Design and synthesis of boron-containing diphenylpyrimidines as potent BTK and JAK3 dual inhibitors

Design and synthesis of boron-containing diphenylpyrimidines as potent BTK and JAK3 dual inhibitors

Journal Pre-proofs Design and Synthesis of Boron-containing Diphenylpyrimidines as Potent BTK and JAK3 Dual Inhibitors Jing Ren, Wei Shi, Damin Zhao, ...

1MB Sizes 0 Downloads 6 Views

Journal Pre-proofs Design and Synthesis of Boron-containing Diphenylpyrimidines as Potent BTK and JAK3 Dual Inhibitors Jing Ren, Wei Shi, Damin Zhao, Qinglin Wang, Xiayun Chang, Xiangyi He, Xiaojin Wang, Yong Gao, Peng Lu, Xiquan Zhang, Hongjiang Xu, Yinsheng Zhang PII: DOI: Reference:

S0968-0896(19)31649-9 https://doi.org/10.1016/j.bmc.2019.115236 BMC 115236

To appear in:

Bioorganic & Medicinal Chemistry

Received Date: Revised Date: Accepted Date:

23 September 2019 21 November 2019 23 November 2019

Please cite this article as: J. Ren, W. Shi, D. Zhao, Q. Wang, X. Chang, X. He, X. Wang, Y. Gao, P. Lu, X. Zhang, H. Xu, Y. Zhang, Design and Synthesis of Boron-containing Diphenylpyrimidines as Potent BTK and JAK3 Dual Inhibitors, Bioorganic & Medicinal Chemistry (2019), doi: https://doi.org/10.1016/j.bmc. 2019.115236

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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.

© 2019 Published by Elsevier Ltd.

Design and Synthesis of Boron-containing Diphenylpyrimidines as Potent BTK and JAK3 Dual Inhibitors Jing Ren, Wei Shi, Damin Zhao, Qinglin Wang, Xiayun Chang, Xiangyi He, Xiaojin Wang, Yong Gao, Peng Lu, Xiquan Zhang, Hongjiang Xu*, Yinsheng Zhang* Institute for Innovative Drug Discovery Chia Tai Tianqing Pharmaceutical Group Co., Ltd 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China *Correspondence to: Yinsheng Zhang ([email protected]), Institute for Innovative Drug Discovery, Hongjing Xu ([email protected]), Institute for Pharmacological Evaluation, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China

ABSTRACT: Bruton's tyrosine kinase (BTK) and Janus kinase 3 (JAK3) are very promising targets for hematological malignancies and autoimmune diseases. In recent years, a few compounds have been approved as a marketed medicine, and several are undergoing clinical trials. By recombining the dominant backbone of known active compounds, constructing a foused library, and screening a broad panel of kinases, we found a class of compounds with dual activities of anti-BTK and anti-JAK3. Some of the compounds have shown 10-folds more active in the enzyme and cell-based assays than a known active compound. Furthermore, liver microsome stability experiments show that these compounds have better stability than ibrutinib. These explorations offered new clues to discover benzoxaborole fragment and pyrimidine scaffold as more effective BTK and JAK3 dual inhibitors. Keywords; BTK, JAK3, dual inhibitor, benzoxaborole, pyrimidine, hematological, autoimmune diseases

1. Introduction Bruton’s tyrosine kinase (BTK) is a non-receptor Tec family tyrosine kinase that is broadly expressed in hematopoietic cells, with the exception of T cells. B cells are

essential to the pathogenesis of autoimmune diseases such as rheumatoid arthritis (RA). BTK plays a crucial role in signaling through the B-cell antigen receptor (BCR) and the Fcγ receptor (FcγR) in B cells and myeloid cells, respectively [1,2]. It is necessary for BCR-dependent proliferation of B cells as well as the production of pro-inflammatory cytokines and co-stimulatory molecules [3,4,5]. BTK deregulation has been observed in numerous B-cell-derived malignancies, including acute lymphoblastic leukemia (ALL), chronic lymphocytic Leukemia (CLL), Non Hodgkin Lymphoma (NHL), mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM) and multiple myeloma (MM) [6,7]. Taken together, BTK could be a potential target for the treatment of autoimmune diseases and hematologic malignancies. A few inhibitors are launched such as ibrutinb [8,9] and acalabrutinib [10,11] and several are under clinical trials for the treatment of CLL, MCL, WM, rheumatoid arthritis, etc.

The Janus kinases (JAKs) are a family of non-receptor tyrosine protein kinases crucially involved in immune signaling [12]. All four JAK family members, namely JAK1, JAK2, JAK3 and tyrosine kinase (TYK) 2 signal via the JAK/STAT (signal transducers and activators of transcription pathway). JAK3 was found to play an important role in cytokine-induced proliferation and survival of normal and leukemic B-cell precursors [13,14]. JAK3 also regulates the anti-apoptotic PI3K-AKT pathway and their downstream targets, some of which have been implicated as important oncogenic proteins [15]. Furthermore, selective JAK3 inhibitors mediate cytokine signaling through the γ-common chain receptors [16]. As a result, JAK3 is an attractive target for hematologic malignant and autoimmune disease. Currently, the selective JAK3 inhibitor PF-06651600 is in the clinical phase II study for the treatment of rheumatoid arthritis

(NCT02969044),

alopecia

(NCT02974868)

and

ulcerative

colitis

(NCT02958865).

The 2,4 di-substituted pyrimidine is a class of dominant scaffold with broad biological activity in multiple kinase targets, such as EGFR, ALK, CDK, BCR-ABL and so on. The nitrogen atom on pyrimidine ring as a hydrogen bond acceptor and the common 2-

or 4-substituted amino group in the pyrimidine core as an H bond donor can form strong hydrogen bonds with ATP pocket hinge region. It is noticed that 22 of the 48 kinase inhibitors currently on the market owns the pyrimidine core. Some represent compounds are shown in Figure 1.

N

O

H N

N

O H N

NH O N

O S OH O

N N

N

N N H

2

Brigatinib, ALK

Cl

O

N N H

N N

N

N

N H

N

O S OH O

N F

3 Abemaciclib, CDK4\6

O

N

F

N

N

N

P

N

1 Osimertinib, EGFR

N

N

H N

N

4 Imatinib,BCR-ABL, PDGFR, c-KIT

Figure 1. The examples of the marked drugs with a pyrimidine core.

In recent years, the application of boron atoms in drug development has received more and more attention. The boron atom is located at the 5th position of the periodic table as a non-metallic element and is a necessary trace element for plants grown in some natural environments. Boron is in Group 3 of the periodic table, and the outer electrons have a p-orbital orbital that forms a coordination bond with the lone pair of O and N. Many boron-containing compounds take advantage of this feature to form covalent bonds with the amino acid residues of the receptor protein, thereby increasing their binding activity [17]. There are currently four boron-containing drug molecules on the market, and many are at different stages of clinical research. Figure 2 shows some active compounds containing a common boron heterocycle, benzoxaboroles, such as crisaborole 5 [18], GSK565 6 [19], antimalarial agent 7 [20] and anti-Wolbachia agent 8 [21]. Some studies on benzoxaboroles show they have drugable properties and could be a class of dominant pharmacophore [22, 23, 24, 25].

HO

O

OH B O

O O B OH

NC

5 crisaborole,PDE-4

HO

N N

6 GSK656,Leucyl-tRNA Synthetase (Mycobacterium tuberculosis) Inhibitors

OH B O

O

N

OH HO B O

O 7

HCl NH2

Cl

F

O O

O

O AN11251,anti-Wolbachia activity 8

antimalarial activity

Figure 2. The boron-containing drugs on the market and in clinical trials.

To create a new chemical entity we combined these two types of dominant pharmacophores by introducing the benzoxaboroles into the 2- or 4-position of pyrimidine. A total of 31 compounds were synthesized to construct a small focused library. Then, we performed a broad kinases screening (23 kinases) of these compounds and found seventeen hit compounds for eight targets (Table 1), among which the IC50 of compound 9 is 2.97 nM for BTK and 0.50 nM for JAK3 (Figure 3).

Table 1. Number of hits with >50% inhibition rate at 100 nM Targets

BTK

ALK

EGFR

JAK1

JAK3

BLK

ABL1

FLT3

(L858R\ T790M) Number of hits

1

6

4

2

1

1

1

1

Inhibition rate

94%

54-63%

51-98%

66-89%

98%

68%

69%

82%

at 100 nM

R N R

O N

2, 4-substituted pyrimidine

HN

+ R R

X

N O B OH

benzoxaboroles

N H

O B OH

N

N H

NH 3

O B HO

N H

4

N 2

5

N

CF3

6

1

9 31 compounds focused library

BTK IC50= 2.97 nM JAK3 IC50=0.50 nM

Figure 3. Design of a focused library and results of a broad anti-kinases screening.

Inspired by the above results, we then designed and synthesized a series of derivatives of compound 9 containing an organic boron fragment based on the known structureactivity relationships (SARs) of effective BTK inhibitors. Interestingly, these newly designed boron-containing compounds exhibited potent inhibitory activity against both BTK and JAK3 enzymes. In this paper, we describe these newly discovered BTK and JAK3 dual inhibitors, including their synthesis, biological evaluation in vitro and SARs.

2. Results and Discussion 2.1. Molecular docking of compound 9 with BTK and JAK3 To provide a direction for subsequent structural optimization we first probed the binding mode of the compound 9 with BTK using MOE (Figure 4a). As shown in Figure 4a, besides a covalent bond force between the essential acrylamide functionality of 9 with the Cys481 residue, compound 9 forms several important hydrogen bonds with the ATP pocket of BTK kinase domain. The NH at 2-position and the nitrogen atom at 1-position of the pyrimidine ring make two hydrogen bonds with the backbone carbonyl and NH of Met477 residue, respectively. Additionally, the hydroxyl group of boric acid acts as an extra hydrogen bond donor to form another hydrogen bond with the carbonyl group of Ala478 residue. The phenyl ring attached to the amino group at 2-position of the pyrimidine and the pyrimidine ring itself make a favorable hydrophobic interaction with Leu408 and Leu528. The binding mode of the known molecule, CC292, is similar to compound 9 as a whole, but CC292 does not form the hydrogen bond with Ala478 residue (Figure 4b).

Figure 4. a) Binding mode of compound 9 with BTK kinase domain generated by MOE (PDB ID: 5P9J). b) The overlay of CC292 (red,co-crystal) and compound 9 (yellow) in ATP pocket of BTK kinase domain. c) Binding mode of compound 9 with JAK3 kinase domain generated by MOE (PDB ID: 5TOZ). d) The overlay of PF-06651600 (blue,co-crystal) and compound 9 (yellow) in ATP pocket of JAK3 kinase domain.

We also probed the binding mode of the compound 9 with JAK3 using MOE (Figure 4c). As shown in Figure 4c, compound 9 forms several important biding forces with the ATP pocket of the JAK3 protein.

