in Liver Microsome and Cytochrome P450 Enzyme Inhibition of Compound 7a-l.

Compd. No. R1 R2 Cytochrome P450 inhibition (% of control) Liver microsomal stability (% remaining after 30 min) cLog Pa

1A2 2C9 2C19 2D6 3A4 Human Dog Rat Mouse
7a H graphic file with name nihms878864t1.jpg 80 90 87 67 40 5 48 22 8 3.9
7b F graphic file with name nihms878864t1.jpg 91 94 93 95 51 26 27 11 15 4.1
7c Cl graphic file with name nihms878864t1.jpg 89 83 86 90 44 16 17 11 12 4.7
7d I graphic file with name nihms878864t1.jpg 81 88 86 82 47 14 19 14 10 5.1
7e H graphic file with name nihms878864t2.jpg 84 90 >100 87 89 70 73 78 76 2.0
7f F graphic file with name nihms878864t2.jpg >100 >100 >100 >100 >100 84 87 87 80 2.2
7g Cl graphic file with name nihms878864t2.jpg 93 >100 98 85 91 61 77 76 66 2.8
7h I graphic file with name nihms878864t2.jpg 91 >100 99 80 88 65 93 81 57 3.2
7i H graphic file with name nihms878864t3.jpg 82 88 85 79 56 10 50 6 5 3.9
7j F graphic file with name nihms878864t3.jpg >100 88 91 >100 71 9 33 7 10 4.1
7k Cl graphic file with name nihms878864t3.jpg 87 59 75 88 39 11 31 13 23 4.6
7l I graphic file with name nihms878864t3.jpg 79 85 67 93 54 13 35 12 12 5.0
a

Calculated log P using ChemDraw Professional version 16.0.0.82.

7c displayed reasonable mouse pharmacokinetic properties with a moderate T1/2 of 1.25 hour and AUClast value of 1551 ng·h/mL following an oral dose of 10 mg/Kg (Table 4A). Furthermore, 2 hours after an intravenous (2 mg/Kg) or an oral (10 mg/Kg) dose, 7c showed higher lung exposure (2.36 μM and 4.64 μM) than plasma exposure (0.10 μM and 0.15 μM) which lasted for 8 hours (0.11 μM and 0.74 μM versus 0.00 μM and 0.01 μM). Despite low bioavailability (24 %), the preferential accumulation of 7c in lung tissue over plasma would be advantageous to use in studying the effects of dual ALK/EGFR inhibition in murine models of lung cancer (Table 4B).

Table 4.

(A) Pharmacokinetic Properties and (B) In vivo Lung Tissue Availability of Compound 7c.

(A)
ID route dose (mg/Kg) matrix T1/2 (hr) Tmax (hr) Cmax (ng/mL) AUClast (hr·ng/mL) Cl_obs (mL/min/Kg) Vss_obs (L/Kg) F (%)
7c IV 2 plasma 1.25 882 1287 28.99 2.66
PO 10 plasma 1.17 520 1551 114.54 24
(B)
ID matrix route dose (mg/Kg) time (hr) conc. (μM)
7c plasma IV 2 2 0.10
PO 10 2 0.15
IV 2 8 0.00
PO 10 8 0.01
7c lung IV 2 2 2.36
PO 10 2 4.64
IV 2 8 0.11
PO 10 8 0.74

2.4 Chemistry

The synthesis, which is described in scheme 1, followed well established synthetic routes for similar compounds [13, 21, 23]. All variations were made at the first nucleophilic aromatic substitution (SNAr) reaction step using commercially available 2,4-dichloropyrimidine cores and three different anilines. Then, we performed a four-step reaction sequence to synthesize the isopropylsulfone and dimethylsulfoamide analogs (7a – d and 7i – l) including two SNAr reactions, nitro-reduction and acrylamide formation to install the C2-aniline substituent. The dimethylphosphine oxide analogs (7e – h) were also made via similar route except for a Buchwald coupling which was employed to install the 4-fluoro-2-methoxy-5-nitroaniline at C2-position of pyrimidine.

Scheme 1.

Scheme 1

Synthetic Routes for Compound 7a-l.

(i) 2-1 and 2-3 : NaH, DMF/DMSO (9:1), 0 °C; 2-2 : K2CO3, DMF, 70 °C; (ii) 3a – d and 3i – l : 4-fluoro-2-methoxy-5-nitroaniline, TFA, 2-BuOH, 100 °C; 3e – h : 4-fluoro-2-methoxy-5-nitroaniline, Pd2dba3, Xphos, K2CO3, 2-BuOH; (iii) N1,N1,N2-trimethylethane-1,2-diamine, 1,4-dioxane, 80 °C; (iv) 5a – d and 5i – l : SnCl2·2H2O, HCl, EtOAc; 5e – h : Pd/C, H2 gas, MeOH; (v) acryloyl chloride, sat. NaHCO3/THF (1:1).

3. Conclusion

Based on our previous effort developing covalent T790M EGFR inhibitor, WZ4002, we have rationally designed and synthesized the potent dual inhibitor of ALK and EGFR, particularly the T790M resistant mutant, by combining the crucial functional groups of known ALK inhibitors ceritinib and brigatinib, and EGFR inhibitors WZ4002 and osimertinib, in the 2,4-diarylaminopyrimidine scaffold. This effort resulted in the identification of 7c as a potent dual inhibitor of ALK and EGFRs, including T790M and L858R/T790M mutants, in biochemical assays with low nanomolar IC50 values, which was well correlated with cell growth inhibitory activities against EML4-ALK, wild type EGFR, L858R, exon19del, L858R/T790M or exon19del/T790M EGFR transformed Ba/F3 cells. Moreover, compound 7c recapitulated the cell growth inhibitory activity observed upon combinatorial treatment with both ceritinib and osimertinib against the patient-derived NSCLC cell line, DFCI032 which harbors both EML4-ALK and activated EGFR, and inhibited the cellular phosphorylation of ALK and EGFR, as well as their downstream signaling proteins Akt and ERK. These results suggest that 7c is a promising lead compound to develop advanced ALK and EGFR dual inhibitors. Further optimization of this scaffold to improve metabolic stability and kinome-wide selectivity is currently underway.

4. Experimental protocols

4.1 Chemistry

Starting materials, reagents and solvents were purchased from commercial suppliers and were used without further purification unless otherwise noted. All reactions were monitored using a Waters Acquity UPLC/MS system (Waters PDA eλ Detector, QDa Detector, Sample manager – FL, Binary Solvent Manager) using Acquity UPLC® BEH C18 column (2.1 × 50 mm, 1.7 μm particle size): solvent gradient = 90 % A at 0 min, 1 % A at 1.6 min; solvent A = 0.1 % formic acid in Water; solvent B = 0.1 % formic acid in Acetonitrile; flow rate : 0.6 mL/min. Reaction products were purified by flash column chromatography using CombiFlash®Rf with Teledyne Isco RediSep®Rf High Performance Gold or Silicycle SiliaSep High Performance columns (4 g, 12 g, 24 g, 40 g, or 80 g) and Waters HPLC system using SunFire Prep C18 column (19 × 100 mm, 5 μm particle size): solvent gradient = 80 % A at 0 min, 10 % A at 25 min; solvent A = 0.035 % TFA in Water; solvent B = 0.035 % TFA in MeOH; flow rate : 25 mL/min. 1H NMR spectra were recorded on 400 MHz and 500 MHz Bruker Avance III spectrometers and 13C NMR spectrum was recorded on 125 MHz Bruker Avance III spectrometer. The mass spectrometry data were measured in positive electrospray ionization (ESI) mode on LCMS-2020 system (Shimadzu, Tokyo, Japan). Chemical shifts are reported relative to methanol (δ = 3.30), chloroform (δ = 7.24) or dimethyl sulfoxide (δ = 2.50) for 1H NMR and 13C NMR. Data are reported as (br = broad, s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet).

