Вы находитесь на странице: 1из 11

Bioorganic Chemistry 81 (2018) 689–699

Contents lists available at ScienceDirect

Bioorganic Chemistry
journal homepage: www.elsevier.com/locate/bioorg

Design, synthesis and biological evaluation N2-(2-alkyoxy-6-aliphatic T


aminopyridin-3-yl)-2,4-diaminepyrimidine derivatives bearing acylamino or
DBTD ‘head’ as potential ALK inhibitors
Lingyun Xing, Tongfei Jing, Junlong Zhang, Ming Guo, Xiuqi Miao, Feng Jiang, Xin Zhai

Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China

ARTICLE INFO ABSTRACT

Keywords: Aiming to develop promising ALK inhibitors, two series of N2-(2-alkyoxy-6-aliphatic aminopyridin-3-yl)-2,4-
ALK inhibitor diaminepyrimidine derivatives (22a-x and 23a-d) were designed according to scaffold hopping and bioisos-
2,4-diarylaminopyrimidine terism principles. All compounds were efficiently synthesized by concise reactions and anti-proliferative activ-
Pyridinyl group ities on ALK-addicted H2228, Karpas299 cells and EGFR-expressive A549 cell were evaluated by MTT assay.
DBTD
Several compounds exhibited potential cytotoxic activities with IC50 values below 0.10 μM. Five compounds
(22g, 22h, 22l, 22s and 23a) were selected for further enzymatic determination, resulting in the discovery of 22l
against ALK and ALKL1196M with IC50 values of 2.1 nM and 3.8 nM. Particularly, western blot and cell apoptosis
assays identified 22l as a promising ALK inhibitor, which was capable of obviously inhibiting cellular ALK
activity and inducing cell apoptosis. Eventually, molecular docking modes of 22l with ALK confirmed structural
basis in accordance with the SARs analysis.

1. Introduction occupied the kinase domain and formed interactions with amino acid
residues [14]. Thus, this structural domain should be remained for
Anaplastic lymphoma kinase (ALK) was a single stranded trans- potent activity and selectivity [15]. While hydrophilic piperidinyl
membrane protein belonging to the insulin receptor tyrosine kinase group (‘tail’) extended to solvent and was responsible for a salt-bridge
family [1]. The oncogenic nucleophosmin (NPM)-ALK fusion gene was interaction with Glu1210, tolerating to further modification with var-
discovered as an ALK translocation product occurring in 80% of ana- ious aliphatic amines [16,17]. The isopropoxy group on ceritinib ske-
plastic large-cell lymphoma [2]. Echinoderm microtubule-associated leton exhibited higher kinase selectivity on account of combination
protein-like-4 (EML4)-ALK was the first targetable fusion kinase to be with protein pockets. To our disappointed, it had been reported that
identified in 4–6% of lung adenocarcinomas [3,4]. ALK fusion genes ceritinib was ineffective for ALKG1202 mutation duing to large steric
had generated substantial interest for the development of potent and hindrance of isopropoxy moiety [18]. Therefore, isopropoxy group was
specific ALK inhibitors for treatment of EML4-ALK rearrangement [5]. replaced with fragments of methoxyl, isopropylthio and hydrogen atom
Recently, a few ALK inhibitors have been successfully approved or in aiming to investigate the effect of steric hindrance. Additionally, the
clinical study (Fig. 1). The first-generation ALK inhibitor crizotinib was replacement of benzene group with pyridinyl group in aromatic ring
approved in 2011 which had demonstrated clear clinical benefits to system has shown many important effects on molecular which could
treat ALK-positive NSCLC patients [6]. However, a majority of patients properly tune the lipophilicity and improve the metabolism [19–21].
developed resistance to crizotinib treatment successively [7,8]. There- Hence, pyridinyl group as a high-impact moiety has been applied in
fore, several more potent second- and third-generation inhibitors have drug design [22,23].
been identified to combat disease resistance, including ceritinib [9] and Optimization towards different R1 substituted (‘head’) was an ef-
alectinib [10], brigatinib [11], the pre-registered lorlatinib [12] and the fective approach to enrich structural diversity. Beyond intramolecular
phase III investigational agent ensartinib [13]. hydrogen bond on isopropyl sulfonyl group, acylamino group was re-
The co-crystal structure of ceritinib with ALK revealed interactions garded as a favorable design strategy which could form hydrogen bond
between ligand and molecular (Fig. 2). The 4-arylaminopyrimidine and with receptors either [24]. Meanwhile, the acylamino ‘head’ could
Cl atom on pyrimidine ring were indispensable for activity which enhance the affinity between molecules and enzymes to increase


Corresponding author.
E-mail address: zhaixin_syphu@126.com (X. Zhai).

https://doi.org/10.1016/j.bioorg.2018.09.019
Received 22 June 2018; Received in revised form 29 August 2018; Accepted 11 September 2018
Available online 12 September 2018
0045-2068/ © 2018 Elsevier Inc. All rights reserved.
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

NH O
Cl N Cl N
N N
O NH H
F N N
N N N
Cl H H
H2N N S O O
O N
O

crizotinib (PF-02341066) ceritinib (LDK378) alectinib (CH5424802)


launched 2011 launched 2014 launched 2014

N O
F Cl
N N O
Cl N N F O N
N
O N Cl O NH
N
N N N H
H H N
P O O H2N N H2 N N
N

brigatinib (AP26113) lorlatinib (PF-06463922) ensartinib (X396)


launched 2017 pre-registered phaseIII

Fig. 1. Structures of ALK inhibitors.

Fig. 2. Design strategies for target compounds (22a-x and 23a-d) based on co-crystal structure of ceritinib with ALK (PDB ID code: 4MKC). (A) The co-crystal
structure of ceritinib with ALK. (B) Structure design strategies of 22a-x and 23a-d.

inhibitory activity. In addition, the rigid 3,4-dihydro-2H-benzo[1,4-b] 96.5% yield. 3 was acylated by different acyl chloride in the presence of
thiazine-1,1-dioxide (DBTD) fragment was built as a new structural triethylamine to afford the key intermediates 4a-c [26]. Meanwhile,
motif by scaffold hopping [25], which could enhance the rigidity. Thus, substitution reaction of compound 2-fluoronitrobenzene (5) with mer-
compounds 23a-d with DBTD ‘head’ were designed successfully to captoethanol gave 6, which was chlorinated by thionyl chloride to
discuss the binding ability to protein and evaluate its effectivity. generate 2-chloroethyl-2-nitrophenyl thioether (7) as a yellow solid. 7
Herein, two series of N2-(2-alkyoxy-6-aliphatic aminopyridin-3-yl)- was oxidized by hydrogen peroxide to yeild 8, which was subjected to
2,4-diaminepyrimidine derivatives (22a-x and 23a-d) bearing acyla- reduction by iron powder in the presence of hydrochloric acid to gain 9
mino or DBTD ‘head’ were designed and synthesized. Compounds were in good yield. Intramolecular N-alkylation of 9 in dimethyl formamide
assayed for the anti-proliferative activity in vitro against three cancer gave desired DBTD analog 10, which was concentrated with 2,4,5-tri-
cell lines H2228, Karpas299 and A549. Subsequently, five compounds chloropyrimidine to yield the key intermediate 11
were selected to test in vitro enzymatic evaluation on ALK and Compound 12 reacted with methanol (isopropanol or isopropyl
ALKL1196M. To further study, the promising compound 22l was eval- mercaptan) in the company of sodium hydride to obtain 13b-d without
uated for western blot assay and cell apoptosis. Finally, molecular further purification. Subsequently, amination of 13a-d by corre-
docking studies were carried out to identify SARs. sponding aliphatic amines with potassium carbonate furnished target
intermediates 14a-c, 14e-g, 15a-g, 16a-c or 17a-c in satisfied yields.
2. Results and discussion Treatment 14a-c, 14e-g, 15a-15g, 16a-16c or 17a-17c with hydro-
chloric acid as catalysis under activated iron powder in ethanol/water
2.1. Chemistry (9:1) gave rise to 18a-c, 18e-g, 19a-g, 20a-c or 21a-c, which were
reacted with 4a-c and purified on silica gel column to get target pro-
The synthetic routes to compounds 22a-x and 23a-d were outlined ducts 22a-x. Similarly, the desired compounds 23a-d were accom-
in Scheme 1. The commercially available 2,4,5-trichloropyrimidine (1) plished using the same method as preparation of 22a-x.
was reacted with benzene-1,2-diamine (2) to provide intermediate 3 in

690
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

Scheme 1. Reagents and conditions: (i) DIPEA, ethanol, 80 °C; (ii) the acyl chloride of R1, triethylamine, DCM, rt; (iii) 2-mercaptoethanol, K2CO3, DMF, 110 °C; (iv)
thionyl chloride, toluene, 80 °C; (v) hydrogen peroxide, glacial acetic acid, 100 °C; (vi) iron powder, hydrochloric acid, ethanol/water(9:1), 80 °C; (vii) K2CO3, DMF,
100 °C; (viii) NaH (60%), DMF, 0 ∼ 5 °C; (ix) NaH (60%), methanol(/IPA/isopropyl mercaptan), THF, rt; (x) aliphatic amines, K2CO3, THF, 50 °C; (xi) iron powder,
hydrochloric acid, ethanol/water(9:1), 80 °C; (xii) 4a-d, p-toluenesulfonic acid, IPA, 80 °C.

