Sei sulla pagina 1di 4

Bioorganic & Medicinal Chemistry Letters xxx (2016) xxx–xxx

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry Letters


journal homepage: www.elsevier.com/locate/bmcl

Modification on the O-glucoside of Sergliflozin-A: A new strategy for


SGLT2 inhibitor design
Xuefeng Cao a,y, Wenpeng Zhang b,y, Xu Yan a, Zhi Huang a, Zhenqing Zhang b, Peng Wang a,⇑, Jie Shen a,⇑
a
State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, PR China
b
State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Poor pharmacokinetic stability is one of the issues of O-glucoside SGLT2 inhibitors in clinical trials, hence
Received 18 February 2016 C-glucoside inhibitors have been developed and extensively applied. Herein, we provided an alternative
Revised 14 March 2016 approach to improve the pharmacokinetic stability of such inhibitors. Nine derivatives of Sergliflozin-A
Accepted 16 March 2016
with modifications on the O-glucoside fragment were prepared, among which the 4-O-methyl derivative
Available online xxxx
exhibited similar pharmacodynamics potency in excreted glucose urine test. Most attractively, signifi-
cantly increased pharmacokinetic stability was observed for 4-O-methyl derivative of O-glucosides.
Keywords:
This work proved that modification on the O-glucoside fragment could be a promising approach to the
SGLT2 inhibitor
Sergliflozin-A
future SGLT2 inhibitor design.
O-Glucoside Ó 2016 Elsevier Ltd. All rights reserved.
Pharmacokinetic stability

During the past few years, great efforts have been devoted to mide donor A0 was coupled with the aglycon acceptor S219 to
develop sodium glucose co-transporter 2 (SGLT2) inhibitors as a afford the intermediate A1 which was deacetylated to give the
new class of drugs for the treatment of diabetes.1–8 These com- compound A, Sergliflozin-A. Promoted by boron trifluoride ether-
pounds (Fig. 1) could effectively relieve diabetic hyperglycemia ate, the glycosyl donor B019 was coupled with S2 to provide the
via lowering the renal glucose reabsorption.9–11 Phlorizin, isolated intermediate B1, which was sequentially deacetylated and methy-
from the bark of apple roots, was the first O-glucoside compound lated at its O2 position to afford the 2-O-methyl 3,4,6-tri-O-benzyl
which can inhibit both SGLT1 and SGLT2.12,13 Other O-glucoside derivative B3. Pd-catalyzed debenzylation of B3 gave the com-
inhibitors with high selectivity towards SGLT2 were also devel- pound B. The 2-fluorinated glucosyl Schimidt donor C019 was cou-
oped. However, most of O-glucoside inhibitors, for example, T- pled with S2 to provide the intermediate C1 as an unseparable a/b
109514 and Sergliflozin,15,16 were suspended. Currently, majority mixture.19 After the sequential debenzylation, acetylation and flash
of the launched SGLT2 inhibitors, e.g., Dapagliflozin,4 chromatography separation, C1 was converted to the intermediate
Empagliflozin6,17 and Canagliflozin,7,18 are C-glucosides. This could C3 as a pure b isomer, which was converted to the compound C by
at least partially be attributed to the poor metabolic stability of O- the global deacetylation. The compounds D, E, I were prepared
glucosides, arising from the glucosidase mediated hydrolysis. from the 1-acetylated donors D0,19 E019 and I019 respectively via
Alternatively, it is understandable that modification on the O- a similar two-step procedure, including a glycosylation reaction
glucoside fragment may affect the substrate-glucosidase binding, followed by a global deacetylation. The compound F was prepared
thus could also help to improve in vivo stability. In this work, tak- from compound A in 5 steps. Specifically, after the 4,6-benzylide-
ing Sergliflozin-A (active form of Sergliflozin) as a parent structure, nation and 2,3-benzylation reactions, A was converted to the inter-
a series of derivatives with modification on hydroxyl groups (e.g., mediate F2. Regioselective and reductive ring opening of the 4,6-
methylated, fluorine-substituted, deoxidized) of its O-glycoside benzylidene of F2 gave the intermediate F3 whose 4-hydroxyl
were synthesized (Fig. 2) and evaluated. group was then methylated with MeI. Then the obtained interme-
The synthesis of Sergliflozin-A and its nine derivatives were diate was converted to the product F via global debenzylation.
illustrated in Scheme 1. Briefly, the peracetylated a-glucosyl bro- Regioselective benzoylation of the thioglycoside G019 gave the
intermediate G1 whose 4-hydroxyl group was thiocarbonylated
to give the intermediate G2. Barton–McCombie radical reduction
⇑ Corresponding authors. was then performed to convert G2 to the 4-deoxy intermediate,
E-mail addresses: pwang@nankai.edu.cn (P. Wang), jieshen@nankai.edu.cn (J. which was converted to 1-acetylated donor compound G3. Then
Shen). G3 was coupled with S2 to afford the glycosylation product G4,
y
Authors contributed equally to this work.