4.1.1 Representative procedure for the synthesis of compounds 7a7d and 7i7l

2,5-Dichloro-N-(2-(isopropylsulfonyl)phenyl)pyrimidin-4-amine (3c)

To a solution of 2-(isopropylsulfonyl)aniline (3.0 g, 15.1 mmol) in DMF (80 mL) was added sodium hydride (1.2 g, 30.1 mmol, 60 % in mineral oil) at 0 °C. After stirring for 30 min, 2,4,5-trichloropyrimidine was added to the reaction mixture followed by warming the mixture to room temperature. After stirring for 2 h, the reaction mixture was quenched with ice and diluted with excess water. The precipitate was filtered and the solid was dried by blowing nitrogen gas to obtain 3c as an off-white solid (3.75 g, 72 %). 1H NMR (500 MHz, DMSO-d6) δ 9.81 (s, 1H), 8.56 (s, 1H), 8.33 – 8.31 (m, 1H), 7.89 (dd, J = 7.9, 1.5 Hz, 1H), 7.88 – 7.84 (m, 1H), 7.48 (td, J = 7.6, 1.2 Hz, 1H), 3.58 – 3.48 (m, 1H), 1.16 (d, J = 6.7, 6H); LC/MS (ESI) m/z 346.18 [M+H]+.

5-Chloro-N2-(4-fluoro-2-methoxy-5-nitrophenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (4c)

To a suspension of 2,5-dichloro-N-(2-(isopropylsulfonyl)phenyl)pyrimidin-4-amine (3c) (1.0 g, 2.89 mmol) and 4-fluoro-2-methoxy-5-nitroaniline (0.565 g, 3.03 mmol) in 2-butanol (10 mL) was added trifluoroacetic acid (1.1 mL, 14.5 mmol). Then, the mixture was stirred at 80 °C for 6 h followed by cooling to room temperature. The resulting mixture was concentrated under reduced pressure and the residue was purified by flash column chromatography on silica gel (0 to 10 % MeOH in DCM) to give 4c as a brown solid (1.23 g, 86 %). 1H NMR (500 MHz, DMSO-d6) δ 9.53 (s, 1H), 8.72 (s, 1H), 8.54 (d, J = 8.2 Hz, 1H), 8.44 (d, J = 7.3 Hz, 1H), 8.33 (s, 1H), 7.82 (dd, J = 7.9, 1.5 Hz, 1H), 7.62 – 7.55 (m, 1H), 7.37 – 7.31 (m, 2H), 3.95 (s, 3H), 3.50 – 3.40 (m, 1H), 1.16 (d, J = 6.7, 6H); LC/MS (ESI) m/z 496.36 [M+H]+.

5-Chloro-N2-(4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (5c)

To a solution of 5-chloro-N2-(4-fluoro-2-methoxy-5-nitrophenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (4c) (0.800 g, 1.61 mmol) in dioxane (5 mL) was added N1,N1,N2-trimethylethane-1,2-diamine (0.640 mL, 4.84 mmol). The reaction mixture was stirred at 100 °C for 2 h. After completion of the reaction, the resulting mixture was concentrated under reduced pressure and purified by flash column chromatography on silica gel (0 to 20 % MeOH in DCM) to give 5c as a sticky liquid (838 mg, 90 %) 1H NMR (500 MHz, DMSO-d6) δ 9.53 (s, 1H), 8.55 (s, 1H), 8.49 (br, 1H), 8.27 (s, 1H), 8.17 (br, 1H), 7.81 (dd, J = 7.9, 1.5 Hz, 1H), 7.60 – 7.51 (m, 1H), 7.34 – 7.29 (m, 1H), 6.84 (s, 1H), 3.90 (s, 3H), 3.49 – 3.40 (m, 1H), 3.40 – 3.24 (m, 2H), 2.88 (br, 2H), 2.83 (s, 1H), 2.47 (br, 6H), 1.16 (d, J = 6.7, 6H); LC/MS (ESI) m/z 578.59 [M+H]+.

N4-(5-Chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)-N1-(2-(dimethylamino)ethyl)-5-methoxy-N1-methylbenzene-1,2,4-triamine (6c)

To a suspension of 5-chloro-N2-(4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (5c) (0.274 g, 0.473 mmol) and tin (II) chloride dihydrate (1.00 g, 4.74 mmol) in ethyl acetate (7 mL) was added conc. HCl (1 mL). Then, the resulting mixture was stirred at 50 °C for 5 h and quenched with NH4OH (pH 5), Na2CO3 solid (pH 7) and the resulting suspension was filtered through Celite. The filtrate was concentrated under reduced pressure to afford 6c as a brown solid (217 mg, 92 %). 1H NMR (500 MHz, DMSO-d6) δ 9.58 (s, 1H), 8.67 (d, J = 8.6 Hz, 1H), 8.32 (s, 1H), 8.20 (s, 1H), 7.80 (dd, J = 7.9, 1.5 Hz, 1H), 7.69 – 7.63 (m, 1H), 7.33 – 7.27 (m, 1H), 7.08 (bs, 1H), 6.81 (s, 1H), 3.70 (s, 3H), 3.49 – 3.39 (m, 1H), 3.27 (br t, J = 6.0 Hz, 2H), 3.19 (br t, J = 5.8 Hz, 2H), 2.80 (s, 6H), 2.55 (s, 3H), 1.17 (d, J = 6.7 Hz, 6H); LC/MS (ESI) m/z 548.58 [M+H]+.

N-(5-((5-Chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide (7c)

To a solution of N4-(5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)-N1-(2-(dimethylamino)ethyl)-5-methoxy-N1-methylbenzene-1,2,4-triamine (6c) (120 mg, 0.219 mmol) in THF (2 mL) and aq.NaHCO3 (2 mL) was added dropwise acryloyl chloride (0.027 mL, 0.329 mmol) at 0 °C. After stirring at 0 °C for 10 min, the reaction mixture was diluted with excess DCM and washed with brine. The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative HPLC to give 7c as an off-white solid (52 mg, 53 %). 1H NMR (500 MHz, DMSO-d6) δ 10.02 (br s, 1H), 9.52 (s, 1H), 8.57 – 8.46 (m, 2H), 8.39 (s, 1H), 8.22 (s, 1H), 7.76 (dd, J = 7.9, 1.5 Hz, 1H), 7.47 (t, J = 7.0 Hz, 1H), 7.23 (t, J = 7.5 Hz, 1H), 7.00 (s, 1H), 6.48 – 6.30 (m, 1H), 6.15 (dd, J = 16.8, 1.8 Hz, 1H), 5.72 (dd, J = 10.2, 1.8 Hz, 1H), 3.76 (s, 3H), 3.47 – 3.38 (m, 1H), 2.90 (br s, 2H), 2.71 (s, 3H), 2.36 (br s, 2H), 2.24 (br s, 6H), 1.16 (d, J = 6.7 Hz, 6H); 13C NMR (125 MHz, DMSO-d6) δ 162.5, 158.5, 155.4, 154.5, 148.8, 140.1, 138.0, 134.5, 132.1, 130.8, 126.9, 126.2, 123.8, 123.4, 122.9, 122.9, 117.8, 105.4, 104.1, 56.7, 55.7, 54.9, 54.9, 45.1, 45.1, 42.2, 14.9, 14.9; LC/MS (ESI) m/z 602.69 [M+H]+; HRMS (ESI) calcd for C28H37ClN7O4S [M+H]+ 602.2311; found 602.2304.

