OA18588A - 1,1,1-Trifluoro-3-Hydroxypropan-2-yl Carbamate Derivatives and 1,1,1-Trifluoro-4Hydroxybutan-2-yl Carbamate Derivatives as Magl Inhibitors. - Google Patents

1,1,1-Trifluoro-3-Hydroxypropan-2-yl Carbamate Derivatives and 1,1,1-Trifluoro-4Hydroxybutan-2-yl Carbamate Derivatives as Magl Inhibitors. Download PDF

Info

Publication number
OA18588A
OA18588A OA1201800030 OA18588A OA 18588 A OA18588 A OA 18588A OA 1201800030 OA1201800030 OA 1201800030 OA 18588 A OA18588 A OA 18588A
Authority
OA
OAPI
Prior art keywords
alkyl
trifluoro
formula
oxa
cycloalkyl
Prior art date
Application number
OA1201800030
Inventor
Bruce Nelsen Rogers
Patrick Robert Verhoest
Christopher John Helal
Douglas Scott Johnson
Steven Victor O'neil
Michael Aaron Brodney
Christopher Ryan BUTLER
Adam Matthew Gilbert
Elizabeth Mary BECK
Laura Ann Mcallister
Justin Ian Montgomery
Damien Webb
Original Assignee
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc. filed Critical Pfizer Inc.
Publication of OA18588A publication Critical patent/OA18588A/en

Links

Abstract

The present invention provides, in part, compounds of Formula I

Description

1.1.1-TRIFLUORO-3-HYDROXYPROPAN-2-YL
CARBAMATE DERIVATIVES AND 1,1,1-TRIFLUORO-4HYDROXYBUTAN-2-YL CARBAMATE DERIVATIVES AS MAGL INHIBITORS
FIELDOFTHE INVENTION
The présent invention relates to novel 1,1,1-trifluoro-3-hydroxypropan-2-yl carbamate dérivatives and 1,1,1-trifluoro-4-hydroxybutan-2-yl carbamate dérivatives, which are monoacylglycerol lipase (MAGL) inhibitors, pharmaceutically compositions thereof, and uses thereof in the treatment of MAGL-mediated disorders such as pain, an inflammatory disorder, traumatic brain injury, dépréssion, anxiety, Alzheimeris disease, a metabolic disorder, stroke, or cancer.
BACKGROUND OF THE INVENTION
MAGL Is the principal enzyme responsable for the in vivo dégradation of 2-arachidonoyl glycerol (2-AG), an endogenous ligand of the cannabinoid receptors (e.g., CB1 and CB2). See e.g,, Patel, J. Z. et al., Loratadine analogues as MAGL inhibitors, Bioorg. Med. Chem. Lett., 2015, 25(7): 1436-42; Mechoulam, R. et al., Identification of an endogenous 2-monoglyceride, présent In canine gut, that blnds to cannabinoid receptors* Biochem. Pharmacol., 50 (1995), 8390; Suglura, T. et al., “2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand In brain, Biochem. Biophys. Res. Commun., 215 (1995), 89-97.
There continues to be a need for alternative MAGL inhibitors.
SUMMARY OF THE INVENTION
The présent invention provides, in part, a novel compound of Formula I:
or a pharmaceutically acceptable sait thereof, wherein:
each of R1 and R2 is independently Cm alkyl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and C>7 cycloalkyl, wherein the Cj.7 cycloalkyl is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkyl, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy;
or R1 and R2, together with the N atom to which they are attached, form 4- to 14membered heterocycloalkyl that Is optionally substituted with Ra and optionally substituted with one or more independently selected R® or R30;
each of R3 and R4 Is independently H, halogen, OH, Cm alkyl, or C».7cycloalkyl, wherein the Cm alkyl of R3 and R4 Is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and Cm cycloalkyl, and wherein the C3-7 cycloalkyl of R3 and R4 is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Ci4 alkyi, Cm haloalkyi, Cm alkoxy, and Cm haloalkoxy;
or R3 and R4, togetherwith the C atom to which they are attached, form C3.7 cycloalkyl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkyi, Cm haloalkyi, Cm alkoxy, and Cm haloalkoxy;
each of Rsand Re is independently H, Cm alkyl, or C3-7cycloalkyl, wherein the Cm alkyi of R5 and R9 is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and CMcycioalkyl, and wherein the C3-7 cycloalkyl of R5 and R9 is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkyl, Cm haloalkyi, Cm alkoxy, and Cm haloalkoxy;
or R’and R®, together with the C atom to which they are attached, form C3-7cycloalkyl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkyl, Cm haloalkyi, Cm alkoxy, and Cm haloalkoxy;
R7 is H, Cm alkyl, C3.7cycloalkyl, or R10, wherein the Cm alkyl of R7 is optionally substituted with one or more substituents each Independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and Cmcycloalkyl, and wherein the C3-7 cycloalkyl of R7 is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkyl, Cm haloalkyi, Cm alkoxy, and Ci4 haloalkoxy;
or R7and R’, togetherwith the intervening moiety of *C(RS)-O to which they are attached, form 4- to 7-membered heterocycloalkyi or 5- to 10-membered heteroaryl that Is optionally substituted with one or more substituents each independently selected from the group consisting of OH, oxo, halogen, Cm alkyl, Cm haloalkyi, Cm alkoxy, and Cm haloalkoxy, and wherein each of the ring-formlng atoms of the 4- to 7-membered heterocycloalkyi is independently C, N, O, S, or P and wherein each of the ring-forming atoms of the 5- to 10membered heteroaryl is C, N, O, or S;
or R7 and R3, together with the intervening moiety of “C(R4)-C(R’R®)-0 to which they are attached, form a 5- to 7-membered heterocycloalkyi or 5- to 10-membered heteroaryl that is optionally substituted with one or more substituents each Independently selected from the group consisting of OH, oxo, halogen, Cm alkyl, Cm haloalkyi, Cm alkoxy, and Cm haloalkoxy, and wherein each of the ring-forming atoms of the 5- to 7-membered heterocycloalkyi is independently C, N, O, S, or P, and wherein each ofthe ring-forming atoms ofthe 5- to 10membered heteroaryl is C, N, O, or S;
R® is -L’-R11, -L2-R12, -L3-R13, -L4-R14, -C(R15)(Cy’HCy2}, -C(R15)(Cy’)[-NR23-S(=O)2Cy2], or-L’-N(-L9-Cy3)(-L7-C/);
each R9 is independently OH, oxo, halogen, optionally substituted Cm alkyl, optionally substituted Cu alkoxy, or optionally substituted C» cycloalkyl;
R10 is -P(=O)(OR9,)(ORB2) or-S(=O)2OR»;
each of L1, L2, L3, and L4 is Independently absent, -(CR21R22)m-, -NR23-, -O-, -C(=O)-, S(=0)r, -S(=O)2-(CR21R22)n-, -C(=O)-(CR21R22)n-, -S(=O)rNR23-, -C(=O)-NR23-, -(CR^R22),,NR23-(CR21R22)f2-, -(CR21R22)frO-(CR21R22)f2·, -C(=O)-NR23-(CR2,R22)P-, or-S(=O)2-NR23(CR21R22)P-;
L9 Is absent or -(CR21R22)-;
L’Is absent or -(CR21R22)-;
L7 is absent, -(CR21Ri2)-, or -S(=O)2-;
R is 5- to 10-membered heteroaryl optionally substituted with one or more independently selected R31;
R12 is 4- to 14-membered heterocycloalkyl optionally substituted with one or more independently selected R32;
R13 is Ce-toaryl optionally substituted with one or more independently selected R33;
R14 is C3.U cycloalkyl optionally substituted with one or more independently selected R34;
R13 is H, OH, halogen, Cm alkoxy, Cm alkyl, or cyclopropyl;
each of R21 and R22 Is independently H, OH, halogen, Cm alkyl, or cyclopropyl, wherein the Ci-3 alkyl Is optionally substituted with one or more substituents each Independently selected from the group consisting of OH, halogen, C1-3 alkoxy, C1-3 haloalkoxy, and cyclopropyl;
R23 is H, Cm alkyl, or cyclopropyl;
each of R“, R3’, R32, R33, and R34 is independently selected from the group consisting of halogen, -N(R*)(Rb), -N(Rc)(C(=O)Rd), -N(Re)(S(=O)2Rd), -C(=O)-N(R')(Rb), -C(=O)-Rd, -C(=O)ORd, -OC(=O)-Rd, -N(Rq)(S(=O)2Rd), -S(=O)2-N(R*)(Rb), -SRd, -S(=O)2Rd, -ORd, -OR33. -CN, C,. e alkyl, Cm alkenyl, Cm alkynyl, C3-10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl,
5- to 10-membered heteroaryl, (C3.10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-w aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3-10 cycloalkyl, 4- to 10membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-to aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-CM alkyl- is optionally substituted with one or more independently selected R35; and wherein each of the Cm alkyl, C3.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl·, and (5- to 10-membered heteroaryl)-CM alkyl- is further optionally substituted one or more oxo;
each R33 is independently selected from the group consisting of H, Cm alkyl, C3-10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-io aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-Ci-4 alkyl-, wherein each of the Cm alkyl, Cno cycloalkyl, 4- to 10-membered heterocycloalkyl, Cmo aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)Cu alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-Ci-4 alkyl- is optionally substituted with one or more substituents independently selected from the group consisting of halogen, -CN, -C(=0)Cm alkyl, -C(=O)OH, -C(=0)0-Cm alkyl, -C(=O)NHCm alkyl, -C(=O)N(Cm alkyl)2, oxo, -OH, -OC(=O)-Cm alkyl, 0C(=0)0-Cm alkyl, -NH2, -NH(Cm alkyl), -N(Cm alkyl)2, -NHC(=O)Cm alkyl, -NHC(=O)OCm alkyl, -NHC(=O)NHCm alkyl, and Cm alkoxy:
each R36 Is Independently selected from the group consisting of halogen, -OH, -NO2, CN, -SFs, Cm alkyl, Cm haloalkyl, Cm haloalkoxy, Cm alkenyl, Cm alkynyl, C3.7 cycloalkyl, a 4to 10-membered heterocycloalkyl, -N(Re)(Rb), -N(Re)(C(=O)Rd), -C(“O)-N(R')(Rb), -C(=O)-Rd, C(=O)-ORd, -OC(=O)-Rd, -N(Re)(S(=O)2Rd), -S(=O)2-N(R*)(Rb), -SRd, -S(=O)2Rd, and -ORd, wherein each of the Cm alkyl, C3.7 cycloalkyl, and heterocycloalkyl is optionally substituted with one or more substituents each Independently selected from the group consisting of halogen, CN, -OH, Cm alkyl, Cm alkoxy, Cm haloalkyl, Cm haloalkoxy, Cm cycloalkyl, -N(R*)(Rb), N(Re)(C(=O)Rd), -C(=O)-ORd, -C(=O)H, -C(=O)Rd, -C(=O)N(R‘)(Rb), -N(Re)(S(=O)2Rd), -S(=O)zN(Ri)(Rb), -SRd, -S(=O)2Rd, and -ORd;
each of R81, RBZ, and R90 Is Independently selected from the group consisting of H, Cm alkyl, C3.7 cycloalkyl, and (C3-7 cycloalkyl)-CM alkyl-, wherein each of the Cm alkyl, C3.7 cycloalkyl, and (C3.7 cycloalkyl)-CM alkyl- is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CN, -OH, oxo, -NH2, NH(Cm alkyl), -N(Cm alkyl)2, Cm alkyl, Cm alkoxy, Cm haloalkyl, Cm haloalkoxy, Cm cycloalkyl;
or OR81 and OR82, together with the P(=O) to which they are attached, form 4- to 10membered heterocycloalkyl that is further optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CN, -OH, oxo, -NH2, NH(Cm alkyl), -N(Cm alkyl)2, Cm alkyl, Cm alkoxy, Cm haloalkyl, Cm haloalkoxy, Cm cycloalkyl;
each of Cy1, Cy2, Cy3, and Cy4 is independently selected from the group consisting of R11, R12, R13, and R14;
each R· is independently H, Cm alkyl, Cm haloalkyl, Cj.? cycloalkyl, or (C3.7 cycloalkyl)Cm alkyl-;
each Rb Is Independently H or selected from the group consisting of Cm alkyl, Cm haloalkyl, C3-7 cycloalkyl, a 4- to 10-membered heterocycloalkyl, Ce-n aryl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-w aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, wherein each of the sélections from the group is optionally substituted with one or more substituents each Independently selected from the group consisting of -OH, -CN, Cm alkyl, C3-7 cycloalkyl, Cm hydroxylalkyl, -SCm alkyl, -C(=O)H, -C(=0)-Cm alkyl, -C(=0)-0-Cm alkyl, -C(=O)-NH2, -C(=0)-N(Cm alkyl)2, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy;
or R* and Rb, together with the N atom to which they are attached, form a 4- to 10membered heterocycloalkyl or a 5- to 10-membered heteroaryl, each optionally substituted with one or more substituents each independently selected from the group consisting of halogen, OH, oxo, -C(=O)H, -C(=O)OH, -C(=0)-Cm alkyl, -C(=O)-NH2, -C(=0)-N(Cm alkyl)2, -CN, Cm alkyl, Cm cycloalkyl, (CM cycloalkyl)-Ci-2 alkyl-, Cm alkoxy, Cm hydroxylalkyl, Cm haloalkyl, and Cm haloalkoxy;
each Rc is independently selected from the group consisting of H, Cm alkyl, C3.7 cycloalkyl, and (C3.7 cycloalkyl)-CM alkyl-;
each Rd is independently selected from the group consisting of Cm alkyl, C3.7 cycloalkyl, a 4- to 14-membered heterocycloalkyl, Ce-ioaryl, a 5- to 10-membered heteroaryl, (Ca-7 cycloalkyl)-Cm alkyl-, (4-to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, wherein each of the sélections from the group Is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CF31 -CN, -OH, oxo, -S-Cm alkyl, Cm alkyl, Cm haloalkyl, C2_e alkenyl, C2_e alkynyl, C3.7 cycloalkyl, Cm alkoxy, and Cm haloalkoxy;
each of f1 and f2 is independently 0,1, or 2, provided that the sum of f1 and f2 is 1, 2, or 3;
m is 1,2, or 3;
n is 1, 2, or 3;
p is 1, or 2; and ris 0 or 1, provided that when r is 1 and each of R3, R4, Rs and R® is H, then the moiety of *N(R1)(R2) is other than optionally substituted 4-oxo-3H-5,6,7,8-tetrahydropyrido[3,4dJpyrimÎdin-7-yl.
In some embodiments, the compound of Formula l or pharmaceutically acceptable sait thereof is a compound of Formula 1-1:
or pharmaceutically acceptable sait thereof.
In some embodiments, the compound of Formula I or pharmaceutically acceptable sait thereof is a compound of Formula l-a:
l-a or pharmaceutically acceptable sait thereof.
In some embodiments, the compound of Formula I (or 1-1 or l-a) or pharmaceutically acceptable sait thereof is a compound of Formula l-a1 :
or pharmaceutically acceptable sait thereof.
In some embodiments, the compound of Formula I or pharmaceutically acceptable sait thereof Is a compound of Formula l-b:
or pharmaceutically acceptable sait thereof.
In some embodiments, the compound of Formula I (or 1-1 or l-b) or pharmaceutically acceptable sait thereof Is a compound of Formula l-b 1 :
l-b1 or pharmaceutically acceptable sait thereof.
Unless otherwise specified, the compound of Formula I or a sait thereof described In the 20 following embodiments can be a compound of Formula 1-1, l-a, l-a1, l-b, or l-b1, or a sait thereof.
In some embodiments, R1 and R2, together with the N atom to which they are attached, form 4- to 14-membered heterocycloalkyl that Is optionally substituted with R’ and optionally substituted with one or more independently selected R9 or R30.
In some embodiments, R1 and R2, together with the N atom to which they are attached, form 4- to 14-membered heterocycloalkyl that is optionaiiy substituted with R8 and optionally substituted with one or more independently selected Re.
In some embodiments, R1 and R2, together with the N atom to which they are attached, form 4- to 14-membered heterocycloalkyl that is substituted with Re and optionally substituted with one or more independently selected R®.
In some embodiments:
the moiety of “-N(R1)(R2) is a moiety of Formula a-1, a-2, a-3, a-4, a-5, or a-6:
a-5 a-6 ring A1 is 4- to 7-membered cycloalkyl or heterocycloalkyl;
t1 fsO, 1, 2, or 3;
is 0,1, 2, or 3;
t3 is 0,1, 2, or 3;
s1 is 1 or 2; and s2 is 1 or 2.
In some embodiments:
the moiety of Formula a-6 is a moiety of Formula a-6-1 or a-6-2:
X1 Is O, NR41, or C(R42)2;
each of R41 and R42 Is Independently H or R9;
s3 Is 0,1, or 2, provided that when s3 Is 0, then X1 is C(R42)2; and s4 is 0,1, or 2.
ln some embodiments:
the molety of “-NtR’XR2) Is a moiety of Formula a-11, a-12, a-13, a-14, a-15, a-16-1, or
a-16-1 a-16-2
X1 is O, NR41, or C(R42)2;
each of R41 and R42 is independently H or R9;
t1 is 0,1,2, or 3;
isO, 1,2, or 3;
t3, is 0,1, 2, or 3;
s1 is 1 or 2;
s2 is 1 or 2;
s3 is 0,1, or 2, provided that when s3 is 0, then X1 is C(R42)2; and s4 is 0,1, or 2.
In some embodiments:
the moiety of Formula a-11 is a moiety of Formula a-11-1 :
R9· is H, OH, optionally substituted Cm alkoxy, Cm alkyl, or cyclopropyl, or cyclobutyl; and t1a is 0,1, or 2.
In some embodiments:
the moiety of Formula a-12 is a moiety of Formula a-12-1:
(R%.
a-12-1
R9a is H, OH or optionally substituted Cm alkoxy; and t1 a is 0,1, or 2.
In some embodiments:
the moiety of ”-N(R1)(R2)’ Is a moiety of Formula a-26:
ring A2 is 5- or 6- membered cycloalkyl or heterocycloalkyl;
t2 is 0,1,2, or 3; and t3 is 0,1,2, or 3.
In some embodiments, ring A2 is 5- or 6-membered heterocycloalkyl and wherein at least one of the rlng-forming atoms of ring A2 is O.
In some embodiments:
the moiety of Formula a-26 is a moiety of Formula a-36:
a-36 ring A3 is 5- or 6-membered heterocycloalkyl (wherein the O atom shown in the ring is linked directly to the carbon bridge-head);
t2 is 0,1,2, or 3; and t3 isO, 1,2, or 3,
In some embodiments:
the moiety of Formula a-26 is a moiety of Formula a-46-1, a-46-2, a-46-3, or a-46-4, a46-5, a-46-6, or a-46-7:
a-46-6 a-46-7 t2 is 0,1,2, or 3; t3 is 0,1, or 2;
t4 is 0,1, or 2; and each Rw is independently F, Cl, methyl, or Ci fluoroalkyl.In some embodiments, the moiety of Formula a-26 is a moiety of Formula a-46-1 or a-46-2.
In some embodiments, the moiety of Formula a-26 is a moiety of Formula a-46-1.
In some embodiments, the moiety of Formula a-26 is a moiety of Formula a-46-2.
In some embodiments, the moiety of Formula a-26 is a moiety of Formula a-46-4.
In some embodiments, the moiety of Formula a-26 is a moiety of Formula a-46-6.
In some embodiments, the moiety of Formula a-26 is a moiety of Formula a-46-7. In some embodiments:
the moiety of *-N(R1)(R2)' is a moiety of Formula b-6:
b-6 ring A1 is 4- to 7-membered cycloalkyl or heterocycloalkyl; t11 is 0, 1,2, or 3;
t3 is 0,1,2, or 3;
s1is1or2;and s2 Is 1 or 2.
In some embodiments:
the moiety of Formula b-6 is a moiety of Formula b-6-1 or b-6-2:
b-6-1
X1 Is O, NRS1, or C(R52)2;
b-6-2;
each of RS1 and RS2 is independently H or R30;
s3 is 0,1, or 2, provided that when s3 Is 0, then X1 is C(RS2)2! and s4 is 0,1, or 2.
In some embodiments:
the moiety of “-NiR^fR2) is a molety of Formula b-26:
b-26 b-36 ring A2 Is 5- or 6-membered cycloalkyl or heterocycloalkyl;
ring A3 Is 5- or 6-membered heterocycloalkyl;
t11 is 0,1,2, or 3; and t3 is 0, 1,2, or 3.
In some embodiments wherein the moiety of “-N(R1)(R2)' is a moiety of Formula b-26, ring A2 Is 5- or 6-membered heterocycloalkyl and at least one of the ring-forming atoms of ring A2 is O.
In some embodiments wherein the moiety of “-N(R1)(R2) Is a moiety of Formula b-26: the moiety of Formula b-26 is a moiety of Formula b-46-1, b-46-2, b-46-3, b-46-4, b-465, b-46-6, or b-47:
b-46-1 b-46-2 b-46-3
b-46-5
‘‘V'PC \NJ (R9b)t3 b-46-7 t3 isO, 1, or 2; and each R9b is independently F, Cl, methyl, or Ci fluoroalkyl.
In some embodiments wherein the moiety of ‘-NiR’XR2)* is a moiety of Formula b-26, and the moiety of Formula b-26 is a moiety of Formula b-46-1, b-46-2, or b-46-7.
in some embodiments, the moiety of'-N(R1)(R2)’ is a moiety of Formula b-46-1. In some further embodiments, the moiety of Formula b-46-1 is a moiety of Formula b-46-1 a:
d30 b-46-1 a each R30* is independently halogen, Cm alkyl, Cm haloalkyl, Cm alkoxy, or Cm haloalkoxy; and t12 is 0,1,or2.
In some embodiments, the moiety of Formula b-46-1 a is a moiety of Formula b-46-1 a-1 or b-46-1 a-2:
b-46-1 a-1 or b-46-1 a-2;
t12 Is 0,1, or 2; and each R30A is Independently F, Cl, methyl, Ci fluoroalkyl, methoxy, or Ci fluoroalkoxy. In some embodiments, the moiety of “-NiR’XR2) is a moiety of Formula b-46-1 a-1. In some embodiments, the moiety of -N(R1)(R2) Is a moiety of Formula b-46-1 a-2. In some embodiments, the moiety of -NtR’XR2) is a moiety of Formula b-46-2. In some embodiments:
the moiety of -N(R1)(R2) is a moiety of Formula b-46-2; the moiety of Formula b-46 2 io a moiety of Formu'a b-4ô-2a:
(R30*)^
b-46-2a
each R30A Is Independently halogen, Cu alkyl, Cm haloalkyl, Cm alkoxy, or Cm haloalkoxy; and t12 is 0,1, or 2.
In some further embodiments, the moiety of Formula b-46-2a is a moiety of Formula b
46-2a-1, b-46-2a-2, or b-46-2a-3:
Rc Is Ci-3 alkyl or cyclopropyl;
t12 is 0,1, or 2; and each R30A is independently F, Cl, methyl, Ci fluoroalkyl, methoxy, or Ci fluoroalkoxy. In some further embodiments, Rc is Cm alkyl. In some yet further embodiments, R° Is methyl.
In some embodiments, the moiety of -NtR’JfR2) is a moiety of Formula b-46-2a-1; Rc is Cm alkyl; and each R30A is independently F, Cl, methyl, or Ci fluoroalkyl. In some further embodiments, Rc Is methyl.
In some embodiments, the moiety of ‘-NtR’JiR2) is a moiety of Formula b-46-2a-2; Re is
Cm alkyl; and each RMA Is independently F, Cl, methyl, or Ci fluoroalkyl. In some further embodiments, Rc Is methyl.
In some embodiments, the moiety of “-NtR’XR2) is a moiety of Formula b-46-2a-3.
In some embodiments, the moiety of -NtR’JfR2) is a moiety of Formula b-46-3.