Besides a covalent bond force between the essential

acrylamide functionality of 9 with the Cys356 residue, the NH at 2-position and the nitrogen atom at 1-position of the pyrimidine ring make two hydrogen bonds with the backbone carbonyl and NH of Leu323 residue, respectively. Two aromatic ring systems

of 9 are engaged in a hydrophobic association with leu73 and leu497. In addition, the hydroxyl group of boric acid acts as an extra hydrogen bond donor to form another hydrogen bond with the carbonyl group of Pro324 residue. The binding mode of the known compound, PF-06651600, at ATP pocket of JAK3 kinase domainat is not exactly the same as that of compound 9, but both have a covalent bond with the Cys356 residue and a hydrogen bond with Leu323 residue (Figure 4d).

2.2. Chemistry Scheme 1. Synthesis of compounds 14a-p

R

O N H

Cl

1

X1

N Cl

R1

O R2

N

a

Cl 10 10a, X1= NH2, R1=H 10b, X1=NH2, R1=OMe 10c, X1=OH, R1=H

11 11a,R2=CF3, 11b,R2=F, 11c,R2=Cl, 11d,R2=Me, 11e,R2=OMe

R

N

2

N

12 12a, R1=H,R2=CF3, X1=NH 12b, R1=H,R2=F, X1=NH 12c, R1=H,R2=Cl, X1=NH 12d, R1=H,R2=Me, X1=NH 12e, R1=H,R2=OMe, X1=NH 12f, R1=H,R2=OMe, X1=O 12g, R1=OMe, R2=CF3, X1=NH

b 13a, R3=

X1

N H

13

X1

N H

R1

O

R3 NH2

R O

3

N H

R2

N N

14a-e (Table 1) 14f-n (Table 2) 14o-p (Table 3)

B HO

13b, R3= O

B HO

B HO

3 14l, R =

B HO

13g, R =

B HO O

B O

c

B HO

3 14m, R =

HO

B HO

B OH

3

HO

14m', R3=

O

13f, R3= O

B O

c

3 13d, R = O

13e, R3=

O

14l', R3=

13c, R3= O

F

O

B HO

13h, R3=

O

13i,

R 3=

O

13j,

R 3=

B O

B O NH

Reagents and conditions: (a) DIPEA, n-BuOH, rt; (b): (1) TFA, n-BuOH, reflux, for compound 14a-p except for 14n; (2) Pd2(dba)3, X-PHOS, K2CO3, t- BuOH, MW, 120oC, 30 min, for compound 14n; (c) Isobutylboronic, HCl, MeOH, heptane, rt, 1.5h.

The syntheses of compounds 14a-p were convenient, and the general routes are shown in Scheme 1. Generally, to synthesize these boron-containing molecules, the key intermediates, 3- or 4-subtituted N-phenylacrylamide (10) and animo-substituted benzoxaboroles (13) were first prepared according to the know methods [24, 25, 26, 27] or purchased in case they are commercially available. Substitution of 2,4-dichloropyrimidine (11a-e) with aniline or phenol derivatives (10a-c) at the 2-position in the presence of DIPEA gave 2-substituted chloro-pyrimidine derivatives (12a-g), which were further substituted with boron-containing anilines (13a-j) at the 4-position in the

presence of TFA to afford the final compounds (14a-m, 14o-p, Scheme 1). Intermediate 12a reacted with 13j under Buchwald–Hartwig condition to give compound 14n (Scheme 1).

In some cases, the boron-containing anilines had to selectively substitute 2-chloro group of 2,4-dichloro-5-(trifluoromethyl)pyrimidine (11a) first using ZnCl2 as a catalyst. Then, the further substitution of the intermediate (15a-b) with anilines or phenol (10a, 10c-e) at the 4-position under microwave heating condition furnished the final compounds (16a-d, Scheme 2). Scheme 2. Synthesis of compounds 16a-d

Cl 13b,k

13b, R3= O B 4 HO X2=NH2

CF3

N

R3 X2

N R3 2 X N

a

N 11a

CF3

R4 1 X N R3 2 X N

b

15a-b

15a, R3-X2=

O B 3 HO

O

2

15b, R -X =

O B 4 HO

CF3

16a (Table 1) 16b-d (Table 3)

10a, R4= H N

3

13k, R3= O B 3 HO 2 X = OH

R4-X1 10

Cl

Cl

O

1

X =NH2 10c, R4=

N H

10d, R4= Boc X1=NH2

X =NH2

16c, R4= N H

N

N H

N H

10e, R4= 1

H 2N

c O

O

X1=OH

16c', R4=

N O

Reagents and conditions: (a) ZnCl2, Et3N, 1,2-Dichloroethane, t-BuOH, 0oC-rt, 16h; (b) EtOH, MW, 100oC, 30 min; (c) trans-4-Dimethylaminocrotonic acid hydrochloride, HATU, DIPEA, DMF, rt, 1h.

2.3. Biological activity against BTK Based on the molecular docking of compound 14a with BTK, We first conducted a preliminary modification at the three positions of R1, R2, and X2 of structure A (Table 2, structure A). We replaced the trifluoromethyl group (R2=CF3) with other hydrophobic groups such as F, Cl, and Me. The results showed that the activity of compound 14a with a trifluoromethyl group was the best, and the activity of other comppounds (14b, 14c, 14d) with F, Cl and Me group was slightly decreased (compound 14a→ 14b, 14c, 14d). Our results indicate that the R2 group may be located in a conservative hydrophobic pocket, which is consistant with the result of the

molecular docking. After replacing the NH at the X2 position of structure A with an oxygen atom (compound 14a→16a), we found that the activity of compound 16a was basically lost. This indicates that NH at this position acts as a hydrogen bond donor required for keeping the activity, which verifies the result of the docking (Figure 4). Introduction of a methoxy group (R1=OMe) resulted in the loss of activity, probably because the steric hindrance of methoxy group prevented the compound 14e from entering the covalent binding capsule of protein, thus losing the covalent binding effect.

Table 2 The inhibitory activities of compound 14a-e and 16a against BTK R1

O N H

NH

B HO

Compounds

R1

R2

R2

N

O X2

N

A

X2

IC50 (nM) a BTK

a

14ab

H

CF3

NH

2.9

14b

H

F

NH

8.5

14c

H

Cl

NH

4.7

14d

H

Me

NH

6.1

14e

OMe

CF3

NH

>1000

16a

H

CF3

O

>1000

IC50 stands for 50% inhibition concentration. Dose-response curves were determined at five concentrations.

b14a

=9

Based on the structure-activity relationship (SAR) results from Table 1, we then fixed R1 group as hydrogen, R2 as trifluoromethyl and X2 as NH, and only examined the effect of R3 on the activity (Table 3, structure B). We tried many different types of substituents, including the boron-containing moieties and simple substituted-benzenes. First of all, the study on compounds 14a, 14f-14h with a boron atom at different positions of the benzene ring shows that the potency of compound 14h with boron substitution at 4-position is better than one at 3-position (compound 14a) and

significantly better than those at 2 and 5 positions (14f, 14g). Modifications of the fivemembered ring to the six-membered ring (14i) and dimethyl-substituted five-membered ring (14j) maintained the activity. However, the introduction of a fluoro group into the benzoxoborole at 6-position resulted in the significant decrease of the activity. The boron-containing heterocycle was opened to obtain simple boronic acid derivatives (compound 14a→14l, compound 14h→14m), and as a result their activity decreased about 2 times. We further introduced a moiety of phenylhydrazine (14n) as R3 group instead of the boron-containing moieties, and found that the activity of 14n was substantially lost (Table 2, case 14n). The activity loss possibly related to a big change of the electronic property of the compound and lack of a hydrogen bond donor (R2BOH). Table 3 The inhibitory activities of compound 14a and 14f-n against BTK O N H

NH CF3

N R3 B

R3

Compounds

IC50 (nM)

N H

N

R3

Compounds

IC50 (nM) a

BTK O

14a 14f

2.9

B 3 HO

O

BTK 2.0 14j

267.8

14k

B 2 HO

14g

O B HO F O B HO

63.0

14l

HO

O B 5 HO

14h

O B 4 HO

156.2

0.6

6.5 B OH

14m

1.5 HO

B OH

14i

a

O

1.9

14n

>1000

NH

B OH

IC50 stands for 50% inhibition concentration. Dose-response curves were determined at five concentrations.

According to the result of molecular docking, the X1 moiety (Table 4, structure C ) did not form a hydrogen bond with the hinge region. Referring to the structure-activity relationship of the compound (AC0058) currently studied in clinical phase [28], we replaced NH with an oxygen atom (compound 14h→16b) and found that its activity decreased about 10 folds (Table 4, structure C). Furthermore, when R2 moiety was changed from an electron-withdrawing group (CF3) to an electron-donating group (OMe), the activity of the compound 14o decreased about 10 folds, too. Interestingly, once both X1 and R2 were simultaneously modified from NH and CF3 to O and OMe, respectively, the activity of the resulted compound 14p was 4-5 times higher than that of the single modified compound (16b or 14o), indicating that the two groups had a certain synergic effect.

With reference to the covalent inhibitors currently reported, such as afatinib and ibrutinib, a few warheads (Michael addition acceptor) were selected to explore the effect of R4 moiety (compounds 16c and 16d). It was found that the compounds 16c & 16d displayed much lower activity (11.8 nM and 36.8 nM, respectively) than compound 14h (0.62 nM).

Table 4 The inhibitory activities of compounds 14h, 14o-p and 16b-d against BTK R4

HO

Compounds

R2

X1 R2

N B O

N H C

R4

N

X1

IC50 (nM) a BTK

14h

CF3

O

NH

0.6

NH

8.2

O

1.7

O

6.5

NH

11.8

NH

36.8

N H

14o

OMe

O N H

14p

OMe

O N H

16b

CF3

O N H

16c

CF3

O N

16d

N H

CF3 N O

a

IC50 stands for 50% inhibition concentration. Dose-response curves were determined at five concentrations.

2.4. Biological activity against BTK and JAKs Similar to the BTK protein, the ATP pocket of the JAK3 protein also has a homologous cysteine residue that can form irreversible covalent binding to small molecule inhibitors. In addition to the separate application of BTK inhibitors, JAK3 inhibitors are also used in the field of immunotherapy. Both inhibitors of the BTK pathway and the JAK3 pathway might have synergistic effects in clinical applications [29, 30]. Therefore, we selected some compounds with better BTK inhibitory activity and tested their inhibitory activity against JAK family kinases (Table 5). From the results of the activity tests, the compounds showed potent inhibitory activity against JAK3 and most of the compounds had less than 1 nM of IC50, while the inhibitory activity against JAK1, JAK2 and TYK2 was very low. The selectivity of JAK3 over JAK1, JAK2 and TYK2 was great (>1000 folds).