2.2. Biological evaluation fragment (22f). Meanwhile, significant potency loss was detected on
compounds 22a-c with diethylamino, piperidyl and pyrrolidinyl groups
2.2.1. In vitro anti-proliferative activity and SARs study due to the lack of hydrophilicity.
MTT assay was employed to evaluate the anti-proliferative char- As a general trend, among diverse chain R1-moiety, compounds
acteristics of target compounds to screen the active compounds pre- bearing sulfonyl (22l) and acetyl (22g) showed better activities against
liminarily. Compounds 22a-x and 23a-d were investigated on cyto- H2228 with IC50 values less than 0.1 μM than compound bearing butyl-
toxicity against ALK-addicted H2228 (Human NSCLC cells), Karpas299 2-acrylamide substitution (22h). Generally, introduction of butyl-2-
(Human transsexual cell lymphoma cell lines) cells and EGFR-ex- acrylamide moiety would result in unsatisfactory activities as indicated
pressive A549 (Human EGFR-positive NSCL cell lines) cell. Using cer- in 22h-k, which illustrated the effect of steric on cellular potency.
itinib as the positive control, the results expressed as IC50 values were Notably, it was found that compound 22l bearing sulfonyl ‘head’ in-
shown in Tables 1 and 2. creased ALK potency significantly.
The pharmacological data indicated that most compounds exhibited The IC50 values of 22l-x showed that variations of R2 group had a
moderate to excellent cytotoxic activities against ALK-addicted H2228 marked impact on activity. The R2 moiety was introduced with lipo-
and Karpas299 cells. Compound 22l displayed significant activity philic substituents which occupied the ALK protein lipophilic pocket to
against H2228 and Karpas299 with IC50 values of 0.030 μM and improve the activity. Derivatives 22m-o with isopropyl provided
0.036 μM. Meanwhile, 22l showed poor inhibitory activity against moderate potency, however, shifting isopropyl to bulk isopropylthio led
A549 with IC50 values of 0.65 μM. Compared with 22a-x, compounds to poor activity deducing to the steric hindrance within the hydro-
23a-d exhibited obviously reduced activities against H2228 and phobic pocket. For example, the inhibitory activities of 22q and 22r
Karpas299 cells, indicating the negative effect of DBTD group on cy- were decreased extremely with IC50 values more than 1 μM against
totoxicity. As expected, all compounds showed weak potency against tested cell lines. Replacement of isopropyl with hydrogen atom resulted
A549 cells, suggesting the selectivity on ALK cell lines. in compounds 22s-x, which decreased activity, indicating R2-hydro-
As shown in Table 1, anti-proliferative activity of target compounds phobic pocket should be inserted the motif with appropriate volume.
would preliminarily define the structure–activity relationships. A small Thus, methoxyl on R2 moiety (22l) was regarded as the optimal sub-
set of compounds 22a-g with different R3-moiety ‘tail’ were evaluated stituent.
for cytotoxic activities, wherein compound 22g bearing morpholinyl To integrate above factors and process further explore, the effect of
moiety showed the best potency with IC50 values of 0.09 μM, 0.068 μM rigid sulfonyl ‘head’ group was studied by scaffold hopping principle.
and 0.95 μM against H2228, Karpas299 and A549, which identified As illustrated in Table 2, compounds with rigid DBTD ‘head’ were in-
polarity ‘tails’ with good hydrophilia property being beneficial for in- ferior to corresponding compounds bearing acylamino ‘head’. Ex-
creased potency. Compounds containing azetidin-3-ol (22d) and hy- tremely, 23d demonstrated dramatic poor effect on H2228 and
droxyethyl piperazinyl (22e) were 2.4- and 6.3-fold more potent Karpas299 cells with IC50 values more than 5 μM. Further analysis on
against H2228 than those inserting methyl group in piperazinyl SARs would be detailed description on molecular docking studies.

691
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

Table 1 Table 2
Cytotoxicity of 22a-x against H2228, Karpas299 and A549 cell lines in vitro Cytotoxicity of 23a-d against H2228, Karpas299 and A549 cell lines in vitro

. .

Compd. R1 R2 R3 IC50 (µM) Compd. R2 R3 IC50 (µM)

H2228 Karpas299 A549 H2228 Karpas299 A549

22a 0.92 0.16 2.16 23a 2.05 0.82 1.62

22b 0.51 0.094 1.06 23b 2.33 1.28 2.84

23c 3.41 2.68 0.44


22c 0.80 0.19 1.23
23d >5 >5 2.26
22d 0.62 0.06 0.54
ceritinib 0.032 0.048 0.79

22e 0.24 0.052 0.18

22f 1.51 1.28 3.11 Table 3


Enzymatic inhibition of five compounds against ALK and ALKL1196M in vitro.
22g 0.09 0.068 0.95 Compd. IC50 (nM)

22h 0.19 0.09 1.12 ALK ALKL1196M

22i 0.98 0.23 1.88 22g 3.6 5.4


22h 5.8 12.8
22j 0.78 0.12 0.97
22l 2.1 3.8
22k 0.37 0.08 0.66 22s 8.3 20.3
23a > 3000 > 3000
22l 0.030 0.036 0.65 ceritinib 2.3 8.9

22m 0.054 0.04 0.32


compound 22l turned out to be superior to ceritinib (IC50 = 2.3 nM)
22n 0.65 0.87 0.62 with IC50 value of 2.1 nM. Likewise, compounds 22g and 22l showed
significant inhibitory activity to ceritinib (IC50 = 8.9 nM) against
22o 0.74 1.66 2.51
ALKL1196M with IC50 values 5.4 nM and 3.8 nM, respectively. Paralleling
22p 0.66 0.84 3.23 with cellular results, 22l was found to be 1.4- and 2.3-fold more potent
than 22h with butyl-2-acrylamide ‘head’ and 22s with hydrogen atom
22q 1.12 1.19 5.22 substitution on pyridinyl group against ALKL1196M, which revealed that
appropriate steric hindrance was favor to improve the activities.
22r 1.47 1.08 4.69

22s H 0.20 0.038 0.287 2.2.3. In western blot assay


Western blot assay was an important method to elucidate the
22t H 0.68 0.37 1.21 modulation of target phosphorylation effect of 22l in the in vitro assay.
Using ceritinib as the positive control, compound 22l and ceritinib were
22u H 0.94 0.61 2.81
examined to determine the abilities to inhibit Karpas299 cell lysates
22v H 0.92 0.11 0.58 (Fig. 3).
As demonstrated in Fig. 3, 22l suppressed ALK expression and re-
22w H 0.67 0.096 0.66 duced phosphorylation of ALK in a dose-dependent suppression manner
from 25 nM to 100 nM, which was comparable to ceritinib. Slight in-
22x H 1.26 1.79 4.23
hibition of ALK expression and phosphorylation could be observed at a
dose of 50 nM, meanwhile, obvious inhibitions were discovered a
ceritinib 0.032 0.048 0.79
concentration of 100 nM. These results addressed 22l did potently in-
hibit protein activity.

2.2.4. Apoptosis analysis


2.2.2. In vitro enzymatic assays Apoptosis assay elucidated the process for the observed induction of
Based on the cellular assays, further enzymatic assays in vitro were cellular death, so it played a necessary role to assess the potency of
of particular importance to evaluate antitumor potential. Five re- compound 22l. The cytotoxicity of 22l was further confirmed by an
presentative compounds (22g, 22h, 22l, 22s and 23a) were selected acridine orange (AO)/ethidium bromide (EB) fluorescent staining assay
against ALK and ALKL1196M inhibitory studies. The results were sum- against H2228 and Karpas299 cell lines (Fig. 4). This staining solution
marized in Table 3, which were comparable to the positive control was a mixture of AO and EB, in which AO was used for live cell iden-
ceritinib. tification and EB for identification of necrotic cells. The AO permeated
As shown in Table 3, compared with 22l, compound 23a was ob- into the intact cell membrane and thus stains live cells to appear green,
served a loss in activity against ALK and ALKL1196M, which indicated whereas, EB stained necrotic cells to appear orange when visualized by
that DBTD group was unfavorable to inhibitory activity. However, op- fluorescent microscopy due to disruption of membrane integrity.
timal compounds with chain R1-moiety displayed good inhibitory po- Therefore, early apoptotic cells contained bright green condensed
tency against ALK with IC50 values ranging from 2.1 to 8.3 nM. Notably, bodies and later apoptotic cells presented colored orange nuclei

692
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

Fig. 3. Mechanism of action of compound 22l and ceritinib in Karpas299 cell lines. Cells were treated with 22l or ceritinib at the indicated concentrations for 6 h and
the levels of protein were evaluated by western blot analysis of cell lysates using specific antibodies.

indicating. engaged in carbon hydrogen bond interactions with Asp1203 and


As shown in Fig. 4, H2228 and Karpas299 cells were observed cell Leu1122, which was similar to ceritinib. These simulation models
apoptosis in 22l treated groups. It was observed that control cells dis- proved that 22l had familiar contact to cerinitib with ALK.
played normal cell morphological features with uniformly stained green It was available from Fig. 5E and F that 23a vanished interactions
nuclei, while compound 22l at the concentration of 160 nM started to with Lys1150 and Gly1125 deducing to the rigid DBTD ‘head’, which
induce morphological changes. At the concentration of 320 nM, the explained the reason of poor activities and the important of the chain
number of green cells was reduced and orange cells were increased ‘head’. Hence, the results from the docking study were in accordance
indicating cells underwent apoptotic stage. Simultaneously, apoptotic with the SARs analysis.
cells were clearly observed at 640 nM. These results explored 22l in-
duced dose-dependent apoptosis of H2228 and Karpas299 cells as evi-
dent by observed hall marks of apoptosis. 3. Conclusions

2.2.5. Molecular docking studies In this paper, two series of N2-(2-alkyoxy-6-aliphatic aminopyridin-
Computer aided molecular docking was used to simulate interac- 3-yl)-2,4-diaminepyrimidine derivatives (22a-x and 23a-d) bearing
tions and evaluate active sites. In order to detail the binding mode, acylamino or DBTD ‘head’ were designed, synthesized and evaluated for
molecular dockings of 22l and 23a were performed based on co-crystal biological activities. Compared with compounds 22a-x bearing acyla-
structures of ALK (PDB ID code: 4MKC) by Discovery Studio 2017 mino group, compounds with rigid DBTD ‘head’ (23a-d) decreased the
(Fig. 5). potency relative. Importantly, 22l possessed remarkable antitumor
As shown in Fig. 5A, 22l occupied the same kinase domain with potency against H2228 and Karpas299 cells with IC50 values of
ceritinib. Both cerinitib and 22l were found to form two hydrogen 0.030 μM and 0.036 μM as well as excellent activity on ALK and
bonds via the amino and pyrimidine nitrogen atoms onto the backbone ALKL1196M with IC50 of 2.1 nM and 3.8 nM, respectively. Consistently,
oxygen and nitrogen of Met1199 (Fig. 5B and C), which were essential western blot and cell apoptosis tests illustrated 22l could inhibite the
for binding to ALK. The Cl atom on pyrimidine ring of 22l formed cellular ALK activity and induce cell apoptosis in a dose-dependent
hydrophobic interactions with Leu1196 and Leu1256. Regardless of the manner. Moreover, molecular docking modes detailed that 22l could
size of the aromatic ring, cerinitib and 22l formed alkyl interactions not only form critical hydrogen bonding interactions and hydrophobic
with Leu1122. Meanwhile, the acylamino group of compound 22l not interactions but also overlap well with ceritinib in 3D model, which was
only formed hydrogen bond interactions with Lys1150, but also carbon consistent with observed SARs for this class of compounds. In conclu-
hydrogen bond interactions with Gly1125 (Fig. 5D), as illustrated the sion, the target compound 22l was identified as a promising ALK in-
reasonability of the chain ‘head’. The water-soluble ‘tail’ of 22l was hibitor for cancer treatments.

