ACS Medicinal Chemistry Letters
Letter
Table 5. Pharmacokinetics in Rat
IV
PO
compd
cLogP
Cl (L/h/kg)
Vss (L/kg)
T1/2 (h)
Cmax (ng/mL)
AUClast (ng/mL·h)
T1/2 (h)
F (%)
a
4
2.24
1.90
2.40
1.68
2.48
3.26
0.2
3.9
5.42
2.78
5.89
4.55
6.17
12.2
1.02
4.22
2.4
175
86
880
2.8
1.5
3.3
1.6
2.4
4.4
1.9
3.7
35
12
48
28
97
89
<1
b
9
3.67
5.44
5.04
2.40
2.16
1.65
4.39
0.8
249
a
10
1.2
198
291
1250
256
3.52
103
896
a
18
1.1
541
c
27
1.83
4.72
2.48
0.8
3980
1610
19.5
209
d
29
d
30
a
31
1.11
9
a
b
c
d
Dose: po at 10 mg/kg; iv at 5 mg/kg (n = 3). Dose: po at 8 mg/kg; iv at 4 mg/kg (n = 3). Dose: po at 10 mg/kg; iv at 10 mg/kg (n = 3). Dose:
po at 4 mg/kg; iv at 2 mg/kg (n = 3).
Carbonyl oxygen of imidazolone serves as a H-bond acceptor
and forms a specific H-bond with the backbone NH of Tyr631.
The flat pyridinylimidazolone core also anchors two side
moieties into their corresponding pockets well. Benzyl side
moiety fits nicely in a tight hydrophobic groove, where it forms
π−π interactions with its surrounding residues Trp659 and
Tyr666. Nitrogen atom of benzyl cyano group functions as an
H-bond acceptor to Arg125. However, the amino group from
piperidine site moiety is protonated under physiological pH
and forms an essential salt bridge with the backbone
carboxylate of Glu205 and Glu206. It should be mentioned
that this type of salt bridge interaction between enzyme and
ligand is a key feature of exopeptidase such as DPP-4. Naturally,
all marketed DPP-4 inhibitors contain such a protonable amino
group.
A large number of compounds identified with good potency
were selected for rat pharmacokinetics (PK) evaluation in our
lead optimization. Table 5 summarizes key parameters of some
examples. We had initial difficulty achieving good oral exposure
and bioavailability. As we built up a larger data set, we noticed a
reasonable connection between lipophilicity and PK properties.
Good oral compounds in this class of molecules starts to
populate around cLogP greater than 2, regardless of structural
diversity (Figure 4). We then utilized the SAR knowledge of
On the basis of its attractive in vitro and ex vivo potency (data
not shown), high selectivity over other subfamily enzymes, and
favorable pharmacokinetic profile, compound 27 was advanced
into in vivo efficacy studies. Compound 27 was first evaluated in
a proof-of-concept pharmacodynamics model, assessing its
ability to improve glucose tolerance in mice (OGTT, oral
glucose tolerance test). In the studies, a single dose of
compound 27 (0.1, 0.3, 1.0, and 3.0 mg/kg) was given orally to
C57BL/6 mice 30 min prior to the glucose challenge (3.0 g/
kg). The plasma glucose was then measured at 20, 40, 60, and
120 min after the glucose challenge, while DPP-4 activity, GLP-
1 level, and plasma drug concentration were monitored at 20
min. As shown in Table 7, compound 27 was able to reduce
plasma glucose excursion in a dose-dependent manner with a
minimum effective dose at 0.3 mg/kg (48% inhibition). The
observed glucose reduction tracked very well with drug
exposure, DPP-4 inhibition, and augmented GLP-1 level.
With regard to the comparison of Sitagliptin in the head-to-
head experiment, compound 27 showed better inhibition of
DPP-4 enzyme and comparable glucose reduction effects.9
To further characterize the effects in long-term treatment of
27 in rodent diabetic model, db/db mice were fed continuously
with food formulated-27 for 28 days. The random glucose level,
lipids, oral glucose tolerance test, GLP-1, DPP-4, and insulin
level were measured over the course of the study. Compound
27 has shown strong effects on all treated db/db mice in lowing
glucose level, reducing AUC of oral glucose tolerance test,
decreasing TG and HbA1c levels, and protecting beta-cell.
These effects correlated well with the inhibition of DPP-4 and
elevated the plasma GLP-1 level. In addition, no effects on the
body weights in db/db mice were observed under the test
conditions (data are not shown and will be published elsewhere
separately).
In order to assess the key areas for further development,
compound 27 was profiled extensively in a battery of target
independent drug-like properties and safety assays. Shown in
Table 8 are examples of these assays that include solubility,
permeability, protein binding, CYP450 inhibitions, hERG
channel inhibition/dog QT prolongation, Ames test, and in
vivo tox. These data demonstrate remarkable physicochemical
properties and good safety profile of 27 and strongly support its
developability.
The synthesis of 27 is illustrated in Scheme 1. The process
was well developed and validated at kiloscale to support early
development activities. The linear synthesis began from a
commercially available hydroxypyridone starting material 27−
01. Conversion of 27−01 under the treatment of POCl3
yielded dichloropyridine intermediate 27−02. Subsequently,
Figure 4. Bioavailability and cLogP.
the above-mentioned “sweet spot”, tuned in preferable
lipophilicity, and quickly identified good oral compound such
as 27 and 29. Of particular interest, compound 27
demonstrated a superior PK profile with a moderate clearance,
a high plasma exposure, and near 100% rat oral bioavailability.
These good PK attributes were translated into nonrodent
species of dog and monkey in our further characterizations
(Table 6).
D
dx.doi.org/10.1021/ml5001905 | ACS Med. Chem. Lett. XXXX, XXX, XXX−XXX