In some embodiments, the moiety of “-NtR’XR2)’ Is a moiety of Formula b-46-7. In some further embodiments, the moiety of Formula b-46-7 Is a moiety of Formula b-46-7a:
(2RJ-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{[(4-fluorophenyl)sulfonyi](methyl)amino}-1-oxa-8azaspiro[4.5]decane-8-carboxylate
(2RJ-1,1,1-trifluoro-3-hydroxypropan-2-yi (3R}-3-((4-fluoro-N-methylphenyl)sulfonamido)-1-oxa-8azaspiro[4.5]decane-8-carboxylate »
b-46-7a b-46-7a-1 b-46-7a-2 each R3DA Is Independently halogen, Cm alkyl, Cm haloalkyi, Cm alkoxy, or Cm haloalkoxy; and t12 is 0,1,or2.
in some embodiments, the moiety of ‘‘-NtR’XR2)’ is a moiety of Formula b-46-7a-1 ; and each RMA is Independently F, Cl, methyl, or Ci fluoroalkyi.
in some embodiments, the moiety of “-NfR’JtR2) is a moiety of Formula b-46-7a-2; and each R3DA is independently F, Ci, methyl, or Ci fluoroalkyi. ln some embodiments [e.g. wherein the moiety of -NtR’XR2) is a moiety of Formula a-6, a-6-1, a-6-2, a-16-1, a-16-2, a-26, a-36, a-46-1, a-46-2, a-46-3, a-46-4, a-46-5, a-46-6, a-46-7, b-6, b-6-1, b-6-2, b-26, b-36, b-46-1, b18588
46-2, b-46-3, b-46-4, b-46-5, b-46-6, b-47, b-46-1a-1, b-46-1a-2, b-46-2a-1, b-46-2a-2, b-46-2a3, b-46-7a-1, or b-46-7a-2], t3 is 0 or 1. In some further embodiments, t3 is 0.
In some embodiments [e.g. wherein the moîety of -NtFfffR2)’ Is a mofety of Formula a6, a-6-1, a-6-2, a-16-1, a-16-2, a-26, a-36, a-46-1, a-46-2, a-46-3, a-46-6, or a-46-7], t3 Is 0 or 5 1 ; and t2 Is 0 or 1. In some further embodiments, t3 is 0.
In some embodiments [e.g. wherein the moiety of ”-N(R1)(R2) is a molety of Formula b6, b-6-1, b-6-2, b-26, b-36, b-46-1, b-46-2, b-46-3, b-46-4, b-46-5, b-46-6, or b-47], t3 is 0 or 1; and t11 Is 0,1, or 2. In some further embodiments, t3 Is 0 and 111 Is 1 or 2. In yet further embodiments, 111 is 1.
In some embodiments [e.g. wherein the moiety of N(R’)(R2) is a moiety of Formula b46-1 a-1, b-46-1 a-2, b-46-2a-1, b-46-2a-2, b-46-2a-3, b-46-7a-1, or b-46-7a-2], t3 is 0 or 1; and t12 Is 0 or 1. In some further embodiments, t3 Is 0. In yet further embodiments, t12 Is 0.
In some embodiments wherein the moiety of “-NiR’XR2) is a moiety of Formula b-461a-1, b-46-1 a-2, b-46-2a-1, b-46-2a-2, b-46-2a-3, b-46-7a-1, or b-46-7a-2; each Rd is independently selected from the group consisting of Cm alkyl, C3.7 cycloalkyl, a 4- to 7membered heterocycloalkyl, Ce-waryl, a 5- to 6-membered heteroaryl, (C3-7 cycloalkyl)-Ci-4 alkyl-, (4- to 7-membered heterocycloalkyl)-Ciu alkyl-, (Ce-to aryl)-CM alkyl-, and (5- to 6membered heteroaryl)-Ci-4 alkyl-, wherein each of the sélections from the group is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CFs, -CN, -OH, oxo, -S-Cm alkyl, Cm alkyl, Cm haloalkyl, C^e alkenyl, Cî-β alkynyl, Cj.7 cycloalkyl, Cm alkoxy, and Cm haloalkoxy.
In some embodiments, each of R3 and R4 Is independently H, halogen, or methyl.
In some further embodiments, each of R3and R4 is independently H or halogen (e.g., F). In yet further embodiments, each of R3 and R4 is independently halogen (e.g., F).
In some other embodiments, each of R3 and R4 Is independently H or methyl.
In some embodiments, each of R® and R* is independently H or Cm alkyl (e.g., methyl or ethyl). In some further embodiments, each of R® and R® Is independently H or methyl.
In some embodiments, each of R® and R® is H.
In some embodiments, R7 is H or R10; and R10 is -P(=O)(OR81)(OR82).
In some embodiments, R7 is H.
In some embodiments, R7 is R10; and R10 is -P(=O)(OR8’)(OR82).
In some embodiments, each of R8’ and R82 is independently selected from the group consisting of H, Cm alkyl, and (C3.7 cycloalkyl)-CM alkyl-, wherein each of the Cm alkyl and (C3. 7 cycloalkyl)-CM alkyl- Is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CN, -OH, oxo, -NH2, -NH(Cm alkyi), -N(Cm alkyljî, Cm alkyl, Cm alkoxy, Cm haloalkyl, Cm haloalkoxy, and Cm cycloalkyl. In some further embodiments, each R8’ and R82 is independently H or Cm alkyl. In some yet further embodiments, each R81 and R82 is H.
In some embodiments:
the moîety of -N(R’)(R2)' is a moiety of Formula a-1, a-2, a-3, a-11, a-12, a-13, a-16-2, a-46-2, or a-46-7;
R’ is -L’-R”, -L2-R12, -L’-R”, or-L4-R14;
each of L’, L2, L3, and L4 Is independently absent, -O-, -S(=O)r, -(CR2,R22)- [e.g. -(CHî)-
1, -NR23-, -O-(CR2’R22)-, -(CR2'R22)-O-(CRI1R22)-, -(CR21R22)-S(=O)r [e.g., -(CH2)-S(=O)^ ], NR23-S(=O)r, or -( CR2’R22)-NR23-S(=O)r [e.g. -(CH2)-NR23-S(«O)r];
each of R21 and R22 is independently H, OH, halogen, Cm alkyl, cyclopropylmethyl, or Cis haloalkyl;
R23 is H or Cm alkyl;
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
R12 is 5- to 6-membered heterocycloalkyl optionally substituted with one or more independently selected R31;
R13 is phenyl optionally substituted with one or more Independently selected R33; and
R14 is Cm cycloalkyl optionally substituted with one or more independently selected R34.
In some embodiments, the moiety of *-N(R’)(R2) is a moiety of Formula a-12 or a-13; and R8 is -L’-R11 or-L’-R13. In some further embodiments, Re is -R”.
In some embodiments, the moiety of *-N(R’)(R2) is a moiety of Formula a-12 (or
Formula a-12-1) wherein R8 is -R11 or -R13. In some further embodiments, R8 is -R11.
In some embodiments, the moiety of *-N(R’)(R2)’ is a moiety of Formula a-13.
In some embodiments, the moiety of *-N(R’)(R2) is a moiety of Formula a-13; and R8 is -L’-R” or -L’-R13. In some further embodiments, R8 is -R11 or -R”. In yet further embodiments, R8 Is -R”.
In some embodiments, the moiety of ‘-NiR’XR2) is a moiety of Formula a-46-2.
In some embodiments, the moiety of “-N(R’)(R2) is a moiety of Formula a-46-2, R8 is NR^-S^OrR”, -NR23-S(=O)rR12, -NR23-S(=O)rR”, or-NR23-S(=O)2-R14; and R23 Is C,., alkyl. In some further embodiments, R23 is methyl.
In some embodiments, the moiety of “—N(RT)(R2) is a moiety of Formula a-46-2; R8 Is 30 NR23-C(=O)-R”, -NR23-C(=O)-R12, -NR23-C(=O)-R13, or -NR23-C(=O)-R14; and R23 Is Cm alkyl.
In some further embodiments, R23 is methyl.
In some embodiments, the moiety of *-N(R’)(R2) is a moiety of Formula a-46-2; and R8 is-R”,-R12,-R”, or-R’4. In some further embodiments, Rs is-R” or-R”.ln some embodiments, the moiety of -NfR’XR2)’ is a moiety of Formula a-46-2; and R8 is -L’-R11 or 35 L’-R13. In some further embodiments, R8 is -NR23-S(=O)2-R11 or -NR23-S(=O)2-R”. In some yet further embodiments, R23 is H or Cm alkyl (e.g., methyl). In still further embodiments, R23 is Cm alkyl (e.g., methyl).
In some embodiments, the moiety of “-N(R’)(R2) is a moiety of Formula a-46-2; and Ra is -O-R” or -O-R’3. In some embodiments, the moiety of -N(R’)(R2)’ is a moiety of Formula a-46-2; and Ra Is -L4-R’4. In some further embodiments, Ra is -R’*. In some embodiments:
the moiety of “-N(R’)(R2) is a moiety of Formula a-14, a-15, a-16-1, a-36, a-46-1, a-465 3, a-46-4, a-46-5, or a-46-6;
Ra is -L’-R”, -L2-R12, -L3-R13, or -L4-R14; (for example Ra is -L’-R”, -L2-R12, or -L’R’3);
each of L1, L2, L3, and L4 Is independently absent, -(CR2’R22)- [e.g. -(CH2)-], -C(=O)-, S(=O)2-, -S(=O)2-NR23-, -S(=O)r(CR21R22)-, -S(=O)^NR23-(CR21R22)-, or -S(=O)2-(CR2’R22)2-;
each of R21 and R22 is Independently H, OH, halogen, C1.3 alkyl, cyclopropylmethyl, or Ci.
haloalkyl (for example, H, C1.3 alkyl, or cyclopropyl);
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
R’2 is 5- to 6-membered heterocycloalkyl optionally substituted with one or more independently selected R31;
R’3 is phenyl optionally substituted with one or more independently selected R33; and
R14 is Cs-î cycloalkyl optionally substituted with one or more independently selected R34. In some yet further embodiments, R23 Is H or Cm alkyl (e.g., methyl).
In some embodiments, the moiety of ”-N(R’)(R2) is a moiety of Formula a-14 or a-15; 20 and Ra is-L’-R” or-L3-R’3.
In some embodiments, the moiety of ”-N(R’)(R2)’ is a moiety of Formula a-15; Ra is -L’R” or -L3-R’3. In some further embodiments,each of L1 and L3 Is independently absent, (CR21 R22)-, -S(=O)2-, -S(=O)rNR2\ -S(=O)2-NR23-(CR2’R22)-, -S(=O)2-(CR21R22)-, or-S(=0)2(CR21 R22)2-; and each of R21 and R22 is independently H, C1.3 alkyl, or cyclopropyl. In some yet 25 further embodiments, each of L1 and L3 is independently -(CR21 R22)- or -S(=O)2-.
In some embodiments, the moiety of “-NÎR’JÎR2) is a moiety of Formula a-16-1, a-46-1, a-46-3, a-46-4, or a-46-6; and Ra Is -L’-R” or-L’-R’3.
In some embodiments, the moiety of -NiR’XR2)' is a moiety of Formula a-46-1. in some further embodiments, Ra is -L’-R”, -L2-R’2, -L’-R’3, or-L4-R14; and each of each of L’, 30 L2, L3, and L4 is -S(=O)2- or -C(=O)-. in some yet further embodiments, each of L’, L2, L3, and
L4 is -S(=O)2-.
in some embodiments, the moiety of *-N(R’)(R2) is a moiety of Formula a-46-1; and Ra is -L’-R” or-L3-R13. In some further embodiments, each of L1 and L3 is Independently absent, (CR21 R22)-, or -S(=O)2-. In some yet further embodiments, each of L’and L3 is Independently, 35 (CR21 R22)-, or -S(=O)2-. In some still further embodiments, each of L’ and L3 is -S(=O)2In some embodiments, the moiety of“-N(R’)(R2) is a moiety of Formula a-46-1; Ra is L’-R”; and L’is absent, -(CR21R22)-, or-S(=O)2-. In some further embodiments, L’is absent or -S(=O)2-. In some yet further embodiments, L1 Is -S(=O)2-.
In some embodiments, the moiety of‘-NfR^fR2) is a moiety of Formula a-46-1; RB is L3-R13; and L3 is absent, -(CR21 R22}-, or -S(=O)2-. in some further embodiments, L3 is absent or -S(=O)2-. In some yet further embodiments, L3 is -S(=0)2-.
In some embodiments, the moiety of -N(R1)(R2)' is a moiety of Formula a-46-1; R* is 5 L1-R11 or -L3-R13; and each of L1 and L3 is -C(=O)- or -S(=O)2-. In some further embodiments, each of L1 and L3 is -S(=O)2-. In yet further embodiments, R’ is - S(=O)2-R13; R13 is phenyl optionally substituted with one or more independently selected R33. In still further embodiments, each R33 is independently selected from the group consisting of halogen (e.g. F or Cl), -CN, Cm alkyl, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy.
In some embodiments, the moiety of *-N(R1)(R2)’ is a moiety of Formula a-46-2.
In some embodiments, the moiety of y-N(R1)(R2)' is a moiety of Formula a-46-2; and R8 is -L’-R11 or-L’-R13. In some further embodiments, each of L1 and L3 is independently absent, -O-, -NR23-, -S(=O)2-(CR21R22)- [for example, RB is -(CR21R22)-S(=O)2-R11 or -(CR21R22)-S(=O)2R13], -O-(CR21R22)- [for example, RB is -O-(CR21R22)- -R11 or-O-(CR21 R22)- -R13]. -S(=O)2-NR2315 [for example, RB is -NR23-S(=O)2-R11 or -NR23-S(=O)2-R13]. or-(CR21R22)-O-(CR21R22)-. In some yet further embodiments, R23 Is H or Cm alkyl (e.g., methyl) and each of R21 and R22 is independently H, OH, halogen, Cm alkyl, cyclopropylmethyl, or Cm haloalkyl (for example, H, Cm alkyl, or cyclopropyl).
In some embodiments, the moiety of *-N(R1)(R2) is a moiety of Formula a-46-2; and R8 20 is -L1-R” or -L3-R13. In some further embodiments, each of L1 and L3 is independently -S(=O)2NR23- [For example, R8 Is -NR23-S(=O)2-R11 or -NR23-S(=O)2-R13]· In some yet further embodiments, R23 Is H or Cm alkyl (e g., methyl). In some still further embodiments, R23 is Cm alkyl (e.g., methyl).
in some embodiments, the moiety of -N(R1)(R2) is a moiety of Formula a-46-2; RB is 25 L1-R11 or -L3-R13; and each of L1 and L3 is independently -NR23-. In some further embodiments,
R23 is H or Cm alkyl (e.g., methyl).
In some embodiments, the moiety of “-N(R1)(R2) is a moiety of Formula a-46-2; RB is L’-R11 or -L3-R13; and each of L1 and L3 is -O-.
In some embodiments, the moiety of -N(R1)(R2)' is a moiety of Formula a-46-2; RB is 30 L*-RM; and L4 is -O-, -NR23-, -S(=O)2-(CR21R22)- [for example, RB is -(CR21R22)-S(=O)2-R11 or (CR21R22)-S(=O)2-Rn], or-S(=O)2-NR23- [for example, RB is-NR23-S(=O)2-R or-NR23-S(=O)2R13]. In some further embodiments, L4 is -S(=O)2-NR23- [For example, RB is -NR23-S(=O)2-R14]. In sonie yet further embodiments, R23 is H or Cm alkyl (e.g., methyl).
In some embodiments, the moiety of -NtR’XR2) is a moiety of Formula a-46-2; RB is 35 NR23-C(=O)-R11, -NR23-C(=O)-R12, -NR23-C(=O)-R13, or-NR23-C(=O)-R14; and R23 is Cm alkyl or cyciopropropyl. In some further embodiments, R23 is Cm alkyl (e.g. methyi).
In some embodiments, the moiety of -N(R1)(R2) is a moiety of Formula a-46-2; and R8 is-R11 or-R13.
In some embodiments, the moiety of “-N(R1)(R2) is a moiety of Formula a-46-4 or a-466; and Ra is -L’-R11 or-L3-R13. In some further embodiments, each of L1 and L3 Is Independently -(CR21 R22)- or -S(=O)2-. In some yet further embodiments, each of L’ and L3 is (CR21 R22)-.
In some embodiments, the moiety of -NfR^iR2) is a moiety of Formula a-46-6; and Ra is -L’-R11 or-L3-R13. In some further embodiments, each of L’and L3 Is independently (CR21 R22)- or -S(=O)î-. In some yet further embodiments, each of L1 and L3 is -(CR21R22)-; and each of R21 and R22 Is independently H or Ci.3 alkyl.
In some embodiments, the moiety of “-N(R1)(R2) is a-46-7; R* is -L1-R11, -L2-R12, -L310 R13, or -L4-R14; and each of L1, L2, L3, and L4 is -C(=O>- or -S (=0)2- [e.g. -C(=O)-]· In some further embodiments, Ra is -L1-R” or -L3-R”; and each of L1 and L3 is -C(=O)-.
In some embodiments, each Ra is independently OH, oxo, halogen, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm alkoxy, Cm haloalkoxy, or cyclopropyl. In some further embodiments, each R9 Is independently OH, oxo, or methyl. In some yet further embodiments, 15 each R9 is independently OH or methyt. In some still further embodiments, each R9 Is OH.
In some embodiments of the compound of Formula l-a or a pharmaceutically acceptable sait thereof:
the moiety of ’-NtR’XR2) is a moiety of Formula a-12 (or Formula a-12-1) wherein R8 is -R11 or-R13;
each of Rs and Ra is independently H or methyl;
R7 is H or-P(=O)(OR81)(OR82) [e.g., -P(-O)(OH)(OH)];
R11 Is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
R13 is phenyl optionally substituted with one or more Independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen,
OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, C2_e alkenyl, C2-e alkynyl, C3-10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (Cj.10 cycloalkyl)-CiM alkyl-, (4- to 10-membered heterocycloalkyl)-CiM alkyl-, (Ce-10 aryl)-CiM alkyl-, and (5-to 10membered heteroaryl)-CiM alkyl-, wherein each of the Cm alkyl, C2_e alkenyl, C2.e alkynyl, Cj.» cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (Cj-w cycloalkyl)-CiM alkyl-, (4- to 10-membered heterocycloalkyl)-CiM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- Is optionally substituted with one or more independently selected R35;
each R38 is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, C2-e alkenyl, C2.e alkynyl, and ¢3.7 cycloalkyl. In some further embodiments, each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyl, and Cm cycloalkyl.
In some embodiments of the compound of Formula l-a or a pharmaceutically acceptable sait thereof:
the moiety of ~N(R1)(R2) is a moiety of Formula a-13 wherein R8 Is -R11 or-R13;
each of R’and R8 is Independently H or methyl;
R7 is H or -P(=O)(OR81)(OR82) [e.g., -P(=O)(OH)(OH)];
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R”;
R13 is phenyl optionally substituted with one or more independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm alkenyl, Cm alkynyl, Cmo cycloalkyl, 4- to 10-membered heterocycloalkyl, Cmo aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-Cm alkyl-, (4- to 10-membered heterocycloalkylJ-Cw alkyl-, (Cmo aryl)-Ci-4 alkyl-, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Cmo aryl, 5- to 10-membered heteroaryl, (Cno cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Cmo arylJ-Cw alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- is optionally substituted with one or more independently selected R3e;
each R38 is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, Cm alkenyl, Cm alkynyl, and Cm cycloalkyl. In some further embodiments, each of R31 and R33 is Independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl. Cm cyanoalkyl, Cm haloalkyl, and Cm cycloalkyl. In some further embodiments, R8 is -R1’.
In some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of *-N(R1)(R2) is a moiety of Formula a-15 wherein R8 is -L’-R11 or-L’-R13;
each of L1 and L3 is independently absent, -(CR^R22)-, -S(=O)2-, -S(=O)rNR23-, -S(=O)r NR23-(CR21R22)-, -S(=O)2-(CR21R22)-, or-S(=O)2-(CR2’R22)2- [e.g., each of L1 and L3 Is independently -(CR21R22)- or -S(=O)2-)];
each of R21 and R22 is independently H, Cm alkyl, or cyclopropyl;
each of R9and R* is independently H or methyl;
R7 is H or -P(=O)(OR81)(OR82) [e.g., -P(=O)(OH)(OH)];
R'1 is 5- to 6-membered heteroaryl optionally substituted wilh one or more independently selected R31;
R13 Is phenyl optionally substituted with one or more independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm alkenyl, Cm alkynyl, Cmo cycloalkyl, 4- to 10-membered heterocycloalkyl, Cmo aryl, 5- to 10-membered heteroaryl, (Cmo cycloalkylJ-Cn
alkyl-, (4- to 10-membered heterocycloalkylj-Cm alkyl-, (Cmo aryl)-CM alkyl-, and (5- to 10membered heteroarylj-Cw alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3-10 cycloalkyl, 4-to 10-membered heterocycloalkyl, Ce-ioaryl, 5-to 10-membered heteroaryl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- is optionally substituted with one or more independently selected R3e;
each R30 is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, Cm alkenyl, Cm alkynyl, and C3-7 cycloalkyl. In some further embodiments, each of R31 and R33 is independently selected from 10 the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyl, and Cm cycloalkyl.
In some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of *-N(R’)(R2F is a moiety of Formula a-46-1 wherein R® is -L’-R11 or-L315 R13;
each of L1 and L3 is independently absent, -(CR21R22)-, or -S(=0)r [e.g., each of L1 and L3 Is -S(=O)r];
each of R2’ and R22 Is Independently H, Cm alkyl, or cyclopropyl;
each of Rsand R* Is Independently H or methyl;
R7 is H or -P(=O)(OR’1)(ORe2) [e.g., -P(=O)(OH)(OH)];
R11 Is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31; .
R13 Is phenyl optionally substituted with one or more independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen, 25 OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm alkenyl, Cm alkynyl, C3-10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl·, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3-10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-io aryl, 5- to 10-membered heteroaryl, (C3-10 30 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- is optionally substituted with one or more Independently selected R38;
each R33 is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, Cm alkenyl, Cm alkynyl, and C3-7 35 cycloalkyl. In some further embodiments, each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyl, and Cm cycloalkyl.
In some embodiments of the compound of Formula l-a (induding Formula l-a1 ) or a pharmaceutically acceptable sait thereof:
the moiety of “~N(R1)(R2f Is a moiety of Formula a-46-1 wherein R* is -L1-Rn;
L1 Is independently absent, -(CR21R22)-, or -S(=O>2- [e.g., absent or -S(=O)2J;
each of R21 and R22 is independently H, Cm alkyl, or cyclopropyl;
each of R’and R* is independently H or methyl;
R7 is H or-P(=O)(OR”)(ORe2) [e.g., -P(=O)(OH)(OH)];
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
each of R31 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm alkenyl, Cm alkynyl, Cno cycloalkyl, 4- to 10membered heterocycloalkyl, Ce-io aryl, 5- to 10-membered heteroaryl, (Cno cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkylJ-Cm alkyl-, (Ce.10 aryl)-Ci-4 alkyl-, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Cmo aryl, 5- to 10-membered heteroaryl, (C3.10 cycloalkyl)-Ci-4 alkyl-, (4- to 10-membered heterocycloalkyl)-Ci-4 alkyl-, (Ce-io aryl)-Ci-« alkyl-, and (5- to 10-membered heteroaryl)-Ci·* alkyl- Is optionally substituted with one or more independentty selected R3e;
each R38 is Independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloaikoxy, -CN, Cm alkyl, Cm haloalkyl, C2-e alkenyl, Cm alkynyl, and C3.7 cycloalkyl. In some further embodiments, each of R31 and R33 Is independentty selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyl, and Cm cycloalkyl. In some further embodiments, L1 Is -S(=O)2-.