Fig. 4. AO/EB stained apoptosis of H2228 and Karpas299 cell lines. H2228 and Karpas299 cell were treated with different concentrations of 22l for 24 h, and then
examined by fluorescence microscope.

693
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

Fig. 5. The binding poses of cerinitib, 22l and 23a with ALK (PDB ID code: 4MKC). (A) Composite model of cerinitib (red sticks) and 22l (gray sticks) with ALK. (B)
Docking model of cerinitib with ALK. (C) Docking model of compound 22l with ALK. (D) 2D diagram of the interaction between 22l and ALK. (E) Docking model of
23a (purple sticks) with ALK. (F) 2D diagram of the interaction between 23a and ALK.

4. Experimental section 4.1.2.2. N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)


methanesulfonamide (4b). Yield: 85.8%; MS (ESI) m/z: 333.2 [M+H]+.
4.1. Chemistry
4.1.2.3. (E)-N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)but-2-
All melting points were obtained on a Büchi Melting Point B-540 enamide (4c). Yield: 91.1%; MS (ESI) m/z: 322.9 [M+H]+.
apparatus (Büchi Labortechnik, Flawil, Switzerland) and were un-
corrected. Mass spectra (MS) were taken in ESI mode on Agilent 1100 4.1.3. Preparation of 2-((2-nitrophenyl)thio)ethan-1-ol (6)
LC-MS (Agilent, palo Alto, CA, USA). 1H NMR and 13C NMR spectra To the solution of mercaptoethanol (16.6 g, 0.21 mol) in dry DMF (1
were performed using Bruker spectrometers (Bruker Bioscience, re- l) was added 2-fluoronitrobenzene 5 (30.0 g, 0.21 mol) and K2CO3
spectively, Billerica, MA, USA) with TMS as an internal standard. (58.7 g, 0.42 mol). The reaction mixture was heated with stirring at
Column chromatography was run on silica gel (200–300 mesh) from 110 °C for 6 h. After reacting at room temperature, the reaction mixture
Qingdao Ocean Chemicals (Qingdao, Shandong, China). Unless other- was poured into water and extracted with AcOEt (3 × 100 mL). The
wise noted, all materials were obtained from commercially available combined organic layer was washed with brine, dried over MgSO4,
sources and were used without further purification. filtered, and concentrated under reduced pressure to yield a yellow
solid 6 in 94.2% yield. MS (ESI) m/z(%): 200.1 [M+H]+.
4.1.1. Preparation of N1-(2,5-dichloropyrimidin-4-yl)benzene-1,2-diamine
(3) 4.1.4. Preparation of 2-chloroethyl-2-nitrophenyl thioether (7)
2,4,5-trichloropyrimidine 1 (30.0 g, 0.16 mol), benzene-1,2-diamine Thionyl chloride (17.85 g, 0.15 mol) was added dropwise to a so-
2 (17.7 g, 0.16 mol) and N,N-diisopropylethylamine (54.0 mL, lution of 6 (10.0 g, 0.05 mol) in toluene (100 mL) at 50 °C for 1 h, then
0.33 mol) was added to ethanol (300 mL). The resulting solution was refluxed for 10 h. After cooling, the reaction mixture was concentrated
stirred at 80 °C for 2 h. The reaction mixture was evaporated and the under reduced pressure. The residue was stirred with water. The solid
residue was stirred with aqueous HCl (0.1 M, 10 mL) for 30 min. The was collected by filtration and dried under vacuum to gain 7 as a yellow
solid was collected by filtration and dried under vacuum. The crude solid in 93.4% yield. MS (ESI) m/z(%): 218.1 [M+H]+.
solid was triturated with dichloromethane to give a solid which was
collected by filtration and dried under vacuum to afford a white solid 3 4.1.5. Preparation of 1-((2-chloroethyl)sulfonyl)-2-nitrobenzene (8)
in 96.5% yield. MS (ESI) m/z: [M+H]+ 255.0. To the solution of 7 (10.9 g, 0.05 mol) in CH3COOH (100 mL), 30%
H2O2 (30.5 mL, 0.16 mol) was added dropwise while maintaining the
4.1.2. General procedure for preparation of compounds (4a-c) temperature below 50 °C. After the addition was completed, the mixture
Acetylchloride, methylsulfonyl chloride or (E)-but-2-enoyl chloride was heated to 100 °C for another 13 h. After cooling, the reaction
(0.094 mol) was added to a mixture of 3 (20.0 g, 0.079 mol) and trie- mixture was poured into water to give a light yellow precipitate. This
thylamine (33.0 mL, 0.24 mol) in DCM (100 mL) at 0 °C for 30 min, then was collected by filtration and air-dried to give 8 as a light yellow solid
stirred at room temperature for 2 h. The reaction mixture was evapo- in 89.0% yield. MS (ESI) m/z(%): 250.0 [M+H]+.
rated and the residue was stirred with water (200.0 mL). The solid was
collected by filtration and dried under vacuum to give 4a-c. 4.1.6. Preparation of 2-((2-chloroethyl)sulfonyl)aniline (9)
To the solution of 8 (12.9 g, 0.05 mol) in ethanol/water (9:1, 60 mL)
4.1.2.1. N-(2-((2,5-dichloropyrimidin-4-yl)amino)phenyl)acetamide was added hydrochloric acid (7.0 mL) and iron powder (11.6 g,
(4a). Yield: 89.2%; MS (ESI) m/z: 297.0 [M+H]+. 0.2 mol). The resulting mixture was heated at 80 °C for 3 h, filtered

694
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

through celite and concentrated to remove ethanol. The aqueous solu- 4.1.10.4. 1-(5-nitropyridin-2-yl)azetidin-3-ol (14e). Yield: 89.0%; MS
tion was extracted with ethyl acetate (3 × 100 mL) and the combined (ESI) m/z: 196.1 [M+H]+.
organic layers were washed with water, brine, dried over Na2SO4 and
concentrated to gain 9 in 67.3% yield. MS (ESI) m/z(%): 220.6 [M 4.1.10.5. 2-(4-(5-nitropyridin-2-yl)piperazin-1-yl)ethan-1-ol
+H]+. (14f). Yield: 83.4%; MS (ESI) m/z: 253.2 [M+H]+.

4.1.7. Preparation of 3,4-dihydro-2H-benzo[1,4-b]thiazine-1,1-dioxide 4.1.10.6. 3-methyl-1-(5-nitropyridin-2-yl)piperazine (14g). Yield: 86.7%;


(10) MS (ESI) m/z: 222.2 [M+H]+.
To the solution of 9 (10.0 g, 0.05 mol) in DMF (100 mL), K2CO3
(18.9 g, 0.14 mol) was added. The mixture was stirred at 100 °C for 4.1.10.7. 4-(6-methoxy-5-nitropyridin-2-yl)morpholine (15a). Yield: 90.1%;
10 h. After cooling, the reaction mixture was poured into water to give MS (ESI) m/z: 240.0 [M+H]+.
a light grey precipitate. This was collected by filtration and air-dried to
give 10 as a light grey solid in 55.4% yield. MS (ESI) m/z(%): 184.0 [M 4.1.10.8. 2-methoxy-3-nitro-6-(piperidin-1-yl)pyridine (15b). Yield: 89.4%;
+H]+. MS (ESI) m/z: 238.1 [M+H]+.

4.1.8. Preparation of 4-(2,5-dichlorophenyl)-3,4-dihydro-2H-benzo[1,4-b] 4.1.10.9. N,N-diethyl-6-methoxy-5-nitropyridin-2-amine (15c). Yield:


thiazine-1,1-dioxide (11) 78.6%; MS (ESI) m/z: 226.0 [M+H]+.
To the suspension of 60% NaH (0.66 g, 0.016 mol) in DMF (20 mL)
was added 10 (1.0 g, 0.005 mol) at 0 °C. The mixture was stirred for 4.1.10.10. 2-methoxy-3-nitro-6-(pyrrolidin-1-yl)pyridine (15d). Yield:
30 min, and then 1 (1.2 g, 0.007 mol) diluted in DMF (6 mL) was added 84.2%; MS (ESI) m/z: 224.2 [M+H]+.
slowly. The mixture was warmed to room temperature and stirred
overnight. The reaction mixture was poured into water to give a yellow 4.1.10.11. 1-(6-methoxy-5-nitropyridin-2-yl)azetidin-3-ol (15e). Yield:
precipitate which was collected by filtration. The crude product pur- 80.5%; MS (ESI) m/z: 226.0 [M+H]+.
ified by column chromatography using dichloromethane/methanol as
an eluent to afford 11 a white solid in 41.8% yield. MS (ESI) m/z(%): 4.1.10.12. 2-(4-(6-methoxy-5-nitropyridin-2-yl)piperazin-1-yl)ethan-1-ol
329.8 [M+H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 9.61 (s, 1H), 8.53 (15f). Yield: 79.3%; MS (ESI) m/z: 283.3 [M+H]+.
(d, J = 8.4 Hz, 1H), 8.30 (s, 1H), 8.01 (dd, J = 8.0, 1.4 Hz, 1H),
7.77–7.69 (m, 1H), 7.33 (dd, J = 17.7, 9.8 Hz, 1H), 6.60 (dd, J = 16.5, 4.1.10.13. 1-(6-methoxy-5-nitropyridin-2-yl)-3-methylpiperazine
9.4 Hz, 1H), 6.50 (d, J = 16.5 Hz, 1H), 6.04 (d, J = 9.4 Hz, 1H). (15g). Yield: 71.6%; MS (ESI) m/z: 253.1 [M+H]+.

