Remibrutinib

Discovery of LOU064 (Remibrutinib), a Potent and Highly Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase

Daniela Angst, François Gessier, Philipp Janser, Anna Vulpetti, Rudolf Wälchli, Christian Beerli, Amanda Littlewood-Evans, Janet Dawson, Barbara Nuesslein-

Hildesheim, Grazyna Wieczorek, Sascha Gutmann, Clemens Scheufler, Alexandra Hinniger, Alfred Gilbert Zimmerlin, Enrico G. Funhoff, Robert Pulz, and Bruno Cenni
J. Med. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jmedchem.9b01916 • Publication Date (Web): 21 Feb 2020 Downloaded from pubs.acs.org on February 22, 2020

Just Accepted

“Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

 

48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

 
Discovery of LOU064 (Remibrutinib), a Potent and Highly Selective Covalent Inhibitor of Bruton’s Tyrosine Kinase

Daniela Angst*a, François Gessiera, Philipp Jansera, Anna Vulpettia, Rudolf Wälchlia, Christian Beerlib, Amanda Littlewood-Evansb, Janet Dawsonb, Barbara Nuesslein-Hildesheimb, Grazyna
Wieczorekb, Sascha Gutmannc, Clemens Scheuflerc, Alexandra Hinnigerc, Alfred Zimmerlind, Enrico G. Funhoffe, Robert Pulza, Bruno Cennib

aGlobal Discovery Chemistry, bAutoimmunity, Transplantation & Inflammation, cChemical Biology & Therapeutics, dPK Sciences, ePreclinical Safety, Novartis Institutes for BioMedical
Research, Novartis Campus, CH-4002 Basel, Switzerland

 

 

 

Abstract. Bruton’s tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a central role in immunity and is considered an attractive target for treating autoimmune diseases. The use of currently marketed covalent BTK inhibitors is limited to oncology indications based on their suboptimal kinase selectivity. We describe the discovery and preclinical profile of LOU064 (remibrutinib, 25), a potent, highly selective covalent BTK inhibitor. LOU064 exhibits an exquisite kinase selectivity due to binding to an inactive conformation of BTK and has the potential for a best-in-class covalent BTK inhibitor for the treatment of autoimmune diseases. It

ACS Paragon Plus Environment

demonstrates potent in vivo target occupancy with an EC90 of 1.6 mg/kg and dose-dependent efficacy in rat collagen-induced arthritis. LOU064 is currently being tested in Phase 2 clinical studies for chronic spontaneous urticaria and Sjoegren’s Syndrome.

 

 

INTRODUCTION
Bruton’s tyrosine kinase (BTK) is a cytoplasmic tyrosine kinase selectively expressed in a subset of immune cells, including macrophages, mast cells, basophils, platelets and B cells, but not in mature plasma cells and T cells.1,2 BTK is activated by membrane recruitment via its pleckstrin homology domain and phosphorylation of the activation loop Tyr551 by SYK and SRC family kinases, resulting in autophosphorylation of Tyr223 in the SH3 domain.1,2 BTK is an essential node and rheostat for the signaling of the antigen receptor in B cells (BCR) and of activating Fc receptors for IgG and IgE (FcγR, FcεR) in macrophages and mast cells.3–5 While inborn BTK deficiency does not affect the development of many immune cells, it results in severely impaired B cell development and antibody production.6–8 In humans BTK deficiency is the most frequent cause of the primary immunodeficiency X-linked agammaglobulinemia (XLA), a condition that if untreated is associated with an increased bacterial infection risk.9 Today XLA is treated by immunoglobulin replacement therapy suggesting that the main risk factor for infections is the absence of protective immunoglobulins rather than the deficiency in cellular BTK function.10
The role of BTK appears not to be critical for B cell survival once the B cells have developed, suggesting that BTK inhibition after infancy may not lead to acute B cell and antibody depletion.6

 

 

ACS Paragon Plus Environment

 

The potential role of BTK in autoimmunity, allergy and chronic inflammatory conditions is supported by genetic validation in many rodent models for arthritis, systemic lupus erythematosus and anaphylaxis.3,11–13 More recently, the functional relevance of BTK has also been validated by pharmacological inhibition in various clinical trials and is testament for the activities across many pharmaceutical companies to identify and develop BTK inhibitors, both covalent and reversible.14–22

The kinase domain of BTK contains Cys481 in a position conserved in a group of eleven kinases including the closely related TEC family kinases BMX, TEC, ITK and TXK, as well as in
EGFR, ERBB2, ERBB4, JAK3, BLK and MKK7.23,24 These conserved cysteines have been targeted in the development of covalent inhibitors of BTK, EGFR, JAK3 and MKK7. The concept of selective targeted covalent kinase inhibitors has received more attention due its potential for sustained target inhibition, uncoupled from sustained and high systemic drug exposure. This special pharmacologic feature of covalent inhibitors may lead to high efficacy and an improved safety profile.25,26

The covalent BTK inhibitors ibrutinib (1) and acalabrutinib (2) (Figure 1, a)) have shown efficacy and an attractive risk-benefit profile leading to their approval in several B cell malignancies including chronic lymphocytic leukemia and mantle cell lymphoma.27 Due to its binding to BTK in its active conformation, ibrutinib (1) not only potently inhibits BTK, but irreversibly inhibits all kinases which carry a Cys at the same position as BTK, and in addition inhibits reversibly several other kinases.14,28,29 While the clinical efficacy of 1 is of significant benefit in difficult to treat oncologic indications, its side effect profile precludes its use in rheumatologic indications.30–34 It is likely that its off-target activity is associated with the clinical risk for skin rash and diarrhea (well established for EGFR inhibitors35), or platelet disfunction

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14

 

 

(shown for TEC and SRC kinases36,37). Second generation clinical-stage covalent inhibitors such as acalabrutinib (2), tirabrutinib (3), evobrutinib (4) and branebrutinib (5) (Figure 1, a)) retain a similar binding mode to BTK as 1 and therefore, despite offering an overall improved kinase selectivity profile, still inhibit several Cys-containing kinases.18,22,38–40

a)

15
16
O

O

NH
N
O

17
18
19
20
21
22

 

N

NH2
N

 

N
N

 
N
NH2

N

 

 

N

 

 

O
N
NH2
N
N

N

 

O

23 N N
N

24
O O

25
26
27
28
29
30
1, ibrutinib
2, acalabrutinib
3, tirabrutinib

 
b)

31
32

NH2
O

H2N
F
O

33
34
35
36
37
38
39
N

N N H
4, evobrutinib

 

N

 

 

O

O
HN

 

5, branebrutinib
N
O
NH

 

 

N

 

 

 

O
NH

 

 

N
H
O

 
N

 

 

O

40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
6, CGI-1746
Figure 1. a) Selected examples of covalent BTK inhibitors (1-5); b) Structure of CGI-1746 (6), the first reported compound binding to an inactive BTK conformation.

 
Reversible BTK inhibitors with high kinase selectivity have been described15,16,21,41 and

CGI-1746 (6) (Figure 1, b)) was the first compound reported to bind to an inactive conformation

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

of BTK resulting in an excellent kinase selectivity. This binding mode prevents the phosphorylation of Tyr551in the activation loop of BTK by upstream kinases and possibly contributes to higher cellular potency.42 In contrast to covalent inhibitors, reversible inhibitors require continuous systemic drug exposure over the entire dosing interval in order to maintain a high degree of target inhibition. In order to achieve this, intensive efforts were necessary to optimize the profile of 6, including ADME properties, potency, and safety, while maintaining the specific binding mode, finally leading to GDC-0853 (fenebrutinib), a selective reversible BTK inhibitor currently in Phase 2 clinical trials.21

We were attracted by the concept of covalent BTK inhibition as it allows the optimization of compounds for a potent and sustained pharmacodynamic effect without the need for extended and high systemic compound exposure.25 At the same time, we wanted to differentiate from competitor molecules by aiming for a very high selectivity to minimize kinase off-target reversible and covalent binding. Therefore we selected the strategy of combining the advantages of the covalent MoA with targeting the inactive conformation of BTK.