In some embodiments of the compound of Formula l-a (induding Formula l-a1 ) or a pharmaceuticalty acceptable sait thereof:
the moiety of -NfR’JfR2) is a moiety of Formula a-46-1 wherein R’ Is -L3-R13;
L3 is absent, -(CR21R22)- or-S(=Oh- [e.g., L3 is -S(=0)2-J;
each of R21 and R22 Is independently H, Cm alkyl, or cyclopropyl;
each of R! and R* is independently H or methyl;
R7 is H or-P(=O)(ORei)(OR82) [e.g., -P(=O)(OH)(OH)J;
R13 is phenyl optionally substituted with one or more Independentty selected R33; each R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm alkenyl, Cm alkynyl, Cmo cycloalkyl, 4- to 1035 membered heterocycloalkyl, Cno aryl, 5- to 10-membered heteroaryl, (Cno cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyi)-Ci-4 alkyl-, (Cmo aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-Ci-4 alkyl-, wherein each of the Cm alkyl, C2-e alkenyl, Cm alkynyl, Cmo cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-w aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-Ci_4 alkyl-, (4- to 10-membered heterocycloalkyl)-Ci-4 alkyl-, (Cmo aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- Is optionally substituted with one or more independently selected R3®;
each RM is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyi, Cm haloalkyi, C2-6 alkenyl, C24 alkynyl, and C3.7 cycloalkyl. In some further embodiments, R’3 is phenyi optionally substituted with one or more substituents each independently selected from halogen, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyi, Cm haloalkyi, and Cm cycloalkyl·
In some embodiments of the compound of Formula l-a (including Formula i-a 1 ) or a pharmaceutically acceptable sait thereof:
the moiety of “-N(R’)(R2) is a moiety of Formula a-46-1 wherein Ra Is -L3-R13;
L3 is -S(=O)2-;
each of R® and Re is independently H or methyl;
R7 is H or-P(=O)(OR81)(OR®2) [e.g., -P(=O)(OH)(OH)];
R13 is phenyi optionally substituted with one or more independently selected R33;
each R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyi, and C3. 4 cycloalkyl. In some further embodiments, R13 is phenyi optionally substituted with one or more substituents each independently selected from halogen, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyi, and Cm cycloalkyl.
In some embodiments ofthe compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of -N(R1)(R2f is a moiety of Formula b-26 (e.g. a moiety of Formula b-36); each of R9 and R® is independently H, methyl, or Ci fluoroalkyl (e.g. H or methyl); and R7 is H or -P(=O)(ORei)(ORe2) [e.g., -P(=O)(OH)(OH)]. In some further embodiments, each of R® and R* is independently H or methyl. In some yet further embodiments, the moiety of “-N(R1)(R2)“ is a moiety of Formula b-36.
In some embodiments of the compound of Formula l-a (including Formula l-a 1 ) or a pharmaceutically acceptable sait thereof:
the moiety of *-N(R1)(R2) is a moiety of Formula b-46-1a (e.g. a moiety of Formula b46-1a-1 orb-46-1a-2);
each of R® and R® is independently H, methyl, or Ci fluoroalkyl (e.g. H or methyl); and
R7 is H or -P(=O)(OR31)(OR82) [e.g., -P(=O)(OH)(OH)]. In some further embodiments, the moiety of *-N(R’)(R2) is a moiety of Formula b-46-1 a-1. In some yet further embodiments, each of R®andR® is independently H or methyl.In some embodiments ofthe compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of *-N(R’)(R2) is a moiety of Formula a-46-2 wherein R® is -L’-R11 or -L3R13;
each of L1 and L3 Is Independently absent, -O-, -NR23-, -S(=O)r(CR21R22)- [for example, R8 is -(CR21R22)-S{=O)2-R11 or-(CR21R22)-S(=O)2-R13], or-S(=O)rNR23- [for example, R8 is NR23-S{=O)rR11 or -NR23-S(=O)rR13];
each of R21 and R22 Is independently H, OH, halogen, Ci.j alkyl, cyclopropylmethyl, or Ci. 3 haloalkyl (for example, H, C1.3 alkyl, or cyclopropyl);
R23 is H or Cm alkyl (e.g., methyl);
each of R’and R8 Is Independently H or methyl;
R7 is H or -P(=O)(OR81)(OR82) [e.g., -P(=O)(OH)(OH)];
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
R13 is phenyl optionally substituted with one or more independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, C2^ alkenyl, Cm alkynyl, C3.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-Cm alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Cmo aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Gmo aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- Is optionally substituted with one or more Independently selected R38;
each R38 is Independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, C2^ alkenyl, Cm alkynyl, and C3.7 cycloalkyl. In some further embodiments, each of R31 and R33 Is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyi, Cm cyanoalkyi, Cm haloalkyl, and Cm cycloalkyl.
In some embodiments of the compound of Formula l-a (including Formula l-a1 ) or a pharmaceutically acceptable sait thereof:
the moiety of -N(R’)(R2)' is a moiety of Formula a-46-2 wherein R8 is-U-R11 or-L3R13;
each of L1 and L3 Is -S(=O)2-NR23- [for example, R8 is -NR23-S(=O)rR” or-NR23S(=O)rR13];
R23 is H or Cm alkyl (e.g., methyl);
each of R5 and R3 is independently H or methyl,
R7 is H or -P(=O)(OR8,)(OR82) [e.g, -P(=O)(OH)(OH)];
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
R13 Is phenyi optionally substituted with one or more independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen,
OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, C2-« alkenyl, C2-e alkynyl, Cyw cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-w aryl, 5- to 10-membered heteroaryl, (C>io cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, C2-e alkenyl, C2.e alkynyl, Cj.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, C&.10 aryl, 5- to 10-membered heteroaryl, (C>i0 cycloalkyl)-Ci-4 alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-io arylJ-Cw alkyl-, and (5- to 10-membered heteroaryl)-CM alkyi- is optionally substituted with one or more independently selected R36;
each RM is Independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, C2_e alkenyl, C2.e alkynyl, and C3-7 cycloalkyl. In some further embodiments, each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyl, and CM cycloalkyl.
in some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of -N(R1)(R2)’ is a molety of Formula a-46-2 wherein R® is -L’-R11 or -L3R13;
each of L1 and L3 is -O- or -NR23-;
R23 is H or Cm alkyl (e.g., methyl);
each of R’and R* Is independently H or methyl;
R7 is H or-P(=O)(ORB1)(OR) (e.g., -P(=O)(OH)(OH)];
R11 îs 5- to 6-membered heteroaryl optionally substituted with one or more independently selected R31;
R13 is phenyl optionally substituted with one or more Independently selected R33;
each of R31 and R33 Is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm alkenyl, C2.e alkynyl, C>io cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce.10 aryl, 5- to 10-membered heteroaryl, (C3-10 cydoalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, C2-e alkenyl, C2_e alkynyl, Cj.10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (Cj.10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-w aryl)-CM alkyl-, and (5- to 10-membered heleroaryl)-CM alkyl- is optionally substituted with one or more independently selected R3e;
each R3* is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyi, Cm haloalkyl, C2-e alkenyl, C2_e alkynyl, and Cy? cycloalkyl. In some further embodiments, each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyi, and C» cycloalkyl.
ln some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of -NfR’XR2) Is a moiety of Formula b-46-2 (e.g. a moiety of Formula b-462a, Formula b-46-2a-1, b-46-2a-2, or b-46-2a-3);
each of R’and R® is independently H, methyl, or Ci fluoroalkyi (e.g. H or methyl); and
R7 Is H or -P(=O)(OR”)(OR82) [e.g., -P(=O)(OH)(OH)]. In some further embodiments, the moiety of “-N(R’)(R2)’ is a moiety of Formula b-46-2a; and each of R’and R® is independently H or methyl.
ln some embodiments ofthe compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of -NtR’JfR2) is a moiety of Formula a-46-2;
R® is -NR23-S(sO)2-R11, -NR23-S(=O)2-R,z. -NR23-S(=O)2-R13, -NR23-S(=O)2-R14, -NR23C(=O)-R, -NR23-C(=O)-R12 -NR23-C(=O)-R,3 i or-NR23-C(=O)-R14;
R23 is Ci-3 alkyl (e.g. methyl);
each of R’and R* is independently H, methyl, or Ci fluoroalkyi (e.g. H or methyl); and R7 is H or -P(=O)(OR’1)(OR®2) [e.g., -P(=O)(OH)(OH)]. In some further embodiments, R23 is methyl; and each of R’and R® is independently H or methyl.
ln some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of -NtR’XR2) Is a moiety of Formula a-46-2;
R® is -R, -R’2, -R13, or -R14 (e.g. R® is -R” or -R13);
each of R’and R® is independently H, methyl, or Ci fluoroalkyi (e.g. H or methyl); and
R7 is H or -P(=O)(OR”)(OR®2) [e.g., -P(=O)(OH)(OH)]. In some further embodiments, R23 is -R’4; and each of R’and R® is independently H or methyl.
ln some embodiments of the compound of Formula l-a (including Formula l-a1 ) or a pharmaceutically acceptable sait thereof:
the moiety of ‘-N(R’XR2) is a moiety of Formula a-46-7;
R® is -L’-R11, -LAR12, -L3-R13, or-L4-R14;
each of L1, L2, L3, and L4 is -C(=O)- or -S(=O)2- [e.g. -C(=O)-];
each of R’and R® is Independently H, methyl, or Ci fluoroalkyi (e.g. H or methyl); and
R7 is H or -P(=O)(OR9,)(ORS2) [e.g., -P(=O)(OH)(OH)]. ln some further embodiments, R* is -U-R11 or-L3-R13; each of L1 and L3 is -C(=O)-; and each of R’and R® is independently H or methyl.
ln some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moîety of “-NfR’XR2) is a moiety of Formula b-46-7a (e.g. a moiety of Formula b46-7a-1 or a moiety of Formula b-46-7a-2);
each of R’and R® is independently H, methyl, or Ci fluoroalkyl (e.g. H or methyl); and
R7 is H or -P(=O)(OR®1)(OR®2) [e.g., -P(=O)(OH)(OH)J. In some further embodiments, the moiety of -NfR’XR2) is a moiety of Formula b-46-7a-2; and each of R’and R® is independently H or methyl.
In some embodiments of the compound of Formula l-a (including Formula l-a1) or a pharmaceutically acceptable sait thereof:
the moiety of “-NiR’XR2) is a moiety of Formula a-46-4 or a-46-6 wherein R® is -L1-R’1 or-L’-R13;
each of L1 and L3 is independently -(CR21 R22)- or -S(=O)2-;
each of R21 and R22 is independently H, Cm alkyl, or cyclopropyl;
each of R’and Re is independently H or methyl;
R7 is H or-P(=O)(OR®1)(OR82) [e.g.. -P(=O)(OH)(OH)J;
R11 is 5- to 6-membered heteroaryl optionally substituted with one or more Independently selected R3’;
R13 Is phenyl optionally substituted with one or more independently selected R33;
each of R31 and R33 is independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, C2_e alkenyl, C2.e alkynyl, Cj-w cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-w aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-Ci-< alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-C« alkyl-, and (5- to 10membered heteroaryl)-CM alkyl-, wherein each of the Cm alkyl, Cm alkenyl, Cm alkynyl, C3-10 cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-10 aryl, 5- to 10-membered heteroaryl, (C3-10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce-10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-Ci-* alkyl- is optionally substituted with one or more Independently selected R3®;
each R3® is independently selected from the group consisting of halogen, -OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm haloalkyl, C2< alkenyl, Cm alkynyl, and C3-7 cycloalkyl. In some further embodiments, each of R31 and R33 is Independently selected from the the group consisting of halogen, OH, Cm alkoxy, Cm haloalkoxy, -CN, Cm alkyl, Cm hydroxylalkyl, Cm cyanoalkyl, Cm haloalkyl, and Cm cycloalkyl.
In some embodiments, the présent invention provides a compound selected from Examples 1 to 150 (e.g. Examples 1 to 91) in the EXAMPLES section or a pharmaceutically acceptable sait thereof (or the parent compound thereof where the exemplary compound, for example, Is a sait) herein below.
In some embodiments, the présent Invention provides a compound selected from:
1,1,1-trifluoro-3-hydroxypropan-2-yl 6-[1 -(5-methoxypyridin-2-yl)-l H-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate;
1.1.1- tnfluoro-3-hydroxypropan-2-yl 6-[1-(4-fluorophenyl)-1H-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate;
1.1.1- trifluoro-3-hydroxypropan-2-yl (1 a,5a,6a)-6-[1-{5-methoxypyridin-2-yl)-1 H-pyrazol-
3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxylate;
1,1, 1-trifluoro-3-hydroxypropan-2-yl (1 a,5a,6a)-6-[1-(4-fluorophenyl)-1 H-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate;
1.1.1- trifluoro-3-hydroxypropan-2-yl4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate;
1.1.1- trifluoro-3-hydroxypropan-2-yl 4-(phenylsulfonyl)-1-oxa-4,9- diazasplro[5,5]undecane-9-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[(phenylsulfonyl)amino]-1-oxa-8azaspiro[4,5]decane-8-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[( 3-fluorophenyl) sulfony l]-1 -oxa-4,9diazaspiro[5.5]undecane-9-carboxylate;
1,1,1 -trifluoro-3-hydroxypropan-2-yl 3-[methyl(phenylsulfonyl)amîno]-1-oxa-8azaspiro[4.5]decane-8-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{4-fluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-820 carboxylate;
1,1,1 -trifluoro-3-hydroxypropan-2-yl 4-[(4-fluorophenyl)sulfonyl]-3-hydroxy-1 -oxa-4,9diazaspiro[5.5]undecane-9-carboxylate;
3.3.3- trifluoro-2-[{{3-[methyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]dec-8yl)carbonyl)oxy]propyl dihydrogen phosphate;
3,3,3-trifluoro-2-[{{4-[{4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl dihydrogen phosphate;
3.3.3- trifluoro-2-[((4-[{3-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy] propyl dihydrogen phosphate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[1-{tetrahydro-2H-pyran-4-yl)-1 H-pyrazol-3- yl]pîperidine-1 -carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(4-fluorobenzyl)-1-oxa-4,9diazasplro[5.5]undecane-9-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(3,4-difluorophenyl)sulfonyl]-1*oxa-419diazaspiro[5.5]undecane-9-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl4-[(4-ethynylphenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-(4-fluorobenzyl)-2-oxo-1-oxa-3,8diazaspiro[4.5]decane-8-carboxylate;
3.3.3- trifluorO’2-({[4-(phenylsulfonyl)-1-oxa-4,9-diazaspiro[5.5]undec-9yl]carbonyl}oxy)propyl dihydrogen phosphate;
1.1.1- trifluoro-3’hydroxypropan-2-yl 3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8azaspiro[4.5]decane-8-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[(cyclopropylsulfonyl)(methyl)amino]-1 -oxa-8azasplro[4.5]decane-8-carboxylate;
1.1.1- trifluoro-3-hydroxypropan-2-yl 3-[benzoyl(methyl)amrno]-1-oxa-8azaspiro[4.5]decane-8-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{[(cyclopropylmethyl)sulfonyl](methyl)amino}-1 - oxa-8-azaspiro[4.5]decane-8-carboxylate;
1,1, 1-trifluoro-3-hydroxypropan-2-yl 3-[3-(trifluoromethoxy)phenyl]-1 -oxa-8azaspiro[4.5]decane-8-carboxylate;
1,1,1-trifluoro-3-hydroxypropan-2-yl 2-(cyclopentylcarbonylT2,8-diazaspiro[4.5]decane8-carboxylate;
1,1,1-trifluoro-3'hydroxypropan-2-yl 3-{methyl[(2,2,2-trifluoroethyl)sulfonyl]amino}-1-oxa*
8-aza spiro[4.5]deca n e-8-carboxylate ;
1,1, 1-trifluoro-3-hydroxypropan-2-yl 3-{niethyl[(2-methylpropyl)sulfonyl]amino}-1 -oxa-8azaspiro[4.5]decane-8-carboxylate; and
1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[(cyclopropylacetyl)(methyl)amino]-1-oxa-8- azaspiro[4.5]decane-8-carboxylate, or a pharmaceutically acceptable sait thereof;
or a pharmaceutically acceptable sait selected from:
3,3,3’trifluoro-2-[({3-[methyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]dec-8yl}carbonyl)oxy]propyl phosphate, disodium sait;
3,3,3-trifluoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, disodium sait;
313.3- trifluoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, (bis)-L-lysine sait;
3,3,3’trifluoro-2-[({4-[(3-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-930 yl}carbonyl)oxy]propyl phosphate, disodium sait; and
3.3.3- trifluoro-2-({[4-(phenylsulfonyl)-1-oxa-4,9-diazaspiro[5.5]undec-9yl]carbonyl}oxy)propyl phosphate, disodium sait.
The présent invention includes any subset of any embodiment described herein.
The présent invention includes combinations of two or more embodiments described hereinabove, or any subset thereof.
The présent invention further provides the compound of Formula I or a pharmaceutically acceptable sait thereof (including ali embodiments and combinations of two or more
embodiments described herein or any subcombfnatïon thereof) for use in the treatment of a MAGL-mediated disease or disorder described herein.
The présent invention further provides use of the compound of Formula i or a pharmaceutically acceptable sait thereof (including ail embodiments and combinations of two or 5 more embodiments described herein or any subcombination thereof) for treating a MAGLmediated disease or disorder disorder described herein.
The présent invention further provides a method for treating a MAGL-mediated disease or disorder In a patient (e.g., a mammal such as a human) comprising administering to the patient a therapeutically effective amount of the compound of Formula I or a pharmaceutically 10 acceptable sait thereof (including ail embodiments and combinations of two or more embodiments described herein or any subcombination thereof).
The présent Invention further provides use of the compound of Formula I or a pharmaceutically acceptable sait thereof (including ail embodiments and combinations of two or more embodiments described herein or any subcombination thereof) In the manufacture of a 15 médicament for use In the treatment of a MAGL-mediated disease or disorder described herein.
The compound of Formula I or a pharmaceutically acceptable sait thereof of the présent invention (or a métabolite thereof) is a MAGL Inhibitor. Thus, the présent invention further provides a method for inhibiting MAGL (i.e., an activity of MAGL either in vitro or in vivo), comprising contacting (including incubating) the MAGL with the compound of Formula I or a 20 pharmaceutically acceptable sait thereof (such as one selected from Examples 1-91 herein) described herein.
The amount of the compound of Formula I or a pharmaceutically acceptable sait thereof used in any one of the methods (or uses) of the présent invention is effective in inhibiting MAGL.
MAGL-mediated diseases or disorders include, for example, a metabolic disorder (e.g., obesity); a kidney disease (e.g. acute inflammatory kidney injury and diabetic nephropathy); vomiting or emesls (e.g. chemotherapy induced vomiting); nausea (e.g. refractory nausea or chemotherapy induced nausea); an eating disorder (e.g anorexla or bulimia); neuropathy (e.g., diabetic neuropathy, pellagric neuropathy, alcoholic neuropathy, Béribéri neuropathy); burning feet syndrome; a neurodegenerative disorder [multiple sclerosis (MS), Parkinson’s disease (PD), Huntington's disease, dementia, Alzheimer's disease, amyotrophie latéral sclerosis (ALS), epilepsy, fronto-temporal lobe dementia, a sleep disorder, Creutzfeldt-Jakob disease (CJD), or prion disease], a cardiovascular disease (e.g., hypertension, dyshpidemia, atherosclerosis, cardiac arrhythmias, or cardiac Ischemia); osteoporosis; osteoarthritis; schizophrénie;
dépréssion; bipolar disease; tremor; dyskinesia; dystonia; spastîcîty; Tourette’s syndrome; sleep apnea; hearing loss; an eye disease (e.g., glaucoma, ocular hypertension, macular degeneration, ora disease arisingfromelevated intraocular pressure); cachexia; Insomnia; meningitis; sleeping sickness; progressive multifocal leukoencephalopathy; De Vivo disease;
cérébral edema; cérébral palsy; withdrawal syndrome [alcohol withdrawal syndrome, antidepressant discontinuation syndrome, antipsychotic withdrawal syndrome, benzodiazépine withdrawal syndrome, cannabis withdrawal, néonatal withdrawal, nicotine withdrawal, oroploid withdrawal]; traumatic brain Injury; non-traumatic brain injury; spinal cord injury; setzures; excïtotoxin exposure; ischemia [stroke, hepatic ischemia or reperfuston, CNS ischemia or reperfusion]; liver fibrosis, iron overioad, cirrhosis of the liver; a lung disorder [asthma, allergies, COPD, chronic bronchitis, emphysema, cystic fibrosis, pneumonia, tuberculosis, pulmonary edema, lung cancers, acute respiratory distress syndrome, intersitital lung disease (ILD), sarcoidosis, idiopathic pulmonary fibrosis, pulmonary embolism, pleural effusion, or mesothelioma]; a liver disorder [acute liver failure, Alagille syndrome, hepatitis, enlarged liver, Gilbert’s syndrome, liver cysts, liver hemangioma, fatty liver disease, steatohepatitis, primary sclerosing cholangitis, fascioliasis, primary bilary cirrhosis, Budd-Chiari syndrome, hemochromatosis, Wîlson's disease, or transthyretin-related hereditary amyloidosis], stroke [e.g., ischémie stroke; hémorrhagie stroke]: subarachnoid hemorrhage; intracérébral hemorrhage; vasospasm; AIDS wasting syndrome; rénal ischemia; a disorder associated with abnormal ceîl growth or prolifération [e.g., a benign tumor or cancer such as benign skin tumor, brain tumor, papilloma, prostate tumor, cérébral tumor (glioblastoma, medulloepithelioma, medullobiastoma, neuroblastoma, astrocytoma, astroblastoma, ependymoma, oligodendroglioma, plexus tumor, neuroepithelioma, epiphyseal tumor, ependymoblastoma, malignant meningioma, sarcomatosis, melanoma, schwannoma), melanoma, metastatic tumor, kidney cancer, bladder cancer, brain cancer, glioblastoma (GBM), gastrointestinal cancer, leukemia or blood cancer]; an autoimmune diseas [e.g., psoriasis, lupus erythematosus, Sjogren’s syndrome, ankylosing spondylitis, undifferentiated spondylitis, Behcet’s disease, hemolytic anémia, graft rejection]; an inflammatory disorder [e.g., appendicitis, bursitis, colitis, cystitis, dermatltis, phlebitis, rhinitis, tendonitis, tonsïllitis, vasculitis, acné vulgaris, chronic prostatitis, glomerulonephritis, hypersensitivities, IBS, pelvic inflammatory disease, sarcoidosis, HIV encephalitis, rabies, brain abscess, neuroinflammation, Inflammation in the central nervous system (CNS)]; a disorder of the immune system (e.g., transplant rejection orceliac disease); post-traumatic stress disorder (PTSD); acute stress disorder; panic disorder; substanceinduced anxiety; obsessive-compulsive disorder (OCD); agoraphobia; spécifie phobia; social phobla; anxiety disorder; attention déficit disorder (ADD); attention déficit hyperactivity disorder (ADHD); Aspergeras syndrome; pain [e.g., acute pain; chronic pain; Inflammatory pain; viscéral pain; post-operative pain, migraine; iower back pain; joint pain; abdominal pain; cliest pain; postmastectomy pain syndrome; menstrual pain; endometriosis pain; pain due to physical trauma; headache; sinus headache; tension headache arachnoiditis, herpes virus pain, dlabetic pain; pain due to a disorder selected from: osteoarthritis, rheumatoid arthritis, osteoarthritis, spondylitis, goût, labor, musculoskeletal disease, skin disease, toothache, pyresis, burn, sunbum, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitiel cystitis, urinary tract infection (UTI). rhlnitis, contact dermatitis/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, irritable bowel syndrome (IBS), cholecystitis, and pancreatitis; neuropathie pain (e.g., neuropathie low back pain, complex régional pain syndrome, post trigeminal neuralgia, causalgia, toxic neuropathy, reflex sympathetic dystrophy, diabetic neuropathy, chronic neuropathy from chemotherapeutïc agent, or sciatica pain)]; a demyelinating disease [e.g., multiple sclerosis (MS), Devic's disease, CNS neuropathies, central pontine myelinolysis, syphilitic myelopathy, leukoencephalopathies, leukodystrophies, Guillain-Barre syndrome, chronic inflammatory demyelinating polyneuropathy, anti-myelinassociated glycoprotein (MAG) peripheral neuropathy, Charcot-Marie-Tooth disease, peripheral neuropathy, myelopathy, optic neuropathy, progressive inflammatory neuropathy, optic neuritis, transverse myeîitis]; and cognitive Impairment [e.g., cognitive impairment associated with Down's syndrome; cognitive impairment associated with Alzheîmer*s disease; cognitive impairment associated with PD; mild cognitive impairment (MCI), dementia, post-chemotherapy cognitive impairment (PCCI), postoperative cognitive dysfunction (POCD)].