4.1.1.1 N-(5-((5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)propionamide (7c-R)

7c-R was synthesized using the analog method that used to synthesize 7c. To a solution of N4-(5-chloro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)-N1-(2-(dimethylamino)ethyl)-5-methoxy-N1-methylbenzene-1,2,4-triamine (6c) (35 mg, 0.064 mmol) in THF (1 mL) and aq.NaHCO3 (1 mL) was added dropwise propionyl chloride (0.014 mL, 0.154 mmol) at 0 °C. After stirring at 0 °C for 10 min, the reaction mixture was diluted with excess DCM and washed with brine. The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative HPLC to give 7c-R as an off-white solid (28 mg, 72 %). 1HNMR (500 MHz, DMSO-d6) δ 9.64 (br s, 1H), 9.53 (s, 1H), 8.54 (d, J = 7.9 Hz, 1H), 8.48 (s, 1H), 8.22 (s, 1H), 7.78 (d, J = 7.9 Hz, 1H), 7.52 (br s, 1H), 7.26 (t, J = 7.5 Hz, 1H), 6.96 (s, 1H), 3.76 (s, 3H), 3.47 – 3.40 (m, 1H), 2.97 (br s, 2H), 2.68 (s, 3H), 2.60 – 2.05 (m, 10H), 1.17 (d, J = 6.7 Hz, 6H), 1.04 (t, J = 7.5 Hz, 3H); LC/MS (ESI) m/z 604.35 [M+H]+.

4.1.1.2 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide (7a)

The procedure that used to synthesize 7c was applied to prepare 7a from compound 1-1. 1H NMR (400 MHz, DMSO-d6) δ 10.09 (br s, 1H), 8.87 (br s, 1H), 8.55 (br s, 1H), 8.19 (d, J = 8.2 Hz, 1H), 8.07 (d, J = 5.6 Hz, 1H), 7.97 (s, 1H), 7.77 – 7.75 (m, 1H), 7.54 (br t, J = 8.0 Hz, 1H), 7.24 (t, J = 7.9 Hz, 1H), 6.96 (s, 1H), 6.36 – 6.33 (m, 2H), 6.19 – 6.14 (m, 1H), 5.74 – 5.71 (m, 1H), 3.79 (s, 3H), 3.48 – 3.34 (m, 1H), 2.86 (br t, J = 6.5 Hz, 2H), 2.69 (s, 3H), 2.28 (br t, J = 6.5 Hz, 2H), 2.20 (s, 6H), 1.14 (d, J = 6.8 Hz, 6H); LC/MS (ESI) m/z 568.45 [M+H]+; HRMS (ESI) calcd for C28H38N7O4S [M+H]+ 568.2700; found 568.2695.

4.1.1.3 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-fluoro-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide (7b)

The procedure that used to synthesize 7c was applied to prepare 7b from compound 1-2. 1H NMR (500 MHz, DMSO-d6) δ 9.54 (br s, 1H), 9.45 (d, J = 2.1 Hz, 1H), 8.55 (d, J = 8.5 Hz, 1H), 8.30 (br, 1H), 8.19 (d, J = 3.1 Hz, 1H), 8.00 (s, 1H), 7.80 (dd, J = 7.9, 1.5 Hz, 1H), 7.61 – 7.57 (m, 1H), 7.29 – 7.24 (m, 1H), 6.96 (s, 1H), 6.61 – 6.52 (m, 1H), 6.23 (dd, J = 17.9, 1.5 Hz, 1H), 5.75 (dd, J = 10.4, 1.5 Hz, 1H), 3.84 (s, 3H), 3.50 – 3.42 (m, 1H), 3.16 (br, 3H), 3.01 (br, 2H), 2.69 – 2.62 (s, 8H), 1.18 (d, J = 6.7 Hz, 6H); LC/MS (ESI) m/z 586.72 [M+H]+; HRMS (ESI) calcd for C28H37FN7O4S [M+H]+ 586.2606; found 586.2596.

4.1.1.4 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((5-iodo-4-((2-(isopropylsulfonyl)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide (7d)

The procedure that used to synthesize 7c was applied to prepare 7d from compound 1-4. 1H NMR (400 MHz, DMSO-d6) δ 10.13 (br s, 1H), 9.13 (br s, 1H), 8.46 – 8.42 (m, 4H), 7.81 – 7.79 (m, 1H), 7.50 (br t, J = 7.3 Hz, 1H), 7.31 – 7.24 (m, 1H), 7.05 (s, 1H), 6.45 – 6.38 (m, 1H), 6.18 (m, 1H), 5.77 (m, 1H), 3.81 (s, 3H), 3.51 – 3.41 (m, 1H), 2.94 – 2.91 (m, 2H), 2.77 (s, 3H), 2.39 – 2.35 (m, 2H), 2.26 (s, 6H), 1.24 (d, J = 6.8 Hz, 6H); LC/MS (ESI) m/z 694.30 [M+H]+; HRMS (ESI) calcd for C28H37IN7O4S [M+H]+ 694.1667; found 694.1661.

4.1.1.5 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(N,N-dimethylsulfamoyl)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide (7i)

The procedure that used to synthesize 7c was applied to prepare 7i from compounds 1-1 and 2-3. 1H NMR (400 MHz, CDCl3) δ 10.06 (s, 1H), 8.73 (br s, 1H), 8.58 (br s, 1H), 8.16 (d, J = 8.0 Hz, 1H), 8.06 (d, J = 5.7 Hz, 1H), 7.91 (s, 1H), 7.72 – 7.69 (m, 1H), 7.46 (br t, J = 7.3 Hz, 1H), 7.20 (t, J = 7.4 Hz, 1H), 6.96 (s, 1H), 6.39 – 6.32 (m, 2H), 6.17 – 6.12 (m, 1H), 5.73 – 5.70 (m, 1H), 3.79 (s, 3H), 2.85 (br t, J = 5.2 Hz, 2H), 2.69 (s, 3H), 2.62 (s, 6H), 2.29 (br t, J = 5.2 Hz, 2H), 2.21 (s, 6H); LC/MS (ESI) m/z 569.45 [M+H]+; HRMS (ESI) calcd for C27H37N8O4S [M+H]+ 569.2653; found 569.2649.