4.1.9. General procedure for preparation of compounds (13b-d) 4.1.10.14. 4-(6-isopropoxy-5-nitropyridin-2-yl)morpholine (16a). Yield:
To the suspension of 60% NaH (4.6 g, 0.11 mol) in THF (20 mL) was 89.5%; MS (ESI) m/z: 268.1 [M+H]+.
added 12 (20.0 g, 0.1 mol) at 0 °C. The mixture was stirred at 0 °C for
30 min, and then methanol, IPA or isopropyl mercaptan (0.1 mol) was 4.1.10.15. 2-isopropoxy-3-nitro-6-(piperidin-1-yl)pyridine (16b). Yield:
added slowly. The mixture was warmed to room temperature and 88.4%; MS (ESI) m/z: 266.0 [M+H]+.
stirred for 6 h. The reaction mixture was poured into water to give a
yellow precipitate which was collected by filtration to afford 13b-d. 4.1.10.16. N2,N2-diethyl-6-isopropoxy-5-nitropyridin-2-amine
(16c). Yield: 85.1%; MS (ESI) m/z: 254.1 [M+H]+.
4.1.9.1. 6-chloro-2-methoxy-3-nitropyridine (13b). Yield: 72.7%; MS
4.1.10.17. 4-(6-(isopropylthio)-5-nitropyridin-2-yl)morpholine
(ESI) m/z: 188.9 [M+H]+.
(17a). Yield: 82.9%; MS (ESI) m/z: 284.1 [M+H]+.
4.1.9.2. 6-chloro-2-isopropoxy-3-nitropyridine (13c). Yield: 69.1%; MS
4.1.10.18. 2-(isopropylthio)-3-nitro-6-(piperidin-1-yl)pyridine
(ESI) m/z: 217.1 [M+H]+.
(17b). Yield: 86.2%; MS (ESI) m/z: 282.1 [M+H]+.

4.1.9.3. 6-chloro-2-(isopropylthio)-3-nitropyridine (13d). Yield: 78.2%;


4.1.10.19. N2,N2-diethyl-6-(isopropylthio)-5-nitropyridin-2-amine
MS (ESI) m/z: 233.0 [M+H]+.
(17c). Yield: 86.8%; MS (ESI) m/z: 270.1 [M+H]+.

4.1.10. General procedure for preparation of compounds (14a-c, 14e-g, 4.1.11. General procedure for preparation of compounds (18a-c, 18d-g,
15a-g, 16a-c and 17a-c) 19a-g, 20a-c and 21a-c)
A solution of 13a-d (0.005 mol) in THF (20 mL) was added K2CO3 To the solution of 14a-c, 14e-g, 15a-g, 16a-c or 17a-c (0.005 mol)
(0.011 mol) and various secondary amines (0.011 mol), which was in ethanol/water (9:1, 30 mL) was added hydrochloric acid (1.0 mL)
stirred at 50 °C. After cooling to room temperature, the reaction mixture and iron powder (1.12 g, 0.02 mol). The resulting mixture was heated at
was poured into water. The aqueous solution was extracted with ethyl 80 °C for 3 h, filtered through celite and concentrated to remove
acetate (3 × 100 mL) and the combined organic layers were washed ethanol. The aqueous solution was extracted with ethyl acetate
with water, brine, dried over Na2SO4 and concentrated to gain 14a-c, (3 × 100 mL) and the combined organic layers were washed with
14e-g, 15a-g, 16a-c or 17a-c. water, brine, dried over Na2SO4 and concentrated to gain 18a-c, 18e-g,
19a-g, 20a-c or 21a-c.
4.1.10.1. 4-(5-nitropyridin-2-yl)morpholine (14a). Yield: 89.3%; MS
(ESI) m/z: 210.1 [M+H]+. 4.1.11.1. 6-morpholinopyridin-3-amine (18a). Yield: 79.3%; MS (ESI)
m/z: 180.1 [M+H]+.
4.1.10.2. 5-nitro-2-(piperidin-1-yl)pyridine (14b). Yield: 91.0%; MS
(ESI) m/z: 208.1 [M+H]+. 4.1.11.2. 6-(piperidin-1-yl)pyridin-3-amine (18b). Yield: 92.6%; MS
(ESI) m/z: 178.1 [M+H]+.
4.1.10.3. N,N-diethyl-5-nitropyridin-2-amine (14c). Yield: 87.6%; MS
(ESI) m/z: 196.3 [M+H]+. 4.1.11.3. N2,N2-diethylpyridine-2,5-diamine (18c). Yield: 87.4%; MS

695
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

(ESI) m/z: 166.1 [M+H]+. DMSO‑d6) δH: 10.06 (s, 1H), 8.23 (s, 1H), 7.99 (s, 1H), 7.78 (s, 1H),
7.63 (d, J = 8.2 Hz, 1H), 7.58 (d, J = 8.6 Hz, 1H), 7.24 (dd, J = 8.0,
4.1.11.4. 1-(5-aminopyridin-2-yl)azetidin-3-ol (18e). Yield: 87.3%; MS 2.1 Hz, 1H), 7.30–7.14 (m, 2H), 6.20 (d, J = 8.4 Hz, 1H), 3.84 (d,
(ESI) m/z: 166.1 [M+H]+. J = 2.1 Hz, 4H), 3.77 (s, 3H), 2.06 (s, 3H), 1.27 (s, 6H).

4.1.11.5. 2-(4-(5-aminopyridin-2-yl)piperazin-1-yl)ethan-1-ol (18f). Yield: 4.1.12.2. N-(2-((5-chloro-2-((2-methoxy-4-(piperidin-1-yl)phenyl)