The term therapeutically effective amount as used herein refers to that amount of the compound (including a pharmaceutically acceptable sait thereof) being administered which will relieve to some extent one or more of the symptoms of the disorder being treated. In reference to the treatment of a MAGL-mediated disease or disorder (e.g., Alzheimeris disease, inflammation, or pain), a therapeutically effective amount refers to that amount which has the effect of relieving to some extent (or, for example, eliminating) one or more symptoms associated with the MAGL-mediated disease or disorder (e.g., psychotic symptom of Alzheimeris disease).
The term treating, as used herein, unless otherwise indicated, means reversrng, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term treatment, as used herein, unless otherwise Indicated, refers to the act of treating as treating is defined herein. The term “treating also includes adjuvant and neo-adjuvant treatment of a subject.
As used herein, the term “adjacent* in describing the relative positions of two substituent groups on a ring structure refers to two substituent groups that are respectively attached to two ring-forming atoms of the same ring, wherein the two ring-forming atoms are directly connected through a chemical bond. For example, in each of the following structures:
either of the two R™ groups is an adjacent group of RM0.
As used herein, the term “n-membered, where n is an integer, typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n. For
example, pyridine is an example of a 6-membered heteroaryl ring and thiophene is an example of a 5-membered heteroaryl group,
At various places in the présent spécification, substituents of compounds of the invention are disclosed in groups or in ranges. It Is specifically intended that the invention include each and every individual sub-combination of the members of such groups and ranges. For example, the term Cm alkyl is specifically Intended to include Ci alkyl (methyl), C2 alkyl (ethyl), Ca alkyl, C4 alkyl, C9 alkyl, and Ce alkyl. For another example, the term a 5- to 10membered heteroaryl group is specifically intended to Include any 5-, 6-, 7-, 8-, 9- or 10membered heteroaryl group.
As used herein, the term alkyl Is defined to include saturated aliphatic hydrocarbons including straight chains and branched chains. In some embodiments, the alkyl group has 1 to 20 carbon atoms, 1 to 10 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. For example, the term Cm alkyl, as well as the alkyl moieties of other groups referred to herein (e.g., Cm alkoxy) refers to linear or branched radicals of 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, Isobutyl, seobutyl, tert-butyl, n-pentyl, orn-hexyl). For yet another example, the term Cm alkyl refers to linear or branched aliphatic hydrocarbon chains of 1 to 4 carbon atoms; the term Cm alkyl refers to linear or branched aliphatic hydrocarbon chains of 1 to 3 carbon atoms; the term Cm alkyl refers to methyl and/or ethyl; and the term Ci alkyl refers to methyl. An alkyl group optionally can be substituted by one or more (e.g., 1 to 5) suitable substituents.
As used herein, the term alkenyl refers to aliphatic hydrocarbons having at least one carbon-carbon double bond, including straight chains and branched chains having at least one carbon-carbon double bond. In some embodiments, the alkenyl group has 2 to 20 carbon atoms, 2 to 10 carbon atoms, 2 to 6 carbon atoms, 3 to 6 carbon atoms, or 2 to 4 carbon atoms.
For example, as used herein, the term Cm alkenyl means straight or branched chain unsaturated radicals (having at least one carbon-carbon double bond) of 2 to 6 carbon atoms, Including, but not limited to, ethenyl, 1-propenyl, 2-propenyl (allyl), isopropenyl, 2-methyl-1propenyl, 1-butenyl, 2-butenyl, and the like.. An alkenyl group optionally can be substituted by one or more (e.g., 1 to 5) suitable substituents. When the compounds of Formula I contain an alkenyl group, the alkenyl group may exist as the pure E form, the pure Z form, or any mixture thereof.
As used herein, the term alkynyl refers to aliphatic hydrocarbons having at least one carbon-carbon triple bond, including straight chains and branched chains having at least one carbon-carbon triple bond. In some embodiments, the alkynyl group has 2 to 20, 2 to 10, 2 to 6, 35 or 3 to 6 carbon atoms. For example, as used herein, the term Cm alkynyl refers to straight or branched hydrocarbon chain alkynyl radicals as defined above, having 2 to 6 carbon atoms. An alkynyl group optionally can be substituted by one or more (e.g., 1 to 5) suitable substituents.
As used herein, the term cycloalkyl refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings (e.g., monocyclics such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cydooctyl, cyclononyl, or bicyclics including spiro, fused, or bridged Systems (such as bicyclo[1.1.1]pentanyl, bicyclo[2.2.1]heptanyl, 5 bicyclo[3.2.1]octanyl or bicyclo[5.2.0]nonanyl, decahydronaphthalenyl, etc.). The cycloalkyl group has 3 to 15 carbon atoms. In some embodiments the cycloalkyl may optionally contain one, two or more non-cumulative non-aromatic double or triple bonds and/or one to three oxo groups. In some embodiments, the bicycloalkyl group has 6 to 14 carbon atoms. For example, the term Cj-u cycloalkyl refers to saturated or unsaturated, non-aromatic, monocyclic or 10 polycyclic (such as bicyclic) hydrocarbon rings of 3 to 14 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, blcyclo[1.1.1]pentanyl, or cyclodecanyl); and the term Cm cycloalkyl refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings of 3 to 7 ring-forming carbon atoms (e.g., cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo[1.1.1 ]pentan-1-yl, or bicyclo[1.1.1]pentan-2-yl). For 15 another example, the term Cm cycioalkyl refers to saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon rings of 3 to 6 ring-forming carbon atoms. For yet another example, the term Cm cycloalkyl refers to cyclopropyl or cyclobutyl. Also included in the définition of cycloalkyl are moieties that have one or more aromatic rings (including aryl and heteroaryl) fused to the cycloalkyl ring, for example, benzo or thienyl dérivatives of cyclopentane, cyclopentene, cyclohexane, and the like (e.g., 2,3-dihydro-1Hindene-1-yl, or 1H-inden-2(3H)-one-1-yl). The cycloalkyl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term aryl refers to all-carbon monocyclic or fused-ring polycyclic aromatic groups having a conjugated pl-electron system. The aryl group has 6 or 10 carbon 25 atoms in the ring(s). Most commonly, the aryl group has 6 carbon atoms in the ring. For example, as used herein, the term “C«-io aryl* means aromatic radicals containing from 6 to 10 carbon atoms such as phenyi or naphthyl. The aryl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term “heteroaryl refers to monocyclic or fused-ring polycyclic aromatic heterocyclic groups with one or more heteroatom ring members (ring-forming atoms) each independently selected from O, S and N in at least one ring. The heteroaryl group has 5 to 14 ring-forming atoms, including 1 to 13 carbon atoms, and 1 to 8 heteroatoms selected from O, S, and N. In some embodiments, the heteroaryl group has 5 to 10 ring-forming atoms Including one to four heteroatoms. The heteroaryl group can also contain one to three oxo or thiono (i.e., =S) groups. In some embodiments, the heteroaryl group has 5 to 8 ring-forming atoms Including one, two or three heteroatoms. For example, the term 5-membered heteroaryl refers to a monocyclic heteroaryl group as defined above with 5 ring-forming atoms in the monocyclic heteroaryl ring; the term 6-membered heteroaryl refers to a monocyclic
heteroaryl group as defined above with 6 ring-forming atoms in the monocyclic heteroaryl ring; and the term 5- or 6-membered heteroaryl refers to a monocyclic heteroaryl group as defined above with 5 or 6 ring-forming atoms in the monocyclic heteroaryl ring. For another example, term 5- or 10-membered heteroaryl refers to a monocyclic or bicyclic heteroaryl group as defined above with 5,6, 7,8, 9 or 10 ring-forming atoms in the monocyclic or bicyclic heteroaryl ring. A heteroaryl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents. Examples of monocyclic heteroaryls include those with 5 ring-forming atoms including one to three heteroatoms or those with 6 ring-forming atoms including one, two or three nitrogen heteroatoms. Examples of fused bicyclic heteroaryls Include two fused 5- and/or
6-membered monocyclic rings including one to four heteroatoms.
Examples of heteroaryl groups include pyridinyl, pyrazinyl, pyrimidinyi, pyridazinyl, thienyi, furyi, Imidazolyl, pyrrolyl, oxazolyl (e.g., 1,3-oxazolyl, 1,2-oxazolyi), thlazolyl (e.g., 1,2thiazolyi, 1,3-thiazolyl), pyrazolyl (e.g., pyrazol-1-yl, pyrazol-3-yl, pyrazol-4-yl), tetrazolyl, triazolyl (e.g., 1,2,3-triazolyl, 1,2,4-triazolyl), oxadiazolyl (e.g., 1,2,3-oxadiazolyl), thladiazolyl (e.g., 1,3,4-thladiazolyl), quinolyl, isoquinolyl, benzothienyl, benzofuryl, indolyl, 1H-imidazo[4,5cjpyridinyl, imidazo[1,2-a]pyridinyl, 1H-pyrrolo[3,2-c]pyridinyl, imidazo[1,2-a]pyrazinyl, imidazo[2,1-c][1,2,4]triazïnyl, imidazo[1,5-a]pyrazinyl, imidazo[1,2-a]pyrimidinyl, 1H-indazolyl, 9H-purïnyl, imidazo[1,2-a]pyrimidinyl, [1,2,4]triazolo[1,5-a]pyrimidinyl, [1,2.4]triazolo[4,3bjpyridazinyl, isoxazolo[5,4-c]pyridazinyl, isoxazolo[3,4-c]pyridazinyl, pyridone, pyrimidone, pyrazinone, pyrimidinone, ïH-imidazol-2(3H)-one, ÎH-pyrrole-2,5-dione, 3-oxo-2H-pyridazinyl,
H-2-oxo-pyrimidinyl, 1 H-2-oxo-pyridinyl, 2,4(1 H,3H)-dioxo-pyrimidinyl, 1 H-2-oxo-pyrazinyl, and the like. The heteroaryl group optionally can be substituted by 1 or more (e.g., 1 to 5) suitable substituents.
As used herein, the term heterocycloalkyl refers to a monocyclic or polycyclîc [including 2 or more rings that are fused together, including spiro, fused, or bridged Systems, for example, a bicyclic ring system], saturated or unsaturated, non-aromatic 4- to 15-membered ring System (such as a 4- to 14-membered ring system, 4- to 12-membered ring system, 5- to 10-membered ring system, 4- to 7-membered ring system, 4- to 6-membered ring system, or 5to 6-membered ring system), including 1 to 14 ring-forming carbon atoms and 1 to 10 rlng30 forming heteroatoms each independently selected from O, S and N (and optionally P or B when présent). The heterocycloalkyl group can aiso optionally contain one or more oxo (Le., =0) or thiono (i.e., =S) groups. For example, the term 4- to 12-membered heterocycloalkyl refers to a monocyclic or polycyclîc, saturated or unsaturated, non-aromatic 4- to 12-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from
O, S and N; and the term 4- to 10-membered heterocycloalkyl refers to a monocyclic or polycyclîc, saturated or unsaturated, non-aromatic 4-to 10-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from O, S and N. For another example, the term 4- to 6-membered heterocycloalkyl refers to a monocyclic
or polycyclic, saturated or unsaturated, non-aromatîc 4- to 6-membered ring system that comprises one or more ring-forming heteroatoms each independently selected from O, S and N; and the term 5- to 6-membered heterocycloalkyl refera to a monocyclic or polycyclic, saturated or unsaturated, non-aromatic 5- to 6-membered ring system that comprises one or more ring-forming heteroatoms each Independently selected from O, S and N. Also included in the définition of heterocycloalkyl are moieties that hâve one or more aromatic rings (including aryl and heteroaryl) fused to the nonaromatic heterocycloalkyl ring, for example pyridinyl, pyrimidinyl, thiophenyl, pyrazolyl, phthalimidyl, naphthalimidyl, and benzo dérivatives of the nonaromatic heterocycloalkyl rings. The heterocycloalkyl group optionally can be substituted by 10 1 or more (e.g., 1 to 5) suitable substituents.
Examples of such heterocycloalkyl rings include azetidinyl, tetrahydrofuranyl, imidazolidinyl, pyrrolidinyl, piperidinyl, piperazînyl, oxazolidinyl, thiazolidinyl, pyrazolidinyl, thiomorpholinyl, tetrahydrothiazinyl, tetrahydrothiadiazinyI, morpholinyl, oxetanyl, tetrahydrodiazinyl, oxazinyl, oxathiazinyl, quinuclidinyl, chromanyl, isochromanyl, benzoxazinyl,
2-oxaspiro[3.3]heptyl {e.g., 2-oxaspiro[3.3]hept-6-yl}, 7-azabicyclo[2.2.1]heptan-1-yl, 7azabicyclo[2.2.1]heptan-2-yl, 7-azabicyclo[2.2.1]heptan-7-yl, 2-azabicyclo[2.2.1]heptan-3-on-2yl, 3-azabicyclo[3.1.0]hexanyl, 3-azabîcyclo[4.1.0]heptanyl and the like. Further examples of heterocycloalkyl rings Include tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydropyranyl (e.g., tetrahydro-2H-pyran-4-yl), lmidazolidin-1-yl, imtdazolidin-2-yl, imidazolidin-4-yl, pyrrolidin-1-yl, 20 pyrrolidin-2-yl, pyrrolidin-3-yl, piperidin-1 -yl, piperidin-2-yl, piperidin-3-yl, piperidin-4-yl, piperazin-1-yl, piperazin-2-yl, 1,3-oxazolidin-3-yl, 1,4-oxazepan-1-yl, isothiazolidînyl, 1,3thiazolldin-3-yl, 1,2-pyrazolidin-2-yl, 1,2-tetrahydrothiazin-2-yl, 1,3-thlazinan-3-yl, 1,2tetrahydrodiazin-2-yl, 1t3-tetrahydrodiazin-1-yl, 1,4-oxazin-4-yl, oxazolidinonyl, 2-oxo-piperidinyl (e.g., 2-oxo-piperidin-1-yl), 2-oxoazepan-3-yl, and the like. Some examples of aromatic-fused 25 heterocycloalkyl groups include indolinyl, Isoindolinyl, isoindolin-1-one-3-yl, 5,7-dihydro-6HpyrTolotS^-bJpyrîdin-G-yl, 6,7-dihydro-5H-pyrrolo[3,4-d]pyrimidin-6-yl, 4,5,6,7tetrahydrothienotZ.S-clpyridine-S-yl, 5l6-dihydrothieno[2,3-c]pyridin-7(4H)-one-5-yl11,4,5,6tetrahydropyrrolotS^-clpyrazol-S-yl, and 3,4-dihydroisoquinolin-1(2H)*one-3-yl groups. The heterocycloalkyl group Is optionally substituted by 1 or more (e.g., 1 to 5) suitable substituents. 30 Examples of heterocycloalkyl groups include 5- or 6-membered monocyclic rings and 9- or 10membered fused bicyclic rings.
As used herein, the term halo or halogen group is defined to include fluorine, chlorine, bromine or iodine.
As used herein, the term “haloalkyl refera to an alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom of the alkyl group has been replaced by a halogen atom). For example, the term “Cm haloalkyl refera to a Cm alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e., every hydrogen atom ofthe alkyl group has been replaced by a halogen atom). For another example, the term “Cm haloalkyl
refers to a Cm alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e, every hydrogen atom of the alkyi group has been replaced by a halogen atom); the term “Cm haloalkyl refers to a Cm alkyl group having one or more halogen substituents (up to perhaloalkyl, i.e, every hydrogen atom of the alkyl group has been replaced by a halogen atom); and the term Ci-2 haloalkyl” refera to a Cm alkyl group (i.e, methyl or ethyi) having one or more halogen substituents (up to perhaloalkyl, I.e, every hydrogen atom of the alkyl group has been replaced by a halogen atom). For yet another example, the term Ci haloalkyl refera to a methyl group having one, two, or three halogen substituents. Examples of haloalkyl groups Include CFj, C2FS, CHF2, CH2F, CH2CF3, CH2Ci and the like.
As used herein, the term aikoxy or alkyloxy refera to an -O-alkyl group. For example, the term Cm alkoxy or “Cm alkyioxy refera to an -0-(Cm alkyl) group; and the term “Cm alkoxy or “Cm alkyloxy refera to an -0-(Cm alkyl) group; For another example, the term “Ci-2 alkoxy or Ci-2 alkyloxy refera to an -O-(Ci-2 alkyl) group. Examples of alkoxy Include methoxy, ethoxy, propoxy (e.g, n-propoxy and Isopropoxy), tert-butoxy, and the like. The alkoxy or alkyloxy group optionally can be substituted by 1 or more (e.g, 1 to 5) suitable substituents.
As used here, the term haloalkoxy refera to an -O-haloalkyl group. For example, the term “Cm haloalkoxy* refera to an -O-(Cm haloalkyl) group. For another example, the term “Cm haloalkoxy refera to an -O-(Cm haloalkyl) group; and the term “Ci-2 haloalkoxy refera to an -O(Ci.2 haloalkyl) group. For yet another example, the term Ci haloalkoxy” refera to a methoxy 20 group having one, two, or three halogen substituents. An example of haloalkoxy Is -OCFj or OCHF2.
As used herein, the term fluoroalkyl refera to an alkyl group having one or more fluorine substituents (up to perfluoroalkyl, I.e, every hydrogen atom of the alkyl group has been replaced by fluorine). For example, the term “Ci-2 fluoroalkyl refera to a Ci-2 alkyl group having 25 one or more fluorine substituents (up to perfluoroalkyl, i.e, every hydrogen atom of the Ci.2 alkyl group has been replaced by fluorine). For another example, the term Ci fluoroalkyl refera to a Ci alkyl group (I.e, methyl) having 1,2, or 3 fluorine substituents). Examples of fluoroalkyl groups Include CF3, C2Fs, CH2CF3, CHF2, CH2F, and the like.
As used here, the term “fluoroalkoxy refera to an -O-fluoroalkyi group. For example, the 30 term “Ci.2 fluoroalkoxy refera to an -O-C1.2 fluoroalkyl group. For another example, the term “Ci fluoroalkoxy refera to a methoxy group having one, two, or three fluorine substituents. An example of Ci fluoroalkoxy Is -OCFs or -OCHF2.
As used herein, the term ' hydroxylaikyl” or “hydroxyalkyl refers to an alkyi group having one or more (e.g, 1,2, or 3) OH substituents. The term “Cm hydroxylalkyi or “Cm hydroxyalkyl refera to a Cm alkyl group having one or more (e.g, 1, 2, or 3) OH substituents. The term Cm hydroxylalkyi or Cm hydroxyalkyl refera to a Cm alkyi group having one or more (e.g, 1, 2, or 3) OH substituents; the term C1.3 hydroxylalkyi or Cm hydroxyalkyl refera to a C1-3 alkyl group having one or more (e.g, 1,2, or 3) OH substituents; and the term Cm
hydroxylalkyl or Cm hydroxyalkyl refers to a Cm alkyl group having one or more (e.g., 1,2, or
3) OH substituents. An example of hydroxylalkyl Is -CHjOH or -CH2CH2OH. As used herein, the term “cyanoalkyl refers to an alkyl group having one or more (e.g.,
1,2, or 3) -CN substituents. The term Cm cyanoalkyl refers to a Cm alkyl group having one 5 or more (e.g., 1, 2, or 3) -CN substituents. For example, Ci cyanoalkyl is Ci alkyl (I.e., methyl) having one or more (e.g., one) -CN substituents. An example of cyanoalkyl is -CH2CN or -
CH2CH2CN.
As used herein, the term “oxo refers to =0. When an oxo is substituted on a carbon atom, they together form a carbonyl moiety [-0(=0)-]. When an oxo is substituted on a sulfur 10 atom, they together form a sulfïnyl moiety [-S(=O)-]; when two oxo groups are substituted on a sulfur atom, they together form a sulfonyl moiety [-S(=0H.
As used herein, the term “thiono refers to =S. When an thiono is substituted on a carbon atom, they together form moiety of [-C(=S)-J.
As used herein, the term optionally substituted means that substitution is optional and 15 therefore includes both unsubstituted and substituted atoms and moieties. A substituted atom or moiety Indicates that any hydrogen on the deslgnated atom or moiety can be replaced with a sélection from the indicated substituent group (up to that every hydrogen atom on the designated atom or moiety is replaced with a sélection from the indicated substituent group), provided that the normal valency of the designated atom or moiety Is not exceeded, and that 20 the substitution results in a stable compound. For example, If a methyl group (Le., CHa) Is optionally substituted, then up to 3 hydrogen atoms on the carbon atom can be replaced with substituent groups.
As used herein, the term optionally substituted Cm alkyl refers to Cm alkyl optionally substituted by one or more (e.g., 1 to 5) substituents each independently selected from the group 25 consisting of -OH, halogen, -CN, -NH2, -NH(Cm alkyl), -N(Cm alkyl)2, Cm alkoxy, and Cm haloalkoxy.
As used herein, the term optionally substituted Cm cycloalkyl refers to Cm cycloalkyl optionally substituted by one or more (e.g., 1 to 5) substituents each Independently selected from the group consisting of-OH, halogen, -CN, -NH2, -NH(Cm alkyl), -N(Cm alkyl)2, Cm alkyl, Cm 30 haloalkyl, Cm hydroxylalkyl, Cm alkoxy, and Cm haloalkoxy.
As used herein, the term optionally substituted Cm cycloalkyl-Ci-2 alkyl- refers to Cm cycloalkyl-Ci-2 alkyl- optionally substituted by one or more (e.g., 1 to 5) substituents each independently selected from the group consisting of -OH, halogen, -CN, -NH2, -NH(Cm alkyl), N(Cm alkyl)2, Cm alkyl, Cm haloalkyl, Cm hydroxylalkyl, Cm alkoxy, and Cm haloalkoxy.
As used herein, the term optionally substituted Cm alkoxy refers to Cm alkoxy optionally substituted by one or more (e.g., 1 to 5) substituents each Independently selected from the group consisting of-OH, halogen, -CN, -NH2, -NH(Cm alkyl), -N(Cm alkyl)2, Cm alkoxy, and Cm haloalkoxy.
As used herein, unless specified, the point of attachment of a substituent can be from any suitable position of the substituent. For example, piperidinyl can be piperidin-1-yl (attached through the N atom of the piperidinyl), piperidin-2-yl (attached through the C atom at the 2position of the piperidinyl), piperidin-3-yl (attached through the C atom at the 3-position of the piperidinyl), or piperidin-4-yl (attached through the C atom atthe 4-position of the piperidinyl). For another example, pyridtnyl (or pyridyl) can be 2-pyridinyl (or pyridin-2-yl), 3-pyridinyl (or pyridin-3-yl), or 4-pyridinyl (or pyridin-4-yl).
As used herein, the point of attachment of a substituent can be specified to indicate the position where the substituent is attached to another moiety. For example, '-Cu alkyl-fC» cycloalkyl) means the point of attachment occurs at the Cm alkyl part of the -Cm alkyUCw cycloalkyl). For another example, (Cm cycloalkyl)-CM alkyl-’ also means the point of attachment occurs at the Cm alkyl part of the “(Cm cycloalkyl)-CM alkyl-.
When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any of the ring-forming atoms In that ring that are substitutable (i.e., bonded to one or more hydrogen atoms), unless otherwise specified or otherwise implicit from the context. For example, as shown In the structure of Formula a-6 below, RB may be bonded to any of the ring atoms of ring A1, but not to the ring including the N atom as shown in Formula a-6. For another example, as shown in Formula a-5 below (when t1 is 1 ), the R* group can be bonded to any of the ring carbon atoms or the N atom (of the NH moiety) because the cross-bond is through both rings of the bicyclic structure; on the other hand, RB can only be bonded to the N atom (of the NH moiety) and the two carbon ring atoms that are directly connected to the N atom (of the NH moiety). R8 cannot be bonded to either of the carbon atom of the moiety of CH2CH2” (the H atoms are not shown) of the pyrrolîdine ring of the bicyclic System of Formula a-5 because the bond does not cross the pyrrolidine ring.
a-6
As used herein, unless otherwise specifically indicated, a linkage/linker—a moiety that links two other moieties—can bc attached to the other two moieties in ci’hcr direction, if the linkage/linker Is asymmetric. For example, when R8 is -L’-R11 and L1 is -S(=O)2-NR23-, then R8 can be either -S(=O)rNR23-R11 or -NR23-S(=O)î-R11 (unless otherwise specifically indicated).