We also compared our compounds with two currently marketed drugs and a testing drug in clinical studies. In terms of BTK inhibitory activity, our compounds were much

more active than CC292, but comparable to Ibrutinb. In terms of JAK3 inhibitory activity, our compounds were more active than the three positive control compounds. Overall, our compounds were a class of very potent BTK/JAK3 dual inhibitors.

Table 5 The inhibitory activitiesa of selected compounds against BTK and JAKs Compounds

a

IC50 (nM)

IC50 (nM)

IC50 (nM)

IC50 (nM)

IC50 (nM)

BTK(WT)

JAK1

JAK2

JAK3

TYK2

14a

2.9

>1000

971.6

0.5

>1000

14h

0.6

>1000

657.9

0.4

>1000

14i

1.9

>1000

>1000

1.5

>1000

14j

2.0

>1000

>1000

1.0

>1000

14m

1.5

>1000

891.5

0.3

>1000

14p

1.7

>1000

>1000

0.8

>1000

Ibrutinb

0.3

>1000

>1000

20.3

NT

CC292

9.8

>1000

533.7

8.5

NT

Tofacitinib

>100

8.3

14.3

14.1

235

IC50 stands for 50% inhibition concentration. Dose-response curves were determined at five concentrations. NT =

not test

2.5. Antiproliferative Activities against several hematopoietic cell lines To further validate the anti-proliferative activity of these compounds, the molecules with IC50 values below 5 nM were chosen to examine the effect in cancer cells. Five typical hematopoietic cell lines associated with the BTK signaling pathway were used for the cellular inhibitory activity by CCK-8 assay. The results were depicted in Table 6. For comparison, ibrutinib and CC292 were also tested as reference compounds. Significant cytotoxicity with IC50 in the μM range was determined for most of hematopoietic cell lines employed in the screening program.

In cell-based assays, both 14a and 14h showed inhibition on the proliferations of Raji,

Ramos and HEL, Jeko-1 and OCI-LY-10 cells at concentrations ranging from 0.5 to 10 μmol/L. Compound 14h with the best kinase-based activity displayed better efficacy

than CC292 and Ibrutinib in the four cells of Raji, Romas, HEL, and Jeko-1.

Table 6 Antiproliferative activitiesa of compound 14a and 14h against hematopoietic cells Compounds

a

IC50 (μM)

IC50 (μM)

IC50 (μM)

IC50 (μM)

IC50 (μM)

Raji

Romas

HEL

Jeko-1

OCI-LY10

14a

6.8

2.3

5.1

1.2

>10

14h

1.7

0.5

1.1

0.5

1-10

CC292

17.6

3.9

5.3

1.3

>10

Ibrutinb

>20

2.4

>20

3.0

1-10

IC50 stands for 50% inhibition concentration. Dose-response curves were determined at five concentrations.

2.6 Human liver microsome stability The selected compounds 14a and 14h along with two know drugs, ibrutinb and CC292 were then submitted to metabolic stability studies in human liver microsome. Our tests (Table 7) showed that the metabolic stability of the compound 14a and 14h in the liver microsome was very close (T1/2 56 vs. 60 min), however, better than CC292 (T1/2 43 min) and significantly better than ibrutinib (T1/2 5.5 min).

Table 7 Human liver microsomal stability assay of compounds 14a, 14h and the known drugs Compounds

T1/2

Clint

Remaining

(min)

(µL/min/mg)

(T=60min)

14a

55.9

24.8

45.0%

14h

58.8

23.6

47.2%

CC292

43.4

31.9

38.8%

Ibrutinb

5.5

254.2

0.04%

3. Conclusion Both BTK and JAK3 kinase are very popular targets in hematological malignancies and autoimmune diseases. We recombined two types of dominant scaffold, the 2,4disubstituted pyrimidine, and benzoxaboroles, and constructed a small library of 31 compounds. By screening 23 kinases, we found a class of compounds with a potent BTK inhibitory activity. Through structural optimization, we obtained more active compounds 14h, 14i, 14m and 14p with IC50 < 2 nM against both BTK and JAK3 kinase. Therefore, a new class of compounds may be considered as a dual inhibitor of BTK and JAK3. The cell activity screen showed that the compound 14h was more active than CC292 and Ibrutinib by 10 folds. Metabolic stability experiment in human liver microsome exhibited that the compounds 14h had better stability than the know drugs. As a dual-target inhibitor, these compounds are expected to exert better effects in hematological and immune diseases through synergy. The further pre-clinical evaluation of the compound 14h is underway.

4. Experimental section 4.1. Chemistry. All reagents were purchased from commercial sources and were used as received. Routine monitoring of reactions was performed by thin layer chromatography (TLC) using pre-coated Haiyang GF254 silica gel TLC plates. NMR spectra were recorded on a Bruker AVANCE 500 spectrometer at 500 MHz with tetramethylsilane used as an internal reference. High resolution mass spectra (HRMS) were performed on Agilent Acrrurate-Mass Q-ToF LC/MS 6520 mass spectrometer with electron spray ionization (ESI) mode. Microwave reactions were done on a CEM Discover SP. 4.1.1 Intermadiates 10a-10e & 11a-d Intermediates 10a, 10c-d, 11a-e were purchased from Nanjing Ally Chemical S&T Co., Ltd, Shanghai biochempartner Co., Ltd and Sigama-aldrich. Intermediates 10b [27] and 10e [28] were prepared according to the literatures’ methods. 4.1.2 General procedure A for preparation of intermediates 12a-g

Intermediate 10 (1.2 eq), 11 (1.0 eq), n-butanol and DIPEA (1.5 eq) were added to the reaction flask at room temperature. The resulted mixture was stirred at room temperature until the TLC plates showed the completion of the reactions. Most of the reaction solvent was evaporated. Then, ethyl acetate and water were added to the residue. The organic phase was washed with saturated brine and dried over anhydrous sodium sulfate, filtered and then evaporated to dryness. Purification by column chromatography (low, medium and high pressure preparative chromatography) gave the desired compounds. N-(3-((2-chloro-5-(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (12a) Yield 61%; white solid; 1H NMR (300 MHz, DMSO-d6) δ [ppm] : 10.22 (s, 1H), 9.57 (s, 1H), 8.58 (s, 1H), 7.79 (s, 1H), 7.49-7.52 (m, 1H), 7.33-7.38 (m, 1H), 7.13-7.16 (m, 1H), 6.40-6.49 (m, 1H), 6.23-6.29 (d, 1H), 5.75-5.78 (m, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm]: 163.7, 162.9, 158.1, 157.1 (q, J = 5.0 Hz), 139.8, 137.6, 132.2, 129.3, 127.5, 123.7 (q, J = 270.9 Hz, CF3), 121.5, 117.6, 117.2, 106.2 (q, J = 32.8 Hz, CCF3). HRMS (ESI) m/z calculated for C14H11ClF3N4O [M+H]+: 343.0573, found: 343.0566. N-(3-((2-chloro-5-fluoropyrimidin-4-yl)amino)phenyl)acrylamide(12b) Yield 93%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.20 (s, 1H), 10.02 (s, 1H), 8.31 (d, J = 3.5 Hz, 1H), 8.01 (s, 1H), 7.44 (s, 1H), 7.42 (s, 1H), 7.33 (t, J = 8.5 Hz, 1H), 6.45-6.51 (m, 1H), 6.28 (dd,J = 17.0, 1.5 Hz, 1H), 5.76 (dd, J = 10.0, 1.5 Hz, 1H); 13C NMR (126 MHz, DMSO-d6) δ 163.7, 153.4 (q, J = 3.2 Hz), 151.7 (q, J = 11.7 Hz), 145.6 (q, J = 259.4 Hz), 142.0 (q, J = 21.0 Hz), 139.8, 138.4, 132.3, 129.3, 127.4, 117.7, 116.1, 113.6. HRMS (ESI) m/z calculated for C13H10ClFN4O [M+H]+: 293.0605, found: 293.0598. N-(3-((2,5-dichloropyrimidin-4-yl)amino)phenyl)acrylamide(12c). Yield 89%; white solid; 1H-NMR (500 MHz, DMSO-d6) δ [ppm] : 10.20 (s, 1H), 9.55 (s, 1H), 8.38 (s, 1H), 7.92 (s, 1H), 7.46 (d, J = 8.0 Hz, 1H), 7.34 (t, J = 8.0 Hz, 1H), 7.28-7.30 (d, J = 8.0 Hz, 1H), 6.44-6.50 (m, 1H), 6.26-6.29 (dd, J = 17.0 Hz, 1.5 Hz, 1H), 5.77 (dd, J = 10.0 Hz, 1.5 Hz, 1H);

13C

NMR (126 MHz, DMSO-d6) δ [ppm]:

163.7, 157.8, 157.4, 155.9, 139.7, 138.1, 132.3, 129.2, 127.4, 119.6, 116.8, 115.4, 114.1. HRMS (ESI) m/z calculated for C13H11Cl2N4O [M+H]+ : 309.0310, found: 309.0305. N-(3-((2-chloro-5-methylpyrimidin-4-yl)amino)phenyl)acrylamide(12d). Yield 57%; white solid; 1H-NMR (500 MHz, DMSO-d6) δ [ppm] : 10.16 (s, 1H), 8.89

(s, 1H), 8.04 (s, 1H), 7.97 (s, 1H), 7.38 (t, J = 7.0 Hz, 2H), 7.32 (t, J = 8.0 Hz, 1H), 6.45-6.51(m, 1H), 6.28 (dd, J = 17.0, 1.5 Hz, 1H), 5.76 (dd ,J = 10.0, 1.5 Hz, 1H), 2.18 (s, 3H);

13C

NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 160.9, 157.3, 156.7,

139.6, 139.3, 132.4, 129.1, 127.3, 118.5, 115.8, 114.9, 114.4, 14.0. HRMS (ESI) m/z calculated for C14H14ClN4O [M+H]+ : 289.0856, found: 289.0845. N-(3-((2-chloro-5-methoxypyrimidin-4-yl)amino)phenyl)acrylamide(12e). Yield 81%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.15 (s, 1H), 9.28 (s, 1H), 8.01 (s, 1H), 7.94 (s, 1H), 7.48 - 7.38 (m, 2H), 7.29 (t, J = 8.1 Hz, 1H), 6.48 (dd, J = 17.0, 10.2 Hz, 1H), 6.27 (dd, J = 17.0, 2.0 Hz, 1H), 5.76 (dd, J = 10.1, 2.0 Hz, 1H), 3.95 (s, 3H); 13C NMR (126 MHz, DMSO-d6) δ 163.6, 153.1, 150.0, 140.3, 139.6, 139.1, 136.2, 132.4, 129.1, 127.3, 117.8, 115.7, 113.7, 57.0. ESI-MS: m/z 305.1 [M+H]+. N-(3-((2-chloro-5-methoxypyrimidin-4-yl)oxy)phenyl)acrylamide (12f). Yield 64%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.33 (s, 1H), 8.40 (s, 1H), 7.67 (s, 1H), 7.50 (d, J = 8.2 Hz, 1H), 7.42 (t, J = 8.1 Hz, 1H), 6.96 (ddd, J = 8.1, 2.4, 1.0 Hz, 1H), 6.44 (dd, J = 17.0, 10.1 Hz, 1H), 6.27 (dd, J = 17.0, 1.9 Hz, 1H), 5.79 (dd, J = 10.1, 1.9 Hz, 1H), 3.99 (s, 3H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] :163.8, 160.1, 152.3, 148.3, 142.3, 142.2, 140.8, 132.1, 130.5, 127.8, 117.2, 116.9, 112.7, 57.4. ESI-MS: m/z 306.1 [M+H]+. N-(3-((2-chloro-5-(trifluoromethyl)pyrimidin-4-yl)amino)-4methoxyphenyl)acrylamide (12g). Yield 16%; white solid; 1H-NMR (500 MHz, DMSO-d6) δ [ppm] : 10.14 (s, 1H), 9.15 (s, 1H), 8.56 (s, 1H), 7.78 (s, 1H), 7.56-7.58 (dd, J = 9.0, 2.5 Hz, 1H), 7.11-7.13 (d, J = 8.5 Hz, 1H), 6.41-6.46 (m, 1H), 6.22-6.26 (dd, J = 17.0, 1.5 Hz, 1H), 5.73-5.75 (dd, J = 10.0, 1.5 Hz, 1H), 3.76 (s, 3H);