80.7%; MS (ESI) m/z: 223.2 [M+H]+. amino)pyrimidin-4-yl)amino)phenyl)acetamide (22b). Yield: 84.5%;
m.p.: 178.3–179.6 °C; MS (ESI) m/z: 468.4 [M+H]+; 1H NMR
4.1.11.6. 6-(3-methylpiperazin-1-yl)pyridin-3-amine (18g). Yield: 82.4%; (400 MHz, DMSO‑d6) δH: 10.03 (s, 1H), 8.36 (s, 1H), 8.02 (s, 1H),
MS (ESI) m/z: 192.2 [M+H]+. 7.81 (s, 1H), 7.73 (d, J = 8.0 Hz, 1H), 7.57 (d, J = 8.4 Hz, 1H), 7.25
(dd, J = 7.2, 2.0 Hz, 1H), 7.22–7.08 (m, 2H), 6.18 (d, J = 8.5 Hz, 1H),
4.1.11.7. 2-methoxy-6-morpholinopyridin-3-amine (19a). Yield: 90.1%; 3.77 (s, 3H), 3.44 (d, J = 5.1 Hz, 4H), 2.09 (s, 3H), 1.57 (s, 6H).
MS (ESI) m/z: 210.1 [M+H]+.
4.1.12.3. N-(2-((5-chloro-2-((2-methoxy-4-(pyrrolidin-1-yl)phenyl)
4.1.11.8. 2-methoxy-6-(piperidin-1-yl)pyridin-3-amine (19b). Yield: amino)pyrimidin-4-yl)amino)phenyl)acetamide (22c). Yield: 80.4%;
89.5%; MS (ESI) m/z: 208.1 [M+H]+. m.p.: 183.8–185.6 °C; MS (ESI) m/z: 454.4 [M+H]+; 1H NMR
(400 MHz, DMSO‑d6) δH: 10.08 (s, 1H), 8.32 (s, 1H), 8.00 (s, 1H),
4.1.11.9. N2,N2-diethyl-6-methoxypyridine-2,5-diamine (19c). Yield: 7.80 (s, 1H), 7.77–7.72 (m, 1H), 7.51 (d, J = 8.4 Hz, 1H), 7.29–7.21
87.3%; MS (ESI) m/z: 196.1 [M+H]+. (m, 1H), 7.17–7.10 (m, 2H), 5.84 (d, J = 8.2 Hz, 1H), 3.79 (s, 3H), 3.36
(s, 4H), 2.09 (s, 3H), 1.94 (s, 4H).
4.1.11.10. 2-methoxy-6-(pyrrolidin-1-yl)pyridin-3-amine (19d). Yield:
86.1%; MS (ESI) m/z: 194.1 [M+H]+. 4.1.12.4. N-(2-((5-chloro-2-((4-(3-hydroxyazetidin-1-yl)-2-
methoxyphenyl)amino)pyrimidin-4-yl)amino)phenyl)acetamide
4.1.11.11. 1-(5-amino-6-methoxypyridin-2-yl)azetidin-3-ol (19e). Yield: (22d). Yield: 85.2%; m.p.: 183.8–190.6 °C; MS (ESI) m/z: 454.4
80.6%; MS (ESI) m/z: 196.0 [M+H]+. [M−H]−; 1H NMR (400 MHz, DMSO‑d6) δH: 10.01 (s, 1H), 8.31 (s,
1H), 7.99 (s, 1H), 7.79 (s, 1H), 7.73 (s, 1H), 7.41 (d, J = 8.3 Hz, 1H),
4.1.11.12. 2-(4-(5-amino-6-methoxypyridin-2-yl)piperazin-1-yl)ethan-1- 7.24 (d, J = 9.3 Hz, 1H), 7.18–7.06 (m, 2H), 6.36 (t, J = 6.0 Hz, 1H),
ol (19f). Yield: 79.4%; MS (ESI) m/z: 253.1 [M+H]+. 5.33 (s, 1H), 3.89 (s, 1H), 3.77 (s, 3H), 3.66 (dd, J = 11.1, 4.5 Hz, 1H),
3.56 (dd, J = 11.0, 5.7 Hz, 1H), 3.41 (dd, J = 13.0, 6.5 Hz, 1H), 3.24
4.1.11.13. 2-methoxy-6-(3-methylpiperazin-1-yl)pyridin-3-amine (dd, J = 13.3, 5.9 Hz, 1H), 2.09 (s, 3H).
(19g). Yield: 82.1%; MS (ESI) m/z: 223.2 [M+H]+.
4.1.12.5. N-(2-((5-chloro-2-((4-(4-(2-hydroxyethyl)piperazin-1-yl)-2-
4.1.11.14. 2-isopropoxy-6-morpholinopyridin-3-amine (20a). Yield: methoxyphenyl)amino)pyrimidin-4-yl)amino)phenyl)acetamide
86.4%; MS (ESI) m/z: 238.2 [M+H]+. (22e). Yield: 79.8%; m.p.:150.1–155.5 °C; MS (ESI) m/z: 513.4 [M
+H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 10.01 (s, 1H), 8.37 (s, 1H),
4.1.11.15. 2-isopropoxy-6-(piperidin-1-yl)pyridin-3-amine (20b). Yield: 8.01 (s, 1H), 7.83 (s, 1H), 7.73 (d, J = 6.2 Hz, 1H), 7.61 (d, J = 8.4 Hz,
84.2%; MS (ESI) m/z: 236.2 [M+H]+. 1H), 7.26 (d, J = 7.8 Hz, 1H), 7.16 (s, 2H), 6.19 (d, J = 8.3 Hz, 1H),
4.46 (s, 1H), 3.78 (s, 3H), 3.54 (t, J = 5.8 Hz, 2H), 3.40 (s, 4H), 2.53 (s,
4.1.11.16. N2,N2-diethyl-6-isopropoxypyridine-2,5-diamine (20c). Yield: 4H), 2.45 (t, J = 6.0 Hz, 2H), 2.09 (s, 3H). 13C NMR (100 MHz,
85.7%; MS (ESI) m/z: 224.0 [M+H]+. DMSO‑d6) δC: 170.07, 159.21, 156.06, 154.95, 154.42, 135.09,
132.32, 131.14, 126.42, 125.74, 125.15 (2C), 112.98, 104.01, 98.18,
4.1.11.17. 2-(isopropylthio)-6-morpholinopyridin-3-amine (21a). Yield: 60.77, 58.95, 53.34 (2C), 53.09, 45.63 (2C), 29.46, 23.46.
86.1%; MS (ESI) m/z: 254.1 [M+H]+.
4.1.12.6. N-(2-((5-chloro-2-((2-methoxy-4-(3-methylpiperazin-1-yl)
4.1.11.18. 2-(isopropylthio)-6-(piperidin-1-yl)pyridin-3-amine phenyl)amino)pyrimidin-4-yl)amino)phenyl)acetamide (22f). Yield:
(21b). Yield: 82.2%; MS (ESI) m/z: 252.2 [M+H]+. 84.5%; m.p.:164.1–164.8 °C; MS (ESI) m/z: 483.4 [M+H]+; 1H NMR
(400 MHz, DMSO‑d6) δH: 10.03 (s, 1H), 8.35 (s, 1H), 8.12 (s, 1H), 7.87
4.1.11.19. N2,N2-diethyl-6-(isopropylthio)pyridine-2,5-diamine (s, 1H), 7.72 (d, J = 6.2 Hz, 1H), 7.52 (d, J = 8.4 Hz, 1H), 7.22 (d,
(21c). Yield: 86.5%; MS (ESI) m/z: 240.2 [M+H]+. J = 6.9 Hz, 1H), 7.18 (s, 2H), 6.23 (d, J = 8.4 Hz, 1H), 3.78 (s, 3H),
3.40 (s, 4H), 2.53 (s, 4H), 2.09 (s, 3H) 1.39 (s, 3H).
4.1.12. General procedure for preparation of compounds (22a-x and 23a-
d) 4.1.12.7. N-(2-((5-chloro-2-((2-methoxy-4-morpholinophenyl)amino)
To the solution of 18a-c, 18e-g, 19a-g, 20a-c or 21a-c (0.005 mol) pyrimidin-4-yl)amino)phenyl)acetamide (22g). Yield: 81.3%;
in isopropanol was added 4a-c or 11(0.005 mol) and p-toluenesulfonic m.p.:154.1–159.5 °C; MS (ESI) m/z: 468.4 [M−H]−; 1H NMR (400 MHz,
(0.005 mol). The resulting mixture was heated at 80 °C for 24 h. After DMSO‑d6) δH: 10.04 (s, 1H), 8.39 (s, 1H), 8.03 (s, 1H), 7.85 (s, 1H), 7.72
completion of the reaction as indicated by TLC, the mixture was filtered (d, J = 8.2 Hz, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.27 (dd, J = 7.2 Hz, 2.1HZ,
immediately. Then the residue was stirred with aqueous NaHCO3. The 1H), 7.23–7.09 (m, 2H), 6.20 (d, J = 8.5 Hz, 1H), 3.79 (s, 3H), 3.72–3.69
aqueous solution was extracted with DCM (3 × 50 mL) and combined (m, 4H), 3.37–3.34 (m, 4H), 2.09 (s, 3H).
organic layers. The mixture was evaporated and purified on silica gel
(200–300 mesh) using dichloromethane/methanol (2500:1). Then 4.1.12.8. (E)-N-(2-((5-chloro-2-((2-methoxy-4-morpholinophenyl)amino)
evaporate the mixture. The solid was dried under vacuum and gained pyrimidin-4-yl)amino)phenyl)but-2-enamide (22h). Yield: 78.3%;
pure compounds 22a-x and 23a-d. m.p.:172.3–178.6 °C; MS (ESI) m/z: 496.6 [M+H]+; 1H NMR
(400 MHz, DMSO‑d6) δH: 10.07 (s, 1H), 8.51 (s, 1H), 8.01 (s, 1H),
4.1.12.1. N-(2-((5-chloro-2-((4-(diethylamino)-2-methoxyphenyl)amino) 7.83 (s, 1H), 7.75–7.69 (m, J = 13.9, 1H), 7.65 (d, 1H), 7.38–7.27 (m,
pyrimidin-4-yl)amino)phenyl)acetamide (22a). Yield: 82.0%; m.p.: J = 9.0 Hz, 1H), 7.24–7.14 (m, 2H), 6.87 (dq, J = 13.9, 6.9 Hz, 1H),
172.3–178.6 °C; MS (ESI) m/z: 454.4 [M−H]−; 1H NMR(400 MHz, 6.19 (d, J = 8.8 Hz, 2H), 3.79 (s, 3H), 3.76–3.67 (m, 4H), 3.39–3.33

696
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

(m, 4H), 1.23 (t, J = 7.2 Hz, 3H). J = 6.9 Hz, 4H), 2.96 (s, 3H), 1.20 (d, J = 6.2 Hz, 6H), 1.13 (t,
J = 6.9 Hz, 6H).
4.1.12.9. (E)-N-(2-((5-chloro-2-((6-(diethylamino)-2-methoxypyridin-3-yl)
amino)pyrimidin-4-yl)amino)phenyl)but-2-enamide (22i). Yield: 84.8%; 4.1.12.16. N-(2-((5-chloro-2-((2-(isopropylthio)-4-morpholinophenyl)
m.p.:193.8–194.6 °C; MS (ESI) m/z: 480.5 [M−H]−; 1H NMR (400 MHz, amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22p). Yield:
DMSO‑d6) δH: 10.07 (s, 1H), 8.33 (s, 1H), 7.90 (s, 1H), 7.83 (s, 1H), 80.7%; m.p.: 166.9–168.3 °C; MS (ESI) m/z: 548.5 [M−H]−; 1H NMR
7.75–7.67 (m, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.38–7.27 (m, J = 8.7 Hz, (400 MHz, DMSO‑d6) δH: 9.32 (s, 1H), 8.41 (s, 2H), 8.05 (s, 2H), 7.32 (t,
1H), 7.24–7.14 (m, 2H), 6.87 (dq, J = 12.9, 6.9 Hz, 1H), 6.19 (d, J = 8.5 Hz, 2H), 7.11 (s, 2H), 6.52 (d, J = 8.7 Hz, 1H), 3.83–3.75 (dt,
J = 8.4 Hz, 2H), 3.79 (s, 3H), 3.67–3.56 (m, 4H), 1.26 (t, 6H), 1.43 (t, J = 13.6, 6.8 Hz, 1H), 3.73 (m, J = 13.6, 4H), 3.44 (m, 4H), 2.98 (s,
J = 6.9 Hz, 3H). 3H), 1.28 (d, J = 6.8 Hz, 6H).