When a substituted or optionally substituted moiety is described without indicating the atom via which such moiety is bonded to a substituent, then the substituent may be bonded via any appropriate atom in such moiety. For example in a substituted arylalkyl, a substituent on
the arylalkyl [e.g., (Ce-ίο aryl)-Cw alkyl-] can be bonded to any carbon atom on the alkyl part or on the aryl part of the arylalkyl. Combinations of substituents and/or variables are permissible only if such combinations resuit ln stable compounds.
As noted above, the compounds of Formula I may exist ln the form of pharmaceutically acceptable salts such as acid addition salts and/or base addition salts of the compounds of Formula I. The phrase pharmaceutically acceptable salt(s), as used herein, unless otherwise Indicated, includes acid addition or base salts which may be présent in the compounds of Formula I.
Pharmaceutically acceptable salts of the compounds of Formula I Include the acid addition and base salts thereof.
Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulfate/sulfate, borate, camphorsulfonate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, giucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromîde/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulfate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stéarate, succinate, tannate, tartrate, tosylate, trifiuoroacetate and xînafoate salts.
Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnésium, meglumine, olamlne, potassium, sodium, tromethamine and zinc saîts.
H émisait s of acids and bases may also be formed, for example, hemlsulfate and hemicalcium salts.
For a review on suitable salts, see “Handbook of Pharmaceutical Salts: Properties,
Sélection, and Use by Stahl and Wermuth (Wiiey-VCH, 2002), Methods for making pharmaceutically acceptable salts of compounds of Formula I are known to one of skill in the art.
As used herein the terms Formula Γ or “Formula I or a pharmaceutically acceptable sait thereof are defined to Include ali forms of the compound of Formula I or pharmaceutically sait thereof, including hydrates, solvatés, isomers (including for example rotational stereoisomers), crystalline and non-crystalllne forms, isomorphs, polymorphs, métabolites, and prodrugs thereof.
As is known to the person skilled ln the art, amine compounds (Le., those comprising one or more nitrogen atoms), for example tertiary amines, can form W-oxides (also known as amine oxides or amine N-oxides). An W-oxide has the formula of (R1M)(R200)(R’00)N*-O· wherein the parent amine ^100)^200)^300^ can be, for example, a tertiary amine (for example, each of R100, R200, R300 is independently alkyl, arylalkyl, aryl, heteroaryl, or the like), a heterocyclic or heteroaromatic amine [for example, (R100)(R200)(R*’0)N together forms 1alkylpipendine, 1-alkylpyrrolidine, 1-benzylpyrrolidme, orpyndine]. For Instance, an Imine nitrogen, especlally a heterocyclic or heteroaromatic Imine nitrogen, or pyridine-type nitrogen ( 4=Ν-ί) gtorn [such as a nitrogen atom in pyridine, pyridazine, or pyrazine], can be N-oxidized 0 to form the N-oxlde comprising the group ( ΉΗ-). Thus, a compound according tothe présent Invention comprising one or more nitrogen atoms (e.g., an imine nitrogen atom) may be capable of forming an N-oxide thereof (e.g., mono-N-oxides, bis-N-oxides or multi-N-oxides, or mixtures thereof depending on the number of nitrogen atoms suitable to form stable N-oxides).
As used herein, the term N-oxide(s) refer to ail possible, and in particular ail stable, Noxide forms of the amine compounds (e.g., compounds comprising one or more imine nitrogen atoms) described herein, such as mono-N-oxides (including different isomers when more than one nitrogen atom of an amine compound can form a mono-N-oxide) or multi-N-oxides (e.g., bis-N-oxides), or mixtures thereof In any ratio.
Compounds of Formula I and their salts described herein further include N-oxides thereof.
In the description herein below, unless otherwise specified, compounds of Formula I (or compounds of the Invention) Include salts of the compounds and the N-oxides of the compounds or the salts.
As Is also known to the person skilled in the art, tertiary amine compounds (I.e., those comprising one or more tertiary amine nitrogen atoms) can form quatemary ammonium salts. In the description herein below, unless otherwise specified, compounds of Formula I (or compounds of the invention) further include their quaternary ammonium salts.
Compounds of Formula I may exist In a continuum of solid states ranging from fully amorphous to fully crystalline. The term 'amorphous* refers to a state In which the material lacks long-rangé order at the molecular level and, depending upon température, may exhibit the physical propertîes of a solid or a liquid. Typically such materials do not give distinctive X-ray diffraction patterns and, while exhibiting the propertîes of a solid, are moreformally described as a liquid. Upon heating, a change from apparent solid to a material with liquid propertîes occurs, which is characterised by a change of state, typically second order (’glass transition'). The term 'crystalline' refers to a solid phase in which the material has a regular ordered internai structure at the molecular level and gives a distinctive X-ray diffraction pattern with defined peaks. Such materials when heated sufficîently will also exhibit the propertîes of a liquid, but the change from solid to liquid is characterized by a phase change, typically first order (’meltîng point').
Compounds of Formula I may exist in unsolvated and solvated forms. When the solvent or water is tightly bound, the complex will hâve a well-defined stoichiometry independent of humldity. When, however, the solvent or water Is weakly bound, as in channel solvatés and
hygroscopic compounds, the water/solvent content will be dépendent on humidity and drying conditions. In such cases, non-stoichiometry will be the norm.
The compounds of Formula I may exist as clathrates or other complexes (e.g., cocrystals). Included within the scope of the invention are complexes such as clathrates, drug5 host inclusion complexes wherein the drug and host are présent in stoichiometric or nonstoichiometric amounts. Also Included are complexes of the compounds of Formula I containing two or more organic and/or inorganic components, which may be in stoichiometric or nonstoichiometric amounts. The resulting complexes may be ionized, partially ionized, or nonionized. Co-crystals are typically defined as crystalline complexes of neutral molecular constltuents that are bound together through non-covalent interactions, but could also be a complex of a neutral molécule with a sait. Co-crystals may be prepared by melt crystallization, by recrystallization from solvents, or by physically grinding the components together; see O. Almarsson and M. J. Zaworotko, Chem. Commun. 2004,17,1889-1896. Fora general review of multi-component complexes, see J. K. Haleblian, J. Pharm. Sel. 1975, 64,1269-1288.
The compounds of the invention may also exist in a mesomorphic state (mesophase or liquid crystal) when subjected to suitable conditions. The mesomorphic state is intermediate between the true crystalline state and the true liquid state (either melt or solution). Mesomorphism arising as the resuit of a change in température is described as ’thermotropic’ and that resulting from the addition of a second component, such as water or another solvent, is 20 described as ’lyotropic*. Compounds that hâve the potential to form lyotropic mesophases are described as 'amphiphilic' and constat of molécules which possess an Ionie (such as -COONa*, -COO K*, or -SO3’Na+) or non-ionic (such as -NN*(CH3)3) polar head group. For more information, see Crystals and the Polarizing Microscope by N. H. Hartshome and A. Stuart, 401 Edition (Edward Arnold, 1970).
The invention also relates to prodrugs of the compounds of Formula I, Thus certain dérivatives of compounds of Formula I which may hâve little or no pharmacologlcal activity themselves can, when administered into or onto the body, be converted into compounds of Formula I having the desired activity, for example, by hydrolytic cleavage. Such dérivatives are referred to as prodrugs. Further information on the use of prodrugs may be found in Pro-drugs 30 as Novel Delivery Systems, Vol, 14, ACS Symposium Sériés (T. Higuchi and W. Stella) and Bioreversible Carriers in Drug Design, Pergamon Press, 1987 (Ed. E. B. Roche, American Pharmaceutical Association).
Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities présent in the compounds of Formula I with certain moieties known 35 to those skilled in the art as 'pro-moieties' as described, for example, in Design of Prodrugs by H. Bundgaard (Elsevler, 1985), or in Prodrugs: Challenges and Reward, 2007 édition, edited by Valentino Stella, Ronald Borchardt, Michael Hageman, Reza Oliyai, Hans Maag, Jefferson Tilley, pages 134-175 (Springer, 2007).
Moreover, certain compounds of Formula I may themselves act as prodrugs of other compounds of Formula I.
Also included within the scope of the invention are métabolites of compounds of Formula I, that is, compounds formed in vivo upon administration of the drug.
The compounds of Formula I include all stereoisomers and tautomers. Stereoisomers of
Formula I include cis and trans isomers, optical isomers such as R and S enantiomers, diastereomers, géométrie isomers, rotational isomers, atropisomers, and conformational isomers of the compounds of Formula I, including compounds exhibiting more than one type of isomerism; and mixtures thereof (such as racemates and diastereomeric pairs). Also included 10 are acid addition or base addition salts wherein the counterion is optically active, for example, D-lactate or L-lysine, or racemic, for example, DL-tartrate or DL-arginine.
In some embodiments, the compounds of Formula I (including salts thereof) may hâve asymmetric carbon atoms. The carbon-carbon bonds of the compounds of Formula I may be depicted herein using a solid line (------), a wavy line (-------), a solid wedge ( ), or a dotted wedge ( ). The use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that ali possible stereoisomers (e.g., spécifie enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of either a solid or dotted wedge to depict bonds to asymmetric carbon atoms is meant to indicate that only the stereolsomer shown is meant to be included. The use of a wavy iine to depict bonds to asymmetric carbon atoms is 20 meant to indicate that the stereochemistry is unknown (unless otherwise specified). It is possible that compounds of Formula I may contain more than one asymmetric carbon atom. In those compounds, the use of a solid line to depict bonds to asymmetric carbon atoms is meant to indicate that all possible stereoisomers are meant to be included. For example, unless stated otherwise, it is intended that the compounds of Formula i can exist as enantiomers and 25 diastereomers or as racemates and mixtures thereof. The use of a solid line to depict bonds to one or more asymmetric carbon atoms in a compound of Formula I and the use of a solid or dotted wedge to depict bonds to other asymmetric carbon atoms in the same compound is meant to Indicate that a mixture of diastereomers is présent.
In some embodiments, the compounds of Formula I may exist in and/or be isolated as 30 atropisomers (e.g., one or more atropenantiomers). Those skilled in the art would recognize that atropisomerism may exist in a compound that has two or more aromatic rings (for example, two aromatic rings linked through a single bond). See e.g., Freedman, T. B. et al., Absolute Configuration Détermination of Chiral Molécules in the Solution State Using Vibrational Circuler Dichroism. Chirality 2003,15, 743-758; and Bringmann, G. et al., Atroposelective Synthesis of 35 Axially Chiral Biaryl Compounds. Angew. Chem., Int. Ed. 2005,44,5384-5427.
When any racemate crystallizes, crystals of different types are possible. One type is the racemic compound (true racemate) wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. Another type is a racemic mixture or
conglomerate wherein two forms of crystal are produced in equal or different molar amounts each comprising a single enantiomer.
The compounds of Formula I may exhibit the phenomena of tautomerism and structural isomerlsm. For example, the compounds of Formula I may exist in several tautomeric forms, 5 Including the enol and Imine form, the amide and Imidîc acid form, and the keto and enamine form and géométrie isomers and mixtures thereof. Ail such lautomeric forms are inciuded within the scope of the compounds of Formula I. Tautomers may exist as mixtures of a tautomeric set in solution. In solid form, usually one tautomer prédominâtes. Even though one tautomer may be described, the présent Invention includes ail tautomers of the compounds of 10 Formula I. For example, when one of the following two tautomers (wherein R can be, for example, phenyl that Is further substituted) Is disclosed, those skilled In the art wouid readily recognize the other tautomer.
The présent Invention Includes ail pharmaceutically acceptable isotopically labelled compounds of Formula I or salts thereof wherein one or more atoms are repiaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which prédominâtes in nature.
Exemples of Isotopes suitable for inclusion In the compounds of the Invention include isotopes of hydrogen, such as 2H and ’H, carbon, such as nC, 13C and 14C, chlorine, such as 20 “Cl, fluorine, such as 18F, iodine, such as 123l and 12SI, nitrogen, such as 13N and 15N, oxygen, such as 1SO, 17O and 18O, phosphorus, such as “P, and sulphur, such as 35S.
Certain isotopically labelled compounds of Formula I, for example, those Incorporating a radioactive isotope, are useful In drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e., 3H, and carbon-14, I.e., 14C, are particularly useful for this 25 purpose in view of theîr ease of Incorporation and ready means of détection.
Substitution with heavier isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for exampie, Increased in vivo haif-life or reduced dosage requirements, and hence may be preferred in some circumstances.
Substitution with positron-emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful 30 in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
Isotopically iabeled compounds of Formula I can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Préparations using an approprlate isotopically Iabeled reagent in place of the non-labeied reagent previously employed.
The présent invention also provides compositions (e.g., pharmaceutical compositions) comprising a novel compound of Formula I. Accordingly, In one embodiment, the invention
provides a pharmaceutical composition comprising (a therapeutically effective amount of) a novel compound of Formula I or a pharmaceutically acceptable sait thereof and optionally comprising a pharmaceutically acceptable carrier. In one further embodiment, the invention provides a pharmaceutical composition comprising (a therapeutically effective amount of) a 5 compound of Formula I or a pharmaceutically acceptable sait thereof, optionally comprising a pharmaceutically acceptable carrier and, optionally, at least one additional médicinal or pharmaceutical agent (such as an antipsychotic agent or antl-schlzophrenla agent described below). In one embodiment, the additional médicinal or pharmaceutical agent Is an antischizophrenia agent as described below.
The pharmaceutically acceptable carrier may comprise any conventional pharmaceutical carrier or excipient. Suitable pharmaceutical carriers include inert diluents or fillers, water and various organic solvents (such as hydrates and solvatés). The pharmaceutical compositions may, If desired, contain additional Ingrédients such as flavorings, blnders, excipients and the like. Thus for oral administration, tablets containing various excipients, such as citric acid, may be employed together with various désintégrants such as starch, alginic acid and certain complex silicates and with binding agents such as sucrose, gelatin and acacia. Additionally, lubricating agents such as magnésium stéarate, sodium lauryl sulfate and talc are often useful for tableting purposes. Solid compositions of a similar type may also be employed In soft and hard filled gelatin capsules. Non-limiting examples of matériels, therefore, include lactose or milk sugar and high molecular weight polyethylene glycols. When aqueous suspensions or élixirs are desired for oral administration, the active compound therein may be combined with various sweetening orflavoring agents, coloring matters or dyes and, If desired, emulsifytng agents or suspending agents, together with diluents such as water, éthanol, propylene glycol, glycerin, or combinations thereof.
The pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulation, solution or suspension, for parentéral injection as a stérile solution, suspension or émulsion, for topical administration as an ointment or cream or for rectal administration as a suppository.
Exemplary parentéral administration forms include solutions or suspensions of active 30 compounds In stérile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms may be suitably buffered, If desired.
The pharmaceutical composition may be In unit dosage forms suitable for single administration of précisé dosages. One of ordmary skill In the art would appreciate that the composition may be formulated In sub-therapeutic dosage such that multiple doses are 35 envîsioned.
In one embodiment the composition comprises a therapeutically effective amount of a compound of Formula I or sait thereof and a pharmaceutically acceptable carrier.
Compounds of Formula I (including salts thereof) are MAGL Inhibitors. In some embodiments, the IC» of a compound of Formula I (or Its métabolite) Is less than about 10 μΜ, 5 μΜ, 2 μΜ, 1 μΜ, 500 nM, 200 nM, 100 nM, 50,40,30, 20,10, 5, 2, or 1 nM as determined by the method in Exampie AA described herein below.
Administration of the compounds of Formula I (including salts therof) may be effected by any methodthat enables deliveryofthe compoundstothe siteofaction. These methods Include, for example, enterai routes (e.g, oral routes, buccal routes, sublabial routes, sublingual routes), oral routes, Intranasal routes, Inhaled routes, Intraduodenal routes, parentéral Injection (including intravenous, subcutaneous, Intramuscular, intravascular or Infusion), Intrathecal routes, épidural routes, intracérébral routes, Intracerbroventricular routes, topical, and rectal administration.
In one embodiment of the présent invention, the compounds of Formula I may be administered/effected by parentéral Injection routes (e.g, intravenous injection route).
In one embodiment ofthe présent Invention, the compounds of Formula I may be administered/effected by oral routes.
Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be adminîstered over time or the dose may be proportionally reduced or Increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parentéral compositions In 20 dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refera to physically discrète units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The spécifications for the dosage unit forms of the invention are dictated 25 by a variety of factors such as the unique characteristics of the therapeutic agent and the particular therapeutic or prophylactîc effect to be achieved. In one embodiment of the présent invention, the compounds of Formula I may be used to treat humans.
It is to be noted that dosage values may vary with the type and severity ofthe condition to be alleviated, and may include single or multiple doses. It is to be further understood that for 30 any particular subject, spécifie dosage regimens shouid be adjusted over time according to the individuai need and the professional judgment ofthe person administering or supervising the administration ofthe compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limît the scope or practice of the claimed composition. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamie parametere, which 35 may include clinical effects such as toxic effects and/or iaboratory values. Thus, the présent invention encompasses intra-patient dose-escalation as determined by the skilled artisan. Determining appropriate dosages and regimens for administration ofthe chemotherapeutic
agent is weli-known in the relevant art and would be understood to be encompassed by the skilled artisan once provided the teachings disclosed herein.
The amount of the compound of Formula I administered will be dépendent on the subject being treated, the severity of the disorder or condition, the rate of administration, the 5 disposition of the compound and the discrétion of the prescribing physician. Generally, an effective dosage is in the range of about 0.0001 to about 50 mg per kg body weight per day, for example about 0.01 to about 10 mg/kg/day, In single or divided doses. For a 70 kg human, this would amount to about 0.007 mg to about 3500 mg/day, for example about 0.7 mg to about 700 mg/day. In some instances, dosage levels below the lower lîmit of the aforesaid range may be 10 more than adéquate, while In other cases still larger doses may be employed without causing any harmfui side effect, provided that such larger doses are first divided into several smali doses for administration throughout the day.
As used herein, the term combination therapy refers to the administration of a compound of Formula i or a pharmaceutically acceptable sait thereof together with an at ieast 15 one additional pharmaceutical or médicinal agent (e.g., an anti-schizophrenia agent), either sequentially or simultaneously.
The présent invention Includes the use of a combination of a compound of Formula I (Including a sait thereof) and one or more additional pharmaceutically active agent(s). If a combination of active agents is administered, then they may be administered sequentially or 20 simultaneously, in separate dosage forms or combined in a single dosage form. Accordingly, the présent invention also includes pharmaceutical compositions comprising an amount of: (a) a first agent comprising a compound of Formula I (including a pharmaceutically acceptable sait thereof); (b) a second pharmaceutically active agent; and (c) a pharmaceutically acceptable carrier, vehicle or diluent.
Various pharmaceutically active agents may be selected for use In conjunction with the compounds of Formula I, depending on the dîsease, disorder, or condition to be treated. Pharmaceutically active agents that may be used In combination with the compositions of the présent invention Include, without limitation:
(i) acetylcholinesterase inhibitors such as donepezil hydrochloride (ARICEPT, MEMAC); or 30 Adenosine Αςα receptor antagonîsts such as Preladenant (SCH 420814) or SCH 412348;
(ii) amyloid-β (or fragments thereof), such as ABi-isConjugated to pan HLA DR-binding epitope (PADRE) and ACC-001 (Elan/Wyeth);
(iii) antibodies to amyloid-β (or fragments thereof), such as bapineuzumab (also known as AAB001) and AAB-002 (Wyeth/Elan);
(iv) amylold-lowering or -Inhibiting agents (including those that reduce amyloid production, accumulation and fibrillization) such as colostrinîn and bisnorcymserine (also known as BNC);
(v) alpha-adrenergic receptor agonists such as clonidine (CATAPRES);
(vi) beta-adrenergic receptor blocking agents (beta blockers) such as carteolol;
(vii) anticholinergics such as amitriptyline (ELAVIL, ENDEP);
(viii) anticonvulsants such as carbamazepine (TEGRETOL, CARBATROL);
(Ix) antipsychotics, such as lurasldone (also known as SM-13496; Dalnippon Sumitomo);
(x) calcium channel blockers such as nilvadipine (ESCOR, NIVADIL);
(xi) catechol O-methyltransferase (COMT) inhibitors such as tolcapone (TASMAR);
(xii) central nervous system stimulants such as caffeine;
(xiii) corticosteroids such as prednisone (STERAPRED, DELTASONE);
(xiv) dopamine receptor agonists such as apomorphlne (APOKYN);
(xv) dopamine receptor antagoniste such as tetrabenazine (NITOMAN, XENAZINE, dopamine 10 D2 antagonist such as Quetiapine);
(xvl) dopamine reuptake Inhibitors such as nomlfensine maleate (MERITAL);
(xvii) gamma-aminobutyric acid (GABA) receptor agonists such as baclofen (LIORESAL, KEMSTRO);
(xviii) histamine 3 (Ha) antagonists such as ciproxifan;
(xix) immunomodulators such as glatiramer acetate (also known as copolymer-1 ; COPAXONE);
(xx) immunosuppressants such as methotrexate (TREXALL, RHEUMATREX);
(xxi) interferons, including interferon beta-1a (AVONEX, REBIF) and Interferon beta-1b (BETASERON, BETAFERON);
(xxii) levodopa (or its methyl or ethyl ester), alone or in combination with a DOPA decarboxylase inhibitor (e.g., carbidopa (SINEMET, CARBILEV, PARCOPA));
(xxiii) N-methyl-D-aspartate (NMDA) receptor antagonists such as memantine (NAMENDA, AXURA, EBIXA);
(xxiv) monoamine oxidase (MAO) inhibitors such as selegiline (EMSAM);
(xxv) muscarinic receptor (particularly M1 subtype) agonists such as bethanechol chloride 25 (DUVOID, URECHOLINE);
(xxvi) neuroprotective drugs such as 2,3,4,9-tetrahydro-1H-carbazol-3-one oxime;
(xxvii) nicotinic receptor agonists such as epibatidine;
(xxviii) norepinephrine (noradrenaline) reuptake inhibitors such as atomoxetine (STRATTERA);
(xxix) phosphodiesterase (PDE) inhibitors, for example,PDE9 inhibitors such as BAY 73-6691 30 (Bayer AG) and PDE 10 (e.g., PDE1 OA) inhibitors such as papaverine;
(xxx) other PDE inhibitors including (a) PDE1 inhibitors (e.g., vinpocetine), (b) PDE2 inhibitors (e.g., erythro-9-(2-hydroxy-3-nonyl)aden!ne (EHNA)), (c) PDE4 inhibitors (e.g., rolipram), and (d) PDE5 inhibitors (e.g., sildenafil (VIAGRA, REVAI 10)), (xxxi) quinolines such as quinine (including its hydrochloride, dihydrochloride, sulfate, bisulfate 35 and gluconate salts);
(xxxii) β-secretase inhibitors such as WY-25105;
(xxxiii) γ-secretase inhibitors such as LY-411575 (Lilly);
(xxxiv) serotonin (5-hydroxytryptamine) 1A (5-ΗΊΊα) receptor antagonists such as spiperone;
(xxxv) serotonin (5-hydroxytryptamine) 4 (5-HT<) receptor agonists such as PRX-03140 (Epix); (xxxvi) serotonin (5-hydroxytryptamine) 6 (5-HTe) receptor antagonists such as mianserin (TORVOL, BOLViDON, NORVAL);
(xxxvii) serotonin (5-HT) reuptake inhibitors such as alaproclate, citalopram (CELEXA,
CIPRAMIL);
(xxxviii) trophic factors, such as nerve growth factor (NGF), basic fibroblast growth factor (bFGF; ERSOFERMIN), neurotrophin-3 (NT-3), cardiotrophin-1, brain-derived neurotrophic factor (BDNF), neublastin, meteorin, and glial-derived neurotrophic factor (GDNF), and agents that stimulate production of trophic factors, such as propentofylline;
(xxxix) antihemorrhagic (i.e., hemostatic) agents such as rivaroxaban or apixaban; and the like.