13C

NMR (126 MHz, DMSO) δ [ppm] : 163.3,

163.0, 158.4, 156.8 (q, J = 5.0 Hz), 150.4, 132.6, 132.3, 127.0, 125.9, 123.8 (q, J = 270.9 Hz, CF3), 119.3, 119.2, 112.9, 105.9 (q, J = 32.8 Hz, CCF3), 56.5. HRMS (ESI) m/z calculated for C15H13ClF3N4O2 [M+H]+: 373.0679, found: 373.0743. 4.1.3 Intermadiates 13a-13k. Intermediates 13a, 13e-j were purchased from Nanjing Ally Chemical S&T Co. Ltd. Intermediates 13b, 13c and 13d were prepared according to the literatures’ methods [24, 25].

4.1.4 General procedure B for preparation of compound 14a-m, 14o-p Intermediate 12 (1 eq), substituted aniline intermediate 13 (1.2 eq), n-butanol and trifluoroacetic acid (3 eq) were placed in a flask. The resulting mixture was heated to reflux until the TLC plates monitored the completion of the reactions. Most of the reaction solvent was evaporated under reduced pressure. The pH of remaining residue was adjusted to 9 with a saturated aqueous solution of sodium bicarbonate. The basic mixture was extracted with EtOAc. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, evaporated to dryness. Purification by column chromatography (low, medium and high pressure preparative chromatography) gave the desired compounds. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide(14a) Yield 44%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.24 (s, 1H), 9.75 (brs, 1H), 9.13 (s, 1H), 8.73 (brs, 1H), 8.36 (s, 1H), 7.82 (d, J = 8.5 Hz, 2H), 7.54 (d, J = 8.5 Hz, 2H), 7.38-7.26 (m, 1H), 7.21 (d, J = 8.0 Hz, 1H), 7.05 (brs, 1H), 6.45 (dd, J = 16.9, 10.1 Hz, 1H), 6.24 (dd, J = 16.9, 2.1 Hz, 1H), 5.74 (dd, J = 10.1, 2.1 Hz, 1H), 4.88 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 161.3, 157.8, 156.2 (q, J = 5.0 Hz), 148.2, 139.6, 138.9, 138.8, 132.3, 129.1, 129.0, 127.3, 125.2 (q, J = 270.9 Hz, CF3), 123.8, 122.2, 121.5, 121.3, 117.2, 116.6, 98.3 (q, J = 32.8 Hz, CCF3), 70.1; HRMS (ESI) m/z calculated for C21H18BF3N5O3 [M+H]+: 456.1455, found: 456.1452. N-(3-((5-fluoro-2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6yl)amino)pyrimidin-4-yl)amino)phenyl)acrylamide (14b) Yield 39%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.06 (s , 1H), 9.38 (s , 1H), 9.12 (s, 1H), 9.07 (s, 1H), 8.10 (d, J =3.5 Hz, 1H), 7.94 (s, 1H), 7.87 (s, 1H), 7.83-7.82 (m, 1H), 7.62 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.27 (t, J = 8.0 Hz, 1H), 7.20 (d, J = 8.0 Hz, 1H), 6.48-6.43 (m, 1H), 6.25 (dd, J = 16.0, 1.5Hz, 1H), 5.75 (dd, J = 10.0, 1.5 Hz, 1H), 4.91 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 156.1, 150.3, 147.2, 142.1, 141.1, 140.9, 140.1, 139.6, 139.5, 132.4, 129.1, 127.2, 122.9, 121.4, 121.1, 117.5, 115.1, 113.4, 70.1; 19F NMR (471 MHz, DMSO-d6) δ [ppm] : -164.13; HRMS (ESI) m/z calculated for C20H18BFN5O3 [M+H]+: 406.1487, found: 406.1494. N-(3-((5-chloro-2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6yl)amino)pyrimidin-4-yl)amino)phenyl)acrylamide (14c) Yield 51%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.29 (s, 1H), 9.40

(s, 1H), 9.09 (s, 1H), 8.82 (s, 1H), 8.15 (s, 1H), 8.02 (s, 1H), 7.85 (dd, J = 8.0, 1.5 Hz, 1H), 7.82 (s, 1H), 7.52 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 9.0 Hz, 1H), 7.29 (t, J = 8.0 Hz, 1H), 7.15 (d, J = 8.5 Hz, 1H), 6.50 (dd, J = 17.0, 10.0 Hz, 1H), 6.25 (dd, J = 17.0, 2.0 Hz, 1H), 5.75 (dd, J = 10.0, 2.0 Hz, 1H), 4.90 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 158.3, 156.4, 154.9, 147.4, 139.7, 139.6, 139.4, 132.5, 131.0, 129.0, 127.0, 123.3, 121.5, 121.3, 118.8, 115.5, 114.7, 104.5, 70.1; HRMS (ESI) m/z calculated for C20H18BClN5O3 [M+H]+: 422.1191, found: 422.1200. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6-yl)amino)-5methylpyrimidin-4-yl)amino)phenyl)acrylamide (14d). Yield 32%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.06 (s, 1H), 9.06 (s, 1H), 8.91 (s, 1H), 8.33 (s, 1H), 7.97 (s, 1H), 7.92-7.90 (m, 2H), 7.83 (s, 1H), 7.52 (d, J = 8.0 Hz, 1H), 7.38 (d, J = 8.5 Hz, 1H), 7.27 (t, J = 8.0 Hz, 1H), 7.15 (d, J =8.5 Hz, 1H), 6.45 (dd, J = 17.0, 10.5 Hz, 1H), 6.25 (dd, J = 17.0, 2.0 Hz,1H), 5.75 (m, 1H), 4.90 (s, 2H), 2.13 (s, 3H);

13C

NMR (126 MHz, DMSO-d6) δ [ppm] : 163.5, 159.7,

158.7, 156.0, 146.6, 140.6, 140.5, 139.4, 132.5, 130.9, 128.9, 127.1, 122.8, 121.3, 120.8, 118.2, 114.7, 114.1, 106.4, 70.1, 14.0; HRMS (ESI) m/z calculated for C21H21BN5O3 [M+H]+: 402.1737, found: 402.1742. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)-4-methoxyphenyl)acrylamide (14e). Yield 54%; white solid; 1H NMR(500 MHz, DMSO-d6) δ [ppm] : 10.06 (s, 1H), 9.75 (s, 1H), 9.10 (s, 1H), 8.36 (s, 1H), 8.18 (brs, 1H), 7.98 (brs, 1H), 7.80-7.82 (dd, J = 8.5, 1.5 Hz, 1H), 7.61-7.63 (d, J = 8.5 Hz, 1H), 7.10-7.12 (d, J = 8.5 Hz, 1H), 7.05 (s, 1H), 6.40-6.45 (m, 1H), 6.20-6.24 (dd,J = 17.0, 1.5 Hz, 1H), 5.71-5.74 (dd, J = 10.0, 1.5 Hz, 1H), 4.88 (s, 2H), 3.78 (s, 3H), 3.39-3.40 (m, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.2, 161.3, 157.9, 155.9 (q, J = 5.0 Hz), 149.6, 148.0, 139.0, 132.4, 132.3, 131.0, 127.2, 126.8, 125.4 (q, J = 270.9 Hz, CF3), 123.3, 121.9, 121.3, 119.1, 118.1, 112.2, 98.1 (q, J = 32.8 Hz, CCF3), 70.1, 56.4; HRMS (ESI) m/z calculated for C22H20BF3N5O4 [M+H]+ : 486.1560, found: 486.1568. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-7-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (14f). Yield 30%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.91 (s, 1H), 10.36 (s, 1H), 9.47 (s, 1H), 8.68 (s, 1H), 7.86 (s, 1H), 7.70 (d, J = 8.0 Hz, 1H), 7.42 (t, J = 8.0 Hz, 1H), 7.29 (d, J = 8.0 Hz, 1H), 7.01-6.98 (m, 2H), 6.70 (d, J = 6.5 Hz, 1H), 6.51 (dd, J = 16.5, 10.0 Hz, 1H), 6.28 (d, J = 17.0 Hz, 1H), 5.78 (d, J = 10.5 Hz, 1H),