 

 

RESULTS AND DISCUSSION
The concept of combining a reversible BTK inhibitor binding to an inactive conformation of BTK with a warhead leading to highly selective, covalent BTK inhibitors has been recently published by us and others. 43,44 We disclosed the discovery of a series of pyrrolopyrimidines. Using compound 7 (BTK enzyme IC50 of 0.013 ± 0.0078 M, Figure 2) as starting point, a reversible BTK inhibitor binding to the same inactive BTK conformation as 6, we designed inhibitors forming a covalent bond with Cys481. The compounds of this series, represented by 8
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40

 

 

(BTK enzyme IC50 of 0.0009 ± 0.0005 M, Figure 2), are potent inhibitors of BTK in vitro and we were able to demonstrate BTK occupancy after oral dosing in a PK/PD model. However, the pyrrolopyrimidine series suffered from very high in vivo clearance, making it challenging to achieve in vivo efficacy at low doses. Thorough investigation of the pyrrolopyrimidine SAR did not lead to a significantly improved PK profile. Therefore, we decided to explore alternative hinge binders with the aim of improving the ADME properties, while keeping the key interactions with BTK, as displayed in the X-ray cocrystal structure of 7 (Figure 3). Replacing the pyrrolopyrimidine moiety by 6-aminopyrimidine as hinge binder motif provided 9 (Figure 2) as a reversible prototype. Figure 3 shows the model of the aminopyrimidine 9 bound to the inactive conformation of BTK based on the solved crystal structure with 7. In the model, the aminopyrimidine scaffold forms the same key interactions with BTK as the pyrrolopyrimidine 7 with the exception of the contacts at the entrance of the ATP binding pocket. The removed phenyl at position 6 of compound 7 is nicely sandwiched between the side chains of Leu408 and Gly480. For that reason, some loss of affinity was expected.

41 O O O O

42
43
44

HN

HN

F

N

HN

F

45
46

N
4

6

O

N

5

N

5

47
48

N
N
H

N

N
N
H

NNH2

49
50
51
52
7
BTK IC50 0.013 M
N
8
BTK IC50 0.0009 M
9
BTK IC50 7.3 M

53
54
55
56
57
58
59
60
Figure 2. Morphing of the pyrrolopyrimidine scaffold to an aminopyrimidine scaffold. BTK enzyme inhibition in Caliper assay, IC50 shown.

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9

 

 

Thr474

 

 
Lys430

10
11
Met477

12
13
14
15
Tyr551

16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
Cys481
Figure 3. Crystal structure of 7 (PDB 6S90; in cyan) in complex with human BTK overlaid with the binding model of compound 9. H-bonds of the pyrrolopyrimidine and 6-aminopyrimidine scaffold (in purple) to Met477 and a water molecule (red sphere) are highlighted (black dashed lines), as well as the hydrogen bond between the carbonyl of the amide of the ligand and Lys430 side chain.

 
Indeed, compound 9 inhibited BTK in an enzymatic assay with an IC50 of 7.3 ± 0.35 M, confirming our morphing idea. Similarily to what we described for the pyrrolopyrimidine scaffold,43 we envisaged to generate potent and selective covalent inhibitors by introducing a linker with a warhead at position 5 of the pyrimidine moiety to reach Cys481. We started with acrylamide warheads, as they have been used successfully in covalent BTK inhibitors such as 1 and 4.45

SAR: Optimizing in vivo BTK Occupancy. We first investigated alkene based linkers between the pyrimidine core and the warhead providing a somewhat rigidified exit vector. Introducing a three carbon linker (10) and a four carbon linker (11) led to moderately potent

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

compounds in the BCR/CD69 assay in blood (Table 1). However, 10 and 11 suffered from very high clearance in rat liver microsomes (CLint 924 and 815 L/min/mg, respectively). Replacing the carbon linkers with 3-oxypyrrolidine (12) was not tolerated with respect to BTK inhibition, but the metabolic stability significantly increased (CLint 135 L/min/mg). Switching to a
3-oxyazetidine linker (13) led to an improvement of blood BCR/CD69 potency and good metabolic stability (CLint 25 L/min/mg). Having succesfully combined these two properties, we sought to assess the in vivo PD effect of 13 by measuring BTK occupancy in rat spleen after a single oral dose (Experimental Section). Administration of 13 (10 mg/kg po) provided 36 % BTK occupancy and blood levels of 13 nM 5 h post dose. A rat PK of 13 revealed a bioavailability of only 5 %, likely limited by the low permeability. Therefore we set out to optimize the permeability, since improved oral absorption should subsequently increase target occupancy. We envisioned that introducing a fluoro substituent ortho to the amide on the phenyl ring occupying the H3 pocket could form an internal hydrogen bond in solution, masking the amide NH and leading to improved permeability.46 The fluoro substituent was tolerated with respect to potency and 14 indeed displayed improved permeability by more than one log unit compared to 13. This led to a higher bioavailability of 51 % in a rat PK study and resulted in
96% target occupancy after a 10 mg/kg po dose. Lowering the dose of 14 to 3 mg/kg po and 1 mg/kg po still provided BTK occupancy of 79 % and 44 %, respectively. In an effort to further improve the potency we investigated two alternative moieties in the H3 pocket, which in previous studies on a related reversible series (not shown) had provided a positive effect on the potency. Surprisingly, replacing the cyclopropyl substituent in 14 with 2-hydroxypropanyl (15) led to a decreased potency in the blood BCR/CD69 assay. Introducing a second fluoro substituent (16) recovered the blood potency, however the permeability of 16 was poor,

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

translating into a modest target occupancy of only 32 % after a dose of 3 mg/kg po. Based on the promising results of 14 in the BTK occupancy assay, we profiled the compound more broadly. However, the stability of 14 in human plasma turned out to be low (t1/2 35 min). Plasma instability could be an indication of an undesired covalent binding to plasma proteins or enzymatic cleavage, therefore we used this assay to optimize our scaffold.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 
ACS Paragon Plus Environment

 

1
2
3
4

 

 

Table 1. Linker and tail modifications.

5
6
7
8
9
10
11
12
13
R2 O
HN

 

N
F
R1

N NH2

14
15
16
17
18
19
R1
R2

BTK IC50
[nM]a

FcR/IL8 IC50 [nM]b

BCR/CD69 IC50 [nM]c

Permeability logPe pH 6.8d
BTK occupancy [%] (Dose [mg/kg])e

20 O

21
22
23
24
25
10

 

11

N
N
3.8 ± 0.32

 

1.8e
11± 6

 

15 ± 4
106 ± 51

 

223 ± 18
-5.0

 

-4.6
n.t.

 

n.t.

26
27
28
29
30
31
32
33

 

12

 

13

 

O

 

O

 
N

 

N
O
O

 

3651 ± 3619 1437 ± 309

 

3.6± 0.66 17 ± 3

 

n.t.

 

95± 4

 

-6.2

 

<-6.7

 

n.t.

 

36 (10)

34
35
O

36
37

O
96(10)

38 14 N 4.5 ± 1.6 20 ± 13 86 ± 30 -5.2 79 (3)

39
40
41
O

F
44 (1)

42 O HO

43
44
45
46
15
N

O
F
23± 10
108 ± 78
250 ± 0
-6.6
n.t.

47 O HO

48
49
50
16
N

O

F
F
2.3 ± 1.1
24± 7
55 ± 29
<-6.6
32 (3)

51
52
53
54
55
56
57
58
59
60
a) BTK enzyme inhibition in Caliper assay IC50 shown as means ± SD (n ≥ 2); b) FcγR induced IL8 IC50 shown as means ± SD (n ≥ 2); c) anti-IgM/IL-4 induced CD69 expression on human blood B cells IC50 shown as means ± SD (n ≥ 2); d) PAMPA permeability, logPe at pH 6.8; e) BTK occupancy in rat spleen homogenate 5 h after po dose of compounds compared to vehicle control, determined as mean (n = 3); e) n = 1; f) n.t.: not tested.