The compound of Formula I (including a sait thereof) is optionally used in combination with another active agent. Such an active agent may be, for example, an atypical antipsychotic or an anti-Parkinson’s disease agent or an anti-Alzheimer*s agent. Accordingly, another 15 embodiment of the invention provides methods of treating a MAGL-mediated disease or disorder in a mammal, comprising administering to the mammal an effective amount of a compound of Formula I (including a pharmaceutically acceptable sait thereof) and further comprising administering another active agent.
As used herein, the term ‘another active agent refers to any therapeutic agent, other than the compound of Formula I (including or a pharmaceutically acceptable sait thereof) that is useful for the treatment of a subject disorder. Examples of additional therapeutic agents include antidepressants, antipsychotics (such as anti-schizophrenla), anti-pain, anti-Parkinson‘s disease agents, anti-LID (levodopa-induced dyskinesia), anti-Alzheimer’s, anti-anxiety, and antihemorrhagic agents. Examples of particular classes of antidepressants that can be used in combination with the compounds of the invention include norepinephrine reuptake inhibitors, sélective serotonin reuptake inhibitors (SSRIs), NK-1 receptor antagonists, monoamine oxidase inhibitors (MAOIs), réversible inhibitors of monoamine oxidase (RIMAs), serotonin and noradrenaline reuptake inhibitors (SNRIs), corticotropin releasing factor (CRF) antagonists, aadrenoreceptor antagonists, and atypical antidepressants. Suitable norepinephrine reuptake inhibitors include tertiary amine tricyclics and secondary amine tricyclics. Examples of suitable tertiary amine tricyclics and secondary amine tricyclics include amitriptyline, clomipramine, doxepin, imipramine, trimipramine, dothiepin, butriptyline, iprindole, lofepramine, nortriptyline, protriptylme, amoxapine, desipramine and maprotiline. Examples of suitable sélective serotonin reuptake inhibitors include fluoxetine, fluvoxamine, paroxetine, and sertraline. Examples of monoamtne oxidase Inhibitors include isocarboxazid, phenelzine, and tranylcyclopramine. Examples of suitable réversible inhibitors of monoamine oxidase include moclobemide. Examples of suitable serotonin and noradrenaline reuptake inhibitors of use in the présent invention include venlafaxîne. Examples of suitable atypical antidepressants include bupropion,
lithium, nefazodone, trazodone and viloxazine. Examples of anti-Alzheimeris agents Include
Dimebon, NMDA receptor antagonists such as memantine; and cholinestérase inhibitors such as donepezil and galantamine. Examples of suitable classes of anti-anxiety agents that can be used in combination with the compounds of the Invention include benzodiazépines and serotonin 1A (5-HT1A) agonlsts or antagonists, especially 5-HT1A partial agonists, and corticotropin releasing factor (CRF) antagonists. Suitable benzodiazépines include alprazolam, chlordiazepoxide, clonazepam, chlorazepate, diazepam, halazepam, lorazépam, oxazepam, and prazepam. Suitable 5-HT1A receptor agonists or antagonists include buspirone, flesinoxan, gepirone, and ipsapirone. Suitable atypical antipsychotics include paliperidone, 10 bifeprunox, ziprasidone, rispéridone, aripiprazole, olanzapine, and quetiapine. Suitable nicotine acétylcholine agonists include ispronicline, varenicline and MEM 3454. Anti-pain agents Include pregabalin, gabapentin, clonidine, neostigmine, baclofen, midazolam, ketamine and ziconotide. Examples of suitable anti-Parkinson's disease agents include L-DOPA (or its methyl or ethyl ester), a DOPA decarboxylase inhibitor (e.g., carbidopa (SINEMET, CARBILEV, PARCOPA), 15 an Adenosine A» receptor antagonist [e.g., Preladenant (SCH 420814) or SCH 412348], benserazide (MADOPAR), α-methyldopa, monofluoromethyldopa, difluoromethyldopa, brocresine, or m-hydroxybenzylhydrazlne), a dopamine agonist [such as apomorphine (APOKYN), bromocriptine (PARLODEL), cabergoline (DOSTINEX), dihydrexidine, dihydroergocryptine, fenoldopam (CORLOPAM), lisuride (DOPERGIN), pergolide (PERMAX), 20 piribedil (TRIVASTAL, TRASTAL), pramipexole (MIRAPEX), quinpirole, ropinirole (REQUIP), rotigotine (NEUPRO), SKF-82958 (GlaxoSmithKIine), and sarizotan], a monoamine oxidase (MAO) inhibitor [such as selegiline (EMSAM), selegiline hydrochlorlde (L-deprenyl, ELDEPRYL, ZELAPAR), dimethylselegilene, brofaromine, phenelzine (NARDIL), tranylcypromine (PARNATE), moclobemide (AURORIX, MANERIX), befloxatone, safinamide, isocarboxazid 25 (MARPLAN), nialamide (NIAMID), rasagiline (AZILECT), ipronîazide (MARSILID, IPROZID,
IPRONID), CHF-3381 (Chiesl Farmaceutici), iproclozide, toloxatone (HUMORYL, PERENUM), bifemelane, desoxypeganine, harmine (also known as telepathine or banasterine), harmaline, linezolid (ZYVOX, ZYVOXID), and pargyline (EUDATIN, SUPIRDYL)], a catechol Omethyltransferase (COMT) inhibitor [such as tolcapone (TASMAR), entacapone (COMTAN), 30 and tropolone], an W-methyl-D-aspartate (NMDA) receptor antagonist [such as amantadine (SYMMETREL)], anticholinergics [such as amitriptyline (ELAVIL, ENDEP), butriptyline, benztropine mesylate (COGENTIN), trihexyphenidyl (ARTANE), diphenhydramine (BENADRYL), orphenadrine (NORFLEX), hyoscyamine, atropine (ATROPEN), scopolamine (TRANSDERM-SCOP), scopolamine methylbromide (PARMINE), dicydoverine (BENTYL, 35 BYCLOMINE, DIBENT, DILOMINE, tolterodine (DETROL), oxybutynin (DITROPAN, LYRINEL
XL, OXYTROL), penthienate bromide, propantheline (PRO-BANTHINE), cyclizine, imipramine hydrochloride (TOFRANIL), imipramine maleate (SURMONTIL), lofepramine, deslpramlne (NORPRAMIN), doxepin (SINEQUAN, ZONALON), trimipramine (SURMONTIL), and
glycopyrrolate (ROBINUL)], or a combination thereof. Examples of anti-schizophrenia agents include ziprasidone, rispéridone, olanzapine, quetiapine, aripiprazole, asenapine, blonanserin, or lloperidone. Some additional another active agent examples Include rivastigmine (Exelon), Clozapine, Levodopa, Rotigotine, Aricept, Methylphenidate, memantine. milnacipran, guanfacine, bupropîon, and atomoxetine. Examples of antihemorrhagic agents (including, e.g., coagulation factors, activators, or stabilizers) include Factor Xa Inhibitors (e.g., rivaroxaban or apixaban) and recombinant Coagulation Factor Vlla (e.g., NovoSeven®).
As noted above, the compounds of Formula I or salts thereof may be used ln combination with one or more additional antFAlzheimeris agents which are described herein.
When a combination therapy is used, the one or more additional anti-Alzheimeris agents may be administered sequentially or simultaneously with the compound of the invention. In one embodiment, the additional anti-Alzheimer's agent(s) is(are) administered to a mammal (e.g., a human) prior to administration of the compound of the invention. In another embodiment, the additional anti-Alzheimer’s agent(s) Is(are) administered to the mammal after administration of the compound of the invention. In another embodiment, the additional anti-Alzheimer’s agent(s) Is(are) administered to the mammal (e.g., a human) simultaneously with the administration of the compound of the invention (or a pharmaceutically acceptable sait thereof).
The invention also provides a pharmaceutical composition for the treatment of an inflammatory disorder (e.g., nueroinflammation) in a mammal, including a human, which 20 comprises an amount of a compound of Formula I (including a sait thereof), as defined above (including hydrates, solvatés and polymorphs of said compound or pharmaceutically acceptable salts thereof), in combination with one or more (for example one to three) anti-inflammation agents, wherein the amounts of the active agent and the combination when taken as a whole are therapeutically effective for treating the inflammatory disorder.
The invention also provides a pharmaceutical composition for treating a MAGL-mediated disease or disorder ln a mammal, Including a human, which comprises an amount of a compound of Formula I (including a sait thereof), as defined above (including hydrates, solvatés and polymorphs of said compound or a sait thereof), in combination with one or more (for example one to three) other agents for treating the MAGL-mediated disease or disorder, wherein the amount of the active agents and the combination when taken as a whole are therapeutically effective for treating the MAGL-mediated disease or disorder.
It will be understood that the compounds of Formula I depicted above are not limited to a particular stereoisomer (e.g., enantiomer or diasteroisomer) shown, but also include ail stereoisomers and mixtures thereof.
DETAILED DESCRIPTION OF THE INVENTION
Compounds ofthe Invention, including salts ofthe compounds, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes. The reactions for preparing compounds of the invention can be
carried out in suitable solvents, which can be readily selected by one of ski II in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the températures at which the reactions are carried out, e.g., températures that can range from the solvent's freezing température to the solvent's boiling température. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Préparation of compounds of the invention can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the sélection of 10 appropriate protecting groups, can be readily determined by one skilled in the art. The chemlstry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Pmtective Groups in Organic Synthesis, 3ri Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in Its entirety.
Reactions can be monitored according to any suitable method known in the art. For 15 example, product formation can be monitored by spectroscopic means, such as nuclear magnetic résonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographie methods such as hrghperformance liquid chromatography (HPLC) or thin layer chromatography (TLC).
Compounds of Formula i and intermediates thereof may be prepared according to the 20 following reaction schemes and accompanying discussion. Unless otherwise Indicated, R1, R2, R3, R4, Rs, R®, R7, r and structural Formula I (Including l-a) in the réaction schemes and discussion that follow are as defined above. In general, the compounds of this Invention may be made by processes which include processes analogous to those known In the chemical arts, particularly in light of the description contained herein. Certain processes for the manufacture of 25 the compounds of this Invention and intermediates thereof are provided as further features of the invention and are illustrated by the following réaction schemes. Other processes are described in the experimental section. The schemes and examples provided herein (including the corresponding description) are for Illustration only, and not Intended to limit the scope of the présent invention.
Scheme 1 refera to the synthesis of compounds of Formula I. Referring to Scheme 1, a compound of Formula 1-3 [wherein Pg1 is an alcohol protecting group such as tert-butyldimethyl sllyl (TBDMS) or p-methoxbenzyl] can be prepared by reacting an amine of Formula 1-1 with a compound of Formula 1-2 using standard methods of carbamate formation well known to those skilled in the art [for example, in the presence of phosgene, triphosgene, or a suitably activated carbonate reagent such as bis(pentafluorophenyl)carbonate or Ν,Ν'-disuccinimidyl carbonate]. Amines of Formula 1-1 may be obtained commerclally, synthesized by methods described herein, or made by other methods well known to those skilled In the art. Carbamate formation may be accomplished In the presence of a base (such as triethylamine or hunigs base). A compound of Formula 1-4 may be obtained by deprotecting the compounds of Formula 1-3, using appropriate conditions depending on the sélection of the Pg1 group. For example, where Pg1 is TBDMS, treatment with an acid such as trifluoroacetic acid In aprotic solvent such as dichloromethane may be employed. The compound of Formula 1-4 (which is a compound of Formula I wherein R7 is H) may optionally be converted to a compound of Formula I wherein R7 Is other than H. For example, an alkylation reaction of the compound of Formula 1-4 with a halide compound (alkyl halide or cycloalkyl halide) can provide a compound of Formula I wherein R7 Is Cm alkyl, C3-7 cycloalkyl. As another example, reaction of the alcohol of Formula 1-4 with diphosphoryl tetrachloride In a suitable solvent such as acetonitrile affords compounds of Formula I where R7 is -P(=O)(OH)î or a sait thereof. For yet another example, reaction of the alcohol of Formula 1-4 with a sulfating agent [e.g. SO3, sulfamic acid H2N-S(=O)î(OH), chlorosulfonic acid HO-S(=O)2(CI)] under suitable conditions can afford a compound of Formula I wherein R7 is -S(=O)2(OH) or a sait thereof.
Scheme 2 refers to synthesis of compounds of Formula l-a. An amine of Formula 1-1 may be reacted with a compound of Formula 2-2 [where Pg1 is a suitable alcohol protecting group, such as TBDMS or p-methoxybenzyl], using methods analogous to those described In Scheme 1, to form a carbamate of Formula 2-3. The compound of Formula 2-3 may be deprotected using appropriate conditions depending of the sélection of Pg1 to give a compound of Formula 2-4. Similar to the discussions in Scheme 1, the compound of Formula 2-4 (which Is a compound of Formula l-a wherein R7 is H) may optionally be converted to a compound of Formula l-a wherein R7 is other than H.
Scheme 3 refera to the préparation of compounds of Formula 3-4 [wherein Pg1 is an alcohol protecting group such TBDMS orp-methoxbenzyl], which can be used as a compound 5 of Formula 1-2 In Scheme 1 [wherein r is 1; and both Rs and R® are H]. Referring to Scheme 3, a compound of Formula 3-3 may be prepared by treatment of compound 3-1 with a base (such as n-butyllithium) followed by addition to formaldéhyde 3-2 (or its équivalent such as paraformaldéhyde) in the presence of a reducing agent such as sodium borohydrlde. Protection of the alcohol moiety in the compound of Formula 3-3 may be achieved by methods known to those skilled in the art. For example, where the Pg1 is TBDMS, the protection can be achieved by treatment of the compound of Formula 3-3 with an actîvated silyl reagent [such as tertbutyl(dimethyl)silyl chloride] in the presence of a base (such as 1H-imidazole) In a suitable nonprotic solvent (such as THF or DMF) at a suitable température (e.g., ambient température).
Scheme 3
Pg1
Scheme 4 refera to a synthesis of compounds of Formula 4-3 [wherein Pg2 is an alcohol protecting group such p-methoxbenzyl], which can be used as a compound of Formula 1-2 in Scheme 1 [wherein r is 0]. Referring to Scheme 4, reaction of an epoxide of Formula 4-1 with an alcohol of Formula 4-2, in the presence of a base [e.g., NaN(TMS)2) In a in non-protic solvent (e.g., THF or DMF), affords a compound of Formula 4-3.
Scheme 4
+ Pg2—OH
4-2
4-3
Scheme 4A refers to a synthesis of a compound of Formula 4A-5 or a sait thereof [i.e., a compound of Formula l-a or sait thereof, wherein R7 Is -P(=O)(OH)2]. Referring to Scheme 4A, reaction of an epoxide of Formula 4A-1 with a phosphorus compound of Formula 4A-2 [wherein each of Pg2* is a hydroxyl protecting group such as benzyl], optionally In the presence of a base [e.g., NaN(TMS)2] In a in non-protic solvent (e.g., THF or DMF), affords a compound of Formula 4A-3. Similar to the carbamate formation reaction described In Schemes 1 and 2, reaction of the compound of Formula 4A-3 and an amine of Formula 1-1 affords a compound of Formula 4A-4. Depending on the cholce of the Pg2* groups, removal of the protecting Pg2* groups under suitable conditions will afford a compound of Formula 4A-5 or a sait thereof.
Scheme 4A
A R^OPg2*
R® HOZ ^OPg2*
R5 +
4A-1 4A-2
OH
R” R
II
Pç-OPg
OPg2*
2A
H
1-1
4A-3
R2
Scheme 5 refers to the préparation of amines of Formula 5-8 (wherein R31 Is aryl or heteroaryl that are optionally substituted), which can be used as a spécifie type of amine of Formula 1-1 for the préparation of compounds of Formula I or l-a In Schemes 1 and 2. The Weinrcb amide of Formula 5-2 [where Pg3 is an amine protecting group such as tertbutoxycarbonyl (BOC)] can be prepared by coupling N-methoxymethanamine with a carboxylic acid of Formula 5-1 using a suitable coupling agent [e.g., O-(7-azabenzotriazol-1-yl)-N,N,N',Ntetramethyluronium hexafluorophosphate (HATU)]. Addition of a Grignard reagent (e.g., méthylmagnésium bromide) to the Weinreb amide of Formula 5-2 results in a ketone of Formula 5-3. Treatment of the ketone of Formula 5-3 with Λ/,Ν-dimethylformamide dimethyl a ce ta I at elevated températures results in an enamine of Formula 5-4. Subséquent treatment with
hydrazine (or its équivalent) in a protic solvent such as éthanol affords a pyrazole of Formula 5
5. A compound of Formula 5-7 can be obtained by reacting the pyrazole of Formula 5-5 with a (hetero)aryi boronic acid of Formula 5-6 in the presence of a catalyst (such as copper acetate) and a base (e.g., pyrldine) in a suitable solvent (such as dichloromethane). Altematively, the pyrazole of Formula 5-5 can be transformed into the compound of Formula 5-7 by palladiumcatalyzed coupling with a suitable (hetero)aryl halide of Formula 5-9 wherein X is a suitable halide such are Cl, Br or I. Coupling can be achieved by reaction of the pyrazole of Formula 5-5 and (hetero)aryl halide of Formula 5-9 in the presence of a palladium catalyst such as [1,1 'bis(diphenylphosphino)ferrocene]dichloropalladium(ll) [Pd(dppf)Ch] together with a base such 10 as potassium acetate at an elevated température in a non-protic solvent such as toluene. A compound of Formula 5-8 can be prepared by removal of the protecting group Pg3. For example, wherein the Pg3 is fert-butoxycarbonyl (BOC), cleavage can be achieved under acidic conditions by treatment with, for example, trifluoroacetic acid.
Scheme 5
5-9
Schéma 6 refers to a synthesis of a spiromorpholine of Formula 6-6 (wherein Pg4 Is a suitable amine protecting group such as BOC), which can be used as a starting material in Scheme 7. Referring to Schéma 6, reaction of a suitably protected piperidine of Formula 6-1 with nitromethane In the presence of a mild base such as triethylamine affords a compound of
Formula 6-2. Réduction of the nitro moiety of the compound of Formula 6-2 to obtain an aminoalcohol of Formula 6-3 can be achleved by using methods such as palladium-catalyzed hydrogénation, for example utilizing 10% palladium on carbon in an alcoholic solvent under an atmosphère of hydrogen. Acétylation of the compound of Formula 6-3 can be achieved by treatment with chloroacetyl chloride in the presence of a suitable base such as potassium carbonate. Ring closure of the chloride compound of Formula 6-4 can be achieved by treatment with a suitable base (e.g., potassium fert-butoxide) In a non-protic solvent (e.g., THF) under reflux conditions to fumish a compound of Formula 6-5. A spiromorpholine compound of Formula 6-6 may be obtained by réduction ofthe amide functionality in the compound of Formula 6-5 using a suitable reducing agent (e.g.. borane-dimethyl sulfide complex In THF).
Scheme 6
Scheme 7 refers to the synthesis of compounds of Formula 7-4,7-7,7-10, or 7-13 from an amine of Formula 6-6. A compound of Formula 7-3 [wherein R70 can be, for example, R11, 20 R, R13, or R14] can be prepared by reacting the amine of Formula 6-6 with an aldéhyde of
Formula 7-2 using reductive amination conditions well known to ttioso skilled in the art. For example, treatment with titanium(IV) Isopropoxide and a reducing agent such as sodium borohydrlde can be employed. Reaction of an amine of Formula 6-6 with sulfonyl chlorides of
Formula 7-5 [wherein R70 can be, for example, R11, R12, R13, or R14] in the presence of a suitable 25 base (such as pyridine or sodium bicarbonate) results in a sulfonamide of Formula 7-6. An amine 6-6 can be treated with a suitably activated compound of Formula 7-8 (wherein Lg1 Is a leaving group such as Cl) to give a compound of Formula 7-9 [wherein R71 can be, for example,
R23; and R72 can be, for example, R11, R12, R13, R14, -(CR21R22)P-R11, -(CR21R22)P-R12, (CR21R22)j,-R13, or -(CR21R22)P-R14; or R71 and R72, together with the N atom to which they are attached, form 4- to 14-membered heterocycloalkyl optionally substituted with RB and one or more independently selected R9]. A compound of Formula 7-12 [wherein R73 can be, for example, R11 or R12] can be prepared by metal-catalyzed coupling of compounds of Formula 6-6 with a compound of Formula 7-11 (wherein X is a halogen atom such as Cl or Br). A compound of Formula 7-3,7-6, 7-9, or 7-12 can be converted to a compound of Formula 7-4,7-7, 7-10, or
7-13, respectively, by appropriate deprotection. For example, when Pg4 Is BOC, the deprotection can be achieved by treatment with an acid such as trifluoroacetic acid. A compound of Formula 7-4, 7-7, 7-10, or 7-13 can each be used as starting material [as a spécifie amine of Formula 1-1] for synthesis of compounds of Formula I (e.g. Formula l-a or l-b) as described In Schemes 1 and 2.
Scheme7
O=S=O O=S=O
R70 7-7 A 7'10
R71 XR72
Scheme 8 refers to a synthesis of compounds of Formula 8-6 [where each t2a Is independently 0 or 1; and R8* can be, for example, R11, R12, R13, or R14]. A compound of Formula 8-3 can be prepared by treatment of the aminoalcohol of Formula 8-1 (which can be prepared using the method as described in Scheme 6 for the amtnoalcohol of Formula 6-3) with a sulfonyl chloride of Formula 8-2 in the presence of a suitable base (e.g., pyridine). Reaction of the compound of Formula 8-3 with a compound of Formula 8-4 (wherein each X is independently a suitable leaving group such as Br or Cl), in the presence of a base such as potassium carbonate in a polar aprotic solvent such as DMF, results in a compound of Formula 8-5. Removal ofthe protecting group results in a compound ofFormula 8-6, which can be used as starting material [as a spécifie amine of Formula 1-1] in Schemes 1 and 2 for the préparation of compounds of Formula I (including compounds of Formula l-a or l-b).
Scheme 8
9)t2a
X X
8-4
Scheme 9 refers to a préparation of compounds of Formula 9-3 [where R“* can be. for example, R11, R12, R13, or R14]. A compound of formula 9-1 [where Pg4 is an amine protecting group (e.g., BOC)] can be obtained commercially or be readily synthesized by methods well known to those skilled in the art. A compound of Formula 9-2 can be obtained by reaction of a compound of Formula 9-1 with sulfonyl chlorides of Formula 8-2 in a suitable solvent (e.g., dichloromethane) in the presence of a suitable base (e g., sodium bicarbonate). Deprotection of compounds of Formula 9-2 using appropriate conditions well known to those skilled in the art provîdes a compound of Formula 9-3. The compound of Formula 9-3 can be used as starting material [as a spécifie amine of Formula 1-1] in Schemes 1 and 2 for the préparation of compounds of Formula I (including compounds of Formula l-a or l-b).
Scheme 9
Additional starting materials and intermediates useful for making the compounds of the présent invention can be obtained from chemical vendors such as Sigma-Aldrich or can be made according to methods described in the chemical art.
Those skilled in the art can recognïze that in ail of the schemes described herein, if there are functional (reactive) groups présent on a part of the compound structure such as a substituent group, for example R1, R2, R3, R4, Rs, R®, R7, etc., further modification can be made if appropriate and/or desired, using methods well known to those skilled in the art. For example, a -CN group can be hydrolyzed to afford an amide group; a carboxylic acid can be converted to an amide; a carboxylic acid can be converted to an ester, which in tum can be reduced to an alcohol, which in tum can be further modified. For another example, an OH group can be converted into a better leavïng group such as a methanesulfonate, which in tum is suitable for nucleophilic substitution, such as by a cyanide ion (CN ). For another example, an 15 S- can be oxidized to -S(=O)- and/or -S(=O)2-. For yet another example, an unsaturated bond such as C=C or C=C can be reduced to a saturated bond by hydrogénation. One skilled in the art will recognize further such modifications. Thus, a compound of Formula I having a substituent that contains a functional group can be converted to another compound of Formula I having a different substituent group.