4.1.1.6 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(N,N-dimethylsulfamoyl)phenyl)amino)-5-fluoropyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide (7j)

The procedure that used to synthesize 7c was applied to prepare 7j from compounds 1-2 and 2-3. 1H NMR (400 MHz, DMSO-d6) δ 10.09 (br s, 1H), 9.28 (br s, 1H), 8.56 (d, J = 8.3 Hz, 1H), 8.50 (br s, 1H), 8.25 (s, 1H), 8.20 (s, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.47 (br t, J = 7.6 Hz, 1H), 7.22 (br t, J = 7.6 Hz, 1H), 6.99 (s, 1H), 6.44 – 6.37 (m, 1H), 6.16 (br d, J = 16.9 Hz, 1H), 5.74 (br d, J = 10.2. Hz, 1H) 3.78 (s, 3H), 2.89 (m, 2H), 2.70 (s, 3H), 2.51 (d, J = 1.48 Hz, 6H), 2.35 (m, 2H), 2.24 (s, 6H); LC/MS (ESI) m/z 587.20 [M+H]+; HRMS (ESI) calcd for C27H36FN8O4S [M+H]+ 587.2559; found 587.2551.

4.1.1.7 N-(5-((5-Chloro-4-((2-(N,N-dimethylsulfamoyl)phenyl)amino)pyrimidin-2-yl)amino)-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide (7k)

The procedure that used to synthesize 7c was applied to prepare 7k from compounds 1-3 and 2-3. 1H NMR (400 MHz, DMSO-d6) δ 10.08 (br s, 1H), 9.37 (br s, 1H), 8.51 – 8.48 (m, 2H) 8.42 (s, 1H), 8.22 (s, 1H), 7.73 – 7.71 (m, 1H), 7.41 (br t, J = 8.0 Hz, 1H), 7.20 (t, J = 7.8 Hz, 1H), 7.01 (s, 1H), 6.40 – 6.33 (m, 1H), 6.17 – 6.12 (m, 1H), 5.73 – 5.70 (m, 1H), 3.76 (s, 3H), 2.87 (br t, J = 5.6 Hz, 2H), 2.72 (s, 3H), 2.64 (s, 6H), 2.31 (br t, J = 5.6 Hz, 2H), 2.21 (s, 6H); LC/MS (ESI) m/z 603.45 [M+H]+; HRMS (ESI) calcd for C27H36ClN8O4S [M+H]+ 603.2263; found 603.2256.

4.1.1.8 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(N,N-dimethylsulfamoyl)phenyl)amino)-5-iodopyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide (7l)

The procedure that used to synthesize 7c was applied to prepare 7l from compounds 1-4 and 2-3. 1H NMR (400 MHz, DMSO-d6) δ 9.91 (s, 1H), 9.01 (s, 1H), 8.98 – 8.92 (m, 4H), 7.74 – 7.67 (m, 1H), 7.43 (br t, J = 7.7 Hz, 1H), 7.23 (t, J = 7.1 Hz, 1H), 6.94 (s, 1H), 6.63 – 6.60 (m, 1H), 6.15 – 6.11 (m, 1H), 5.73 – 5.69 (m, 1H), 3.77 (s, 3H), 3.08 – 3.06 (m, 2H), 2.80 (s, 3H), 2.40 – 2.38 (m, 2H), 2.20 (s, 6H), 1.23 (s, 6H); LC/MS (ESI) m/z 695.30 [M+H]+; HRMS (ESI) calcd for C27H36IN8O4S [M+H]+ 695.1619; found 695.1606.

4.1.2 Representative procedure for the synthesis of compounds 7e7h

(2-((2-Chloro-5-fluoropyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (3f)

To a solution of 2,4-dichloro-5-fluoropyrimidine (0.5 g, 2.99 mmol) and (2-aminophenyl)dimethylphosphine oxide (0.323 g, 1.907 mmol) in DMF (6.36 mL) was added potassium carbonate (0.828 g, 5.99 mmol). The reaction mixture was stirred at 70 °C for overnight. After completion of the reaction, the resulting mixture was cooled to room temperature, quenched with water, extracted with DCM and washed with brine. The combined organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (0 to 20 % MeOH in DCM) to give 3f as a yellowish solid (0.178 g, 31 %). 1H NMR (400 MHz, DMSO-d6) δ 12.09 (s, 1H), 8.54 – 8.51 (m, 1H), 8.37 (br d, J = 3.0 Hz, 1H), 7.68 – 7.60 (m, 2H), 7.23 (br t, J = 6.7 Hz, 1H), 1.83 (s, 3H), 1.80 (s, 3H); LC/MS (ESI) m/z 300.00 [M+H]+.

(2-((5-Fluoro-2-((4-fluoro-2-methoxy-5-nitrophenyl)amino)pyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (4f)

To a solution of (2-((2-chloro-5-fluoropyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (3f) (0.1 g, 0.334 mmol) and 4-fluoro-2-methoxy-5-nitroaniline (0.124 g, 0.667 mmol) in sec-butanol (2 mL) was added potassium carbonate (0.231 g, 1.669 mmol). The reaction mixture was stirred at 80 °C for 10 min and then Pd2(dba)3 (0.031 g, 0.033 mmol) and Xphos (0.016 g, 0.033 mmol) were added rapidly. The reaction mixture was stirred at 100 °C for 2 h. After completion of the reaction, the resulting mixture was hot-filtered through Celite and washed with DCM. The filtrate was concentrated under reduced pressure and then purified by flash column chromatography on silica gel (0 to 20 % MeOH in DCM) to afford 4f as a yellowish solid (0.128 g, 85 %). 1H NMR (400 MHz, DMSO-d6) δ 11.64 (s, 1H), 8.74 (d, J = 8.4 Hz, 1H), 8.63 – 8.60 (m, 1H), 8.38 (s, 1H), 8.18 (br d, J = 3.3 Hz, 1H), 7.60 – 7.55 (m, 1H), 7.40 (br t, J = 8.1 Hz, 1H), 7.33 (d, J = 13.4 Hz, 1H), 7.12 (br t, J = 7.2 Hz, 1H), 3.97 (s, 3H), 1.81 (s, 3H), 1.78 (s, 3H); LC/MS (ESI) m/z 450.05 [M+H]+.

(2-((2-((4-((2-(Dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)amino)-5-fluoropyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (5f)

To a solution of (2-((5-fluoro-2-((4-fluoro-2-methoxy-5-nitrophenyl)amino)pyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (4f) (0.128 g, 0.285 mmol) in dioxane (3 mL) was added N1,N1,N2-trimethylethane-1,2-diamine (0.362 mL, 2.85 mmol). After stirring at 100 °C for 2 h, the resulting mixture was concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel to give 5f as a yellowish solid (0.151g, quantitative).1H NMR (400 MHz, DMSO-d6) δ 11.55 (br s, 1H), 8.58 – 8.57 (m, 1H), 8.22 (s, 1H), 8.15 (s, 1H), 8.10 (br d, J = 3.3 Hz, 1H), 7.58 – 7.52 (m, 1H), 7.33 (br t, J = 7.7 Hz, 1H), 7.08 (br t, J = 6.7 Hz, 1H), 6.79 (s, 1H), 3.89 (s, 3H), 3.23 (br t, J = 6.8 Hz, 2H), 2.82 (s, 3H), 2.46 – 2.41 (m, 2H), 2.13 (s, 6H), 1.79 (s, 3H), 1.76 (s, 3H); LC/MS (ESI) m/z 532.15 [M+H]+.