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

 

We sought to investigate the linker to the warhead more broadly with the goal to increase the plasma stability and in addition to further improve blood potency and in vivo PD. We combined various linkers with the most attractive H3 pocket moiety, the fluorinated cyclopropylphenyl tail. The propenyl linker (17) led to a two fold increase in potency in the blood BCR/CD69 assay compared to 14 and good plasma stability (Table 2). However, the increased blood potency did not translate into an improved PD effect, 17 performed similarly to 14 in vivo, providing 77 % BTK occupancy, probably due to the high in vivo clearance (307 ml/min/kg). The oxygen linked open-chain analogue 18 was in the same potency range as 14, and gave high target occupancy of 84 % despite its only moderate permeability. Replacing the linker oxygen by a nitrogen (19) was not tolerated leading to a roughly 10-fold loss of potency in the human blood assay. Despite a more than two-fold reduced blood potency compared to 14, introduction of a piperidine linker gave similar target occupancy, due to high exposure (207 nM 5 h post dose). Whereas the (R)- pyrrolidine linker (21) was not tolerated with respect to BTK inhibition, the (S)-derivative 22 potently inhibited the blood BCR/CD69 assay and BTK occupancy of 90 % was achieved. However, 22 suffered from a strong hERG flag (hERG dofetilide binding IC50 49 ± 12 nM (n = 2)). Replacing the pyrrolidine moiety by an azetidine provided 23, which achieved 80 % BTK occupancy. However, as in the case of azetidine 14, plasma stability of 23 was low. An enhanced reactivity of N-acryloyl azetidines towards cysteines has been reported and might explain the
low plasma stability of compounds 14 and 23.47 The corresponding (R)-azetidine 24 was less potent than 23 in the blood assay. The N-methylated analogue of 18 (25) was nearly 5-fold more potent than 14 in the blood BCR/CD69 assay and displayed 94 % BTK occupancy. Increasing the length of the open chain linker to three carbons led to 26 which was significantly less potent than 25 in the blood assay. Modifying the N-methyl substituent on the acrylamide to an ethyl

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

group provided 27, which was comparable to 25 with respect to blood potency and target occupancy. However, 27 had a higher logP compared to 25 (3.6 vs 2.9) and interestingly inhibited CYP2D6 with an IC50 of 1.26 M. Overall, 27 did not seem to offer any advantage over 25. With the exception of azetidine acrylamide 23 all compounds from Table 2 tested for stability in human plasma showed plasma half-lives of >120 min.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6

 

 

Table 2. Linker modifications.

7
8
O

9
10
11
F
HN
F

12
13
14
15
16
17
18
19
20
21

 

 

 

R1

 

 
BTK IC50
(nM)a

 

 
FcR/IL8 IC50 (nM)b
R1
N

N NH2
BCR/CD69 IC50 (nM)c

 

 
Permeability logPe pH 6.8d

 

 

Stability in
human plasma, t1/2
[min]

 

 

BTK occupancy
[%] after a 3 mg/kg dosee

22 O

23
24
25
17

N
1.9 ± 1.2
11 ± 3
44 ± 4
-4.5
>120
77

26
O

27
28
29
30
31
32
33
34
35
36
37
38
18

 

19

 

20
O

 

HN
O

N
H

 

N
H

 

N

 
O

 

 
O
8.7 ± 4.6

 

51 ± 38

 

3.7± 4.5
18 ± 13 105 ± 13

 

671 ± 690 1245 ± 350

 

12± 2 201 ± 63
-5.4

 

-5.8

 

-4.5
>120

 

n.t.

 

>120
84

 

n.t.

 

77

39
40
41
42
43
44

21

O

N

O

1290 ± 916 1869 ± 87

n.t.

-3.9

n.t.

n.t.

45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
22

 
23

 

24
O

 
O

 
O
N

O
N

O
N

O
1.4 ± 0.4

 
3.1 ± 0.5

 

7.5 ± 6.3
9f

 
5f

 

60 ± 14
57 ± 8

 
50 ± 3

 

354 ± 59
-4.1

 
-3.9

 

-4.1
>120

 
42

 

n.t.
90

 
80

 

n.t.

60
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7

 

 
25

 

 

 

O

 

 

 

N

 

 

O

 

 
1.3± 0.9

 

 
2.5 ± 0.7

 

 
18 ± 5

 

 
-4.3

 

 
>120

 

 
94

8
9

26 O
N
3.6 ± 1.3
17± 6.5
91 ± 9
-4.0
n.t.
n.t.

10
11
12
O

O

13
14
15
27
O
N
0.9 ± 0.2 5f
28 ± 4 -4.2 >120 93

16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
a) BTK enzyme IC50 shown as means ± SD (n ≥ 2); b) FcγR induced IL8 IC50 shown as means ± SD (n ≥ 2); c) anti-IgM/IL-4 induced CD69 expression on human blood B cells IC50 shown as means ± SD (n ≥ 2); d) PAMPA permeability, logPe at pH 6.8; e) BTK occupancy in rat spleen homogenate 5 h after po dose of compounds compared to vehicle control, determined as mean (n = 3); f) n = 1; g) n.t.: not tested.

 
Having identified the highly potent acrylamide 25, we wanted to assess if an alternative warhead could provide an overall advantage. Generally, a highly reactive moiety could lead to increased binding to off-targets, whereas too low intrinsic reactivity could lower the reaction rate to the Cys of interest. To probe the intrinsic reactivity of different warheads we investigated the stability of the compounds towards an excess of glutathione (GSH) as a surrogate for Cys- containing nucleophiles and proteins (Table 3). In the presence of GSH, 25 showed a slow decrease with 68.5 % remaining parent after 24 h. For the sterically hindered methyl substituted
acrylamide 28, a significantly decreased reactivity towards GSH was observed. However, 28 was also significantly less active in the blood BCR/CD69 assay. The 4-(dimethylamino)but-2- enamide 29 showed a slow decrease in the presence of GSH over 24 h, similar to 25. However, the permeability of 29 was very low (log Pe pH 6.8 <-6.5) and together with the reduced blood BCR/CD69 potency compared to 25 we did not pursue this compound further. The propynamide 30 was highly unstable in the presence of GSH with complete disappearance of the parent within 24 h. In addition, only limited stability (t1/2 96 min) in human plasma was observed. For the but-
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

2-yneamide 31, decrease of parent was slow, but due to the decreased blood BCR/CD69 assay potency this compound was also not considered further. In summary, compound 25 (LOU064, remibrutinib) exhibited the best overall profile and was selected for further characterization.

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7

 

 

Table 3. Warhead modifications.

8
9
O

10
11
12
F
HN
F

13
14
15
16
17
18
19
20
21
22
23

 

 

 

 
25

 

 

 

 
O

 

 

R1

 
N

 

 

 

 

O

 

 

BTK IC50
(nM)a
1.3 ± 0.9
R1
N

N NH2

FcR/IL8 IC50 (nM)b
2.5 ± 0.7

 

 

BCR/CD69 IC50 (nM)c
18± 5

 
Stability in
presence of GSH
at 24 hd

68.5

24
O

25
26
27
28
29
30
28
29

 
O
O

 

 

N

N

O

 
N
104 ± 77
11 ± 5
263 ± 119 983 ± 617
76 ± 3 151 ± 69
95.8
60.8

31
32
33

30

O

N
O

1.8 ± 0.2

6.5 ± 2

49 ± 23

0.1

34
35
36
37

31

O
N
O

5.2 ± 2

32 ± 7

161 ± 44

79.6

38
39
40

32

O

N
O
1’300 ± 424
>10’000
>20’000

n.t.

41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
a) BTK enzyme IC50 shown as means ± SD (n ≥ 2); b) FcγR induced IL8 IC50 shown as means ± SD (n ≥ 2); c) anti-IgM/IL-4 induced CD69 expression on human blood B cells IC50 shown as means ± SD (n ≥ 2); d) The stability in the presence of GSH is given as ratio area (analyte/internal standard) in %; e) n.t.: not tested.

 

 

 

 

 

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

The melting point of crystalline LOU064 is 194 °C. It has a low thermodynamic solubility of

<0.003 g/L at pH 6.8, but solubility increases to >1.5 g/L at pH 1. The permeability is high (PAMPA: log Pe pH 6.8 -4.3; Caco Papp(A-B) 15.9, Papp(B-A) 10.2) suggesting good absorption. The half-maximum inhibition of the CYP enzymes 3A4, 2C9 and 2D6 was ≥ 5 M (see Supporting Information). LOU064 showed some activity in an in vitro PXR reporter gene assay (EC50 >5.48 M and Emax 94.7 %) indicating a risk for CYP induction and drug-drug interactions.

 

 

LOU064 Binds Covalently to BTK. The covalent character of BTK inhibition by LOU064 was confirmed in a series of studies. First, the kinetics of BTK inhibition were assessed in an enzyme dilution experiment.48 The biochemical BTK enzyme assay (Supporting Information) was modified to include a one hour preincubation of BTK protein at a concentration of 4.7 µM with a 10 % molar excess of compound. The compound/enzyme mixture was then diluted into kinase assay buffer containing ATP and peptide substrate to reach a final concentration of 1 nM BTK and 1.1 nM compound and enzyme activity was monitored by measuring substrate phosphorylation (Figure 4). The increase in phosphorylated substrate peptide over time indicated recovery of BTK kinase activity by the dilution step for the DMSO solvent control, the reversible BTK inhibitor staurosporine, as well as the inactive non-covalent ethylamide analog 32 (Table 3). In line with expectations for a covalent binder, LOU064 showed no recovery of BTK activity after the dilution step.