http://dx.doi.org/10.1016/j.bmcl.2016.03.065
0960-894X/Ó 2016 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Cao, X.; et al. Bioorg. Med. Chem. Lett. (2016), http://dx.doi.org/10.1016/j.bmcl.2016.03.065
2 X. Cao et al. / Bioorg. Med. Chem. Lett. xxx (2016) xxx–xxx

OH

HO OH OH O OMe

O O O O O O O O
HO RO RO

HO OH HO OH HO OH
OH OH OH
Phlorizin R=-H, T-1095A R=-H, Sergliflozin-A
R=CH3OCO-, T-1095 R=CH3CH2OCO-, Sergliflozin

Cl O Cl O

O O O HO O
HO HO S

HO OH HO OH HO OH F
OH OH OH
Dapagliflozin Empagliflozin Canagliflozin

Figure 1. Important SGLT2 inhibitors.

OH OH OH OH
HO O HO O HO O HO O
HO OR HO OR HO OR H3 CO OR
OH OCH 3 F OH
A B C D
(Sergliflozin-A) (2-OMe) (2-F) (3-OMe)
OH OH OH OCH 3
HO O H CO O H O HO O
H OR 3 HO OR HO OR HO OR
OH OH OH OH
E F G H
(3-deoxy) (4-OMe) (4-deoxy) (6-OMe)
H F
HO O HO O OMe
HO OR HO OR
OH OH R=
I J
(6-deoxy) (6-F)

Figure 2. Sergliflozin-A and its 9 derivatives.

which was converted to the compound G via the global de-esteri- cation at these two hydroxyl groups (compounds B, C, D, E)
fication. Syntheses of the last two compounds H and J shared the almost completely abolished the hypoglycemic effect. In contrast,
same intermediated H1, which was prepared from A via regioselec- 4-methylated derivative F and the 6-deoxy derivative H are the
tive protection of 6-hydrxoyl group with trityl chloride. For the two most potent derivatives, which maintained 73% and 31%
synthesis of H, H1 was per-benzylated to afford the intermediate in vivo potency compare with the parent compound A. These
H2, whose trityl group at O6 was replaced with a methyl group observations were in good consistence with structural data, where
in two steps. The therefore obtained intermediated H4 was con- both 2- and 3-hydroxyl groups formed at least two hydrogen
verted to the compound H via global debenzylation. For the syn- bonds with the bacterial vSGLT (PDB# 3DH4)20,21 or human
thesis of J, H1 was per-acetylated to afford the intermediate J2. SGLT222 in their substrate recognition pocket. On the contrary,
After the trityl group of J2 was removed, DAST reagent was utilized the 4- and 6-hydroxyl groups bound much loosely to the pocket
to accomplish the fluorine substitution reaction at C6, so as to con- and each of them formed only one hydrogen bond in both
vert J3 to J4, whose global deacetylation led to the product J. structures.
As for the glycosylation reactions that promoted by boron triflu- We also envisioned that pharmacokinetic factors might also
oride etherate to give products B1, D1, E1, I1 and G4, product iso- benefit the in vivo potency of the compound F. To compare the
merization at 0 °C was observed for B1 which is an armed metabolic stability of glucoside and its 4-methylated derivative,
glycoside. As the kinetically favored b-isomer, B1 was formed as two fluorescent compounds K and L were prepared and treated
the major product initially with little amount of a-isomer. How- in rat liver homogenate for 8 h. Whereas over 80% glucoside K
ever, it was gradually converted to its more stable a-isomer. There- was hydrolyzed, neglected amount of hydrolysis product could
fore whereas the glycosylation of the disarmed D1 was performed be detected for the corresponding 4-methylated compound L
at 0 °C, the other the other four reactions were performed at (Fig. 4). It should be addressed that the poor metabolic stability
20 °C. was one of the reasons for the failure of O-glucoside SGLT2 inhibi-
Rat urinary glucose excretion experiments were then per- tors in clinical trials.
formed to evaluate the in vivo hypoglycemic effects of the ten It is now well accepted that the topological polar surface area
compounds A–J (Fig. 3). The results clearly indicated that the 2- (tPSA) of an orally administrated drug should be less than
hydroxyl group and 3-hydroxyl group were essential to the inhibi- 140 Å2. Considering the tPSA value of a glucoside fragment is
tion potency of the compound A against SGLT2, since any modifi- around 100 Å2, it is of no wonder that the glycons of all SGLT2 inhi-