(2-((2-((5-Amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)amino)-5-fluoropyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (6f)

To a solution of 2-((2-((4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxy-5-nitrophenyl)amino)-5-fluoropyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (5f) (0.151 g, 0.284 mmol) in MeOH (20 mL) was added 10 % Pd/C (0.02 g, 0.019 mmol). The reaction mixture was stirred at room temperature under an atmosphere of H2 for overnight. After completion of the reaction, the resulting mixture was filtered through Celite, and was washed with methanol. The filtrate was concentrated under reduced pressure to afford 6f as a yellowish solid (0.142 g, quantitative). 1H NMR (400 MHz, DMSO-d6) δ 11.46 (br s, 1H), 8.68 – 8.65 (m, 1H), 8.04 (br d, J = 3.4 Hz, 1H), 7.82 (s, 1H), 7.57 – 7.51 (m, 1H), 7.45 (br t, J = 8.0 Hz, 1H), 7.13 (s, 1H), 7.08 (br t, J = 7.4 Hz, 1H), 6.72 (s, 1H), 3.68 (s, 3H), 2.97 – 2.94 (m, 2H), 2.57 (s, 3H), 2.35 (br s, 6H), 1.79 (s, 3H), 1.75 (s, 3H); LC/MS (ESI) m/z 502.15 [M+H]+.

N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(dimethylphosphoryl)phenyl)amino)-5-fluoropyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide trifluoroacetate salt (7f)

To a solution of (2-((2-((5-amino-4-((2-(dimethylamino)ethyl)(methyl)amino)-2-methoxyphenyl)amino)-5-fluoropyrimidin-4-yl)amino)phenyl)dimethylphosphine oxide (6f) (0.142 g, 0.283 mmol) in THF (5 mL) and aq.NaHCO3 (5 mL) was added dropwise acryloyl chloride (0.034 mL, 0.425 mmol) at 0 °C. After stirring for 10 min, the resulting mixture was diluted with DCM and washed with brine. The organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by preparative HPLC to give 7f as a yellowish solid (0.037 g, 23 %). 1H NMR (400 MHz, DMSO-d6) δ 11.83 (br d, J = 19.5 Hz, 1H), 9.58 (s, 1H), 9.25 (br s, 1H), 8.64 (br s, 1H), 8.53 – 8.51 (m, 1H), 8.15 – 8.13 (m, 2H), 7.60 – 7.55 (m, 1H), 7.37 (br t, J = 7.8 Hz, 1H), 7.10 (br t, J = 7.7 Hz, 1H), 6.98 (s, 1H), 6.63 – 6.56 (m, 1H), 6.28 (d, J = 16.8 Hz, 1H), 5.79 (d, J = 10.2 Hz, 1H), 3.83 (s, 3H), 3.29 – 3.24 (m, 4H), 2.78 (s, 6H), 2.59 (s, 3H), 1.80 (s, 3H), 1.77 (s, 3H); LC/MS (ESI) m/z 556.30 [M+H]+; HRMS (ESI) calcd for C27H36FN7O3P [M+H]+ 556.2596; found 556.2592.

4.1.2.1 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide trifluoroacetate salt (7e)

The procedure that used to synthesize 7f was applied to prepare 7e from compound 1-1. 1H NMR (400 MHz, CD3OD) δ 8.04 (br s, 1H), 7.90 (br d, J = 6.0 Hz, 1H), 7.72 – 7.67 (m, 2H), 7.56 (br t, J = 7.5 Hz, 1H), 7.37 (br t, J = 7.4 Hz, 1H), 6.99 (s, 1H), 6.54 – 6.49 (m, 2H), 6.41 (d, J = 7.2Hz, 1H), 5.92 – 5.90 (m, 1H), 3.95 (s, 3H), 3.51 – 3.47 (m, 2H), 3.28 – 3.25 (m, 2H),) 2.87 (s, 6H), 2.71 (s, 3H), 1.86 (s, 3H), 1.83 (s, 3H); LC/MS (ESI) m/z 538.15 [M+H]+; HRMS (ESI) calcd for C27H37N7O3P [M+H]+ 538.2690; found 538.2689.

4.1.2.2 N-(5-((5-Chloro-4-((2-(dimethylphosphoryl)phenyl)amino)pyrimidin-2-yl)amino)-2-((2-(dimethylamino)ethyl)(methyl)amino)-4-methoxyphenyl)acrylamide trifluoroacetate salt (7g)

The procedure that used to synthesize 7f was applied to prepare 7g from compound 1-3. 1H NMR (400 MHz, DMSO-d6) δ 11.30 (s, 1H), 9.57 (s, 1H), 9.20 (br s, 1H), 8.43 – 8.41 (m, 2H), 8.15 (s, 1H), 8.08 (s, 1H), 7.57 – 7.51 (m, 1H), 7.33 (br t, J = 7.8 Hz, 1H), 7.06 (br t, J = 7.2 Hz, 1H), 6.98 (s, 1H), 6.62 – 6.56 (m, 1H), 6.31 – 6.26 (m, 1H), 5.81 – 5.78 (m, 1H), 3.83 (s, 3H), 3.30 – 3.24 (m, 4H), 2.79 (br s, 6H), 2.59 (s, 3H), 1.78 (s, 3H), 1.74 (s, 3H); LC/MS (ESI) m/z 573.10 [M+H]+; HRMS (ESI) calcd for C27H36ClN7O3P [M+H]+ 572.2300; found 572.2294.

4.1.2.3 N-(2-((2-(Dimethylamino)ethyl)(methyl)amino)-5-((4-((2-(dimethylphosphoryl)phenyl)amino)-5-iodopyrimidin-2-yl)amino)-4-methoxyphenyl)acrylamide trifluoroacetate salt (7h)

The procedure that used to synthesize 7f was applied to prepare 7h from compound 1-4. 1H NMR (400 MHz, DMSO-d6) δ 10.70 (br s, 1H), 9.55 (s, 1H), 9.24 (br s, 1H), 8.45 – 8.43 (m, 1H), 8.33 (s, 1H), 8.13 – 8.10 (m, 1H), 8.05 (s, 1H), 7.55 – 7.50 (m, 1H), 7.33 (br t, J = 7.8 Hz, 1H), 7.07 (br t, J = 8.0 Hz, 1H), 6.96 (s, 1H), 6.64 – 6.57 (m, 1H), 6.31 – 6.26 (m, 1H), 5.82 – 5.79 (m, 1H), 3.82 (s, 3H), 3.28 – 3.22 (m, 4H), 2.78 (br d, J = 4.6 Hz, 6H), 2.57 (s, 3H), 1.76 (s, 3H), 1.73 (s, 3H); LC/MS (ESI) m/z 664.00 [M+H]+; HRMS (ESI) calcd for C27H36IN7O3P [M+H]+ 664.1656; found 664.1645.

4.2 Pharmacology

4.2.1 Enzymatic assays

The enzymatic activity against ALK was tested using Z′-Lyte assay with the ATP concentration equal to the apparent Km. The protocol is available from Life Technologies. All EGFR kinase inhibition assays were conducted by using Promega ADP-Glo Kinase assay systems (Catalog number; V4507, V5325, and V9261). Final compound concentrations were 50 uM, 5 uM, 500 nM, 50 nM, 5 nM, 500 pM, 50 pM, 5 pM, and 0.5 pM and performed at each concentration with osimertinib as a positive control. Each compound was mixed with 0.2 ug/uL of substrate (Poly G:T (4:1) (Signal Chem, Richmond, BC, Canada)), 5 μmol/L ATP (Invitrogen, Carlsbad, CA), 5 ng of EGFR enzyme, and kinase reaction buffer (40 mmol/L TrisHCl, 10 mmol/L MgCl2, and 0.1 μg/μL BSA(bovine serum albumin)) in 384-well plate. After 30 min reaction at room temperature, ADP-Glo (Promega, Madison, WI) reagent was added and incubated at room temperature for 40 min. Finally, Kinase Detection Reagent was added and reacted at room temperature for 30 min. The luminescence signals were detected by using a microplate ELISA reader (Bio-Tek). Compound inhibition curves were fitted using Graphpad Prism 6.0 software.