4.1.12.10. (E)-N-(2-((5-chloro-2-((6-(3-hydroxyazetidin-1-yl)-2- 4.1.12.17. N-(2-((5-chloro-2-((2-(isopropylthio)-4-(piperidin-1-yl)


methoxypyridin-3-yl)amino)pyrimidin-4-yl)amino)phenyl)but-2-enamide phenyl)amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide
(22j). Yield: 82.5%; m.p.:161.0–162.1 °C; MS (ESI) m/z: 482.4 [M (22q). Yield: 86.4%; m.p.: 186.4–192.1 °C; MS (ESI) m/z: 548.4 [M
+H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 10.01 (s, 1H), 8.31 (s, 1H), +H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 9.79 (s, 1H), 8.69 (s, 1H),
7.99 (s, 1H), 7.73 (s, 1H), 7.75–7.67 (m, 1H), 7.56 (d, J = 8.4 Hz, 1H), 8.29 (s, 1H), 8.08 (s, 1H), 8.00 (s, 1H), 7.26 (t, J = 9.5 Hz, 2H), 6.96 (t,
7.38–7.27 (m, J = 8.4 Hz, 1H), 7.24–7.14 (m, 2H), 6.87 (dq, J = 13.3, J = 7.1 Hz, 1H), 6.85 (s, 1H), 6.51 (d, J = 8.7 Hz, 1H), 3.82 (dt,
6.3 Hz, 1H), 6.19 (d, J = 8.5 Hz, 2H), 5.33 (s, 1H), 4.33 (m, 1H), 3.98 J = 13.6, 6.9 Hz, 1H), 3.55 (d, J = 5.0 Hz, 4H), 2.86 (s, 3H),
(dd, J = 11.1, 4.5 Hz, 2H), 3.79 (s, 3H), 3.68 (dd, J = 11.1, 4.5 Hz, 2H), 1.71–1.50 (m, 6H), 1.29 (d, J = 6.8 Hz, 6H).
1.52 (t, J = 6.8 Hz, 3H).
4.1.12.18. N-(2-((5-chloro-2-((4-(diethylamino)-2-(isopropylthio)
4.1.12.11. (E)-N-(2-((5-chloro-2-((6-(4-(2-hydroxyethyl)piperazin-1-yl)- phenyl)amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide
2-methoxypyridin-3-yl)amino)pyrimidin-4-yl)amino)phenyl)but-2-enamide (22r). Yield: 82.3%; m.p.: 173.0–176.2 °C; MS (ESI) m/z: 536.2 [M
(22k). Yield: 81.6%; m.p.:161.0–162.1 °C; MS (ESI) m/z: 539.4 [M +H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 9.29 (s, 1H), 9.08 (s, 1H),
+H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 10.02 (s, 1H), 8.33 (s, 1H), 8.49 (s, 1H), 8.18 (s, 1H), 8.12 (s, 1H), 7.99 (d, J = 7.9 Hz, 1H), 7.70
7.83 (s, 1H), 7.77 (s, 1H), 7.75–7.67 (m, J = 13.8, 1H), 7.56 (d, (d, J = 9.4 Hz, 1H), 7.39 (dd, J = 7.8, 1.4 Hz, 1H), 7.31 (td, J = 7.8,
J = 8.4 Hz, 1H), 7.38–7.27 (m, 1H), 7.24–7.18 (m, 2H), 6.86 (dq, 1.4 Hz, 1H), 7.22 (td, J = 7.7, 1.4 Hz, 1H), 6.69 (d, J = 8.4 Hz, 1H),
J = 13.8, 6.9 Hz, 1H), 6.19 (d, J = 8.4 Hz, 2H), 4.46 (s, 1H), 3.79 (s, 3.43–3.40 (m, 4H), 2.96 (s, 3H), 1.64–1.49 (m, 6H), 1.23 (s, 6H).
3H), 3.55 (t, J = 5.8 Hz, 2H), 3.40 (s, 4H), 2.62 (s, 4H), 2.43 (t,
J = 5.8 Hz, 2H), 1.52 (t, J = 6.9 Hz, 3H). 4.1.12.19. N-(2-((5-chloro-2-((4-morpholinophenyl)amino)pyrimidin-4-
yl)amino)phenyl)methanesulfonamide (22s). Yield: 83.4%; m.p.:
4.1.12.12. N-(2-((5-chloro-2-((2-methoxy-4-morpholinophenyl)amino) 201.2–202.8 °C; MS (ESI) m/z: 476.3 [M+H]+; 1H NMR (400 MHz,
pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22l). Yield: 85.3%; DMSO‑d6) δH: 9.29 (s, 1H), 9.15 (s, 1H), 8.51 (s, 1H), 8.24 (s, 1H), 8.13
m.p.: 181.0–182.1 °C; MS (ESI) m/z: 506.4 [M+H]+; 1H NMR (s, 1H), 7.97 (d, J = 7.8 Hz, 1H), 7.76 (d, J = 9.9 Hz, 1H), 7.39 (dd,
(400 MHz, DMSO‑d6) δH: 9.32 (s, 1H), 8.46 (s, 1H), 8.12 (s, 1H), 8.08 J = 7.9, 1.4 Hz, 1H), 7.32 (t, J = 7.1 Hz, 1H), 7.23 (td, J = 7.7, 1H),
(s, 1H), 7.98 (s, 1H), 7.60 (d, J = 8.3 Hz, 1H), 7.34 (d, J = 8.1 Hz, 1H), 6.69 (d, J = 9.1 Hz, 1H), 3.70 (m, 4H), 3.32 (s, 4H), 2.95 (s, 3H).
7.18 (t, J = 7.8 Hz, 2H), 6.25 (d, J = 8.4 Hz, 1H), 3.78 (s, 3H),
3.75–3.69 (m, 4H), 3.53 (s, 4H), 2.96 (s, 3H). 13C NMR (100 MHz, 4.1.12.20. N-(2-((5-chloro-2-((4-(piperidin-1-yl)phenyl)amino)
DMSO‑d6) δC: 159.44, 155.80, 155.58, 155.05, 154.85, 135.97, 128.89, pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22t). Yield: 82.6%;
127.52, 127.31, 124.80, 124.53, 113.25, 104.06, 98.16, 66.41 (2C), m.p.: 164.3–167.3 °C; MS (ESI) m/z: 474.4 [M+H]+; 1H NMR
53.13, 46.05 (2C), 29.16. (400 MHz, DMSO‑d6) δH: 9.30 (s, 1H), 9.07 (s, 1H), 8.50 (s, 1H), 8.18
(d, 1H), 8.11 (s, 1H), 8.00 (s, 1H), 7.68 (dd, J = 9.3, 1.5 Hz, 1H), 7.37
4.1.12.13. N-(2-((5-chloro-2-((2-isopropoxy-4-morpholinophenyl)amino) (dd, J = 7.8, 1.3 Hz, 1H), 7.27 (t, J = 7.2 Hz, 1H), 7.19 (t, J = 7.8 Hz,
pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22m). Yield: 78.5%; 1H), 6.67 (d, J = 8.9 Hz, 1H), 3.41 (d, J = 5.4 Hz, 4H), 2.95 (s, 3H),
m.p.: 176.7–178.2 °C; MS (ESI) m/z: 532.5 [M−H]−; 1H NMR 1.55 (m, 6H).
(400 MHz, DMSO‑d6) δH: 9.34 (s, 1H), 8.48 (s, 1H), 8.08 (s, 1H), 7.93
(d, J = 6.6 Hz, 1H), 7.87 (s, 1H), 7.64 (d, J = 8.4 Hz, 1H), 7.35 (dd, 4.1.12.21. N-(2-((5-chloro-2-((4-(diethylamino)phenyl)amino)pyrimidin-
J = 7.4, 1.9 Hz, 1H), 7.21–7.15 (m, 2H), 6.19 (d, J = 8.5 Hz, 1H), 5.14 4-yl)amino)phenyl)methanesulfonamide (22u). Yield: 81.4%; m.p.:
(dt, J = 12.3, 6.2 Hz, 1H), 3. 37–3.70 (m, 4H), 3.36 (s, 4H), 2.96 (s, 183.6–186.1 °C; MS (ESI) m/z: 462.4 [M+H]+; 1H NMR (400 MHz,
3H), 1.22 (d, J = 6.2 Hz, 6H). DMSO‑d6) δH: 8.95 (s, 1H), 8.56 (s, 1H), 8.05 (m, 3H), 7.60 (d,
J = 8.5 Hz, 1H), 7.36 (dd, J = 7.7, 1.5 Hz, 1H), 7.16 (m, 2H), 7.12
4.1.12.14. N-(2-((5-chloro-2-((2-isopropoxy-4-(piperidin-1-yl)phenyl) (d, 1H), 6.46 (d, J = 9.1 Hz, 1H), 3.45 (q, J = 7.0 Hz, 4H), 2.93 (s, 3H),
amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22n). Yield: 1.09 (t, J = 7.0 Hz, 6H).
84.9%; m.p.: 211.0–212.1 °C; MS (ESI) m/z: 530.5 [M−H]−; 1H NMR
(400 MHz, DMSO‑d6) δH: 9.33 (s, 1H), 8.59 (s, 1H), 8.08–8.03 (m, 1H), 4.1.12.22. N-(2-((5-chloro-2-((6-(3-hydroxyazetidin-1-yl)pyridin-3-yl)
7.99 (d, J = 5.8 Hz, 1H), 7.82 (s, 1H), 7.58 (d, J = 8.5 Hz, 1H), 7.31 amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22v). Yield:
(dd, J = 7.3, 2.0 Hz, 1H), 7.10–7.05 (m, 2H), 6.19 (d, J = 8.5 Hz, 1H), 84.3%; m.p.: 169.5–172.2 °C; MS (ESI) m/z: 462.0 [M+H]+; 1H NMR
5.13 (dt, J = 12.2, 6.2 Hz, 1H), 3.44 (d, J = 5.1 Hz, 4H), 2.91 (s, 3H), (400 MHz, DMSO‑d6) δH: 8.92 (s, 1H), 8.59 (s, 1H), 8.34–7.84 (m, 3H),
1.58 (s, 6H), 1.23 (d, J = 4.0 Hz, 6H). 7.61 (d, J = 9.1 Hz, 1H), 7.35 (dd, J = 9.5 Hz, 4.5 Hz, 1H), 7.27–7.05
(m, 2H), 7.15 (d, 1H), 6.46 (d, J = 3.1 Hz, 1H), 5.38 (s, 1H), 4.43 (m,
4.1.12.15. N-(2-((5-chloro-2-((4-(diethylamino)-2-isopropoxyphenyl) 1H), 3.98 (dd, J = 8.3, 4.5 Hz, 2H), 3.68 (dd, J = 6.8, 2H), 2.93 (s, 3H).
amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22o). Yield:
82.2%; m.p.: 196.3–202.8 °C; MS (ESI) m/z: 518.5 [M−H]−; 1H NMR 4.1.12.23. N-(2-((5-chloro-2-((6-(4-(2-hydroxyethyl)piperazin-1-yl)
(400 MHz, DMSO‑d6) δH: 9.29 (s, 1H), 8.42 (s, 1H), 8.05 (s, 1H), 8.00 (s, pyridin-3-yl)amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide
1H), 7.86 (s, 1H), 7.45 (d, J = 8.5 Hz, 1H), 7.33 (m, 1H), 7.13 (m, (22w). Yield: 83.9%; m.p.: 181.0–185.1 °C; MS (ESI) m/z: 519.4 [M
J = 6.8 Hz, 2H), 5.97 (d, J = 8.5 Hz, 1H), 5.17–5.07 (m, 1H), 3.44 (q, +H]+; 1H NMR (400 MHz, DMSO‑d6) δH: 9.25 (s, 1H), 8.86 (s, 1H),