Please cite this article in press as: Cao, X.; et al. Bioorg. Med. Chem. Lett. (2016), http://dx.doi.org/10.1016/j.bmcl.2016.03.065
X. Cao et al. / Bioorg. Med. Chem. Lett. xxx (2016) xxx–xxx 3

OAc OAc OH OMe OMe


O O O
(1) AcO
AcO a AcO
AcO OR b HO
HO OR R=
AcO OAc OH
Br OH
S2
A0 A1 A
OBn OBn OBn OBn OH
O O O O O
(2) BnO
BnO OAc
c BnO
BnO OR
b BnO
BnO OR d BnO OR e HO
HO OR
BnO
OAc OAc OH OMe OMe
B0 B1 B2 B3 B

OBn NH OBn OH OAc OH


(3) BnO O BnO O HO O AcO O HO O
BnO O CCl3 f BnO OR e HO OR g AcO OR b HO OR
F F F F F
C0 C1 C2 C3 C
R2 R2 R2
O O O
(4) AcO 1 OAc c for E1,I1 AcO 1 OR b HO 1 OR
R R R
OAc h for D1 OAc OH
R1 =-OMe,R 2 =-OAc,D0 R1 =-OMe,R 2 =-OAc,D1 R1 =-OMe,R 2 =-OH,D
R1 =-H,R 2=-OAc,E0 R1 =-H,R 2=-OAc, E1 R1 =-H,R 2=-OH,E
R1 =-OAc,R 2 =-H,I0 R1 =-OAc, R 2=-H, I1 R1 =-OH,R2 =-H,I
OH Ph OBn OH
Ph O O
O O O O O
(5) HO
HO OR i O
HO OR j O
BnO OR k HO
BnO OR d,e MeO
HO OR
OH OH OBn OBn OH
A F1 F2 F3 F
S
OH OBz OBz OBz OBz
O HO O O H O O
(6) HO
AcO STol l AcO STol m
PhO O
AcO STol n, o AcO OAc c
H
AcO OR
OAc OAc OAc OAc OAc
G0 G1 G2 G3 G4
OH
H O
b HO OR
OH
G
OH OTrt OTrt OH R4
O O O R 3O O 3 O
HO
OR p ORHOj for H2 R 3O
OR q OR d for H4 RRO OR
(7) HO HO R3 O R3 O 3
O
OH OH g for J2 OR3 OR 3 r f or J4 OR 3
A H1 R =-OBn, H2
3 R =-OBn,H3
3
R =-OBn, R =-OMe,H4
3 4