 

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

 

 

 

 

 

 

 

 

 

Figure 4. Rescue of BTK enzymatic activity by dilution. The increase of phosphorylated substrate peptide over time after the dilution step is indicated for the DMSO solvent control (black dash and triangles), for the reversible BTK inhibitor staurosporine (blue dash and triangles), for the non- covalent ethylamide analog 32 (red dash and empty circles) and for LOU064 (red line and filled circles).

 
Secondly, we assessed covalent binding of LOU064 to the BTK kinase domain by analysis with LC/MS (Experimental details see Supporting Information), which revealed the presence of a main peak with 32’222.3 Da mass, closely corresponding to the predicted BTK/LOU064 1:1 adduct (based on the masses of 507.5 and 31’714.5 Da, respectively for LOU064 and the kinase domain construct). This provided further evidence for a specific and single covalent modification of BTK by LOU064 (Figure 5).

 

 

 

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

 

 

 

 

 

 

 

 
Figure 5. Deconvoluted mass spectrum of covalent BTK kinase domain (encompassing amino acids 386-659) adduct with LOU064. The peak with a corresponding mass of 32222.3 Da is indicative of the presence of one covalent adduct of LOU064 (507.5 Da) to the BTK kinase domain (31’714.5 Da).

 
Thirdly, solving the complex structure revealed the atomic details of LOU064 binding to BTK and confirmed the design hypothesis. As expected, the inhibitor is covalently attached to Cys481 (Figure 6), with the carbonyl of the acrylamide making a hydrogen bond with the NH of the backbone of Cys481. The amino-pyrimidine moiety forms a bidentate hydrogen bonding interaction with the backbone of Met477 of the kinase hinge region. The phenyl group of LOU064 at position 4 of the 6-aminopyrimidine scaffold makes hydrophobic contacts with the Val416 side chain while the fluorine makes a multipolar interaction with the carbonyl of Gly409 of the Gly- rich loop (Figure S-1, Supporting Information).49 The fluoro-phenyl-cyclopropyl-group occupies the H3 pocket and stabilizes it in an inactive state. The H3 pocket16 is formed by the side chains of Gln412, Phe413 (both Gly-rich loop), Asp521, Asn526 (both part of the catalytic loop), Asp539 (DFG), Leu542, Ser543, Val546, and Tyr551 (belonging to the activation loop) (Figure S-1). The

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

fluorine in this pocket points towards the cavity, instead of towards the NH of the amide of the ligand, most likely to avoid the unfavorable vicinity to the carbonyl of Asn526 (Figure S-1) .

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 
Figure 6. X-ray complex structure of BTK kinase domain bound to LOU064 (PDB 6TFP, protein chain A with ligand chain L). The molecular surface of LOU064 is colored in orange and key residues lining up the binding site are shown as labelled stick model. The main polar interactions between protein and inhibitor are indicated with dotted green lines. For improved clarity water molecules are omitted.

 

Selectivity of LOU064. Our aim was to combine covalent BTK binding and the high selectivity associated with binding to the inactive BTK conformation. The selectivity of LOU064 was assessed not only in a selected panel of enzymatic kinase assays, but also in a competition binding

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

assay50 against 395 human non-mutant kinases and 61 atypical, mutant or pathogen kinases. In this screen (KINOMEscan™, DiscoverX), LOU064 at a concentration of 1 M showed full inhibition of probe binding to BTK (0.25 % remaining probe binding) and partial inhibition of TEC (24 % remaining probe binding) and BMX (53 % remaining probe binding), indicating an excellent kinase selectivity profile (TREEspotTM Kinase Dendogram of LOU064 (Figure S-2) and KINOMEscan data in Supporting Information).
Based on this kinome-wide screen, the selectivity of LOU064 was further assessed and compared to clinical covalent BTK inhibitors. As the determination of IC50 values in biochemical enzyme assays for covalent inhibitors suffers from time-dependent effects48 we determined the binding constants (Kd) in a competition binding assay50,51 for a set of Cys-containing kinases (Table 4). LOU064 showed very potent affinity to BTK with a subnanomolar Kd of 0.63 nM and with a selectivity of 175 fold against TEC (Kd of 110 nM) and 857 fold against BMX (Kd of 540 nM). LOU064 did not show binding to ITK, EGFR, ERBB2, ERBB4 and JAK3 up to 10 M. By contrast, ibrutinib (1) showed similar high affinity for BTK, but poor selectivity against BMX, TEC, EGFR, ERBB2 and ERBB4. Of note, acalabrutinib (2), tirabrutinib (3) and evobrutinib (4) are significantly less potent than LOU064 on BTK and less selective. Branebrutinib (5)
displayed high affinity for BTK, comparable to LOU064 and ibrutinib, with an improved selectivity profile compared to ibrutinib, however inferior to LOU064, potently inhibiting BMX and TEC. These binding data are in line with recent publications in other assay systems39 and our data in biochemical enzyme assays (not shown). Overall, in addition to being amongst the most potent inhibitors for BTK, LOU064 differentiates with its excellent, best-in-class kinase selectivity from other clinical-stage covalent inhibitors.

 

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7

 

 

Table 4. Binding constants (Kd in nM) of covalent BTK inhibitors for selected kinases containing a conserved cysteine and selectivity vs off-target kinases (Kd, off-target/Kd, BTK).

8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39

40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

We evaluated the cellular selectivity of LOU064 in a panel of culture systems with primary human cells representative for several tissues, including fibroblasts, epithelial cells, keratinocytes and immune cells (BioMAP Diversity plus panel, DiscoverX).52 LOU064 was tested at concentrations of 270, 900, 3000 and 10000 nM, all concentrations well above its cellular BTK inhibition in cell culture conditions (Figure S-3, Supporting Information). The main activity

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

detected was in the BT panel, and represents the intended inhibition of cellular BTK function in B cells stimulated via the B cell receptor and in the presence of T cell co-stimulatory pathways, resulting in the inhibition of several inflammatory readouts such as TNF-α, IL-17A, IL-6 and IL- 2, as well as inhibition of B cell proliferation (indicated by the gray arrow in the BT panel). The response was not concentration dependent as all concentrations tested were well above maximal cellular BTK inhibition. Of note is the absence of a peak in the EGFR signal in the HDF3CGF panel, clearly indicating the lack of any activity of LOU064 in the dermal fibroblast system, an effect which has been shown for BTK inhibitors with off-target effects on EGFR, such as ibrutinib (Figure S-4, Supporting Information).22 Across all these cellular systems, LOU064 showed no cytotoxicity. In addition to the expected effects on B cell proliferation, LOU064 exerted effects on fibroblast proliferation at concentrations of 270 and 10000 nM (gray arrow in the HDF3CGF panel).

Furthermore, the high selectivity of LOU064 was confirmed by testing against a broad panel of in vitro assays (in binding or in functional mode) comprising 85 G-protein coupled receptors, transporters, nuclear receptors, enzymes and ion channels (Supporting Information). LOU064 did not show binding, inhibition, or activation in any of these assays at concentrations up to 10 µM, with the exception of PIK4CB (IC50 3.1 µM) and Bile Salt Export Pump (BSEP, IC50 6.6
µM), thus establishing a more than 2000-fold selectivity for BTK over these off-targets. In the in vitro hERG patch clamp assay an IC50 of 1.4 M was measured. In preclinical invasive and non- invasive dog telemetry studies after single and long-term dosing, no effect on any cardiovascular parameter was detected. As the non-covalent analog 32 showed very similar activity on BSEP (IC50 3.7 µM) and hERG (IC50 2.9 µM in Qpatch assay), it is likely that the interaction of LOU064 with these off-target proteins is non-covalent. No significant inhibition of other cardiac

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42

 

 

ion channels (Nav1.5, Cav1.2, KCNQ1) was observed and LOU064 was found negative in genotoxicity and mutagenicity assays (in vitro Ames, in vitro and in vivo micronucleus tests). Similarly, LOU064 was negative in the 3T3 NRU phototoxicity assay.

 

 

Preclinical PK Profile of LOU064. The in vitro clearance of LOU064 in microsomes was moderate in rat, and high in mouse and dog (Table 5). The in vivo pharmacokinetic properties of LOU064 were assessed in rat, mouse and dog (Table 5). In rats, the compound displayed a moderate clearance, a low volume of distribution, a short half-life, and a moderate bioavailability. LOU064 was mainly cleared via oxidative metabolism and elimination of unchanged LOU064 was not a major pathway in rat. In mice, LOU064 displayed a similar PK profile as for rats with high CL, a low volume of distribution and a short half-life. In dogs, the clearance was high, in the range of the hepatic blood flow, the volume of distribution was low, the half-life was short and the bioavailability was low. Formulations were developed which enabled preclinical toxicology studies to be carried out (not shown).