4.2.2 Cell lines and cell growth inhibition assay

The NSCLC cell lines DFCI032 (EML4-ALK variant 1 E13:A20), and DFCI076 (EML4-ALK variant 3 E6:A20) have been previously published [14, 33]. DFCI032 and DFCI076 were established at Dana-Farber Cancer Institute from patients with crizotinib resistance and were cultured in ACL-4 media (Life Technologies) supplemented with 10% FBS, 100 units/mL penicillin, 100mg/mL streptomycin. EML4-ALK variant 1, wild type EGFR, L858R, exon19del, T790M/L858R and T790M/exon19del transformed Ba/F3 cells were established as previously described [14, 24, 35] and maintained in complete RPMI 1640 media without IL-3. Parental Ba/F3 cells and wildtype EGFR Ba/F3 cells were grown in complete RPMI 1640 media in the presence of IL-3 or EGF, respectively.

Ba/F3 cells and NSCLC cells were treated with dose-escalated drug over the course of 72 hours. Growth and inhibition of growth was assessed by MTS assay according to previously established methods. All experimental treatments were performed in replicates (6–12 wells per treatment) and the presented data are representative of the replicates. The data was graphically displayed using GraphPad Prism version 5.0 for Windows. The EC50 values were fitted using a non-linear regression model with a sigmoidal dose response.

4.2.3 Antibodies and Western blotting

Cell lysis and Western blotting were done as previously described [24]. Antibodies against total-ALK (D5F3), phospho-ALK (Tyr-1604), phospho-EGFR (Tyr-1068; #3777), phospho-Akt (Ser-473; D9E), total-Akt, Total-ERK1/2 and phospho-ERK1/2 (pT185/pY187) were obtained from Cell Signaling Technology; total-EGFR from Bethyl Laboratories; α-tubulin from Sigma Aldrich.

4.2.4 Kinome selectivity profiling

Kinome selectivity profiling was performed using the KINOMEscan method at a compound concentration of 1 μM. Protocols are available from DiscoverX.

4.2.5 Liver microsomal stability test

Acetaminophen, chlorpropamide, dextromethorphan, dextrorphan, phenacetin, and tolbutamide were purchased from Sigma-Aldrich (St. Louis, MO). Pooled human liver microsomes (150 donor) and dog, rat, mouse liver microsomes were obtained from BD Gentest Co. (Woburn, MA). NADPH regeneration system was obtained from Promega. S-Mephenytoin, 4-hydroxymephenytoin, 1′-hydroxymidazolam were purchased from Ultrafine Chemical Co. (Manchester, UK). All other chemicals and solvents were of the highest grade available. The liver microsomal stability assay was performed by incubation of human and selected animal liver microsomes at 37 °C with a test compound at a final concentration of 1 μM, in the presence of 0.5 mg/mL microsomal protein and NADPH regeneration system, in a total volume of 100 μL of 100 mM phosphate buffer, pH 7.4. The incubation was started by the addition of NADPH regeneration system and terminated with 40 μL of ice-cold acetonitrile at 0 and 30 min. Precipitated proteins were removed by centrifugation at 10,000×g for 5 min at 4 °C. Aliquots of the supernatant were injected onto an LC-MS/MS system. Incubations terminated prior to the addition of the NADPH regeneration system (time point 0 min) were used as standards, defined as 100 %. Percent of the parent compound remaining is calculated by comparing peak areas.

4.2.6 Cytochrome P450 inhibition assay

All incubations were performed in duplicate and the mean values were used for analysis. The assays of phenacetin O-deethylase, tolbutamide 4-hydroxylase, S-mephenytoin 4-hydroxylase, dextromethorphan O-demethylase and midazolam 1′-hydroxylase activities were determined as probe activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A, respectively, using cocktail incubation and tandem mass spectrometry. Briefly, incubation reaction was performed with 0.25 mg/mL human liver microsomes in a final incubation volume of 100 μL. The incubation medium contained 100 mM phosphate buffer (pH 7.4) with probe substrates. The incubation mixture containing various inhibitors (10 μM) was pre-incubated for 5 min. After pre-incubation, an NADPH regenerating system was added. After incubation at 37 °C for 15 min, the reaction was stopped by placing the incubation tubes on ice and adding 40 μL of ice-cold acetonitrile. The incubation mixtures were then centrifuged at 10,000×g for 5 min at 4 °C. Aliquots of the supernatant were injected onto an LC-MS/MS system. The CYP-mediated activities in the presence of inhibitors were expressed as percentages of the corresponding control values.

Supplementary Material

supplement

Highlights.

  • Dual inhibitors of ALK/EGFR, particularly the T790M mutant, were rationally designed.

  • Potencies of compounds were assessed using both enzymatic and cellular assays.

  • 7c displayed remarkable inhibitory activities against both ALK and EGFR.

  • 7c inhibited the growth of DFCI032 cells harboring EML4-ALK and activated EGFR.

Acknowledgments

This work was supported by a grant of the Daegu-Gyeongbuk Medical Innovation Foundation New Drug Development Center R&D Project.