R4 R 3=-OAc,J2 R 3=-OAc,J3 R 3=-OAc, R 4 =-F,J4


HO O
e for H HO OR
b for J OH
R 4=-OMe,H
R 4=-F,J

Scheme 1. Chemical synthesis route of compound A–J. Reagents and conditions: (a) S2, NaOH, TBAB, 53%; (b) NaOMe, MeOH, A 87%, B2 53% 2 steps from B0, C 88%, D 79% 2
steps from D0, E 63% 2 steps from E0, I 85%, G 58% 3 steps from G2, J 78%; (c) S2, BF3–Et2O, 20 °C for 24 h. I1 47%, B1, E1, G4 without purification; (d) MeI, NaH, B3 93%, H4
96%, F4 90%; (e) Pd/C, H2, B 83%, C2 without purification, F 80%, H 81%; (f) S2, BF3–Et2O, 40 °C to rt without purification; (g) Ac2O, pyridine, C3 66% 3 steps from C0, J2 91% 2
steps from J0; (h) S2, BF3–Et2O, 0 °C to rt without purification; (i) PhCH(OMe)2, CSA, 80%; (j) NaH, BnBr, F2 95%, H2 96%; (k) Et3SiH, CF3COOH, 80%; (l) BzCl, pyridine, 80%; (m)
carbonochloridothioic acid, O-phenyl ester, DMAP, 85%; (n) Bu3SnO, AIBN, without purification; (o) NIS, AcOH without purification; (p) TrtCl, pyridine, 68%; (q) 33% HBr in
HOAc, H3 85%, J3 74%; (r) DAST, s-collidine, 63%.

bitors in clinical trials have no more than three N and O atoms in


sum. Therefore, depolarization modification on the glucoside
might provide a chance to introduce a polar functional group at
the aglycon fragment without affecting intestinal absorption. As
expected, The log P values of A and F were experimentally deter-
mined to be 1.32 and 1.83, and their tPSA were calculated to be
109 and 98 Å2,23 indicating that 4-methylated derivative F more
suitable to accept a polar functional group onto its aglycon. The
current work therefore provided a new approach to new SGLT2
inhibitors.
Taken together, nine derivatives with modifications on O-glu-
coside fragment of Sergliflozin-A were prepared and the excreted
glucose urine experiments were performed. The results showed
the 2-OH and 3-OH are essential for SGLT2 inhibition potency,
and modifications on the two hydroxyl groups completely abol-
Figure 3. Excreted glucose urine results of ten compounds.
ished the inhibition. In contrast, 4-O-methyl compound F showed
almost equal SGLT2 inhibition compared to Sergliflozin-A. It should

Please cite this article in press as: Cao, X.; et al. Bioorg. Med. Chem. Lett. (2016), http://dx.doi.org/10.1016/j.bmcl.2016.03.065
4 X. Cao et al. / Bioorg. Med. Chem. Lett. xxx (2016) xxx–xxx