 

Table 5. In vitro and in vivo pharmacokinetic parameters of LOU064 in rat, mouse and dog

43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
In vitro CL int (L/min/mg) Hepatic extraction (%)

In vivo Dose
Rat

33

52

Female Lewis
1 mg/kg iv

 

 

ACS Paragon Plus Environment
Mouse

325

93

Female C57BL/6
1 mg/kg iv
Dog

146

87

Male
Beagle

1 mg/kg iv

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25

 

 

 
CL (mL / min / kg) Vss (L / kg)
t1/2 (h)

Cmax d.n. (nM) Tmax (h)
F (%)

AUC po d.n. (nM · h)

 

 

3 mg/kg po 44 ± 12
1.4± 0.3 0.5 ± 0.03 148 ± 33
0.3 29 ± 3
225 ± 27

 

 

3 mg/kg po

97± 11 1.0 ± 0.1 1.0 ± 0.1
53± 14 0.3 ± 0.1
16 ± 4

54± 15

 

 

3 mg/kg po

29 ± 4 1.0 ± 0.1 0.9 ± 0.1
19

0.8

5

55

26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
In essence, the preclinical ADME properties of LOU064 are characterized by excellent permeability, high clearance and fast elimination.

 

 

In vitro BTK Inhibition Kinetics. To further characterize the mechanism of BTK inhibition by LOU064, we tested the kinetics of enzymatic inhibition. In biochemical assays of enzymatic activity with recombinant full-length human BTK, LOU064 showed very fast inhibition of BTK (Figure S-5, Supporting Information). LOU064 showed potent BTK inhibition starting from the earliest time-point and no indication of a time-dependent shift in IC50 over the timeframe that allowed reliable detection of BTK activity or inhibition. The very fast binding kinetics of LOU064 to purified BTK prevented the determination of accurate and meaningful biochemical reaction parameters.
We therefore assessed the engagement of cellular BTK in a clinically relevant matrix – human blood. Blood from healthy volunteers was mixed with serial dilutions of LOU064, incubated at

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

room temperature to allow binding to cellular BTK and at given time points, aliquots of blood were sampled and analyzed for covalent binding of LOU064 to BTK with immunoassays for free BTK (i.e. not covalently occupied by compound) and total BTK protein (see Supporting Information). LOU064 showed very rapid covalent binding to cellular BTK in human blood at concentrations as low as 300 nM (Figure 7, left panel).

 

 

 

 

 

 

 

 

 
Figure 7. Time- and concentration-dependent in vitro engagement of cellular BTK by LOU064 and related inhibitors in human blood. Left panel shows data for LOU064 from two independent experiments with different blood donors (solid lines show non-linear one-phase association curve fits). Right panel shows the association kinetics for a concentration of 300 nM for each compound (solid lines show non-linear one-phase association curve fits). Individual data from two independent experiments with different blood donors.

 

We compared the binding characteristics of LOU064 to related covalent BTK inhibitors in the same type of assay at a concentration of 300 nM (Figure 7, right panel). In contrast to the rapid and complete BTK binding of LOU064, the other covalent BTK inhibitors, with exception of branebrutinib, showed slower binding kinetics. A comparison of the enzymatic and cellular

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14

 

 

potency of LOU064 to these covalent BTK inhibitors across several assays confirmed the very high potency of LOU064 (Table 6). These data suggest that LOU064 has very fast reaction kinetics and rapidly reaches full BTK inhibition in relevant matrices such as human blood.
Table 6. Comparative potency of LOU064 and covalent BTK inhibitors

15
16
17
18
19
20
21
22
23
24
25
26
27
28
29

 
LOU064 (25)

Ibrutinib (1) Acalabrutinib (2)
Tirabrutinib (3) Evobrutinib (4)
Branebrutinib (5)
BTK IC50 (nM)a
1.3 ± 0.9 2.3 ± 2 19 ± 22 5.2 ± 5 62 ± 4
0.1 ± 0.0518
FcR/IL8 IC50 (nM)b
2.5 ± 0.7 16 ± 14 24 ± 24 26 ± 16 61 ± 37
6.0 (n = 1)
BCR/CD69 IC50 (nM)c
18 ± 5

66± 57 203 ± 77 132 ± 46 320 ± 74
18 ± 4
FcεR/CD63 IC50 (nM)d
67± 54 230 ± 210 747 ± 75 363 ± 200 1182 ± 819
40 ± 10

30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
a) BTK enzyme inhibition in Caliper assay IC50 shown as means ± SD (n ≥ 2); b) FcγR induced IL8 IC50 shown as means ± SD (n ≥ 2); c) anti-IgM/IL-4 induced CD69 expression on human blood B cells IC50 shown as means ± SD (n ≥ 2); d) anti-IgE-induced degranulation and CD63 expression on human blood basophils.

 

 

In vivo BTK Engagement. For a covalent inhibitor of BTK, the PD effects are determined by the extent and duration of BTK target occupancy. The initial inactivation of BTK by LOU064 binding is primarily determined by the systemic exposure of the drug. Once the drug is eliminated from systemic circulation the duration of BTK inhibition becomes independent from drug exposure and is determined by the turnover of the inactive drug/protein complex by synthesis of new BTK protein.53 For BTK, the turnover of inactivated protein by newly synthesized protein has been described in preclinical models and ranges from 20 – 40 % of BTK protein being resynthesized after 24 h.18,22,54
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

We set out to determine the detailed PK/PD relationship of LOU064 in rats by using BTK occupancy as the primary PD readout. A series of studies were performed in rats to determine the dose range leading to varying degrees of BTK occupancy in spleen (Figure 8). To minimize the impact of circulating LOU064, the animals were killed 5 hours after oral dosing and tissues were sampled. BTK occupancy in spleen homogenates was measured relative to vehicle controls. A dose-response curve fit indicates an EC50 of 0.6 mg/kg (EC90 1.6 mg/kg). The analysis of LOU064 in blood from these animals showed that for the 1 mg/kg dose, resulting in an average of 78 % BTK occupancy, the systemic exposure was 49.1 nM at 0.5 hours post dose (5.6 nM at 5 hours). This suggests that very low and transient systemic exposures of LOU064 are sufficient for BTK inhibition.

 

 

 

 

 

 

 

 

 

 

 

 

Figure 8. Spleen BTK occupancy relative to vehicle controls was measured 5 hours after oral dosing in female rats combined from two separate studies (with 3 animals per each dose group). Open triangles show the BTK occupancy levels in the combined vehicle dosed animals. Filled

 

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

circles show the BTK occupancy of LOU064 treated animals. The dotted line shows a dose- response curve fit.

 

Preclinical Efficacy in SRBC and CIA Models. To extend the preclinical PK/PD relationship to repeated dosing regimen, we examined the efficacy of LOU064 in a model of B cell dependent antibody formation following immunization with sheep red blood cells (SRBC).55 Female rats were immunized with SRBC and treated for 4 days with one daily oral dose of LOU064 starting on the day of immunization. On day 5, SRBC antibody formation was assessed ex vivo on splenocytes, as well as BTK occupancy in the same tissue (Figure 9, left panel). The IgM antibody response was maximally inhibited (by 96 %) at a dose of 3 mg/kg q.d.. This level of IgM response inhibition was correlated to a spleen BTK occupancy at the 24 h trough of 71.5 % (Figure 9, right panel). Doses as low as 0.3 mg/kg demonstrated a partial, but significant inhibitory effect on SRBC specific antibody response (with a trough BTK occupancy in spleen of 31 %).

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

Figure 9. IgM response inhibition by LOU064 in the rat model of B cell response to SRBC immunization. Left panel: inhibition of anti-IgM response: open triangles show vehicle (V), open circles cyclosporine A (CsA) and closed circles LOU064 treated animals, respectively. The group means are shown as the central bars and the standard deviations are shown as whisker error bars. Statistical significance (ANOVA, followed by Dunnett’s test) is shown as *** for p < 0.001. Right panel: BTK occupancy in spleen 24 hours after the last dose of LOU064: open triangles show vehicle (V), open circles cyclosporine A (CsA) and closed circles LOU064 treated animals relative to the average of the vehicle group. Group means are shown as central bars and standard deviations as whisker error bars. Statistical significance (ANOVA, followed by Dunnett’s test) is shown as *** for p < 0.001.