697
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

8.34–7.84 (m, 3H), 7.60 (d, J = 9.1 Hz, 1H), 7.35 (dd, J = 7.0, 1.5 Hz, absorbency at 492 nm (for absorbance of MTT formazan) and 630 nm
1H), 7.27–7.05 (m, 2H), 7.17 (d, 1H), 6.46 (d, J = 2.8 Hz, 1H), 4.46 (s, (for the reference wavelength) was measured with an ELISA reader. All
1H), 3.45 (t, J = 6.2 Hz, 2H), 3.36 (s, 4H), 2.62 (s, 4H), 2.93 (s, 3H), of the compounds were tested three times in each of the cell lines. The
2.42 (t, J = 5.6 Hz, 2H). results, expressed as IC50 (inhibitory concentration 50%), were the
averages of three determinations and calculated relative to the vehicle
4.1.12.24. N-(2-((5-chloro-2-((6-(3-methylpiperazin-1-yl)pyridin-3-yl) (DMSO) control by the Bacus Laboratories Incorporated Slide Scanner
amino)pyrimidin-4-yl)amino)phenyl)methanesulfonamide (22x). Yield: (Bliss) software.
81.8%; m.p.: 193.2–199.2 °C; MS (ESI) m/z: 489.4 [M+H]+; 1H NMR
(400 MHz, DMSO‑d6) δH: 8.95 (s, 1H), 8.56 (s, 1H), 8.34–7.84 (m, 3H), 4.3. In vitro enzymatic assays
7.61 (d, J = 10.0 Hz, 1H), 7.35 (dd, J = 8.3, 2.6 Hz, 1H), 7.27–7.05 (m,
2H), 7.17 (d, 1H), 6.46 (d, J = 1.8 Hz, 1H), 3.25 (s, 1H), 3.10 (s, 1H), The in vitro enzymatic assays versus ALK and ALKL1196M were
2.93 (s, 3H), 2.88 (s, 1H), 2.79 (s, 1H), 2.73 (s, 1H), 2.72 (s, 1H), 2.68 evaluated by homogeneous time-resolved fluorescence (HTRF) assay. In
(s, 1H), 1.31 (s, 3H), 1.07(s, 1H). enzymatic assay, the solution of peptide substrates, ATP, appropriate
kinase, and diluted compound was mixed with the kinase reaction
4.1.12.25. 4-(5-chloro-2-((6-(4-(2-hydroxyethyl)piperazin-1-yl)-2- buffer (50 mM HEPES, pH 7.5, 0.0015% Brij-35, 10 mM MgCl2, 2 mM
methoxypyridin-3-yl)amino)pyrimidin-4-yl)-3,4-dihydro-2H-benzo[1,4- b] DTT), with blank DMSO solution as the negative control. The kinase
thiazine-1,1-dioxide (23a). Yield: 78.4%; m.p.:190.8–196.0 °C; MS reaction was initiated by the addition of tyrosine kinase proteins diluted
(ESI) m/z: 546.4 [M+H]+; 1H NMR (400 MHz, CDCl3) δH: 9.13 (s, in 39 μL of kinase reaction buffer solution and incubated at 28 °C for
1H), 8.47 (d, J = 8.3 Hz, 1H), 8.17 (d, J = 7.5 Hz, 1H), 8.11 (s, 1H), 60 min. And then add 25 μL of stop buffer (100 mM HEPES, pH 7.5,
8.00 (d, J = 8.3 Hz, 1H), 7.62 (t, J = 7.5 Hz, 1H), 7.28 (d, J = 6.1 Hz, 0.015% Brij-35, 0.2% Coating Reagent #3, 50 mM EDTA) to stop re-
1H), 7.03 (s, 1H), 6.51 (m, 2H), 6.08 (dd, J = 6.1 Hz, 4.9 Hz, 2H), 4.02 action. The plate was read by Caliper at 320 nm and 615 nm. IC50 va-
(s, 1H), 3.94 (s, 3H), 3.74 (s, 2H), 3.55 (s, 4H), 2.76 (s, 4H), 2.71 (d, lues were calculated from the inhibition curves.
J = 4.9 Hz, 2H). 13C NMR (100 MHz, DMSO‑d6) δC: 159.66, 156.37,
155.90, 155.35, 154.90, 137.78, 137.38, 135.20, 129.98, 129.84,
4.4. Western blotting
126.46, 123.92, 123.48, 112.09, 98.28, 70.26, 60.77, 58.96, 53.38
(2C), 53.06, 45.51 (2C), 29.47.
Western blotting analysis was performed as described Karpas299
cells were treated with 22l or ceritinib. After treatment for 24 h, cells
4.1.12.26. 4-(5-chloro-2-((2-methoxy-6-(piperidin-1-yl)pyridin-3-yl)
were collected. The protein in the cytoplasm was extracted, quantified
amino)pyrimidin-4-yl)-3,4-dihydro-2H-benzo[1,4-b]thiazine-1,1-dioxide
with protein assay reagent and electrophoresed on SDS-polyacrylamide
(23b). Yield: 77.8%; m.p.:158.8–166.8 °C; MS (ESI) m/z: 501.3 [M
gel until the proteins of different molecular weights were separated.
+H]+; 1H NMR (400 MHz, CDCl3) δH: 9.17 (s, 1H), 8.46 (s, 1H), 8.00
Then, the different proteins contained in the SDS-polyacrylamide gel
(m, 4H), 7.60 (s, 1H), 7.15 (s, 1H), 6.55 (s, 2H), 6.27 (s, 1H), 6.03 (s,
were transferred to polyvinylidene difluoride (PVDF) membranes. After
1H), 3.94 (s, 3H), 3.46 (s, 4H), 1.71 (s, 4H), 1.26 (s, 2H).
incubated overnight at 4 °C with the respective primary antibodies,
horseradish peroxidase (HRP)-conjugated secondary antibodies (1:
4.1.12.27. 4-(5-chloro-2-((2-methoxy-6-(pyrrolidin-1-yl)pyridin-3-yl)
3000) was added. The Enhanced chemiluminescence reagent was used
amino)pyrimidin-4-yl)-3,4-dihydro-2H-benzo[1,4-b]thiazine-1,1-dioxide
to identify the specific bands. Densitometry analysis on immunoblots
(23c). Yield: 80.5%; m.p.:178.5–181.2 °C; MS (ESI) m/z: 487.4 [M
were performed using ImageJ2 software.
+H]+; 1H NMR (400 MHz, CDCl3) δH: 9.57 (s, 1H), 8.40 (d, J = 8.1 Hz,
1H), 8.01(m, 2H), 7.72 (s, 1H), 7.51 (s, 1H), 7.42 (m, 1H), 7.30 (m,
2H), 6.58–6.50 (m, 2H), 6.06 (d, J = 9.0 Hz, 1H), 5.87 (s, 1H), 3.89 (s, 4.5. Morphology assays of apoptotic cells
3H), 3.45 (s, 4H), 2.02 (s, 4H).
Apoptotic morphological changes of H2228 and Karpas299 cells
4.1.12.28. 4-(5-chloro-2-((2-isopropoxy-6-(piperidin-1-yl)pyridin-3-yl) were detected by AO/EB staining. Briefly, cells were seeded in six-well
amino)pyrimidin-4-yl)-3,4-dihydro-2H-benzo[1,4- b]thiazine-1,1-dioxide plates for 24 h, then treated with ceritinib or different concentrations of
(23d). Yield: 75.3%; m.p.:162.3.5–169.2 °C; MS (ESI) m/z: 529.4 [M 22l for 48 h. The cells were washed with phosphate buffer saline (PBS)
+H]+; 1H NMR (400 MHz, CDCl3) δH: 9.81 (s, 1H), 8.30 (s, 1H), 8.17 and then stained with AO/EB mixed solution (AO: EB ¼ 1: 1) for
(s, 1H), 8.08 (s, 2H), 7.80 (s, 1H), 7.54 (m, 2H), 6.55–6.52 (m, 2H), 15 min. The stained cells were washed twice with PBS and observed by
6.05 (s, 2H), 3.56–3.49 (m, 7H), 2.23 (s, 4H), 1.90 (s, 2H), 1.72 (s, 4H). fluorescence microscope.
13
C NMR (100 MHz, DMSO‑d6) δC: 159.62, 158.74, 155.88, 155.21,
154.89, 137.77, 136.87, 135.10, 129.98, 129.81, 126.56, 123.95, 4.6. Molecular docking
123.64, 111.64, 97.85, 67.88, 51.84, 46.43 (2C), 44.64, 25.39 (2C),
24.75, 22.43 (2C). The molecular modeling studies were performed with Discovery
Studio 2017. The protein coordinates (PDB ID code: 4MKC) were
4.2. MTT assay in vitro downloaded from the Protein Data Bank (http://www.rcsb.org/pdb/).
In the docking process, the protein protocol was prepared by several
The cytotoxic activities of compounds 22a-x and 23a-d were eval- operations, including standardization of atom names and insertion of
uated against H2228, Karpas299 and A549 by the standard MTT assay missing atoms in residues. Then, the receptor model was typed with the
in vitro, with ceritinib as the positive control. The cancer cell lines were CHARMm force field and a binding sphere with radius of 12 Å was
cultured in minimum essential medium (MEM) supplement with 10% defined through the original ligand (ceritinib) as the binding site. The
fetal bovine serum (FBS). Approximate 4 × 103 cells, suspended in ceritinib, 22l and 23a were drawn with ChemSketch and fully mini-
MEM medium, were plated into each well of a 96-well plate and in- mized using the CHARMm force field. Finally, they were docked into
cubated in 5% CO2 at 37 °C for 24 h. The tested compounds at the in- the binding site using the CDOCKER protocol with the default settings.
dicated final concentrations were added to the culture medium and
incubated for 72 h. Fresh MTT was added to each well at the terminal 5. Conflict of interest
concentration of 5 μg/mL, and incubated with cells at 37 °C for 4 h. The
formazan crystals in each well were dissolved in 100 μL DMSO, and the The authors have declared no conflict of interest.