4.43 (d, J = 13.5 Hz, 1H), 4.10 (d, J = 13.5 Hz, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.8, 156.3, 153.3, 146.8, 146.3 (q, J = 4.9 Hz), 140.0, 139.8, 138.0, 133.3, 132.4, 129.4, 127.4, 126.9, 126.2, 123.7 (q, J = 270.9 Hz, CF3), 121.2, 117.5, 117.1, 114.3, 99.7 (q, J = 34.0 Hz, CCF3), 64.8; 19F NMR (471 MHz, DMSO-d6) δ [ppm] : -60.15; HRMS (ESI) m/z calculated for C21H18BF3N5O3 [M+H]+ : 456.1455, found: 456.1485. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-4-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (14g). Yield 30%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.20 (s, 1H), 9.18 (s, 1H), 9.09 (s, 1H), 8.65 (s, 1H), 8.34 (s, 1H), 7.82 (s, 1H), 7.64 (d, J = 7.5 Hz, 1H), 7.48 (d, J = 7.5 Hz, 1H),7.40 (d, J = 6.5 Hz, 1H), 7.24-7.21 (m, 1H), 7.14-7.13 (m, 1H), 6.50-6.44 (m, 1H), 6.25(d, J = 17.0 Hz,1H), 5.75 (d, J = 10.0 Hz,1H), 4.98 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 161.5, 157.7, 156.3 (q, J = 5.0 Hz), 146.7, 139.6, 138.9, 133.3, 132.4, 131.7, 128.9, 127.6, 127.2, 126.4, 125.9, 125.2 (q, J = 270.9 Hz, CF3), 120.8, 116.8, 116.3, 98.3 (q, J = 32.7 Hz, CCF3), 69.5; 19F NMR (471 MHz, DMSO-d6) δ [ppm] : -59.22; HRMS (ESI) m/z calculated for C21H18BF3N5O3 [M+H]+ : 456.1455, found: 456.1465. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-5-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (14h). Yield 19%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.22 (s, 1H), 9.87 (s, 1H), 8.90 (s, 1H), 8.39 (s, 1H), 7.78 (s, 1H), 7.67 (s, 1H), 7.62 (d, J = 7.0 Hz, 1H), 7.46-7.39 (m, 2H),7.33 (m, 1H),7.13 (d, J = 6.5 Hz, 1H), 6.45-6.40 (m,1H), 6.26 (d, J = 16.5 Hz, 1H), 5.75(d, J = 10.0 Hz, 1H), 4.66 (s, 2H); 13C NMR (126 MHz, DMSOd6) δ [ppm] : 163.7, 160.9, 158.3, 156.1 (q, J = 5.0 Hz), 155.4, 142.6, 139.9, 139.1, 132.2, 130.9, 129.4, 129.3, 127.5, 125.2 (q, J = 270.9 Hz, CF3), 122.5, 118.8, 118.0, 117.0, 111.3, 98.0 (q, J = 34.1 Hz, CCF3), 70.2; 19F NMR (471 MHz, DMSO-d6) δ [ppm] : -59.58; HRMS (ESI) m/z calculated for C21H18BF3N5O3 [M+H]+ : 456.1455, found: 456.1470. N-(3-((2-((1-hydroxy-3,4-dihydro-1H-benzo[c][1,2]oxaborinin-7-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (14i). Yield 86%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.14 (s, 1H), 9.51 (s, 1H), 8.67 (s, 1H), 8.33-8.29 (m, 2H), 7.76-7.74 (m, 2H), 7.58-7.53 (m, 2H), 7.31 (m, 1H), 7.20 (m, 1H), 6.86 (s, 1H), 6.44 (dd, J = 17.0, 10.5 Hz, 1H), 6.26 (dd, J = 17.0, 2.0 Hz, 1H), 5.76-5.74 (m ,1H), 4.02 (t, J = 5.9 Hz, 2H), 2.74 (t, J = 5.9 Hz, 2H); 13C

NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 161.2, 157.8, 156.2 (q, J = 5.0 Hz), 140.0, 139.6, 139.0, 137.9, 132.4, 129.1, 129.0, 127.3, 126.8, 125.3 (q, J = 269.6 Hz, CF3), 125.1, 123.4, 121.4, 117.2, 116.5, 98.0 (q, J = 34.5 Hz, CCF3), 63.8, 31.8; HRMS (ESI) m/z calculated for C22H20BF3N5O3 [M+H]+: 470.1611, found: 470.1608. N-(3-((2-((1-hydroxy-3,3-dimethyl-1,3-dihydrobenzo[c][1,2]oxaborol-6-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (14j). Yield 49%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.25 (s, 1H), 9.68 (s, 1H), 8.92 (s, 1H), 8.68 (s, 1H), 8.35 (s, 1H), 7.80 (d, J = 7.0 Hz, 2H), 7.58 (d, J = 8.0 Hz, 2H), 7.32 (t, J = 8.0 Hz,1H), 7.20 (d, J = 7.5 Hz, 1H), 7.05 (s, 1H), 6.47 (dd, J = 17.0, 10.0 Hz, 1H), 6.26 (dd, J = 17.0, 2.0 Hz, 1H), 5.74 (dd, J = 10.0, 2.0 Hz, 1H), 1.39 (s, 6H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.5, 161.2, 157.6, 156.3, 155.5 (q, J = 5.0 Hz), 139.4, 139.2, 132.4, 128.9, 127.1, 125.2 (q, J = 270.9 Hz, CF3), 123.6, 121.8, 120.8, 120.7, 120.5, 116.8, 115.8, 115.4, 98.9 (q, J = 32.0 Hz, CCF3), 82.8, 29.9 (2C); HRMS (ESI) m/z calculated for C23H22BF3N5O3 [M+H]+: 484.1768, found: 484.1768. N-(3-((2-((5-fluoro-1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (14k). Yield 31%; white solid; 1H NMR (500MHz, DMSO-d6) δ [ppm] : 10.03 (s, 1H), 9.24 (s, 1H), 9.16 (s, 1H), 8.51 (s, 1H), 8.31 (s, 1H), 7.73 - 7.63 (m, 2H), 7.36 (d, J = 6.1 Hz, 1H), 7.23 (t, J = 9.3 Hz, 2H), 7.18 - 7.09 (m, 1H), 6.43 (dd, J = 17.0, 10.1 Hz, 1H), 6.25 (dd, J = 17.0, 1.9 Hz, 1H), 5.74 (dd, J = 10.1, 1.9 Hz, 1H), 4.95 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.5, 162.3, 160.1, 158.1, 157.4, 156.4 (q, J = 5.5 Hz), 152.9, 139.3, 132.3, 129.8, 128.7, 127.2, 126.3, 126.3, 126.2, 125.2 (q, J = 270.9 Hz, CF3), 120.3, 116.1, 109.2, 98.6 (q, J = 31.5 Hz, CCF3), 69.9; HRMS (ESI) m/z calculated for C21H17BF4N5O3 [M+H]+ : 474.1361, found: 474.1348. (3-((4-((3-acrylamidophenyl)amino)-5-(trifluoromethyl)pyrimidin-2yl)amino)phenyl)boronic acid (14l) Step 1. The general procedure B was used to prepare N-(3-((2-((3-(4,4,5,5-tetramethyl1,3,2-dioxaborolan-2-yl)phenyl)amino)-5-(trifluoromethyl)pyrimidin-4yl)amino)phenyl)acrylamide (14l’). Yield 39%; yellow solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 9.60 (s, 1H), 9.65 (s, 1H), 8.74 (s, 1H), 8.37 (s, 1 H), 7.92-7.90 (m, 1H), 7.80-7.81 (m, 1H), 7.63 (s, 1H), 7.56-7.54 (m, 1H), 7.34-7.31 (m, 1H), 7.22-7.20 (m, 2H), 7.03-7.02 (m, 1H), 6.46-6.41 (m, 1H), 6.27-6.24 (m, 1 H), 5.76-5.74 (m, 1H), 1.27 (s, 12H); 13C NMR (126 MHz,

DMSO-d6) δ [ppm] :163.6, 161.1, 157.9, 156.2 (q, J = 4.8 Hz), 139.6, 139.0, 132.3, 129.0, 128.5, 128.1, 127.3, 126.3, 126.2, 125.2 (q, J = 270.9 Hz, CF3), 124.1, 123.3, 121.5, 117.3, 116.6, 98.3 (q, J = 30.4 Hz, CCF3), 84.0 (2C), 25.1 (4C). MS (ESI): m/z 526.4 [M+H]+. Step 2. To a solution of compound 14l’ (1000 mg, 1.9 mmol) in a mixed solvent of methanol (4.75 mL) and n-heptane (4.75 mL), were added isobutylboronic acid (776 mg ) and 1 M hydrogen chloride solution (2.5 mL). After the reaction mixture was stirred at room temperature for 1.5 h, a large amount of white solid precipitated from the mixture. The n-heptane layer was discarded, and the pH of methanol layer was adjusted to 7 to obtain a suspension. The solid was collected by filtration and then triturated with methanol to obtain compound 14l as a yellow solid (200 mg, yield 24%). 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.15 (s, 1H), 9.55 (s, 1H), 8.69 (s, 1H), 8.36 (s, 1 H), 7.9 (s, 1H), 7.78-7.75 (m, 2H), 7.66 (s, 1H), 7.55-7.54 (m, 1H), 7.36-7.29 (m, 1 H), 7.24 (s, 1H), 7.24 (s, 1H), 7.01 (brs, 1H), 6.47-6.42 (m, 1H), 6.286.25 (m, 1H), 5.77-5.75 (m, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 161.2, 157.9, 156.2 (q, J = 4.8 Hz), 139.6, 139.2, 139.1, 139.0, 132.3, 129.0, 128.5, 127.6, 127.3, 126.6, 125.2 (q, J = 252 Hz, CF3), 122.5, 121.5, 117.2, 116.5, 98.1 (q, J = 30.4 Hz, CCF3); HRMS (ESI) m/z calculated for C20H18BF3N5O3 [M+H]+ : 444.1455, found: 444.1457. (4-((4-((3-acrylamidophenyl)amino)-5-(trifluoromethyl)pyrimidin-2yl)amino)phenyl)boronic acid (14m). Step 1. The general procedure B was used to prepare N-(3-((2-((4-(4,4,5,5tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)amino)-5-(trifluoromethyl)pyrimidin-4yl)amino)phenyl)acrylamide (14m’). Yield 49%; yellow solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.19-10.17 (m, 1H), 9.81 (s, 1H), 8.85-8.36 (m, 1 H), 7.82-7.79 (m, 1H), 7.64-7.52 (m, 3H), 7.40-7.35 (m, 2H), 7.18-7.17 (m, 1H), 7.18-7.06 (m, 1H), 6.47-6.41 (m, 1H), 6.28-6.27 (m, 1H), 6.24-6.23 (m, 1H), 5.76-5.72 (m, 1H), 1.26 (s, 12H); 13C NMR (126 MHz, DMSO-d6) δ 163.7, 160.9, 158.1, 156.1 (q, J = 5.0 Hz), 143.1, 139.8, 138.9, 135.3 (2C), 132.3, 129.1 (2*C), 128.6, 127.2, 125.1 (q, J = 270.9 Hz, CF3), 121.9, 118.5, 117.6, 116.7, 98.4 (q, J = 30.1 Hz, CCF3), 83.7 (2C), 25.1 (4C). MS (ESI): m/z 526.3 [M+H]+. Step 2. Compound 14m was synthesized starting from 14m’ according to the method to prepare compound 14l. Yield 53%; yellow solid; 1H NMR (500 MHz, DMSO-d6) δ 10.19-10.18 (m, 1H), 9.69 (s, 1H), 8.82 (s, 1H), 8.39 (s, 1H), 7.82-7.75 (m, 1H), 7.707.60 (m, 1H), 7.59-7.53 (m, 5H), 7.40-7.37 (m, 1H), 7.23-7.22 (m, 1H), 6.47-6.42 (m, 1H), 6.28-6.24(m, 1H), 5.75-5.73(m, 1H); 13C NMR (126 MHz, DMSO-d6) δ 163.7,