 

Next we assessed the effects of LOU064 in the inflammatory model of collagen-induced arthritis (CIA) in rats.56,57 Collagen-specific B- and T-lymphocytes are important in the pathogenesis of CIA, with the peak of the T-cell response occurring around the time of disease onset.57,58 Female Lewis rats were immunized with porcine collagen and LOU064 oral dosing, once daily, was started after the onset of arthritis, continuing until day 26 after immunization. The animals treated with LOU064 showed a fast onset of action and within a few days paw swelling was fully reduced (Figure 10, left panel). This was evident in the three highest doses of 3, 10 and 30 mg/kg, whereas the 0.3 mg/kg dose was largely inactive (no 1 mg/kg dose group was included in the study).

 

 

 

 

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

 

 

 

 

 

 

 

 

 

 
Figure 10. Efficacy of LOU064 in the rat model of collagen-induced arthritis. Left panel: Paw swelling scores are shown as means ± SEM. LOU064 treatment once daily with doses of 0.3, 3, 10 and 30 mg/kg from day 12 to 25 post immunization. Statistical significance (ANOVA, followed by Dunnett’s test) is shown as ** p < 0.01. Right panel: BTK occupancy in spleen 24 hours after the last dose of LOU064. Triangles show vehicle (V) and circles represent LOU064 treated animals relative to the average of the vehicle group. Group means are shown as central bars and standard deviations as whiskers. Statistical significance (ANOVA, followed by Dunnett’s test) is shown as *** p < 0.001.

 

 

Histopathological analysis of the 3 and 10 mg/kg groups showed that bone and cartilage erosion, as well as inflammatory cell infiltration, were all significantly inhibited by LOU064 (Figure 11). Dose proportional levels of BTK occupancy in spleen were found on day 26 at trough, 24 hours after the last dose (Figure 10, right panel). Occupancy levels of 80 % (for the 3 mg/kg dose) and higher were associated with significant inhibition of paw swelling.

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Figure 11. Histopathological analysis of arthritis in rat CIA model. Left panel: Histology scores combined for the hind paws of each animal show inflammatory cell infiltrate, joint damage and proteoglycan loss (average +/- SEM of 6 animals per group, ANOVA, followed by Dunnett’s test ** for p < 0.01). Right panel: Photomicrographs of paw sections stained with Giemsa (left column) and Safranin O (right column) illustrate joint tissue inflammation and preservation of joint cartilage, respectively. LOU064 significantly reduced join inflammation and damage as well as proteoglycan loss at 3 and 10 mg/kg q.d. po.

 

 

This study shows that LOU064 potently inhibits a chronic arthritic inflammation in vivo in a therapeutic treatment setting with a fast onset of action. Since we timed the treatment to start

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

after immunization phase, the fast onset of action suggests that the anti-inflammatory effect is due to the inhibition of inflammatory macrophages and of immune complexes in the paws.

 

 

Synthesis of LOU064 (25). The synthesis of 25 is outlined in Schemes 1 and 2. Scheme 1 shows the synthesis of the upper portion of the molecule, intermediate 38. Borylation of 1- bromo-5-fluoro-2-methyl-3-nitrobenzene (33) with bis(pinacolato)diboron in the presence of Pd(dppf)Cl2·DCM and potassium acetate gave boronic ester 34 which upon catalytic hydrogenation provided aniline 35. Suzuki reaction of methyl 4-bromo-2-fluorobenzoate (36) and cyclopropylboronic acid gave cyclopropyl intermediate 37. Boronic ester 38 was then accessed via a NaHMDS mediated amide formation of aniline 35 and ester 37. The synthesis of LOU064 was completed as outlined in Scheme 2. The lower portion of the molecule was assembled starting from 2,4-dichloro-3-methoxypyrimidine (39). Treatment with ammonia in a pressure reactor resulted in aminopyrimidine 40, which was subjected to boron tribromide treatment to give amino-pyrimidinol 41. The linker moiety was introduced via a Mitsunobu reaction with N-Boc-N-methyl-2-hydroxyethylamine in the presence of DIAD and Smopex-301, a convenient polymer supported version of triphenylphosphine, to obtain 42. Boc protected intermediate 43 was assembled via Suzuki coupling of 42 and 38. Finally, Boc deprotection of 43 followed by amide coupling with acrylic acid in the presence of propylphosphonic anhydride (T3P) provided LOU064 (25).

The experimental details are provided in the Experimental Section. For the synthesis of analogues the schemes and synthetic details are provided in the Supporting Information.

Scheme 1. Synthesis of boronic ester 38.a

 

ACS Paragon Plus Environment

 

1
2
3
4
5

 

 
F

 

 
NO2

 

 

 

a

 

 

F

 

 

NO2

 

 

 

b

 

 

F

 

 

NH2

6
7
8
9
10

Br

33
B
OO
34
B
O O
35

11
12

Br
O

13 c d F NH F
14 O O

15
16
17
18
O F

36

O F
37

B
O O

19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
38

aReagents and conditions: (a) BISPIN, Pd(dppf)Cl2·DCM, KOAc, dioxane, 100 °C, 3.5 h, 92 %; (b) H2, Pd/C, MeOH, RT, 7 h, 93 %; (c) cyclopropylboronic acid, Pd(OAc)2, tricyclohexylphosphine, K3PO4, water, toluene, 100 °C, overnight, 99 %; (d) 35, NaHMDS (1 M in THF), THF, RT, 4 h, 76 %.

 

Scheme 2. Synthesis of LOU064.a

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 
ACS Paragon Plus Environment

 

1
2
3

 

 

Cl

 

 

Cl

 

 

Cl

4
5

N
O
a

N
O
b

N
OH
c

6
7
8
9
10
11
N

39
Cl
N
40
NH2
N

41
NH2

12
13

Cl
O

14
15

N
O
N Boc
d
F
NH
F
e

16 N NH2

17
18

42
N
O
N Boc

19
20
21
22
23
24
N
NH2
43

25 O O

26
27
F
NH
F
f
F
NH
F

28
29
30
31
32
N

 
N

O

NH2
N
H
N

 
N

O

NH2
N
O

33
34
35
36
44
LOU064 (25)

37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
aReagents and conditions: (a) NH4OH, 2-propanol, 70 °C, 48 h, 94 %; (b) BBr3, DCM, 40 °C, 3 h, 59 %; (c) N-Boc-N-methyl-2-hydroxyethylamine, DIAD, Smopex-301, THF, 60 °C, 2 h, 53 %; (d) 38, Pd(PPh3)2Cl2, aq. Na2CO3, DME, water, microwave, 110 °C, 25 min, 74 %; (e) TFA, DCM, RT, 12 h; (f) acrylic acid, DIPEA, T3P (50 % in DMF), DMF, RT, 2 h, 45 % over 2 steps.

 

 

 
Conclusions
In summary, we have discovered a covalent BTK inhibitor, LOU064 (remibrutinib, 25), that shows excellent selectivity and potency with potential for best-in-class in vivo profile. By a

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

structure-based approach, starting from a selective, reversible BTK binder, we successfully morphed the hinge binder moiety and introduced a linker with a warhead leading to selective, covalent BTK inhibitors. The drug discovery efforts focused on the optimization of potency, ADME properties and PK/PD using an in vivo target occupancy assay.

The preclinical pharmacokinetic data of LOU064 is favorable for a covalent inhibitor with the potential for once daily dosing. Based on the singular PK/PD profile of a covalent inhibitor LOU064 showed potent BTK occupancy with an EC90 of 1.6 mg/kg after single oral dose with transient low exposure. In addition, the in vivo efficacy on B cell response and the potent inhibition of rat arthritis with a fast onset of action suggest that the compound could have utility in treating autoimmune diseases. The compound displayed a good preclinical tolerability and safety profile in two species at different doses and these findings encouraged us to progress LOU064 into clinical studies in autoimmune diseases. In phase I studies, healthy and atopic subjects have been exposed to LOU064 at doses ranging from 0.5 mg to 600 mg.59 Administration of LOU064 in healthy subjects did not raise any safety signals in any of these studies. With favorable phase I results, LOU064 entered phase 2 clinical studies in several immune disorders including chronic spontaneous urticaria and Sjoegren’s Syndrome. Further details and clinical results will be reported in due course.