References and notes

1. Tsujihara, K.; Hongu, M.; Saito, K.; Kawanishi, H.; Kuriyama, K.; Matsumoto, M.;
Oku, A.; Ueta, K.; Tsuda, M.; Saito, A. J. Med. Chem. 1999, 42, 5311.
2. Ehrenkranz, J. R.; Lewis, N. G.; Ronald Kahn, C.; Roth, J. Diab. Metab. Res. Rev.
2005, 21, 31.
3. Katsuno, K.; Fujimori, Y.; Takemura, Y.; Hiratochi, M.; Itoh, F.; Komatsu, Y.;
Fujikura, H.; Isaji, M. J. Pharmacol. Exp. Ther. 2007, 320, 323.
4. Han, S.; Hagan, D. L.; Taylor, J. R.; Xin, L.; Meng, W.; Biller, S. A.; Wetterau, J. R.;
Washburn, W. N.; Whaley, J. M. Diabetes 2008, 57, 1723.
5. Meng, W.; Ellsworth, B. A.; Nirschl, A. A.; McCann, P. J.; Patel, M.; Girotra, R. N.;
Wu, G.; Sher, P. M.; Morrison, E. P.; Biller, S. A.; Zahler, R. J. Med. Chem. 2008, 51,
1145.
6. Grempler, R.; Thomas, L.; Eckhardt, M.; Himmelsbach, F.; Sauer, A.; Sharp, D. E.;
Bakker, R. A.; Mark, M.; Klein, T.; Eickelmann, P. Diab. Obes. Metab. 2012, 14, 83.
7. Lamos, E. M.; Younk, L. M.; Davis, S. N. Expert Opin. Drug Metab. Toxicol. 2013, 9,
763.
8. Miao, Z.; Nucci, G.; Amin, N.; Sharma, R.; Mascitti, V.; Tugnait, M.; Vaz, A. D.;
Callegari, E.; Kalgutkar, A. S. Drug Metab. Dispos. 2013, 41, 445.
9. Ahmed, D.; Sharma, M.; Kumar, V.; Subhashchandra, Y. P. J. Diab. Metab. 2014,
5, 358.
10. Idris, I.; Donnelly, R. Diab. Obes. Metab. 2009, 11, 79.
11. Verspohl, E. J. Pharmacol. Rev. 2012, 64, 188.
12. Panayotova-Heiermann, M.; Loo, D. D.; Wright, E. M. J. Biol. Chem. 1995, 270,
27099.
13. Kanai, Y.; Lee, W. S.; You, G.; Brown, D.; Hediger, M. A. J. Clin. Invest. 1994, 93,
397.
Figure 4. The pharmacokinetic stability of compound K and compound L. 14. Isaji, M. Curr. Opin. Investig. Drugs 2007, 8, 285.
15. Hussey, E. K.; Clark, R. V.; Amin, D. M.; Kipnes, M. S.; O’Connor-Semmes, R. L.;
O’Driscoll, E. C.; Leong, J.; Murray, S. C.; Dobbins, R. L.; Nunez, D. J. Diabetes
2007, 56, A189.
be emphasized that the stability of 4-O-methyl compound L was 16. Dobbins, R. L.; O’Connor-Semmes, R.; Kapur, A.; Kapitza, C.; Golor, G.;
significantly improved. Meanwhile, due to the decreased molecular Mikoshiba, I.; Tao, W.; Hussey, E. K. Diab. Obes. Metab. 2012, 14, 15.
polarity through such a modification, it provided a chance to 17. Luippold, G.; Klein, T.; Mark, M.; Grempler, R. Diab. Obes. Metab. 2012, 14, 601.
18. Nomura, S.; Sakamaki, S.; Hongu, M.; Kawanishi, E.; Koga, Y.; Sakamoto, T.;
explore SGLT2 inhibitors with a polar functional group at its agly- Yamamoto, Y.; Ueta, K.; Kimata, H.; Nakayama, K.; Tsuda-Tsukimoto, M. J. Med.
con in the future. Chem. 2010, 53, 6355.
19. For preparation details see Supporting information.
20. Watanabe, A.; Choe, S.; Chaptal, V.; Rosenberg, J. M.; Wright, E. M.; Grabe, M.;
Acknowledgment Abramson, J. Nature 2010, 468, 988.
21. Enkavi, G.; Li, J.; Wen, P.; Thangapandian, S.; Moradi, M.; Jiang, T.; Han, W.;
This study was supported by National Natural Science Founda- Tajkhorshid, E. Annu. Rep. Comput. Chem. 2014, 10, 77.
22. Xu, J.; Yuan, H.; Ran, T.; Zhang, Y.; Liu, H.; Lu, S.; Xiong, X.; Xu, A.; Jiang, Y.; Lu,
tion of China (No. 21272127). T.; Chen, Y. J. Mol. Recognit. 2015, 28, 467.
23. Predicted by ChemDraw 12.0.
Supplementary data

Supplementary data associated with this article can be found, in


the online version, at http://dx.doi.org/10.1016/j.bmcl.2016.03.
065.

Please cite this article in press as: Cao, X.; et al. Bioorg. Med. Chem. Lett. (2016), http://dx.doi.org/10.1016/j.bmcl.2016.03.065

Potrebbero piacerti anche