Appendix A. Supplementary data

Supplementary data related to this article can be found at XXXX

Contains the biochemical activities of ceritinib and WZ4002 against EGFR T790M/L858R in the Z′-LYTE assay, the antiproliferative activity of 7c-R against L858R and T790M/L858R EGFR transformed Ba/F3 cells, the biochemical activity of 7c against potential additional targets, and the full KINOMEscan data of 7c.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell. 2010;141:1117–1134. doi: 10.1016/j.cell.2010.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Mok TS. Personalized medicine in lung cancer: what we need to know. Nat Rev Clin Oncol. 2011;8:661–668. doi: 10.1038/nrclinonc.2011.126. [DOI] [PubMed] [Google Scholar]
  • 3.Chan BA, Hughes BG. Targeted therapy for non-small cell lung cancer: current standards and the promise of the future. Transl Lung Cancer Res. 2015;4:36–54. doi: 10.3978/j.issn.2218-6751.2014.05.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kwak EL, Bang YJ, Camidge DR, Shaw AT, Solomon B, Maki RG, Ou SHI, Dezube BJ, Jänne PA, Costa DB, Varella-Garcia M, Kim WH, Lynch TJ, Fidias P, Stubbs H, Engelman JA, Sequist LV, Tan W, Gandhi L, Mino-Kenudson M, Wei GC, Shreeve SM, Ratain MJ, Settleman J, Christensen JG, Haber DA, Wilner K, Salgia R, Shapiro GI, Clark JW, Iafrate AJ. Anaplastic Lymphoma Kinase Inhibition in Non–Small-Cell Lung Cancer. N Engl J Med. 2010;363:1693–1703. doi: 10.1056/NEJMoa1006448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–2139. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
  • 6.Khozin S, Blumenthal GM, Jiang X, He K, Boyd K, Murgo A, Justice R, Keegan P, Pazdur R. U.S. Food and Drug Administration approval summary: Erlotinib for the first-line treatment of metastatic non-small cell lung cancer with epidermal growth factor receptor exon 19 deletions or exon 21 (L858R) substitution mutations. Oncologist. 2014;19:774–779. doi: 10.1634/theoncologist.2014-0089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Solca F, Dahl G, Zoephel A, Bader G, Sanderson M, Klein C, Kraemer O, Himmelsbach F, Haaksma E, Adolf GR. Target binding properties and cellular activity of afatinib (BIBW 2992), an irreversible ErbB family blocker. J Pharmacol Exp Ther. 2012;343:342–350. doi: 10.1124/jpet.112.197756. [DOI] [PubMed] [Google Scholar]
  • 8.Shaw AT, Kim DW, Mehra R, Tan DS, Felip E, Chow LQ, Camidge DR, Vansteenkiste J, Sharma S, De Pas T, Riely GJ, Solomon BJ, Wolf J, Thomas M, Schuler M, Liu G, Santoro A, Lau YY, Goldwasser M, Boral AL, Engelman JA. Ceritinib in ALK-rearranged non-small-cell lung cancer. N Engl J Med. 2014;370:1189–1197. doi: 10.1056/NEJMoa1311107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Larkins E, Blumenthal GM, Chen H, He K, Agarwal R, Gieser G, Stephens O, Zahalka E, Ringgold K, Helms W, Shord S, Yu J, Zhao H, Davis G, McKee AE, Keegan P, Pazdur R. FDA Approval: Alectinib for the Treatment of Metastatic, ALK-Positive Non-Small Cell Lung Cancer Following Crizotinib. Clin Cancer Res. 2016;22:5171–5176. doi: 10.1158/1078-0432.CCR-16-1293. [DOI] [PubMed] [Google Scholar]
  • 10.Gettinger SN, Bazhenova LA, Langer CJ, Salgia R, Gold KA, Rosell R, Shaw AT, Weiss GJ, Tugnait M, Narasimhan NI, Dorer DJ, Kerstein D, Rivera VM, Clackson T, Haluska FG, Camidge DR. Activity and safety of brigatinib in ALK-rearranged non-small-cell lung cancer and other malignancies: a single-arm, open-label, phase 1/2 trial. Lancet Oncol. 2016;17:1683–1696. doi: 10.1016/S1470-2045(16)30392-8. [DOI] [PubMed] [Google Scholar]
  • 11.Jänne PA, Yang JC, Kim DW, Planchard D, Ohe Y, Ramalingam SS, Ahn MJ, Kim SW, Su WC, Horn L, Haggstrom D, Felip E, Kim JH, Frewer P, Cantarini M, Brown KH, Dickinson PA, Ghiorghiu S, Ranson M. AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer. N Engl J Med. 2015;372:1689–1699. doi: 10.1056/NEJMoa1411817. [DOI] [PubMed] [Google Scholar]
  • 12.Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, Orme JP, Finlay MR, Ward RA, Mellor MJ, Hughes G, Rahi A, Jacobs VN, Red Brewer M, Ichihara E, Sun J, Jin H, Ballard P, Al-Kadhimi K, Rowlinson R, Klinowska T, Richmond GH, Cantarini M, Kim DW, Ranson MR, Pao W. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014;4:1046–1061. doi: 10.1158/2159-8290.CD-14-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Finlay MR, Anderton M, Ashton S, Ballard P, Bethel PA, Box MR, Bradbury RH, Brown SJ, Butterworth S, Campbell A, Chorley C, Colclough N, Cross DA, Currie GS, Grist M, Hassall L, Hill GB, James D, James M, Kemmitt P, Klinowska T, Lamont G, Lamont SG, Martin N, McFarland HL, Mellor MJ, Orme JP, Perkins D, Perkins P, Richmond G, Smith P, Ward RA, Waring MJ, Whittaker D, Wells S, Wrigley GL. 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;57:8249–8267. doi: 10.1021/jm500973a. [DOI] [PubMed] [Google Scholar]
  • 14.Sasaki T, Koivunen J, Ogino A, Yanagita M, Nikiforow S, Zheng W, Lathan C, Marcoux JP, Du J, Okuda K, Capelletti M, Shimamura T, Ercan D, Stumpfova M, Xiao Y, Weremowicz S, Butaney M, Heon S, Wilner K, Christensen JG, Eck MJ, Wong KK, Lindeman N, Gray NS, Rodig SJ, Janne PA. A novel ALK secondary mutation and EGFR signaling cause resistance to ALK kinase inhibitors. Cancer Res. 2011;71:6051–6060. doi: 10.1158/0008-5472.CAN-11-1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tani T, Yasuda H, Hamamoto J, Kuroda A, Arai D, Ishioka K, Ohgino K, Miyawaki M, Kawada I, Naoki K, Hayashi Y, Betsuyaku T, Soejima K. Activation of EGFR Bypass Signaling by TGFalpha Overexpression Induces Acquired Resistance to Alectinib in ALK-Translocated Lung Cancer Cells. Mol Cancer Ther. 2016;15:162–171. doi: 10.1158/1535-7163.MCT-15-0084. [DOI] [PubMed] [Google Scholar]
  • 16.Miyawaki M, Yasuda H, Tani T, Hamamoto J, Arai D, Ishioka K, Ohgino K, Nukaga S, Hirano T, Kawada I, Naoki K, Hayashi Y, Betsuyaku T, Soejima K. Overcoming EGFR bypass signal-induced acquired resistance to ALK tyrosine kinase inhibitors in ALK-translocated lung cancer. Mol Cancer Res. 2017;15:106–114. doi: 10.1158/1541-7786.MCR-16-0211. [DOI] [PubMed] [Google Scholar]
  • 17.Yang JJ, Zhang XC, Su J, Xu CR, Zhou Q, Tian HX, Xie Z, Chen HJ, Huang YS, Jiang BY, Wang Z, Wang BC, Yang XN, Zhong WZ, Nie Q, Liao RQ, Mok TS, Wu YL. Lung cancers with concomitant EGFR mutations and ALK rearrangements: diverse responses to EGFR-TKI and crizotinib in relation to diverse receptors phosphorylation. Clin Cancer Res. 2014;20:1383–1392. doi: 10.1158/1078-0432.CCR-13-0699. [DOI] [PubMed] [Google Scholar]