 

 

EXPERIMENTAL SECTION

 

 
Experimental Details

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

All chemicals were used directly as received from commercial suppliers. 1H NMR and 13C NMR spectra were recorded on Bruker 400 or 600 spectrometers. Chemical shifts are expressed in δ ppm referenced to the residual solvent peak (DMSO-d6: δ = 2.50 ppm; CDCl3: δ = 7.27 ppm; CD3OD: δ = 3.30 ppm). Abbreviations used in describing peak signals are: br = broad signal, v = very, s = singlet, d = doublet, dd = doublet of doublets, t = triplet, q = quartet, m = multiplet. All final compounds were purified to > 95% by reverse phase high performance liquid chromatography (HPLC), supercritical fluid chromatography (SFC) or normal phase silica gel column chromatography. The purity was assessed by reverse phase UPLC with a gradient of 5–98 % acetonitrile in water (with either acid or base modifier) and monitored by UV absorption at 210- 450 nM. Purity of all final compounds is ≥ 95 %. Low-resolution mass spectra were recorded on liquid chromatography-mass spectrometer in electrospray positive (ES+) and negative (ES-) mode. The HR-MS analyses were performed by using electrospray ionization in positive ion modus after separation by liquid chromatography (Nexera from shimadzu). The elemental composition was derived from the mass spectra acquired at the high resolution of about 30’000 on an LTQ Orbitrap XL mass spectrometer (Thermo Scientific). The high mass accuracy below 1 ppm was obtained by using a lock mass.
N-(3-(6-Amino-5-(2-(N-methylacrylamido)ethoxy)pyrimidin-4-yl)-5-fluoro-2- methylphenyl)-4-cyclopropyl-2-fluorobenzamide (25, LOU064). Step 1: To a solution of 43 (6.5 g, 11.74 mmol) in DCM (100 mL) was added TFA (9.05 mL, 117 mmol). The reaction mixture was stirred at RT for 12 h. The mixture was concentrated under reduced pressure. The residue was dried in vacuo to obtain the crude amine 44 as TFA salt. [ES-MS] (ESI+): m/z calcd for C24H26F2N5O2 [M + H]+, 454.2; found, 454.3.

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

Step 2: To a solution of acrylic acid (1.10 g, 15.26 mmol) in DMF (50 mL) was added DIPEA (6.15 mL, 35.2 mmol) followed by T3P (50 % in DMF, 7.54 mL, 12.91 mmol). The mixture was stirred at RT for 30 min to form the active ester. The mixture was added dropwise to a solution of crude 44 and DIPEA (6.15 mL, 35.2 mmol) in DMF (50 mL) at 0 °C. The reaction mixture was stirred at RT for 2 h. The mixture was diluted with EtOAc and washed with water (2x) and brine. The organic layer was dried over MgSO4, filtered and concentrated. The residue was purified by column chromatography (SiO2; (DCM containing 2 % NH3)/MeOH gradient) to afford 2.80 g of 25 as a white solid. Yield: 45 % over 2 steps. [ES-MS] (ESI+): m/z calcd for C27H28F2N5O3 [M + H]+, 508.2; found, 508.3. 1H NMR (500 MHz, DMSO-d6) rotamers δ 9.79 and 9.57 (2s, total 1H), 8.22 and 8.19 (2s, total 1H), 7.78 – 7.72 and 7.71 – 7.65 (2m, total 1H), 7.63 – 7.52 (m, 1H), 7.15 – 7.03 (m, 4H), 7.03 – 6.96 (m, 1H), 6.71 – 6.57 (m, 1H), 6.14 – 6.02 (m, 1H), 5.65 – 5.56 (m, 1H), 3.63 – 3.54 (m, 2H), 3.54 – 3.45 (m, 2H), 2.81 and 2.51 (overlapping with DMSO) (2s, total 3H), 2.10 – 2.00 (m, 1H), overlapping with 2.03 and 1.98 (2s, total 3H), 1.11 – 1.03 (m, 2H), 0.86 – 0.78 (m, 2H). 13C NMR (151 MHz, DMSO-d6) rotamers δ 165.58, 165.54, 162.65, 162.40, 160.66, 160.50, 160.16, 160.04, 159.01, 158.85, 158.72, 158.56, 158.44, 153.84, 153.44, 153.35, 152.93, 151.06, 151.01, 150.82, 150.77, 138.75, 138.69, 138.34, 138.28, 137.85, 137.78, 137.70, 136.08, 135.23, 130.55, 130.36, 128.35, 128.32, 127.10, 126.89, 126.20, 126.05, 121.65, 121.59, 120.43, 120.34, 119.99, 119.90, 112.83, 112.78, 112.72, 112.63, 112.57, 112.22, 112.06, 111.86, 70.23, 68.69, 48.25, 47.36, 35.41, 32.80, 15.16, 14.23, 14.19, 10.55, 10.52. HRMS (ESI+): m/z calcd for C27H28F2N5O3 [M + H]+, 508.21547; found, 508.21582.
2-(5-Fluoro-2-methyl-3-nitrophenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (34). To a solution of 1-bromo-5-fluoro-2-methyl-3-nitrobenzene (463 g, 1978 mmol) in dioxane was added bis(pinacolato)diboron (755 g, 2973 mmol) followed by potassium acetate (681 g, 6939 mmol)

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

and PdCl2(dppf)·DCM adduct (85 g, 104 mmol). The reaction mixture was refluxed at 110 °C for 3.5 h. After cooling to RT the mixture was diluted with EtOAc and washed with saturated aqueous NaHCO3 solution. The aqueous layer was back-extracted with EtOAc (2x). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated. The residue was purified by column chromatography (SiO2; heptane/EtOAc 9:1) to obtain 513 g of 34 as a beige solid. Yield: 92 %. 1H NMR (400 MHz, DMSO-d6) δ 7.93 (dd, 1H), 7.62 (dd, 1H), 2.50 (s, 3H, overlapping with DMSO), 1.32 (s, 12H).
5-Fluoro-2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (35). To a solution of 34 (509 g, 1811 mmol) in MeOH (4.6 L) in an autoclave was added Pd-C 10 % (50 g). The mixture was evacuated and purged with hydrogen. The reaction mixture was stirred at RT under a hydrogen atmosphere for 7 h. The mixture was filtered through a suction filter and the filter cake was washed with MeOH. The filtrate was concentrated. The residue was purified by column chromatography (SiO2; heptane/EtOAc gradient) to afford batch 1 (435 g) and batch 2 (5.4 g) of 35. Batch 1 of 35 (435 g) was dissolved in hexane (1400 mL) under reflux and was allowed to cool to RT over 2 h while stirring. The precipitate was filtered off and washed with cold hexane, then dried in vacuo to afford 387 g of 35 as a white solid. The mother liquor was concentrated and combined with batch 2 of 35 (5.4 g). The residue was recrystallised from hexane (100 mL) as described above to afford 36 g of 35 as a white solid. The two batches were combined. Yield: 93
%. [ES-MS] (ESI+): m/z calcd for C13H20BFNO2 [M + H]+, 252.2; found, 252.2. 1H NMR (400 MHz, DMSO-d6) δ 6.52 – 6.45 (m, 2H), 5.09 (s, 2H), 2.17 (s, 3H), 1.28 (s, 12H).
Methyl 4-cyclopropyl-2-fluorobenzoate (37). A solution of methyl 4-bromo-2-fluorobenzoate (20.00 g, 84 mmol) and cyclopropylboronic acid (9.68 g, 109 mmol) in toluene was purged with argon for 5 min. Then, tricyclohexylphosphine (2.36 g, 8.41 mmol) and water (1.82 mL, 101

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

mmol) were added. The mixture was purged with argon for an additional 10 min. Pd(OAc)2 (0.944 g, 4.21 mmol) was added and the reaction mixture was stirred at 100 °C overnight. The mixture was partitioned between EtOAc and water. The black suspension was filtered over a pad of Celite. The layers of the now clear mixture were separated. The aqueous layer was back-extracted with EtOAc. The combined organic layers were washed with saturated aqueous NaHCO3 solution and brine, dried over MgSO4, filtered and concentrated. The residue was purified by column chromatography (SiO2; cyclohexane/EtOAc gradient) to obtain 17.00 g (purity 95 %) of 37 as a brown oil. Yield: 99 %. [ES-MS] (ESI+): m/z calcd for C11H12FO2 [M + H]+, 195.1; found, 195.1. 1H NMR (400 MHz, DMSO-d6) δ 7.79 – 7.69 (m, 1H), 7.04 (s, 1H), 7.03 – 6.99 (m, 1H), 3.81 (s, 3H), 2.06 – 1.95 (m, 1H), 1.10 – 1.00 (m, 2H), 0.85 – 0.73 (m, 2H).
4-Cyclopropyl-2-fluoro-N-(5-fluoro-2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-