Similarty, those skilled in the art can also recognize that in ail of the schemes described herein, If there are functional (reactive) groups présent on a substituent group such as R1, R2, R3, R4, R5, R®, R7, etc., these functional groups can be protected/deprotected in the course of the synthetic scheme described here, if appropriate and/or desired. For example, an OH group can be protected by a benzyl, methyl, or acetyl group, which can be deprotected and converted back to the OH group in a later stage of the synthetic process. For another example, an NH2 group can be protected by a benzyloxycarbonyl (Cbz) or BOC group; conversion back to the NH2 group can be carried out at a later stage of the synthetic process via deprotection.
As used herein, the term reacting (or réaction or reacted) refers to the bringing together of designated chemical reactants such that a chemical transformation takes place generating a compound different from any initially introduced into the System. Reactions can take place in the presence or absence of solvent.
Compounds of Formula I may exist as stereoisomers, such as atropisomers, racemates, enantiomers, or diastereomers. Conventional techniques for the preparation/isolation of Individual enantiomers Include chiral synthesis from a suitable optically pure precursor or resolution of the racemate using, for example, chiral high-performance liquid chromatography (HPLC). Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound contains an acidic or basic moiety, an acid or base such as tartane acid or 1-phenylethylamine. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure 10 enantiomer(s) by means well known to one skilled in the art. Chiral compounds of Formula I (and chiral precursors thereof) may be obtained in enantiomerically enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0% to 50% 2-propanol, typically from 2% to 20%, and from 0% to 5% of an alkylamine, typically 0.1% diethylamine. Concentration of 15 the eluate affords the enriched mixture. Stereoisomeric conglomérâtes may be separated by conventional techniques known to those skilled in the art. See, e.g., Stereochemistry of
Organic Compounds by E. L. Eliel and S. H. Wilen (Wiley, New York, 1994), the disclosure of which is Incorporated herein by reference in its entirety. Suitable stereoselective techniques are well known to those of ordinary ski! I in the art.
Where a compound of Formula I contains an alkenyl or alkenylene (alkylidene) group, géométrie cis/trans (or Z/E) isomers are possible. Cis/trans isomers may be separated by conventional techniques well known to those skilled In the art, for example, chromatography and fractional crystallization. Salts of the présent invention can be prepared according to methods known to those of skill in the art.
The compounds of Formula I that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. Although such salts must be pharmaceutically acceptable for administration to animais, It Is often désirable In practice to inltially isolate the compound of the présent Invention from the reaction mixture as a pharmaceutically unacceptable sait and then sîmply convert the latter back to the free base compound by treatment with an alkaline reagent and subsequently convert the latter free base to a pharmaceutically acceptable acid addition sait. The acid addition salts of the basic compounds of this invention can be prepared by treating the basic compound with a substantially équivalent amount of the selected minerai or organic acid in an aqueous solvent medium or in a suitable organic solvent, such as methanol or éthanol. Upon évaporation of the solvent, the desired solid sait is obtained. The desired acid sait can also be precipitated from a solution of the free base in an organic solvent by adding an appropriate minerai or organic acid to the solution.
If the inventive compound Is a base, the desired pharmaceutically acceptable sait may be prepared by any suitable method available In the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic 5 acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, isonicotinic acid, lactic acid, pantothenic acid, bitartric acid, ascorbic acid, 2,5dihydroxybenzoicacid, gluconlcacid, saccharic acid, formicacid, methanesulfonic acid, ethanesulfonicacid, benzenesulfonicacid, p-toluenesulfonicacid, and pamoic[I e., 4,4'methanediylbis(3-hydroxynaphthalene-2-carboxyiicacid)] acid, a pyranosidyl acid, such as 10 giucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as citric acid or tartaric acid, an amino acid, such as aspartïc acid or glutamic acid, an aromatic acid, such as benzoic acid or cinnamic acid, a sulfonic acid, such as ethanesulfonic acid, or the like.
Those compounds of Formula I that are acidic in nature are capable of forming base salts with various pharmacologically acceptable cations. Examples of such salts include the 15 alkali métal or alkaline earth métal salts, and particularly the sodium and potassium salts.
These salts are ail prepared by conventional techniques. The chemical bases which are used as reagents to préparé the pharmaceutically acceptable base salts of this invention are those which form non-toxic base salts with the acidic compounds of Formula I. These salts may be prepared by any suitable method, for example, treatment of the free acid with an Inorganic or 20 organic base, such as an amine (primary, secondary or tertiary), an alkali métal hydroxide or alkaline earth métal hydroxide, or the like. These salts can also be prepared by treating the corresponding acidic compounds with an aqueous solution containing the desired pharmacologically acceptable cations, and then evaporating the resulting solution to dryness, for example under reduced pressure. Altematively, they may also be prepared by mixing lower 25 alkanolic solutions of the acidic compounds and the desired alkali métal alkoxide together, and then evaporating the resulting solution to dryness in the same manneras before. In either case, stoichiometric quantities of reagents are, for example, employed in order to ensure completeness of reaction and maximum yields of the desired final product.
Pharmaceutically acceptable salts of compounds of Formula I (including compounds of 30 Formula l-a or l-b) may be prepared by, e.g., one or more of three methods:
(i) by reacting the compound of Formula I with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of Formula I or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or (iii) by converting one sait of the compound of Formula I to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column.
Ail three reactions are typically carried out in solution. The resulting sait may précipita te out and be collected by filtration or may be recovered by évaporation of the solvent. The degree of ionizatlon in the resulting sait may vary from completely ionized to almost non-ionized.
Polymorphs can be prepared according to techniques well-known to those skilled In the 5 art, for example, by crystal lization.
When any racemate crystallizes, crystals of two different types are possible. The first type is the racemic compound (true racemate) referred to above wherein one homogeneous form of crystal is produced containing both enantiomers in equimolar amounts. The second type is the racemic mixture or conglomerate wherein two forms of crystal are produced In equimolar 10 amounts each comprising a single enantiomer.
While both of the crystal forms présent in a racemic mixture may hâve almost Identical physical properties. they may hâve different physical properties compared to the true racemate. Racemic mixtures may be separated by conventional techniques known to those skilled In the art - see, for example, Stereochemistry of Organic Compounds by E. L. Eliel and S. H. Wilen 15 (Wiley, New York, 1994).
The invention also includes isotopically labeled compounds of Formula I wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Isotopically labeled compounds of Formula I (or pharmaceutically acceptable salts thereof or N20 oxides thereof) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described herein, using an appropriate isotopically labeled reagent In place of the non-labeled reagent otherwise employed.
Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities présent in the compounds of Formula I with certain moieties known 25 to those skilled in the art as ‘pro-moieties’ as described, for example, In Design of Prodrugs by H. Bundgaard (Eisevier, 1985).
The compounds of Formula I should be assessed for their biopharmaceutlcal properties, such as solubility and solution stability (across pH), permeability, etc., In order to select the most appropriate dosage form and route of administration for treatment of the proposed
Indication.
Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as précipitation, crystaliization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
They may be administered alone or in combination with one or more other compounds of the invention or in combination with one or more other drugs (or as any combination thereof). Generally, they will be administered as a formulation In association with one or more pharmaceutically acceptable excipients. The term “excipient’ is used herein to descrïbe any
ingrédient other than the compound(s) of the invention. The choice of excipient will to a large extent dépend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
Pharmaceutical compositions suitable for the delivery of compounds of the présent invention (or pharmaceutically acceptable salts thereof) and methods for their préparation will be readily apparent to those skilled In the art. Such compositions and methods for their préparation may be found, for example, in Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishlng Company, 1995).
The compounds of the Invention (including pharmaceutically acceptable salts thereof) may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual, or sublingual administration by which the compound enters the bloodstream directly from the mouth.
Formulations suitable for oral administration Include solid, seml-solid and liquid Systems such as tablets; soft or hard capsules containing multi- or nano-particulates, Iiquids, or powders;
lozenges (Including liqurd-filled); chews; gels; fast-dispersing dosage forms; films; ovules; sprays; and buccal/mucoadhesive patches.
Liquid formulations include suspensions, solutions, syrups and élixirs. Such formulations may be employed as filiers in soft or hard capsules (made, for example, from gelatin or hydroxypropyl methyl cellulose) and typically comprise a carrier, for example, water, éthanol, 20 polyethylene glycol, propylene glycol, methyl cellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, fOr example, from a sachet.
The compounds of the invention may also be used in fast-dissolving, fast-disintegratlng dosage forms such as those described by Llang and Chen, Expert Opinion in Therapeutic 25 Patents 2001, 11, 981-986.
For tablet dosage forms, depending on dose, the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form. In addition to the drug, tablets generally contain a disintegrant. Examples of disintegrants Include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl 30 cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl cellulose, starch, pregelatinized starch and sodium alginate. Generally, the disintegrant will comprise from 1 weight % to 25 weight %, for example, from 5 weight % to 20 weight % of the dosage form.
Binders are generaliy used to impart cohesive qualities to a tablet formulation. Suitable binders 35 include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried
monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When présent, surface 5 active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
Tablets also generally contain lubricants such as magnésium stéarate, calcium stéarate, zinc stéarate, sodium stearyl fumarate, and mixtures of magnésium stéarate with sodium lauryl sulfate. Lubricants generally comprise from 0.25 weight % to 10 weight %, for example, from 10 0.5 weight % to 3 weight % of the tablet.
Other possible ingrédients include anti-oxidants, colorants, flavoring agents, preservatives and taste-masking agents,
Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to 15 about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant.
Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt-congealed, or extruded before tabletting. The final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated.
The formulation of tablets Is discussed ln Pharmaceutical Dosage Forms: Tablets, Vol.
1, by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
Consumable oral films for human or veterinary use are typically pliable water-soluble or water-swellable thin film dosage forms which may be rapidly dissolving or mucoadhesive and typically comprise a compound of Formula I, a film-forming polymer, a binder, a solvent, a 25 humectant, a plasticizer, a stabilizer or emulsifier, a viscosity-modifying agent and a solvent. Some components of the formulation may perform more than one function.
The compound of Formula I (or pharmaceutically acceptable salts thereof or N-oxides thereof) may be water-soluble or insoluble. A water-soluble compound typically comprises from 1 weight % to 80 weight %, more typically from 20 weight % to 50 weight %, of the solutés.
Less soluble compounds may comprise a smaller proportion of the composition, typically up to 30 weight % of the solutés. Alternatively, the compound of Formula I may be ln the form of multiparticulate beads.
The film-forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocollolds and Is typically présent in the range 0.01 to 99 weight %, more typically 35 in the range 30 to 80 weight %.
Other possible ingrédients include anti-oxidants, colorants, flavorings and fiavor enhancers, preservatives, salivary stimulating agents, cooling agents, co-solvents (including oils), émollients, bulking agents, anti-foaming agents, surfactants and taste-masking agents.
Films In accordance with the Invention are typically prepared by evaporative drying of thin aqueous films coated onto a peelable backing support or paper. This may be done In a drying oven or tunnel, typically a combined coater dryer, or by freeze-drying or vacuuming.
Solid formulations for oral administration may be formulated to be immédiate and/or modified release. Modified release formulations Include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic and coated particles are to be found in Verma et al, Pharmaceutical 10 Technology On-line, 25(2), 1-14 (2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may also be administered directly into the bloodstream, into muscle, or into an internai organ. Suitable means for parentéral administration include intravenous, intraarterial, intraperitoneal, 15 Intrathecal, intraventricular, Intraurethral, intrastemal, intracranial, intramuscular, intrasynovial and subcutaneous. Suitable devices for parentéral administration include needle (including microneedle) Injectors, needle-free injectors and infusion techniques.
Parentéral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (for example to a pH of from 3 to 9), but, for 20 some applications, they may be more suitably formulated as a stérile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as stérile, pyrogen-free water.
The préparation of parentéral formulations under stérile conditions, for example, by lyophilization, may readily be accomplished using standard pharmaceutical techniques well 25 known to those skilled in the art.
The solubility of compounds of Formula I (including pharmaceutically acceptable salts thereof) used In the préparation of parentéral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents. . Formulations for parentéral administration may be formulated to be Immédiate and/or 30 modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus compounds of the invention may be formulated as a suspension or as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations Include drug-coated stents and semi-solids and suspensions comprising drug35 ioaded poly(DL-lactic-coglycolic acid) (P LG A) mîcrospheres.
The compounds ofthe invention (including pharmaceuticallyacceptable salts thereof) may also be administered topically, (intra)dermally, or transdermally to the skin or mucosa. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams,
ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibers, bandages and microemulsîons. Liposomes may also be used. Typical carriers include alcohol, water, minerai oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Pénétration enhancers may be incorporated. See e.g., Finnin and Morgan, J.
Pharm. Sci. 1999, 88, 955-958.
Other means of topical administration include delivery by electroporation, iontophoresis, phonophoresls, sonophoresis and microneedle or needle-free (e.g., Powdeiject™, Bioject™, etc.) Injection.
Formulations for topical administration may be formulated to be immédiate and/or modified release. Modifîed release formulations Include delayed-, sustained-, pulsed-, controlled-, targeted and programmée! release.
The compounds of the invention (including pharmaceutically acceptable salts thereof) can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone; as a mixture, for example, in a dry blend with lactose; or as a mixed component 15 partie le, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler, as an aérosol spray from a pressurized container, pump, spray, atomizer (for example an atomizer using electrohydrodynamics to produce a fine mist), or nebulizer, with or withoutthe useofasuitable propellant, such as 1,1,1,2-tetrafluoroethaneor 1,1,1,2,3,3,3heptafluoropropane, or as nasal drops. For intranasal use, the powder may comprise a bioadheslve agent, for example, chitosan or cyclodextrln.
The pressurized container, pump, spray, atomizer, or nebulizer contains a solution or suspension ofthe compound(s) of the invention comprising, for example, éthanol, aqueous éthanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic 25 acid, or an oligolactic acid.
Prior to use in a dry powder or suspension formulation, the drug product is micronized to a size suitable for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray 30 drying.
Capsules (made, for example, from gelatin or hydroxypropyl methyl cellulose), blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as L-leucine, mannitol, or magnésium stéarate. The lactose may be 35 anhydrous or in the form of the monohydrate. Other suitable excipients include dextran, glucose, maltose, sorbrtol, xylitol, fructose, sucrose and trehalose.
A suitable solution formulation for use in an atomizer using electrohydrodynamics to produce a fine mist may contain from 1 pg to 20 mg of the compound of the invention per
actuation and the actuation volume may vary from 1 pL to 100 pL. A typical formulation may comprise a compound of Formula I or a pharmaceutically acceptable sait thereof, propylene glycol, stérile water, éthanol and sodium chloride. Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
Suitable flavors, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added tothose formulationsofthe invention intended for inhaled/intranasal administration.
Formulations for inhaled/intranasal administration may be formulated to be Immédiate and/or modified release using, for example, PGLA. Modified release formulations include 10 delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
In the case of dry powder inhafers and aérosols, the dosage unit is determined by means of a valve which delivers a metered amount. Units in accordance with the invention are typically arranged to adminîster a metered dose or ‘pufT containing from 0.01 to 100 mg ofthe compound of Formula I. The overall daily dose will typically be in the range 1 pg to 200 mg, 15 which may be administered in a single dose or, more usually, as divided doses throughout the day.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as 20 appropriate.
Formulations for rectal/vaglnal administration may be formulated to be Immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may also be administered directly to the eye or ear, typically in the form of drops of a micronized suspension or solution in isotonie, pH-adjusted, stérile saline. Other formulations suitable for ocularand aurai administration include ointments, gels, biodégradable (e.g,, absorbable gel sponges, collagen) and non-biodegradable (e.g., silicone) implants, wafers, lenses and particulate or vesicular Systems, such as nlosomes or liposomes. A polymer such as crossed-linked polyacryftc acid, polyvinylalcohol, hyaluronic acid, a cellulosîc polymer, for example, hydroxypropyl methyl cellulose, hydroxyethyl cellulose, or methyl cellulose, or a heteropolysaccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
Formulations for ocular/aural administration may be formulated to be immédiate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted, or programmed release.
The compounds of the invention (including pharmaceutically acceptable salts thereof) may be combined with soluble macromolecular entities, such as cyclodextrin and suitable dérivatives thereof or polyethylene glycol-containing polymère, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use In any ofthe 5 aforementioned modes of administration.
Drug-cyclodextrin complexes, for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non-inclusion complexes may be used. As an alternative to direct complexation with the drug, the cyclodextrin may be used as an auxiliary additive, i.e., as a carrier, diluent, or solubilizer. Most commonly used for these 10 purposes are alpha-, beta- and gamma-cyclodextrins, examples of which may be found in
International Patent Applications Nos. WO 91/11172, WO 94/02518 and WO 98/55148.
Since the présent invention has an aspect that relates to the treatment of the disease/conditions described herein with a combination of active ingrédients which may be administered separately, the invention also relates to combining separate pharmaceutical 15 compositions in kit form. The kit comprises two separate pharmaceutical compositions: a compound of Formula I, a prodrug thereof, or a sait of such compound or prodrug; and a second compound as described above. The kit comprises means for containing the separate compositions such as a container, a divlded bottle or a divided foil packet. Typically the kit comprises directions for the administration of the separate components. The kit form Is 20 particulariy advantageous when the separate components are for example administered in different dosage forms (e.g., oral and parentéral), are administered at different dosage Intervals, or when titration of the Individual components of the combination is desired by the prescribing physician.
An example of such a kit is a so-called blister pack. Blîster packs are well known in the 25 packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a transparent plastic material. During the packaging process recesses are formed in the plasticfoil. The recesses hâve the size and shape ofthetablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet 30 of relatively stiff material Is sealed against the plastic foil at the face of the foil which Is opposite from the direction in which the recesses were formed. As a resuit, the tablets or capsules are sealed In the recesses between the plastic foil and the sheet. In some embodiments, the strength ofthe sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at 35 the place of the recess. The tablet or capsule can then be removed via said opening.
It may be désirable to provide a memory aid on the kit, e.g., In the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen on which the tablets or capsules so specified should be ingested. Another example of such a
memory aid is a calendar printed on the card, e.g., as follows First Week, Monday, Tuesday, etc.... Second Week, Monday, Tuesday,...* etc. Other variations of memory aids will be readily apparent. A daily dose can be a single tablet or capsule or several pills or capsules to be taken on a given day. Also, a daily dose of Formula I compound can consist of one tablet or capsule while a daily dose of the second compound can consist of several tablets or capsules and vice versa. The memory aid should reflect this.
In another spécifie embodiment of the Invention, a dispenser designed to dispense the daily doses one at a time in the order of their Intended use Is provided. For example, the dispenser Is equipped with a memory aid, so as to further facilitate compliance with the regimen. An example of such a memory aid is a mechanical counter which indicates the number of daily doses that has been dispensed. Another example of such a memory aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
The Invention will be described In greater detail by way of spécifie examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non-critical parameters that can be changed or modified to yield essentially the same results. Additional compounds within the scope of this invention may be prepared using the methods illustrated in 20 these Examples, either alone or in combination with techniques generally known in the art. In the following Examples and Préparations, DMSO means dimethyl sulfoxide, “N where referring to concentration means Normal, “M means molar, “mL means milliliter, “mmol means millimoles, “pmol means micromoles, “eq. means équivalent, “’C means degrees Celsius, MHz means mégahertz, “HPLC means high-performance liquid chromatography.
EXAMPLES
The following illustrate the synthesis of various compounds of the présent invention. Additional compounds within the scope of this invention may be prepared using the methods illustrated In these Examples, either alone or in combination with techniques generally known in the art.
Expérimente were generally carried out under inert atmosphère (nitrogen or argon), particularly in cases where oxygen- or moisture-sensitive reagents or Intermediates were employed. Commercial solvents and reagents were generally used without further purification. Anhydrous solvents were empioyed where appropriate, generally AcroSeal® products from Acros Organics or DriSolv® products from EMD Chemicals. In other cases, commercial solvents were passed through columns packed with 4A molecular sieves, until the following QC standards for water were attained: a) <100 ppm for dichloromethane, toluene, Ν,/Ψ dimethylformamide and tetrahydrofuran; b) <180 ppm for methanol, éthanol, 1,4-dioxane and diisopropylamine. For very sensitive réactions, solvents were further treated with metallic
sodium, calcium hydride or molecular sieves, and distilled just prior to use. Products were generally dried under vacuum before being carried on to further reactions or submitted for biological testing. Mass spectrometry data is reported from either liquid chromatography-mass spectrometry (LCMS), atmospheric pressure chemical ionization (APCI) or gas chromatography-mass spectrometry (GCMS) instrumentation. Chemical shifts for nuclear magnetic résonance (NMR) data are expressed in parts per million (ppm, δ) referenced to residual peaks from the deuterated solvents employed. In some examples, chiral séparations were carried out to separate enantiomers or diastereomers of certain compounds of the invention (in some examples, the separated enantiomers are designated as ENT-1 and ENT-2, 10 or the separated diastereomers are designated as DIAST-1 and DIAST-2, according to their order of elution). In some examples, the optical rotation of an enantiomer was measured using a polarimeter. According to its observed rotation data (or its spécifie rotation data), an enantiomer with a clockwise rotation was designated as the (+)-enantiomerand an enantiomer with a counter-clockwise rotation was designated as the (-)-enantiomer. Racemic compounds are indicated by the presence of (+/-) adjacent to the structure; in these cases, indicated stereochemistry represents the relative (ratherthan absoiute) configuration of the compound's substituents.
Reactions proceeding through détectable intermediates were generally followed by LCMS, and allowed to proceed to full conversion prior to addition of subséquent reagents. For 20 synthèses referencing procedures in other Examples or Methods, reaction conditions (reaction time and température) may vary. In general, reactions were followed by thin-layer chromatography or mass spectrometry, and subjected to work-up when appropriate. Purifications may vary between experiments: in general, solvents and the solvent ratios used for eluents/gradients were chosen to provide appropriate RfS or rétention times.
For clarity purposes, the stereochemistry of the substituents on the 3azabicyclo[3.1.0]hexyl skeleton in Examples and intermediates herein is indicated by using Chemical Abstracts nomenclature. The stereochemistry of the other compounds in the Examples and intermediates herein is indicated by using IUPAC nomenclature.
Abbreviations:
BOC - tert-butoxycarbonyl
HPLC - h ig h-performance liquid chromatography NADP - nicotinamide adenine dinucleotide phosphate PMB - para-methoxybenzyl (or 4-methoxybenzyl) p-TsOH -para-toluenesulfonic acid, 4-methylbenzenesulfbnic acid psi - pounds per square inch
Example 1 (2R)-1,1,1-TrifluorO-3-hydroxypropan-2-yl (1a,5a,6a)-6-[1-(5-methoxypyridin-2-yl)-1iï-pyrazol-3yl]-3-azabicyclo[3.1.0]hexana-3-carboxylate (1)
NEt3
CH3MgBr
cf3cooh
CF3COOH
02 NaOH
Pd(dppf)CI2 KOAc
N=< C12 r
O CF3
ΑοΑ^ορμβ
Step 1. Synthesis of(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-ol (C1). (4-Methoxyphenyl)methanol (98%, 1.14 mL, 8.96 mmoî) was slowly added to a 0 ’C solution of sodium bis(trimethylsilyl)amide In tetrahydrofuran (1.0 M, 8.9 mL, 8.9 mmol) in a microwave vial. After the reaction mixture had stirred at 0 *C for 45 minutes, (2R)-2(trifluoromcthyl)oxiranc (500 mg, 4.46 mmol) In tetrahydrofuran (2 mL) was added via syringo, and the vial was sealed and heated at 100 C for 18 hours. The reaction mixture was then cooled to room température and diluted with water; the mixture was extracted twice with tertio butyl methyl ether and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via chromatography on silîca gel (Gradient: 0% to 60% ethyl acetate In heptane)
afforded the product as a pale yellow oil. Yield: 1.09 g, 4.36 mmol, 98%. GCMS m/z 250.1 [M*J. 1H NMR (400 MHz, DMSO-de) δ 7.26 (d, >8.5 Hz, 2H), 6.91 (d, >8.5 Hz, 2H), 6.36 (d, >6.7 Hz. 1H), 4.46 (s, 2H), 4.21-4.09 (m, 1H), 3.74 (s, 3H), 3.58 (dd, half of ABX pattern, >10.6,4.5 Hz, 1H), 3.48 (dd, half of ABX pattern, >10.5, 6.3 Hz, 1H).