161.0, 158.1, 156.1 (q, J = 5.7 Hz), 141.8, 139.8, 139.0, 134.9 (2C), 134.8, 132.3, 129.1 (2C), 127.3, 125.2 (q, J = 269.6 Hz, CF3), 122.0, 118.5, 117.5, 116.8, 98.2 (q, J = 31.1 Hz, CCF3); HRMS (ESI) m/z calculated for C20H18BF3N5O3 [M+H]+ : 444.1455, found: 444.1465. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-5-yl)amino)-5methoxypyrimidin-4-yl)amino)phenyl)acrylamide (14o). Yield 46%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.09 (s, 1H), 9.08 (s, 1H), 8.82-8.80 (m, 2H), 7.96-7.89 (m, 3H), 7.53-7.45 (m, 4H), 7.31-7.28 (m, 1H), 6.48-6.42 (m, 1H), 6.27-6.23 (m, 1H), 5.76-5.73 (m, 1H), 4.81 (s, 2H), 3.88 (s, 3H); 13C

NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 155.6, 153.9, 152.3, 144.3, 140.0,

139.6, 137.1, 135.3, 132.4, 130.9, 129.1 (2C), 127.2, 118.2, 117.6, 114.9, 114.1, 109.7, 70.2, 57.3; HRMS (ESI) m/z calculated for C21H21BN5O4 [M+H]+: 418.1687, found: 418.1711. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-5-yl)amino)-5methoxypyrimidin-4-yl)oxy)phenyl)acrylamide (14p). Yield 48%; brown solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.36 (s, 1H), 9.54 (s, 1H), 8.84 (s, 1H), 8.26 (s, 1H), 7.69 (s, 1H), 7.60-7.58 (m, 1H), 7.52-7.40 (m, 3H), 7.22-7.20 (m, 1H), 6.99-6.97 (m, 1H), 6.46-6.40 (m, 1H), 6.28-6.24 (m, 1H), 5.78-5.75 (m, 1H),4.65 (s, 2H), 3.91 (s, 3H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.8, 159.9, 155.5, 153.5, 153.3, 144.0, 143.6, 140.8, 135.7, 132.1, 130.9, 130.4, 127.8, 117.6, 117.5, 113.3, 112.8, 109.3, 100.0, 70.2, 57.9; HRMS (ESI) m/z calculated for C21H20BN4O5 [M+H]+ : 419.1527, found: 419.1528. N-(3-((2-(2-phenylhydrazinyl)-5-(trifluoromethyl)pyrimidin-4yl)amino)phenyl)acrylamide (14n). A mixture of compound 12a (171 mg, 0.5 mmol), phenylhydrazine (65 mg, 0.6 mmol) and potassium carbonate (276 mg, 2.0 mmol) in tert-butanol (10 mL) were placed in a microwave tube. After nitrogen gas was bubbled through the mixture, 2dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (12 mg, 0.025 mmol) and tris(dibenzylideneacetone)dipalladium (23 mg, 0.025 mmol ) were added to the mixture. Again, after nitrogen gas was bubbled through the mixture, the reaction was carried out at 120 oC for 30 minutes in a microwave reactor. The reaction solution was filtered through a bed of celite, and the filter cake was washed with a small amount of ethyl acetate. After the filtrate was evaporated to dryness, the residue was purified by medium pressure preparative chromatography to give compound 27 as a white solid (80 mg,

yield 38%). 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.06 (s, 1H), 9.20 (d, J = 17.4 Hz, 1H), 8.53-8.18 (m, 2H), 7.90 (s, 1H), 7.75-7.15 (m, 3H), 7.09 (t, J = 7.2 Hz, 2H), 6.97 (d, J = 3.7 Hz, 1H), 6.65 (s, 3H), 6.46 (dd, J = 16.8, 10.2 Hz, 1H), 6.28 (d, J = 16.9 Hz, 1H), 5.77 (d, J = 10.2 Hz, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 164.7, 163.6, 157.1, 156.6 (q, J = 5.0 Hz), 150.0, 138.9, 132.4, 129.1 (2C), 128.8, 127.3, 125.2 (q, J = 269.6 Hz, CF3), 123.7, 119.2, 118.7, 115.3, 114.6, 112.5 (2C), 97.7 (q, J = 34.0 Hz, CCF3); HRMS (ESI) m/z calculated for C20H18F3N6O [M+H]+ : 415.1494, found: 415.1507. 4.1.5 Preparation of Intermediates. 5-((4-chloro-5-(trifluoromethyl)pyrimidin-2-yl)amino)benzo[c][1,2]oxaborol-1(3H)ol (15b). A solution of zinc dichloride (1M, 76 mL, 76 mmol) in diethyl ether was added to a mixture of 2,4-dichloro-5-trifluoromethylpyrimidine (5.5 g, 25.4 mmol) in 1,2dichloroethane and tert-butanol (1:1, 300 ml). The resulting mixture was stirred at 0 ° C for 20 min. Intermediate 13b (4.2 g, 27.9 mmol) in a mixed solution of 1,2dichloroethane and tert-butanol (1:1, 50 ml) was slowly added to the reaction mixture. After stirring for 10 minutes, triethylamine (2.8 g, 27.9 mmol) was then slowly added dropwise. After the completion of the addition, the mixture was stirred at room temperature for 16 hours, and then poured into ethyl acetate. The organic layer was washed with water and brine, dried over anhydrous sodium sulfate, and filtered. After the filtrate was evaporated to dryness, the residue was purified by medium pressure preparative chromatography to give compound 15b as a brown solid (3.5 g, yield 42%). 1H

NMR (500 MHz, DMSO-d6) δ [ppm] : 10.83 (s, 1H), 9.07 (s, 1H), 8.84 (s, 1H),

7.84 (s, 1H), 7.70 (d, J = 8.0 Hz, 1H), 7.64 (d, J = 8.0 Hz, 1H), 4.99 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ 160.9, 158.6 (q, J = 4.8 Hz), 158.1, 155.5, 141.2, 131.4, 125.7, 123.4 (q, J = 270.9 Hz, CF3), 119.7, 112.8, 112.1 (q, J = 33.4 Hz, CCF3), 70.3. MS (ESI): m/z 330.2 [M+H]+. 4.1.6 General procedure C for preparation of compounds 16a-d. A mixture of compound 10 (1.1 eq) and 15 (1.0 eq) in ethanol was placed in a microwave tube. The reaction was carried out at 100 oC for 30 minutes in a microwave reactor. After the reaction solution was evaporated to dryness, the residue was purified by medium pressure preparative chromatography to give the desired compounds. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-6-yl)oxy)-5(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)acrylamide (16a).

Step 1. 6-((4-chloro-5-(trifluoromethyl)pyrimidin-2-yl)oxy)benzo[c][1,2]oxaborol1(3H)-ol (15a). Compound 15a was synthesized starting from intermediate 13k according to the method to prepare compound 15b. A crude product was obtained and used in the next step without further purification. Step 2. The general procedure C was used to prepare compound 16a as a yellow solid (140 mg, yield 9.4 % for two steps). 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.12 (s, 1H),9.19 (s, 1H), 9.15 (s, 1H), 8.51 (s, 1H), 7.73 (s, 1H), 7.42-7.37 (m, 3H), 7.27(d, J = 2.5 Hz, 1H), 7.13-7.07 (m, 2H), 6.47-6.42 (m, 1H), 6.29-6.25(m, 1H), 5.76 (dd, J = 10.0, 2.0 Hz, 1H), 5.00 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 166.6, 163.6, 158.8, 157.8 (q, J = 5.0 Hz), 152.0, 151.1, 139.4, 138.0, 132.4, 132.3, 128.7, 127.4, 125.1, 124.3 (q, J = 270.9 Hz, CF3), 123.4, 122.9, 120.9, 116.8, 116.6, 102.8 (q, J = 32.8 Hz, CCF3), 70.2; 19F NMR (471 MHz, DMSO-d6) δ [ppm] : -60.49; HRMS (ESI) m/z calculated for C21H17BF3N4O4 [M+H]+ : 457.1295, found: 457.1314. N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-5-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)oxy)phenyl)acrylamide (16b). Yield 85%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.44 (br, 1H), 10.40 (s, 1H), 8.97 (br, 1H), 8.74 (s, 1H), 7.76 (s, 1H), 7.62-7.60 (d, J = 8.0 Hz, 1H), 7.537.48 (m, 3H), 7.27 (s, 1H), 7.04 (d, J = 8.0 Hz, 1H), 6.46-6.41 (dd, J = 17.0, 10.0 Hz, 1H), 6.28-6.25 (d, J = 17.0 Hz, 1H), 5.79-5.77 (d, J = 10.0 Hz, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 166.8, 163.9, 162.8, 161.3, 158.3 (q, J = 5.0 Hz), 155.3, 152.5, 141.7, 140.9, 132.0, 131.0, 130.6, 127.9, 124.5, 124.0 (q, J = 269.6 Hz, CF3), 119.0, 117.6, 117.4, 113.2, 101.0 (q, J = 32.8 Hz, CCF3), 70.2; HRMS (ESI) m/z calculated for C21H17BF3N4O4 [M+H]+ : 457.1295, found: 457.1327. (E)-4-(dimethylamino)-N-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-5yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)phenyl)but-2-enamide (16c). Step 1: The general procedure C was used to prepare 5-((4-((3-aminophenyl)amino)-5(trifluoromethyl)pyrimidin-2-yl)amino)benzo[c][1,2]oxaborol-1(3H)-ol (16c’). Yield 70%; grey solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.01 (s, 1H), 8.94 (s, 1H), 8.43 (s, 1H), 7.78-7.65 (m, 1H), 7.54-7.48 (m, 1H), 7.48-7.26 (m, 3H), 7.10 (s, 1H), 4.77 (s, 2H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 167.0, 160.6, 157.8, 156.0 (q, J = 5.0 Hz), 155.5, 147.6, 142.5, 139.9, 135.6, 131.0, 130.0, 125.1 (q, J = 269.6 Hz, CF3), 118.8, 112.4, 111.4, 100.0, 97.3 (q, J = 32.0 Hz, CCF3), 70.3. MS (ESI): m/z 402.2 [M+H]+. Step 2: A mixture of compound 16c' (324 mg, 0.81 mmol) and trans-4-dimethylamino