2-yl)phenyl)benzamide (38). To a suspension of 35 (17.00 g, 64.3 mmol) and 37 (16.24 g, 84.0 mmol) in THF (300 mL) at 0 °C was added NaHMDS (1 M in THF, 96 mL, 96.0 mmol) dropwise. The reaction mixture was stirred at RT for 1 h, then more NaHMDS (1 M in THF, 20 mL, 20.0 mmol) was added. The reaction mixture was stirred at RT for 1 h. Then, a third batch of NaHMDS (1.0 M in THF, 20 mL, 20.0 mmol) was added and the reaction mixture was stirred at RT for another 2 h. The mixture was diluted with EtOAc and washed with saturated aqueous NaHCO3 solution. The aqueous layer was back-extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated. The residue was suspended in EtOAc and stirred at RT for 20 min. The solid was filtered off, washed with EtOAc and dried in vacuo to afford a first batch of 38 (17.8 g) as a beige solid. The filtrate was concentrated and the residue was suspended in EtOAc. The suspension was stirred at RT for 20 min. The resulting solid was filtered off, washed with EtOAc and dried in vacuo to afford a second batch of 38 (3.4 g) as a

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

beige solid. The two batches were combined. Yield: 76 %. [ES-MS] (ESI+): m/z calcd for C23H27BF2NO3 [M + H]+, 414.2; found, 414.2. 1H NMR (400 MHz, DMSO-d6) δ 9.72 (d, J = 2.9 Hz, 1H), 7.67 – 7.59 (m, 1H), 7.52 (d, br, J = 9.9 Hz, 1H), 7.21 (dd, J = 8.7, 2.9 Hz, 1H), 7.10 – 7.03 (m, 2H), 2.38 (s, 3H), 2.08 – 1.97 (m, 1H), 1.32 (s, 12H), 1.10 – 1.01 (m, 2H), 0.83 – 0.75 (m, 2H).
6-Amino-4-chloro-5-methoxypyrimidine (40). To a solution of 4,6-dichloro-5- methoxypyrimidine (5.00 g, 27.9 mmol) in 2-propanol (40 mL) in an autoclave was added ammonium hydroxide (26 % in water, 5.0 mL, 33.5 mmol). The autoclave was sealed and the reaction mixture was stirred at 70 °C for 48 h. After cooling the mixture the precipitate was filtered off and washed with n-pentane and diethylether. The residue was dried in vacuo to obtain 4.40 g of 40 as a white solid. Yield: 94 %. [ES-MS] (ESI+): m/z calcd for C5H7ClN3O [M + H]+, 160.0; found, 160.1. 1H NMR (400 MHz, Chloroform-d) δ 8.12 (s, 1H), 5.29 (s, 2H), 3.89 (s, 3H).
6-Amino-4-chloro-5-pyrimidinol (41). To a solution of 40 (2.30 g, 13.69 mmol) in DCM (100 mL) at RT was slowly added boron tribromide (6.5 mL, 68.5 mmol) and the reaction mixture was stirred at 40 °C for 3 h. The mixture was quenched by slowly adding MeOH (50 mL) at -10 °C, stirred at RT for 12 h, then concentrated under reduced pressure. The residue was taken up in water and EtOAc. The mixture was basified with aqueous sodium carbonate solution (2 M) to pH 9-10. The aqueous layer was washed with EtOAc (discarded), acidified to pH 5-6 with aqueous HCl solution (5 M), then extracted with EtOAc (1x) and DCM (1x). The combined organic layers were washed with brine, dried over MgSO4, filtered and concentrated to afford 1.30 g (purity 90 %) of 41 as a beige solid. Yield: 59 %. [ES-MS] (ESI+): m/z calcd for C4H5ClN3O [M + H]+, 146.0; found, 146.0. 1H NMR (400 MHz, DMSO-d6) δ 9.57 (s, 1H), 7.78 (s, 1H), 6.89 (s, 2H).

 

 

 

ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27

 

 

tert-Butyl (2-((4-amino-6-chloropyrimidin-5-yl)oxy)ethyl)(methyl)carbamate (42). To a solution of 41 (2.00 g, 13.7 mmol) in THF (120 mL) was added a solution of N-Boc-N-methyl-2- hydroxyethylamine (6.74 g, 38.5 mmol) in THF (5 mL) followed by Smopex-301 (1 mmol/g) (34.4 g, 34.4 mmol) and a solution of DIAD (6.68 mL, 34.4 mmol) in THF (15 mL). The reaction mixture was stirred at 60 °C for 2 h. The mixture was filtered through a pad of Celite. The filtrate was concentrated to afford a yellow oil. The residue was treated with EtOAc and sonicated to provide a suspension which was allowed to stand at RT for 1 h. The precipitate was filtered off and dried in vacuo to obtain 2.33 g (purity 95 %) of 42 as a white solid. Yield: 53 %. [ES-MS] (ESI+): m/z calcd for C12H20ClN4O3 [M + H]+, 303.1; found, 303.0. 1H NMR (400 MHz, DMSO-d6) rotamers δ 7.97 (s, 1H), 7.27 (s, br, 2H), 4.04 – 3.89 (m, 2H), 3.59 – 3.48 (m, 2H), 2.97 – 2.81 (m, 3H), 1.47 – 1.30 (m, 9H).

28
29
30
tert-Butyl
(2-((4-amino-6-(3-(4-cyclopropyl-2-fluorobenzamido)-5-fluoro-2-methyl-

31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
phenyl)pyrimidin-5-yl)oxy)ethyl)(methyl)carbamate (43). The reaction was carried out using a Masterwave BTR microwave reactor with a 1000 mL vial. DME and water were purged with argon. To a solution of 42 (17.0 g, 56.2 mmol) and 38 (25.5 g, 61.8 mmol) in DME (264 mL) and water (38 mL) was added PdCl2(PPh3)2 (1.971 g, 2.81 mmol) followed by aqueous Na2CO3 solution (1 M, 168 mL, 168 mmol). The reaction mixture was heated at 110 °C under microwave irradiation for 15 min. More 38 (1.4 g, 3.4 mmol) and PdCl2(PPh3)2 (0.28 g, 0.40 mmol) were added. The reaction mixture was heated at 110 °C under microwave irradiation for an additional 10 min. The mixture was diluted with EtOAc (850 mL) and saturated aqueous NaHCO3 solution (710 mL) and transferred into a separatory funnel. The layers were separated, the aqueous layer was back-extracted with EtOAc (425 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated. The residue was triturated with EtOAc and the

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60

 

 

mixture was stirred at RT for 30 min. The precipitate was filtered off and washed with EtOAc to afford 24.13 g (purity 97 %) of 43 as a light grey solid. Yield: 74 %. [ES-MS] (ESI+): m/z calcd for C29H34F2N5O4 [M + H]+, 554.3; found, 554.3. 1H NMR (400 MHz, DMSO-d6) rotamers δ 9.74 (s, br, 1H), 8.18 (s, 1H), 7.69 – 7.62 (m, 1H), 7.62 – 7.55 (m, 1H), 7.10 – 7.03 (m, 2H), 7.03 – 6.90 (m, 3H), 3.57 – 3.47 (m, 2H), 3.28 – 3.22 (m, 2H), 2.58 (s, 3H), 2.09 – 1.97 (m, 1H) overlapping with 2.04 (s, 3H), 1.33 and 1.29 (2s, br, total 9H), 1.10 – 1.01 (m, 2H), 0.84 – 0.75 (m, 2H).
Kinase Binding Assays

The kinase-binding assays in screening or Kd determination modes have been described50,51 and were as follows: recombinant DNA-tagged kinase proteins were produced from kinase-tagged T7 phage strains or by producing the kinase protein in HEK-293 cells with subsequent tagging with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding. Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in 1x binding buffer (20% SeaBlock, 0.17x PBS, 0.05% Tween 20, 6 mM DTT). Test compounds were prepared as 111X stocks in 100% DMSO. Kds were determined using an 11-point 3-fold compound dilution series with three DMSO control points. All compounds for Kd measurements are distributed by acoustic transfer (non-contact dispensing) in 100% DMSO. The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.9%. All reactions were performed in polypropylene 384-well plates in a final volume of 0.02 ml. The assay plates were incubated at room temperature with shaking

 
ACS Paragon Plus Environment

 

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16

 

 

for 1 hour and the affinity beads were washed with wash buffer (1x PBS, 0.05% Tween 20). The beads were then re-suspended in elution buffer (1x PBS, 0.05% Tween 20, 0.5 μM non- biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.
Binding constants (Kd) were calculated with a standard dose-response curve using the Hill equation:

17
18
19
20
21

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>