  • 18.Tanaka H, Hayashi A, Morimoto T, Taima K, Tanaka Y, Shimada M, Kurosea A, Takanashi S, Okumura K. A case of lung adenocarcinoma harboring EGFR mutation and EML4-ALK fusion gene. BMC Cancer. 2012;12:558–561. doi: 10.1186/1471-2407-12-558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Baik CS, Chamberlain MC, Chow LQ. Targeted Therapy for Brain Metastases in EGFR-Mutated and ALK-Rearranged Non-Small-Cell Lung Cancer. J Thorac Oncol. 2015;10:1268–1278. doi: 10.1097/JTO.0000000000000615. [DOI] [PubMed] [Google Scholar]
  • 20.Cai W, Lin D, Wu C, Li X, Zhao C, Zheng L, Chuai S, Fei K, Zhou C, Hirsch FR. Intratumoral Heterogeneity of ALK-Rearranged and ALK/EGFR Coaltered Lung Adenocarcinoma. J Clin Oncol. 2015;33:3701–3709. doi: 10.1200/JCO.2014.58.8293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Marsilje TH, Pei W, Chen B, Lu W, Uno T, Jin Y, Jiang T, Kim S, Li N, Warmuth M, Sarkisova Y, Sun F, Steffy A, Pferdekamper AC, Li AG, Joseph SB, Kim Y, Liu B, Tuntland T, Cui X, Gray NS, Steensma R, Wan Y, Jiang J, Chopiuk G, Li J, Gordon WP, Richmond W, Johnson K, Chang J, Groessl T, He YQ, Phimister A, Aycinena A, Lee CC, Bursulaya B, Karanewsky DS, Seidel HM, Harris JL, Michellys PY. Synthesis, structure-activity relationships, and in vivo efficacy of the novel potent and selective anaplastic lymphoma kinase (ALK) inhibitor 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulf onyl)phenyl)pyrimidine-2,4-diamine (LDK378) currently in phase 1 and phase 2 clinical trials. J Med Chem. 2013;56:5675–5690. doi: 10.1021/jm400402q. [DOI] [PubMed] [Google Scholar]
  • 22.Galkin AV, Melnick JS, Kim S, Hood TL, Li N, Li L, Xia G, Steensma R, Chopiuk G, Jiang J, Wan Y, Ding P, Liu Y, Sun F, Schultz PG, Gray NS, Warmuth M. Identification of NVP-TAE684, a potent, selective, and efficacious inhibitor of NPM-ALK. Proc Natl Acad Sci U S A. 2007;104:270–275. doi: 10.1073/pnas.0609412103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Huang WS, Liu S, Zou D, Thomas M, Wang Y, Zhou T, Romero J, Kohlmann A, Li F, Qi J, Cai L, Dwight TA, Xu Y, Xu R, Dodd R, Toms A, Parillon L, Lu X, Anjum R, Zhang S, Wang F, Keats J, Wardwell SD, Ning Y, Xu Q, Moran LE, Mohemmad QK, Jang HG, Clackson T, Narasimhan NI, Rivera VM, Zhu X, Dalgarno D, Shakespeare WC. Discovery of Brigatinib (AP26113), a Phosphine Oxide-Containing, Potent, Orally Active Inhibitor of Anaplastic Lymphoma Kinase. J Med Chem. 2016;59:4948–4964. doi: 10.1021/acs.jmedchem.6b00306. [DOI] [PubMed] [Google Scholar]
  • 24.Zhou W, Ercan D, Chen L, Yun CH, Li D, Capelletti M, Cortot AB, Chirieac L, Iacob RE, Padera R, Engen JR, Wong KK, Eck MJ, Gray NS, Janne PA. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M. Nature. 2009;462:1070–1074. doi: 10.1038/nature08622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sequist LV, Soria JC, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, Papadimitrakopoulou V, Solomon BJ, Oxnard GR, Dziadziuszko R, Aisner DL, Doebele RC, Galasso C, Garon EB, Heist RS, Logan J, Neal JW, Mendenhall MA, Nichols S, Piotrowska Z, Wozniak AJ, Raponi M, Karlovich CA, Jaw-Tsai S, Isaacson J, Despain D, Matheny SL, Rolfe L, Allen AR, Camidge DR. Rociletinib in EGFR-mutated non-small-cell lung cancer. N Engl J Med. 2015;372:1700–1709. doi: 10.1056/NEJMoa1413654. [DOI] [PubMed] [Google Scholar]
  • 26.Walter AO, Sjin RT, Haringsma HJ, Ohashi K, Sun J, Lee K, Dubrovskiy A, Labenski M, Zhu Z, Wang Z, Sheets M, St Martin T, Karp R, van Kalken D, Chaturvedi P, Niu D, Nacht M, Petter RC, Westlin W, Lin K, Jaw-Tsai S, Raponi M, Van Dyke T, Etter J, Weaver Z, Pao W, Singh J, Simmons AD, Harding TC, Allen A. Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC. Cancer Discov. 2013;3:1404–1415. doi: 10.1158/2159-8290.CD-13-0314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Warmuth M, Kim S, Gu XJ, Xia G, Adrian F. Ba/F3 cells and their use in kinase drug discovery. Curr Opin Oncol. 2007;19:55–60. doi: 10.1097/CCO.0b013e328011a25f. [DOI] [PubMed] [Google Scholar]
  • 28.Shao H, Shi S, Huang S, Hole AJ, Abbas AY, Baumli S, Liu X, Lam F, Foley DW, Fischer PM, Noble M, Endicott JA, Pepper C, Wang S. Substituted 4-(thiazol-5-yl)-2-(phenylamino)pyrimidines are highly active CDK9 inhibitors: synthesis, X-ray crystal structures, structure-activity relationship, and anticancer activities. J Med Chem. 2013;56:640–659. doi: 10.1021/jm301475f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lange A, Gunther M, Buttner FM, Zimmermann MO, Heidrich J, Hennig S, Zahn S, Schall C, Sievers-Engler A, Ansideri F, Koch P, Laemmerhofer M, Stehle T, Laufer SA, Boeckler FM. Targeting the Gatekeeper MET146 of C-Jun N-Terminal Kinase 3 Induces a Bivalent Halogen/Chalcogen Bond. J Am Chem Soc. 2015;137:14640–14652. doi: 10.1021/jacs.5b07090. [DOI] [PubMed] [Google Scholar]
  • 30.Zhou W, Ercan D, Janne PA, Gray NS. Discovery of selective irreversible inhibitors for EGFR-T790M. Bioorg Med Chem Lett. 2011;21:638–643. doi: 10.1016/j.bmcl.2010.12.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT, Faraoni R, Floyd M, Hunt JP, Lockhart DJ, Milanov ZV, Morrison MJ, Pallares G, Patel HK, Pritchard S, Wodicka LM, Zarrinkar PP. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol. 2008;26:127–132. doi: 10.1038/nbt1358. [DOI] [PubMed] [Google Scholar]
  • 32.Goldstein DM, Gray NS, Zarrinkar PP. High-throughput kinase profiling as a platform for drug discovery. Nat Rev Drug Discov. 2008;7:391–397. doi: 10.1038/nrd2541. [DOI] [PubMed] [Google Scholar]
  • 33.Koivunen JP, Mermel C, Zejnullahu K, Murphy C, Lifshits E, Holmes AJ, Choi HG, Kim J, Chiang D, Thomas R, Lee J, Richards WG, Sugarbaker DJ, Ducko C, Lindeman N, Marcoux JP, Engelman JA, Gray NS, Lee C, Meyerson M, Janne PA. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14:4275–4283. doi: 10.1158/1078-0432.CCR-08-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hatcher JM, Bahcall M, Choi HG, Gao Y, Sim T, George R, Jänne PA, Gray NS. Discovery of Inhibitors That Overcome the G1202R Anaplastic Lymphoma Kinase Resistance Mutation. J Med Chem. 2015;58:9296–9308. doi: 10.1021/acs.jmedchem.5b01136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jiang J, Greulich H, Janne PA, Sellers WR, Meyerson M, Griffin JD. Epidermal growth factor-independent transformation of Ba/F3 cells with cancer-derived epidermal growth factor receptor mutants induces gefitinib-sensitive cell cycle progression. Cancer Res. 2005;65:8968–8974. doi: 10.1158/0008-5472.CAN-05-1829. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplement

RESOURCES