crotonate (174 mg, 1.1 mmol) in DMF (5 mL), DIPEA (423 ul, 2.4 mmol) and HATU (461 mg, 1.2 mmol) were stirred for 1 hour at room temperature. The reaction mixture was diluted with ethyl acetate (20 mL). The organic phase was washed with water, saturated brine, dried over anhydrous sodium sulfate, and filtered. After the filtrate was evaporated to dryness, the residue was purified by medium pressure preparative chromatography to give the desired compound 16c as a white solid (140 mg, yield 34%). 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 10.27 (s, 1H), 9.90 (s, 1H), 8.91-8.87 (m, 2H), 8.39 (s, 1H), 7.83 (s, 1H), 7.68-7.62 (m, 2H), 7.47-7.45 (m, 1H), 7.40-7.35 (m, 2H), 7.12-7.10 (m, 1H), 6.75-6.69 (m, 1H), 6.34-6.30 (m, 1H), 4.67 (s, 2H), 3.17 (s, 2H) , 2.25 (s, 6H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 163.6, 160.9, 158.2, 156.0 (q, J = 5.0 Hz), 155.3, 142.6, 140.6, 140.1, 139.1, 130.9, 130.1, 129.2, 127.3, 125.2 (q, J = 269.6 Hz, CF3), 123.5, 122.0, 118.8, 116.8, 111.3, 98.3 (q, J = 32.0 Hz, CCF3), 70.2, 59.8, 45.1 (2*C); HRMS (ESI) m/z calculated for C24H25BF3N6O3 [M+H]+ : 513.2033, found: 513.2044. 1-(3-((2-((1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborol-5-yl)amino)-5(trifluoromethyl)pyrimidin-4-yl)amino)piperidin-1-yl)prop-2-en-1-one (16d). Yield 35%; white solid; 1H NMR (500 MHz, DMSO-d6) δ [ppm] : 9.85 (s, 1H), 8.978.94 (m, 1H), 8.27 (s, 1H), 7.93-7.85 (m, 1H), 7.59-7.52 (m, 2H), 6.89-6.63 (m, 2H), 6.16-5.97 (m, 1H), 5.74-5.53 (m, 1H), 4.86 (s, 2H), 4.32-4.26 (m, 2H), 3.97 (d, J =11.0 Hz, 1H), 3.25-2.74 (m, 2H), 1.94 (s, 1H), 1.85-1.76 (m, 2H), 1.44 (d, J = 9.0 Hz, 1H); 13C NMR (126 MHz, DMSO-d6) δ [ppm] : 165.2, 161.2, 158.0, 155.4 (q, J = 5.0 Hz), 142.7, 131.1, 128.9, 128.0, 127.5, 125.3 (q, J = 269.6 Hz, CF3), 119.1, 111.8, 111.7, 97.9 (q, J = 32.0 Hz, CCF3), 70.4, 49.5, 47.5, 46.0, 25.4, 24.3; HRMS (ESI) m/z calculated for C20H22BF3N5O3 [M+H]+ : 448.1768, found: 448.1801.

4.2 Biological studies 4.2.1. Kinase enzymatic assay. Recombinant histidine-tagged human BTK enzymes was obtained from Drug Discovery Support Business Division of ThermoFisher Scientific, Inc. 4.2.2.Measurment of BTK inhibitory activity. Measurement of BTK activity was carried out using LANCE Ultra KinaSelectTM Kit (LANCE Ultra KinaSelectTM Kit, PerkinElmer, Inc.). Firstly, we optimized the LanthaScreen™kinase assay for BTK (PerkinElmer, USA) according to the manufacture's specifications. TR-FRET assays were performed by incubating a dilution

series of compound concentrations with ATP (Sigma), LANCE Ultra ULight-Poly GT substrate (PerkinElmer) and BTK Kinase (ThermoFisher Scientific) in kinase reaction buffer (PerkinElmer).The kinase reaction buffer consisted of 50 mM HEPES pH 7.5, 10 mM MgCl2, 2mM DTT, 1 mM EGTA and 0.01% Tween 20. The kinase reaction mixtures were incubated at room temperature (23±2℃) for 2h before stopping the kinase reaction by the addition of 10 mM EDTA . The phosphorylation of the substrate by BTK was detected using Eu-PT66 antibody (PerkinElmer) in TR-FRET LANCETM Detection Buffer at pH 7.5 (PerkinElmer) and then incubated at room temperature (23±2℃) for 1h, finally measured by determining the emission ratio of 665/615 nm on a microplate reader (EnVison, PerkinElmer). IC50 was estimated using the log(inhibitor) vs. response non-linear fit (GraphPad Prism 6.0). Additional assays were similarly carried out to determine selectivity over JAK1/2/3 and TYK2 (Carna Biosciences, Inc). 4.2.3. Cell viability assay 4.2.3.1 Effect of compound on Raji cell viability The Cell Counting Kit-8 (CCK-8) assay was used to assess cell viability after treatment with different doses of compound. Cells were collected and counted with Cell Counter, and then they were seeded in 96-well plates at 8 x 104cells/well and allowed to incubate overnight. Then the compounds were added with Nanoliter pipetting instrument. After another 72 hours, ten microliters of CCK-8 solution were added to each culture well and incubated for 2 hours. The absorbance (OD) of each group was measured with a multifunctional microplate reader (Envision) at a wavelength of 450 nm. The proliferation of Raji cells was observed. Cell proliferation inhibition rate (%) = (control group OD value - experimental group OD value)/(control group OD value blank group OD value) × 100% and calculated IC50 with Graphpad Prism. 4.2.3.2 Effect of compound on Ramos cell viability The Cell Counting Kit-8 (CCK-8) assay was used to assess cell viability after treatment with different doses of compound. Cells were collected and counted with Cell Counter, and then they were seeded in 96-well plates at 5 x104cells/well and allowed to incubate overnight. Then the compounds were added with Nanoliter pipetting instrument. After another 72 hours, ten microliters of CCK-8 reagents was added to

each culture well and incubated for 2 hours. The absorbance (OD) of each group was measured with a multifunctional microplate reader (Envision) at a wavelength of 450 nm. The proliferation of Ramos cells was observed. Cell proliferation inhibition rate (%) = (control group OD value - experimental group OD value)/(control group OD value - blank group OD value) × 100% and calculated IC50 with Graphpad Prism. 4.2.3.3 Effect of compound on HEL cell viability The Cell Counting Kit-8 (CCK-8) assay was used to assess cell viability after treatment with different doses of compound. Cells were collected and counted with Cell Counter, and then they were seeded in 96-well plates at 7.5 × 104cells/well and allowed to incubate overnight. Then the compounds were added with Nanoliter pipetting instrument. After another 72 hours, ten microliters of CCK-8 reagents were added to each culture well and incubated for 2 hours. The absorbance (OD) of each group was measured with a multifunctional microplate reader (Envision) at a wavelength of 450 nm. The proliferation of HEL cells was observed. Cell proliferation inhibition rate (%) = (control group OD value - experimental group OD value)/(control group OD value blank group OD value) × 100% and calculated IC50 with Graphpad Prism. 4.2.3.4 Effect of compound on Jeko-1 cell viability The Cell Counting Kit-8 (CCK-8) assay was used to assess cell viability after treatment with different doses of compound. Cells were collected and counted with Cell Counter, and then they were seeded in 96-well plates at 7.5 × 104cells/well and allowed to incubate overnight. Then the compounds were added with Nanoliter pipetting instrument. After another 72 hours, ten microliters of CCK-8 solution was added to each culture well and incubated for 2 hours. The absorbance (OD) of each group was measured with a multifunctional microplate reader (Envision) at a wavelength of 450 nm. The proliferation of Jeko-1 cells was observed. Cell proliferation inhibition rate (%) = (control group OD value - experimental group OD value)/(control group OD value blank group OD value) × 100% and calculated IC50 with Graphpad Prism. 4.2.3.5 Effect of compound on OCI-LY10 cell viability Diffuse large B-cell lymphoma cells OCI-LY10 was cultured and stored in liquid nitrogen Department of Cancer and Endocrinology, College of Pharmacy, Zhejiang

University. Cells were collected and counted with Cell Counter, and then they were seeded in 96-well plates and allowed to incubate overnight. Then the compounds were treated with a dose range from 0.0001 μM - 10μM. After another 72 hours, the proliferation of OCI-LY10 in vitro was measured by Celltiter. The absorbance (OD) of each group was measured with a multifunctional microplate reader (Envision) at a wavelength of 450 nm. Cell proliferation inhibition rate (%) = (control group OD value - experimental group OD value)/(control group OD value - blank group OD value) ×100% and calculated IC50 with Graphpad Prism. 4.2.4. Human liver microsome stability test Human liver microsomes were obtained from BD Gentest. A typical standard reaction mixture 300 µL consisted of the pooled liver microsomes 0.2 mg/mL, 1 mM NADPH, 5 mM MgCl2, 100 mM potassium phosphate buffer (pH 7.4) and 0.2 µM of test compounds. After a 5-min pre-incubation at 37 °C, the reactions were initiated by addition of NADPH and incubation proceeded for 5, 15, 30, 60 min at 37 °C in a shaking metal bath. The reaction was stopped by transferring 60 µL aliquots to the tubes on ice and adding 120 µL amounts of ice-cold acetonitrile containing internal standards. Concentration of the test compounds was measured by UPLC-MS/MS. 4.3 Molecular docking Molecular docking study was performed by using Molecular Operating Environment (MOE v2018.01). Receptors [BTK (PDB ID: 5P9J) and JAK3 (PDB ID: 5TOZ)] were prepared through QuickPrep protocol in MOE. Ligands were protonated at pH 7 followed by conformation searching. The lowest energy conformation of each Ligand was maintained and covalently docked into the ATP binding site of receptor through specified Cysteine (Cys481 for BTK and Cys356 for JAK3). The GBVI/WSA dG score was selected to evaluate docking poses. The docking models were analyzed and generated by MOE. Acknowledgements The authors would like to thank Jie Wang, Yuhan Li and Jianghao Ma for their generous help on Mass and NMR analysis. Conflict of interest None of the authors of the above manuscript has declared any conflict of interest

which may arise from being named as an author on the manuscript. Author contributions Dr. Y. Zhang and Mr. H. Xu conceived and planned the experiments. J. Ren, D. Zhao, Q. Wang and Y. Gao carried out the synthetic experiments. W. Shi, X. Chang, X. He and X. Wang performed biological experiments. Dr. P. Lu planned and carried out the computational simulations. Dr. Y. Zhang and Dr. X. Zhang contributed to the interpretation of the results. Dr. Y. Zhang took the lead in writing the manuscript. All authors provided critical feedback and helped shape the research, analysis and manuscript. Reference [1] A.J. Mohamed, L. Yu, C.M. Bäckesjö, L. Vargas, R. Faryal, A. Aints, B. Christensson, A. Berglöf, M. Vihinen, B.F. Nore, Bruton’s tyrosine kinase (Btk): function, regulation, and transformation with special emphasis on the PH domain, Immunological reviews, 228 (2009) 5873. [2] A. Mohamed, B. Nore, B. Christensson, C. Smith, Signalling of Bruton's tyrosine kinase, Btk, Scandinavian journal of immunology, 49 (1999) 113-118. [3] J. Jongstra-Bilen, A.P. Cano, M. Hasija, H. Xiao, C.E. Smith, M.I. Cybulsky, Dual functions of Bruton’s tyrosine kinase and tec kinase during Fcγ receptor-induced signaling and phagocytosis, The Journal of Immunology, 181 (2008) 288-298. [4] H.S. Kuehn, M. Rådinger, J.M. Brown, K. Ali, B. Vanhaesebroeck, M.A. Beaven, D.D. Metcalfe, A.M. Gilfillan, Btk-dependent Rac activation and actin rearrangement following FcεRI aggregation promotes enhanced chemotactic responses of mast cells, J Cell Sci, 123 (2010) 2576-2585. [5] S. Tsukada, D.J. Rawlings, O.N. Witte, Role of Bruton's tyrosine kinase in immunodeficiency, Current opinion in immunology, 6 (1994) 623-630. [6] P.J. Mease, B cell-targeted therapy in autoimmune disease: rationale, mechanisms, and clinical application, The Journal of rheumatology, 35 (2008) 1245-1255. [7] A. Sarvaria, J.A. Madrigal, A. Saudemont, B cell regulation in cancer and anti-tumor immunity, Cellular & Molecular Immunology, 14 (2017) 662. [8] J.C. Byrd, R.R. Furman, S.E. Coutre, I.W. Flinn, J.A. Burger, K.A. Blum, B. Grant, J.P. Sharman, M. Coleman, W.G. Wierda, Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia, New England Journal of Medicine, 369 (2013) 32-42.