698
L. Xing et al. Bioorganic Chemistry 81 (2018) 689–699

Acknowledgements lymphoma kinase inhibitor (CH5424802), Bioorg. Med. Chem. 20 (2012)


1271–1280.
[11] W. Huang, S. Liu, D. Zou, M. Thomas, Y. Wang, T. Zhou, J. Romero, A. Kohlmann,
This work was supported by National Natural Science Foundation of F. Li, J. Qi, L. Cai, T.A. Dwight, Y. Xu, R. Xu, R. Dodd, A. Toms, L. Parillon, X. Lu,
China, China (No. 81673308), Natural Science Foundation of Liaoning R. Anjum, S. Zhang, F. Wang, J. Keats, S.D. Wardwell, Y. Ning, Q. Xu, L.E. Moran,
Province, China (No. 201602703) and Development Project of Ministry Q.K. Mohemmad, H.G. Jang, T. Clackson, N.I. Narasimhan, V.M. Rivera, X. Zhu,
D. Dalgarno, W.C. Shakespeare, Discovery of brigatinib (AP26113), a phosphine
of Education Innovation Team, China (No. IRT1073). oxide-containing, potent, orally active inhibitor of anaplastic lymphoma kinase, J.
Med. Chem. 59 (2016) 4948–4964.
References [12] N.R. Infarinato, J.H. Park, K. Krytska, H.T. Ryles, R. Sano, K.M. Szigety, Y. Li,
H.Y. Zou, N.V. Lee, T. Smeal, M.A. Lemmon, Y.P. Mosse, The ALK/ROS1 inhibitor
PF-06463922 overcomes primary resistance to crizotinib in ALK-driven neuro-
[1] S.W. Morris, M.N. Kirstein, M.B. Valentine, K.G. Dittmer, D.N. Shapiro, blastoma, Cancer Discov. 6 (2016) 96–107.
D.L. Saltman, A.T. Look, Fusion of a kinase gene, ALK, to a nucleolar protein gene, [13] C.M. Lovly, J.M. Heuckmann, E. De Stanchina, H. Chen, R.K. Thomas, C. Liang,
NPM, in non-hodgkin's lymphoma, Science 263 (1994) 1281–1284. W. Pao, Insights into ALK-driven cancers revealed through development of novel
[2] X. Lu, K. Ding, Novel anaplastic lymphoma kinase inhibitors targeting clinically ALK tyrosine kinase inhibitors, Cancer Res. 71 (2011) 4920–4931.
acquired resistance, J. Med. Chem. 57 (2014) 1167–1169. [14] M. Latif, Z. Ashraf, S. Basit, A. Ghaffar, M.S. Zafar, A. Saeed, S.A. Meo, Latest
[3] M. Soda, Y.L. Choi, M. Enomoto, S. Takada, Y. Yamashita, S. Ishikawa, S. Fujiwara, perspectives of orally bioavailable 2,4-diarylaminopyrimidine analogues (DAAP
H. Watanabe, K. Kurashina, H. Hatanaka, M. Bando, S. Ohno, Y. Ishikawa, alogues) as anaplastic lymphoma kinase inhibitors: discovery and clinical devel-
H. Aburatani, T. Niki, Y. Sohara, Y. Sugiyama, H. Mano, Identification of the opments, Rsc Adv. 8 (2018) 16470–16493.
transforming EML4–ALK fusion gene in non-small-cell lung cancer, Nature 448 [15] Y. Wang, S. Chen, G. Hu, J. Wang, W.F. Gou, D.Y. Zuo, Y.C. Gu, P. Gong, X. Zhai,
(2007) 561–566. Discovery of novel 2,4-diarylaminopyrimidine analogues as ALK and ROS1 dual
[4] X.C. Zhang, S. Zhang, X.N. Yang, J.J. Yang, Q. Zhou, L. Yin, S.J. An, J.Y. Lin, inhibitors to overcome crizotinib-resistant mutants including G1202R, Eur. J. Med.
S.L. Chen, Z. Xie, M. Zhu, X.L. Zhang, Y.L. Wu, Fusion of EML4 and ALK is asso- Chem. 143 (2018) 123–136.
ciated with development of lung adenocarcinomas lacking EGFR and KRAS muta- [16] G.R. Mathi, C.H. Kang, H.K. Lee, R. Achary, H. Lee, J. Lee, J.D. Ha, S. Ahn,
tions and is correlated with ALK expression, Mol. Cancer. 9 (2010) 188. C.H. Park, C.O. Lee, J.Y. Hwang, C. Yun, H.J. Jung, S.Y. Cho, H.R. Kim, P. Kim,
[5] J. Chen, C. Jiang, S. Wang, LDK378: a promising anaplastic lymphoma kinase (ALK) Replacing the terminal piperidine in ceritinib with aliphatic amines confers activ-
inhibitor, J. Med. Chem. 56 (2013) 5673–5674. ities against crizotinib-resistant mutants including G1202R, Eur. J. Med. Chem. 126
[6] J.J. Cui, M. Tran-Dube, H. Shen, M. Nambu, P. Kung, M. Pairish, L. Jia, J. Meng, (2016) 536–549.
L. Funk, I. Botrous, M. McTigue, N. Grodsky, K. Ryan, E. Padrique, G. Alton, [17] Y. Wang, G. Zhang, G. Hu, Y. Bu, H. Lei, D. Zuo, M. Han, X. Zhai, P. Gong, Design,
S. Timofeevski, S. Yamazaki, Q. Li, H. Zou, J. Christensen, B. Mroczkowski, synthesis and biological evaluation of novel 4-arylaminopyrimidine derivatives
S. Bender, R.S. Kania, M.P. Edwards, Structure based drug design of crizotinib (PF- possessing a hydrazone moiety as dual inhibitors of L1196M ALK andROS1, Eur. J.
02341066), a potent and selective dual inhibitor of mesenchymalepithelial transi- Med. Chem. 123 (2016) 80–89.
tion factor (c-MET) kinase and anaplastic lymphoma kinase (ALK), J. Med. Chem. [18] Z. Song, Y. Yang, Z. Liu, X. Peng, J. Guo, X. Yang, K. Wu, J. Ai, J. Ding, M. Geng,
54 (2011) 6342–6363. A. Zhang, Discovery of novel 2,4-diarylaminopyrimidine analogues (DAAP alogues)
[7] B.J. Solomon, T. Mok, D. Kim, Y. Wu, K. Nakagawa, T. Mekhail, E. Felip, showing potent inhibitory activities against both wild-type and mutant ALK kinases,
F. Cappuzzo, J. Paolini, T. Usari, S. Iyer, A. Reisman, K.D. Wilner, J. Tursi, J. Med. Chem. 58 (2015) 197–211.
F. Blackhall, First-line crizotinib versus chemotherapy in ALK-positive lung cancer, [19] C. Bissantz, B. Kuhn, M. Stahl, A medicinal chemist’s guide to molecular interac-
N. Engl. J. Med. 371 (2014) 2167–2177. tions, J. Med. Chem. 53 (2010) 5061–5084.
[8] M.M. Awad, A.T. Shaw, ALK inhibitors in none small cell lung cancer: crizotinib and [20] D.J. St. Jean Jr., C. Fotsch, Mitigating heterocycle metabolism in drug discovery, J.
beyond, Clin. Adv. Hematol. Oncol. 12 (2014) 429–439. Med. Chem. 55 (2012) 6002–6020.
[9] T.H. Marsilje, W. Pei, B. Chen, W. Lu, T. Uno, Y. Jin, T. Jiang, S. Kim, N. Li, [21] K.W. Ward, Optimizing pharmacokinetic properties and attaining candidate selec-
M. Warmuth, Y. Sarkisova, F. Sun, A. Steffy, A.C. Pferdekamper, A.G. Li, tion, Top. Med. Chem. 11 (2012) 73–95.
S.B. Joseph, Y. Kim, B. Liu, T. Tuntland, X. Cui, N.S. Gray, R. Steensma, Y. Wan, [22] N.A. Meanwell, Synopsis of some recent tactical application of bioisosteres in drug
J. Jiang, G. Chopiuk, J. Li, W.P. Gordon, W. Richmond, K. Johnson, Jo Chang, design, J. Med. Chem. 54 (2011) 2529–2591.
T. Groessl, Y. He, A. Phimister, A. Aycinena, C.C. Lee, B. Bursulaya, [23] N.A. Meanwell, The influence of bioisosteres in drug design: tactical applications to
D.S. Karanewsky, H.M. Seidel, J.L. Harris, P. Michellys, Synthesis, structureactivity address developability problems, Top. Med. Chem. 9 (2013) 283–381.
relationships, and in vivo efficacy of the novel potent and selective anaplastic [24] Z. Liu, J. Ai, X. Peng, Z. Song, K. Wu, J. Zhang, Q. Yao, Y. Chen, Y. Ji, Y. Yang,
lymphoma kinase (ALK) inhibitor 5-chloro-N2-(2-isopropoxy-5-methyl-4-(piperidin- M. Geng, A. Zhang, Novel 2,4-diarylaminopyrimidine analogues (DAAPalogues)
4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (LDK378) showing potent c-Met/ALK multikinase inhibitory activities, Acs. Med. Chem. Lett.
currently in phase 1 and phase 2 clinical trials, J. Med. Chem. 56 (2013) (2014) 304–308.
5675–5690. [25] Y.Q. Wu. Bicyclic anaplastic lymphoma kinase inhibitor, CN 106118029, 2016.
[10] K. Kinoshita, K. Asoh, N. Furuichi, T. Ito, H. Kawada, S. Hara, J. Ohwada, T. Miyagi, [26] J. Major, N. Piton. Sulfonamides and sulfamides as zap-70 inhibitors, WO
T. Kobayashi, K. Takanashi, T. Tsukaguchi, H. Sakamoto, T. Tsukuda, N. Oikawal, 2010146132, 2010.
Design and synthesis of a highly selective, orally active and potent anaplastic

699

Вам также может понравиться