[9] R.H. Advani, J.J. Buggy, J.P. Sharman, S.M. Smith, T.E. Boyd, B. Grant, K.S. Kolibaba, R.R. Furman, S. Rodriguez, B.Y. Chang, Bruton tyrosine kinase inhibitor ibrutinib (PCI-32765) has significant activity in patients with relapsed/refractory B-cell malignancies, Journal of clinical oncology: official journal of the American Society of Clinical Oncology, 31 (2013) 88-94. [10] J. Wu, M. Zhang, D. Liu, Acalabrutinib (ACP-196): a selective second-generation BTK inhibitor, Journal of hematology & oncology, 9 (2016) 21. [11] T. Covey, T. Barf, M. Gulrajani, F. Krantz, B. van Lith, E. Bibikova, B. van de Kar, E. de Zwart, A. Hamdy, R. Izumi, ACP-196: a novel covalent Bruton's tyrosine kinase (Btk) inhibitor with improved selectivity and in vivo target coverage in chronic lymphocytic leukemia (CLL) patients, in, AACR, 2015. [12] J.J. O'Shea, D.M. Schwartz, A.V. Villarino, M. Gadina, I.B. McInnes, A. Laurence, The JAKSTAT pathway: impact on human disease and therapeutic intervention, Annual review of medicine, 66 (2015) 311-328. [13] A.J. Steele, A.G. Prentice, K. Cwynarski, A.V. Hoffbrand, S.M. Hart, M.W. Lowdell, E.R. Samuel, R.G. Wickremasinghe, The JAK3-selective inhibitor PF-956980 reverses the resistance to cytotoxic agents induced by interleukin-4 treatment of chronic lymphocytic leukemia cells: potential for reversal of cytoprotection by the microenvironment, Blood, 116 (2010) 4569-4577. [14] J. Nakayama, M. Yamamoto, K. Hayashi, H. Satoh, K. Bundo, M. Kubo, R. Goitsuka, M.A. Farrar, D. Kitamura, BLNK suppresses pre–B-cell leukemogenesis through inhibition of JAK3, Blood, 113 (2009) 1483-1492. [15] S. Qazi, F.M. Uckun, Gene expression profiles of infant acute lymphoblastic leukaemia and its prognostically distinct subsets, British journal of haematology, 149 (2010) 865-873. [16] J.B. Telliez, M.E. Dowty, L. Wang, J. Jussif, T. Lin, L. Li, E. Moy, P. Balbo, W. Li, Y. Zhao, Discovery of a JAK3-selective inhibitor: functional differentiation of JAK3-selective inhibition over pan-JAK or JAK1-selective inhibition, ACS chemical biology, 11 (2016) 3442-3451. [17] H.S. Ban, H. Nakamura, Boron-Based Drug Design, The Chemical Record, 15 (2015) 616-635. [18] T. Akama, S.J. Baker, Y.K. Zhang, V. Hernandez, H. Zhou, V. Sanders, Y. Freund, R. Kimura, K.R. Maples, J.J. Plattner, Discovery and structure–activity study of a novel benzoxaborole antiinflammatory agent (AN2728) for the potential topical treatment of psoriasis and atopic dermatitis, Bioorganic & medicinal chemistry letters, 19 (2009) 2129-2132.

[19] X. Li, V. Hernandez, F.L. Rock, W. Choi, Y.S.L. Mak, M. Mohan, W. Mao, Y. Zhou, E.E. Easom, J.J. Plattner, W. Zou, E. Pérez-Herrán, I. Giordano, A. Mendoza-Losana, C. Alemparte, J. Rullas, I. Angulo-Barturen, S. Crouch, F. Ortega, D. Barros, M.R.K. Alley, Discovery of a Potent and Specific M. tuberculosis Leucyl-tRNA Synthetase Inhibitor: (S)-3-(Aminomethyl)-4-chloro-7(2-hydroxyethoxy)benzo[c][1,2]oxaborol-1(3H)-ol (GSK656), Journal of Medicinal Chemistry, 60 (2017) 8011-8026. [20] Y.-K. Zhang, J.J. Plattner, E.E. Easom, R.T. Jacobs, D. Guo, Y.R. Freund, P. Berry, V. Ciaravino, J.C.L. Erve, P.J. Rosenthal, B. Campo, F.-J. Gamo, L.M. Sanz, J. Cao, Benzoxaborole Antimalarial Agents. Part 5. Lead Optimization of Novel Amide Pyrazinyloxy Benzoxaboroles and Identification of a Preclinical Candidate, Journal of Medicinal Chemistry, 60 (2017) 5889-5908. [21] R.T. Jacobs, C.S. Lunde, Y.R. Freund, Boron-Pleuromutilins as Anti-Wolbachia Agents with Potential for Treatment of Onchocerciasis and Lymphatic Filariasis. Journal of Medicinal Chemistry, 62 ( 2019) 2521-2540 [22] D. Ding, Y. Zhao, Q. Meng, D. Xie, B. Nare, D. Chen, C.J. Bacchi, N. Yarlett, Y.-K. Zhang, V. Hernandez, Y. Xia, Y. Freund, M. Abdulla, K.-H. Ang, J. Ratnam, J.H. McKerrow, R.T. Jacobs, H. Zhou, J.J. Plattner, Discovery of Novel Benzoxaborole-Based Potent Antitrypanosomal Agents, ACS Medicinal Chemistry Letters, 1 (2010) 165-169. [23] J.W. Tomsho, A. Pal, D.G. Hall, S.J. Benkovic, Ring Structure and Aromatic Substituent Effects on the pKa of the Benzoxaborole Pharmacophore, ACS Medicinal Chemistry Letters, 3 (2012) 48-52. [24] X. Li, S. Zhang, Y.-K. Zhang, Y. Liu, C.Z. Ding, Y. Zhou, J.J. Plattner, S.J. Baker, W. Bu, L. Liu, Synthesis and SAR of acyclic HCV NS3 protease inhibitors with novel P4-benzoxaborole moieties, Bioorganic & medicinal chemistry letters, 21 (2011) 2048-2054. [25] Y.K. Zhang, J.J. Plattner, Y.R. Freund, E.E. Easom, Y. Zhou, J. Gut, P.J. Rosenthal, D. Waterson, F.-J. Gamo, I. Angulo-Barturen, M. Ge, Z. Li, L. Li, Y. Jian, H. Cui, H. Wang, J. Yang, Synthesis and structure–activity relationships of novel benzoxaboroles as a new class of antimalarial agents, Bioorganic & Medicinal Chemistry Letters, 21 (2011) 644-651. [26] Y. Zhang, Y. Gao, J. Ren, Q. Wang, Z. Wang, 2,4-Disubstituted pyrimidine compounds as EGFR or/and ALK inhibitors and their preparation, pharmaceutical compositions and use in the treatment of diseases, CN106905245, 2015.

[27] X. Wang, C. Wu, Y. Xu, C. Shen, L. Li, G. Hu, Y. Yue, J. Li, D. Guo, N. Shi,L. Huang, S. Chen, R. Tu, Z. Yang, X. Zhang, Q. Xiao, H. Tian, Y. Yu, H. Chen, W. Sun, Z. He, J. Shen, J. Yang, J. Tang, W. Zhou, J. Yu, Y. Zhang, Q. Liu, WO2016112637, 2015. [28] X. Xu, X. Wang, L. Mao, L. Zhao, B. Xi, Novel pyrrolopyrimidine compounds as inhibitors of protein kinases, WO2014025486, 2014. [29] Y. Ge, C. Wang, S. Song, J. Huang, Z. Liu, Y. Li, Q. Meng, J. Zhang, J. Yao, K. Liu, X. Ma, X. Sun. Identification of highly potent BTK and JAK3 dual inhibitors with improved activity for the treatment of B-cell lymphoma. European Journal of Medicinal Chemistry, 143 (2018) 18471857. [30] S. A. Ezell, M. Mayo, T. Bihani, S. Tepsuporn, S. Wang, M. Passino, S. E. Grosskurth, M. Collins, J. Parmentier, C. Reimer and K. F. Byth. Synergistic induction of apoptosis by combination of BTK and dual mTORC1/2 inhibitors in diffuse large B cell lymphoma. Oncotarget, 5 (2015) 4990-5001.

Abbreviations: ATP, adenosine triphosphate; HATU, O-(7-aza-1H-benzotriazol-1-yl)N,N,N′,N′-tetramethyluronium hexafluorophosphate; DIPEA, N,Ndiisopropylethylamine; DME, 1,2-dimethoxyethane; DMAP, N,Ndimethylaminopyridine; DMF, N,N-dimethylformamide; DMSO, dimethylsulfoxide; THF, tetrahydrofuran; TFA, trifluoroacetic acid.

Graphical abstract Design and Synthesis of Boron-containing Diphenylpyrimidines as Potent BTK and JAK3 Dual Inhibitors Jing Ren, Wei Shi, Damin Zhao, Qinglin Wang, Xiayun Chang, Xiangyi He, Xiaojin Wang, Yong Gao, Peng Lu, Xiquan Zhang, Hongjiang Xu*, Yinsheng Zhang* Institute for Innovative Drug Discovery Chia Tai Tianqing Pharmaceutical Group Co., Ltd 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China R1 N R2

O N

2, 4-substituted pyrimidine

N H

O B OH

benzoxaboroles

NH

HO

B O

+ R3

dominant pharmacophores re-combination

CF3

N N H

N

14h BTK IC50= 0.6 nM JAK3 IC50= 0.4 nM

Highlights  A dominant pharmacophore re-combination strategy was successfully used to design kinases inhibitors.  A series of boron-containing compounds were synthesized, and some of them exhibited significant inhibitory activity against BTK and JAK3 with both IC50 values below 1 nM.  Several compounds have shown 10-folds more active in the enzyme and cellbased assays than a known active compound, ibrutinib and CC292.

Conflict of interest

None of the authors of the above manuscript has declared any conflict of interest which may arise from being named as an author on the manuscript.

Yinsheng Zhang, Ph.D.