Step 2. Synthesis ofpentafluorophenyl (2R)-1,1,1-trifiuoro-3-[(4-methoxybenzyl)oxyJpropan-2-yl carbonate (C2).
Bis(pentafluorophenyl) carbonate (1.33 g, 3.37 mmol) was added to a 0 °C solution of C1 (929 mg, 3.71 mmol) in acetonitrile (30 mL). Triethylamine (1.71 g, 16.9 mmol) was added in a drop-wise manner, and the reaction was warmed to 25 eC and stirred for 2 hours. The resulting solution of C2 was used directly in Step 11. For subséquent synthèses described herein that utilize C2, this material was generated at the appropriate scale, and the reaction solution of C2 was used directly in the coupling reaction.
Step 3. Synthesis oftert-butyl (1a,5a,6a)-6-[methoxy(methyl)carbamoyl]-3azablcyclo[3.1.0]hexane-3-carboxylate (C3).
1-[3-(Dlmethylamino)propyl]-3-ethylcarbodiimide hydrochloride (10.1 g, 52.7 mmol) and
1H-benzotriazol-1-ol (7.13 g, 52.8 mmol) were added to a 0 eC solution of (1a,5a.6a)-3-(fertbutoxycarbonyl)-3-azabicyclo[3.1.0]hexane-6-carboxylic acid (8.00 g, 35 mmol) in dichloromethane (80 mL), and the reaction mixture was stirred at 0 °C for 30 minutes. A solution of N-methoxymethanamlne hydrochloride (6.87 g, 70.4 mmol) and Λ/,/V-diisopropy le thy lamine 20 (13.6 g, 105 mmol) in dichloromethane (50 mL) was then added drop-wise over a period of 10 minutes, and the reaction mixture was stirred at room température (25 ’C) for 2 hours. After addition of water (100 mL), the mixture was extracted with dichloromethane (3 x 100 mL), and the combined organic layers were washed with water (50 mL) and with saturated aqueous sodium chloride solution (50 mL), dried over sodium sulfate, filtered, and concentrated In vacuo to provide the product as a light yellow oil. Yield: 9.46 g, 35.0 mmol, 100%. 1H NMR (400 MHz, CDCb) δ 3.72 (s, 3H), 3.64 (d, half of AB quartet, >11.2 Hz, 1H), 3.55 (d, half of AB quartet, >11.0 Hz, 1H), 3.49-3.39(m, 2H), 3.18 (s, 3H), 2.11-1.99(m, 2H), 1.99-1.91 (brs, 1H), 1.43 (s, 9H).
Step 4. Synthesis oftert-butyl (1a,5a,6a)-6-acetyl-3-azablcyclo[3.1.0]hexane-3-carboxylate (C4).
Méthylmagnésium bromide (3.0 M solution in tetrahydrofuran: 23.3 mL, 69.9 mmol) was added in a drop-wise manner to a 0 eC solution of C3 (9.46 g, 35.0 mmol) In tetrahydrofuran (100 mL). The reaction mixture was stirred at room température (25 ’C) for 1 hour, whereupon it was quenched with saturated aqueous ammonium chloride solution (200 mL) and extracted with 35 ethyl acetate (3 x 100 mL). The combined organic layers were washed sequentially with water (100 mL) and with saturated aqueous sodium chloride solution (100 mL), dried over sodium sulfate, filtered, and concentrated In vacuo to provide the product as a red solid. Yield: 7.82 g, 34.7 mmol. 99%. ’H NMR (400 MHz, CDCIj) δ 3.62 (d, half of AB quartet, >11.3 Hz, 1H), 3.53
(d, half of AB quartet, 7=11.3 Hz, 1H), 3.41-3.32 (m, 2H), 2.21 (s, 3H), 2.05-2.01 (m, 2H), 1.77 (dd, 7=3.0, 2.9 Hz, 1H), 1.39 (s, 9H).
Step 5. Synthesis ofteft-butyl (1a,5a,6a)-6-[(2E)-3-(dimethylamino)prop-2-enoyl]-3azablcyclo[3.1.0]hexane-3-carboxylate (CS).
To a solution of C4 (7.82 g, 34.7 mmol) in Ν,Ν-dimethylformamÎde (50 mL) was added
Λ/,Ν-dimethylformamide dimethyl acetal (12.4 g, 104 mmol), and the reaction mixture was stirred at 110 ’C for 16 hours. It was then cooled, treated with water (100 mL), and extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed sequentially with water (3 x 100 mL) and with saturated aqueous sodium chloride solution (90 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford the product as a red solid. Yield: 9.20 g, 32.8 mmol, 94%. ’H NMR (400 MHz, CDCh) δ 7.51 (d, 7=12.7 Hz, 1H), 5.13 (d, 7=12.7 Hz, 1H), 3.63 (d, half of AB quartet, <7=11.2 Hz, 1H), 3.54 (d, half of AB quartet, */=11.0 Hz, 1H), 3.44-3.36 (m, 2H), 3.15-2.93 (br s. 3H), 2.93-2.70 (brs, 3H), 2.10-1.97 (m, 2H), 1.60 (dd, 7=2.9, 2.9 Hz, 1H), 1.42 (s, 9H).
Step 6. Synthesis ofteiï-butyl (1a, 5a, 6a)-6-(1iï-pyrazol-3-yl)-3-azabicyclo[3.1.0]hexane-3carboxylate (C6).
Hydrazine hydrate (1.97 g, 39.4 mmol) was added to a solution of C5 (9.20 g, 32.8 mmol) in éthanol (100 mL), and the reaction mixture was stirred at 80 ’Cfor 16 hours. After concentration in vacuo, the residue was purified by chromatography on silica gel (Eluents: 9%, 20 then 17%, then 50% ethyl acetate In diethyl ether) to afford the product as a white solid. Yield: 7.00 g, 28.1 mmol, 86%. LCMS m/z 193.8 [(M - 2-methylprop-1-ene)+H]*. Ή NMR (400 MHz, CDCb) δ 7.47 (d, 7=2.0 Hz, 1 H). 6.01 (br d, 7=1.8 Hz, 1H), 3.78 (d, 7=10.9 Hz, 1H), 3.69 (d, 7=11.0 Hz, 1H), 3.51-3.41 (m, 2H), 1.90-1.83 (m, 2H), 1.80 (dd, 7=3.4, 3.4 Hz, 1H), 1.46 (s, 9H). Step 7. Synthesis oftert-butyl (1a.5a,6a)-6-[1-(5-brOmopyndin-2-yl)-1\-\-pyrazol-3-yl]-325 azabicyclo[3.1.0]hexane-3-carboxylate (C7).
A mixture of C6 (500 mg, 2.01 mmol), 5-bromo-2-fluoropyridine (529 mg, 3.01 mmoi) and césium carbonate (1.96 g, 6.02 mmol) In N,N-dimethylformamide (20 mL) was stirred in a microwave reactor at 160 C for 1 hour. The reaction mixture was then combined with two slmilar reactions carried out on C6 (500 mg, 2.01 mmol, and 350 mg, 1.40 mmol), diiuted with 30 water ( 100 mL), and extracted with ethyl acetate (3x50 mL); the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via silica gel chromatography (Gradient: 0% to 20% ethyl acetate in petroleum ether) afforded the product as a white solid. Yield: 1.25 g, 3.08 mmoi, 57%. ’H NMR (400 MHz, CDCh) δ 8.41 (dd, 7=2.4, 0.6 Hz, 1H), 8.37 (d, 7=2.6 Hz, 1H), 7.87 (dd, half of ABX pattern, 7=8.7, 2.3 Hz, 1H), 7.80 (dd, half 35 of ABX pattern, 7=8.7, 0.7 Hz, 1 H), 6.16 (d, 7=2.6 Hz, 1 H), 3.80 (d, 7=11.0 Hz, 1 H), 3.72 (d,
7=11.0 Hz, 1H), 3.52-3.42 (m, 2H), 1.99-1.91 (m, 2H), 1.85 (dd, 7=3.5, 3.4 Hz, 1H), 1.47 (s, 9H). Step 8. Synthesis oftert-butyl (1a,5a,6a)-6-{1-[5-(4l4,5,5-tetramethyl-1,3,2-dioxaborolan-2yl)pyndin-2-yl]-1 H-pyrazol-3-yl}-3-azabicyclo[3.1.0]hexane-3-carboxylate (C8).
To a suspension of C7 (1,00 g, 2.47 mmol) in toluene (20 mL) were added 4,4,4',4',5,5,5',5'-octamethyl-2,2’-bl-1,3,2-dioxaborolane (940 mg, 3.70 mmol), [1.1’bis(diphenylphosphino)ferrocene]dichloropalladium(ll) (181 mg, 0.247 mmol), and potassium acetate (726 mg, 7.40 mmol), and the mixture was degassed with nitrogen for 5 minutes. The 5 reaction mixture was stirred for 18 hours at 120 ’C, whereupon it was concentrated ln vacuo and purified by chromatography on stlica gel (Gradient: 0% to 20% ethyl acetate in petroleum ether) to afford the product as a white solid. Yield: 1.02 g, 2.25 mmol, 91%. LCMS m/z 453.3 [M+H]*. Ή NMR (400 MHz, CDCh) δ 8.73-8.69 (m, 1H), 8.48 (d, J=2.5 Hz, 1H), 8.14 (dd, J=8.2,
1.8 Hz, 1H), 7.86 (br d, J=8.2 Hz, 1H), 6.16 (d, J=2.5 Hz, 1H), 3.81 (d, J=11 Hz, 1H), 3.73 (d, 10 J=11 Hz, 1H), 3.53-3.42 (m, 2H), 2.01-1.93 (m, 2H), 1.87 (dd, J=3.3, 3.3 Hz, 1H), 1.47 (s, 9H),
1.37 (s, 12H).
Step 9. Synthesis of tert-butyl (1a,5a,6a)-6-[1-(5-hydroxypyridin-2-yl)-1H-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate (C9).
To a 0 ’C mixture of C8 (1.02 g, 2.25 mmol) in tetrahydrofuran and water (1:1 mixture, 15 80 mL) was added aqueous sodium hydroxide solution (6 M, 1 mL, 6 mmol), followed by hydrogen peroxide (30% solution in water, 0.77 g, 6.8 mmol). The reaction mixture was allowed to warm to 25 ’C, and was stirred for 12 hours, whereupon it was quenched with aqueous sodium thiosulfate solution, acidified to pH 6 with aqueous hydrochloric acid, and extracted with dichloromethane (3 x 30 mL). The combined organic layers were concentrated under reduced 20 pressure and purified via chromatography on silica gel (Gradient: 0% to 10% methanol in dichloromethane) to provide the product as a white solid. Yield: 742 mg, 2.17 mmol, 96%. LCMS m/z 343.1 [M+H]*. ’H NMR (400 MHz, CD3OD) δ 8.26 (d, J=2.6 Hz, 1 H), 7.93 (dd, J=2.9, 0.5 Hz, 1H), 7.67 (dd, J=8.8,0.5 Hz, 1H), 7.32 (dd, J=8.8, 2.9 Hz, 1H), 6.21 (d, J-2.5 Hz, 1H), 3.69 (d, J=10.9 Hz, 2H). 3.52-3.42 (m, 2H), 2.02-1.94 (m, 2H), 1.75 (dd, J=3.5, 3.4 Hz, 1H), 1.47 25 (s, 9H).
Step 10. Synthesis of6-{3-[(1a,5a,6a)-3-azabicyclo[3.1.0]hex-6-yl]-1H-pyrazol-1-yl}pyrid/n-3-ol, tris(trifluomacetic acid) sait (C10).
A solution of C9 (742 mg, 2.17 mmol) in dichloromethane (5 mL) was cooled in an ice bath, and then treated with trifluoroacetic acid (3 mL). The reaction mixture was stirred for 30 30 minutes at 25 ’C, whereupon it was concentrated ln vacuo, affording the product (1.27 g) as a yellow gum. LCMS m/z 243.0 [M+H]*.
Step 11. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzy!)oxy]propan-2-yl (1α,5α.6α)-6-[1(5-hydroxypyridin-2-yl)· 1H-pyrazol-3-yl]-3-azabicyclo[3. 1.0]hexane-3-carboxy!ate (C11).
Triethylamine (1.10 g, 10.9 mmol) was slowly added to a 0 ’C solution of C10 (from the 35 previous step, 1.27 g, £2.17 mmol) in acetonitrile (20 mL), whereupon the mixture was stirred for 1 hour. Compound C2 [from step 2, as the crude reaction mixture in acetonitrile (30 mL);
-1.6 g, 3.4 mmol] was added to the 0 ’C reaction mixture, which was then stirred at 28 ’C for 18 hours. It was then cooled in an ice-water bath and slowly treated with a second batch of C2
(-0.74 g, 1.6 mmol). After stirring for 18 hours at 25 *C, the reaction mixture was concentrated in vacuo; the residue was purified via chromatography on silica gel (Gradient: 0% to 30% ethyl acetate In petroleum ether) to provide the product as a white solid. By 1H NMR analysis, this was judged to be a mixture of rotamers. Yield: 430 mg, 0.83 mmol, 38% over two steps. LCMS 5 m/z 519.1 [M+H]*. Ή NMR (400 MHz, CD3OD) δ 8.29-8.25 (m, 1H), 7.93 (d. >2.9 Hz, 1H), [7.69 (d, >8.8 Hz) and 7.68 (d, >8.9 Hz), total 1H], 7.32 (dd, >8.8, 2.9 Hz, 1H), 7.29-7.23 (m, 2H), 6.95-6.88 (m, 2H), [6.24 (d, J*2.5 Hz) and 6.20 (d, >2.5 Hz), total 1 H], 5.53-5.40 (m, 1H), [4.56 (d, half of AB quartet, >11.4 Hz) and 4.54 (d, half of AB quartet, >11.5 Hz), total 1 H],
4.46 (d, half of AB quartet, >11.5 Hz, 1H), 3.84-3.68 (m, 4H), [3.79 (s) and 3.73 (s), total 3H], 10 3.60-3.53 (m, 2H), 2.07-1.99 (m, 2H), [1.77 (dd, >3.5, 3.4 Hz) and 1.74 (dd, >3.5, 3.3 Hz), total 1H],
Step 12. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl(1a,5a,6a)-6-[1(5-methoxypyridln-2-yl)-1H-pyrazol-3-yl]-3-azablcyclo[3.1.0]hexane-3-carboxylate (C12).
To a 0 ’C solution of C11 (90.0 mg, 0.174 mmol) in /V.N-dimethylformamide (1 mL) were 15 added potassium carbonate (36 mg, 0.26 mmol) and iodomethane (25.9 mg, 0.182 mmol). The reaction mixture was stirred at 28 *C for 2 hours, whereupon it was concentrated In vacuo and purified by silica gel chromatography (Gradient: 0% to 30% ethyl acetate in petroleum ether) to afford the product as a colorless gum. By 1H NMR analysis, this was judged to be a mixture of rotamers. Yield: 86 mg, 0.16 mmol, 92%. LCMS m/z 533.3 [M+H]*. Ή NMR (400 MHz, CDCh) δ 20 8.33 (d, >2.4 Hz, 1 H), 8.05 (d, >2.8 Hz, 1 H), 7.82 (d, >8.9 Hz, 1 H), 7.32 (dd, >9.0, 2.9 Hz,
1H), 7.30-7.23 (m, 2H), 6.94-6.86 (m, 2H), [6.14 (d, >2.3 Hz) and 6.13 (d. >2.4 Hz), total 1 H], 5.54-5.43 (m, 1H), [4.57 (d, half of AB quartet, >11.7 Hz) and 4.56 (d, half of AB quartet, >11.7 Hz), total 1 H], 4.48 (d, half of AB quartet, >11.7 Hz, 1H), 3.92-3.65 (m, 4H), 3.87 (s, 3H), [3.81 (s) and 3.78 (s), total 3H], 3.64-3.53 (m, 2H), 2.06-1.97 (m, 2H), 1.89-1.82 (m, 1H).
Step 13. Synthesis of(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (1α,5α,6α)-6-[1-(5methoxypyridin-2-yl)-1 H-pyrazol-3-yl]-3-azablcyclo[3.1.0]hexane-3-carboxylate ( 1).
Trifluoroacetic acid (1 mL) was slowly added to a 0 eC solution of C12 (114 mg, 0.214 mmol) in dichloromethane (2 mL). The reaction mixture was stined at 26 ’C for 30 minutes, whereupon It was cooled in an Ice bath and slowly treated with saturated aqueous sodium 30 bicarbonate solution (20 mL). The mixture was extracted with dichloromethane (3 x 20 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Préparative thîn layer chromatography on silica gel (Eluent: 1:1 petroleum ether: ethyl acetate) provided the product as a white solid. By 1H NMR analysis, this was judged to be a mixture of rotamers. Yield: 65 mg, 0.16 mmol, 75%. LCMS m/z 413.1 [M+H]*. 1H NMR (400 MHz, CD3OD) 35 δ 8.32 (d, >2.5 Hz, 1H), 8.07 (d, >2.9 Hz, 1H), 7.80-7.76 (m, 1H), 7.51 (dd, >9.0, 3.0 Hz, 1H), [6.26 (d, >2.6 Hz) and 6.25 (d, >2.8 Hz), total 1H], 5.34-5.24 (m, 1H), 3.94-3.74 (m, 4H), 3.90 (s, 3H), 3.70-3.57 (m, 2H), 2.11-2.02 (m, 2H), [1.86 (dd, >3.6, 3.5 Hz) and 1.79 (dd, >3.5, 3.4 Hz), total 1H].
Example 2 (2R)-i, ί, 1-Tnfluoro-3-hydroxypropan-2-yl 4-[1-(4-fluorophenyl)-1H-pyrazol-3-ylJpiperidine-1carboxylate (2)
O Cu(OAc)2
N
Et3N
Hz
Pd/C
O ci3coA0,cci3
Step 1. Synthesis oftert-butyl4-(1H-pyrazol-3-yl)piperidine-1-carboxylate (C13).
To a 0 *C mixture of 4-(1H-pyrazol-3-yl)plperidine, dihydrochloride sait (11.3 g, 50.4 mmol) and triethylamine (20.4 g, 202 mmol) In dichloromethane (250 mL) was slowly added ditert-butyl dicarbonate (11.0 g, 50.4 mmol), and the reaction mixture was allowed to stir at room température ovemight. It was then concentrated under reduced pressure and purified using silica gel chromatography (Gradient: 17% to 80% ethyl acetate In petroleum ether), provlding the product as a light yeilow gum. Yield: 9.50 g, 37.8 mmol, 75%. LCMS m/z 195.8 [(M - 2methylprop-1-ene)+H]+. ’H NMR (400 MHz, CDCh) δ 7.50 (brs, 1H), 6.12 (brs, 1H), 4.30-4.03 (br s, 2H), 2.96-2.73 (m, 3H), 2.04-1.86 (m, 2H), 1.73-1.55 (m, 2H), 1.48 (s, 9H).
Step 2. Synthesis ofteü-butyl 4-[1-(4-[luorophenyl)-1iï-pyraz(jl-3-yl]piperidine-1-cai'boxylato (C14).
To a mixture of C13 (700 mg, 2.78 mmol), (4-fluorophenyl)boronic acid (429 mg, 3.07 mmol), and 4A molecular sieves (1.0 g) in dry dichloromethane (40 mL) were added pyridine (441 mg, 5.58 mmol) and copper(ll) acetate (759 mg, 4.18 mmol). The reaction mixture was stined for 48 hours at room température, while open to the air, and was then filtered. The filtrate
was poured Into water and extracted with dichloromethane (3 x 50 mL); the combined organic layers were washed sequentially with water (100 mL) and with saturated aqueous sodium chloride solution (100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via chromatography on silica gel (Eluent: 25% ethyl acetate In petroleum ether) afforded the product as a white solid. Yield: 700 mg, 2.0 mmol, 72%. 1H NMR (400 MHz, CD3OD) δ 8.05 (br d, J=2.5 Hz, 1H), 7.74-7.68 (m, 2H), 7.24-7.17 (m, 2H), 6.37 (br d, J=2.5 Hz. 1H), 4.19-4.10 (m, 2H), 3.02-2.85 (m, 3H), 2.02-1.92 (m, 2H), 1.71-1.58 (m, 2H), 1.48 (s, 9H). Step 3. Synthesis of 4-[1-(4-fluorophenyl)-1H-pyrazol-3-yl]pipendine, hydrochloride sait (C15).
A solution of hydrogen chloride in ethyl acetate (4M, 10 mL, 40 mmol) was added to a 0 10 *C solution of C14 (700 mg, 2.0 mmol) in ethyl acetate (10 mL). After the reaction mixture had been stirred for 1.5 hours at room température (18 eC), it was concentrated in vacuo to provide the product as a white solid. This material was used without further purification. Yield: 560 mg, assumed quantitative. 1H NMR (400 MHz, CD3OD) δ 8.13-8.10 (m, 1H), 7.77-7.70 (m, 2H), 7.26-7.19 (m, 2H), 6.44-6.42 (m, 1H), 3.49 (ddd, J=13,4,4 Hz, 2H), 3.22-3.07 (m, 3H), 2.3115 2.22 (m, 2H), 2.06-1.93 (m, 2H).
Step 4. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(1-(4fluorophenyl)-1 H-pyrazol-3-yl]plperidine-1-carboxylate (C16).
To a 0 eC solution of bis(trichloromethyl) carbonate (27.2 mg, 91.6 pmol) In dichloromethane (5 mL) was added C1 (69.4 mg, 0.277 mmol), followed by N.N20 diisopropylethylamine (36 mg, 0.28 mmol) and 4-(dimethylamino)pyridine (2.0 mg, 16 pmol).
After the reaction mixture had stirred at room température (15 *C) for 7 hours, It was cooled to 0 eC and treated with a solution of C15 (100 mg, 0.408 mmol) and N.AAdiisopropylethylamine (72 mg, 0.56 mmol) in dichloromethane (5 mL). The reaction mixture was then stirred at 15 eC for 16 hours, whereupon it was diluted with dichloromethane (10 mL) and washed sequentially with 25 water (3 x 20 mL) and with saturated aqueous sodium chloride solution (2 x 20 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Eluent: 20% ethyl acetate in petroleum ether) provided the product as a colorless oil, which was not pure via 1H NMR analysis. Yield: 90 mg, £60%. 1H NMR (400 MHz, CD3OD), characteristic peaks: δ 8.04 (d, J=2.5 Hz. 1H), 7.74-7.68 (m, 2H), 7.20 (br dd, J=8.8, 8.8 Hz, 2H), 3.79 (s, 3H), 3.15-2.92 (m, 30 3H), 2.06-1.95 (m, 2H), 1.75-1.61 (m, 2H).
Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[1-(4-fluorophenyl)-1H-pyrazol-
3-yl}piperidine-1-carboxylate (2).
To a solution of C16 (50 mg, 96 pmol) in éthanol (50 mL) was added palladium on carbon (30 mg), and the reaction mixture was stined at 20 *C under hydrogen (40 psi) for 6 35 hours. It was then filtered through a pad of diatomaceous earth, and the filtrate was concentrated in vacuo; purification via préparative thin layer chromatography on silica gel (Eluent: 25% ethyl acetate in petroleum ether) afforded the product as a white solid. Yield: 15 mg, 37 pmol, 38%. LCMS m/z 402.0 [M+H]*. Ή NMR (400 MHz, CD3OD) δ 8.06 (d, J=2.5 Hz,
H), 7.74-7.68 (m, 2H), 7.21 (brdd, >9.0, 8.4 Hz, 2H), 6.38 (d, >2.5 Hz, 1H), 5.32 (dqd, >7,
7,4 Hz, 1H), 4.29-4.16 (brm, 2H), 3.89 (brdd, half of ABX pattern, >12.5, 4 Hz, 1H), 3.79 (br dd. half of ABX pattern, >12.4, 6.9 Hz, 1H), 3.19-3.0 (m, 2H), 2.98 (tt, >11.5,4 Hz, 1H), 2.081.96 (m, 2H), 1.85-1.61 (br m, 2H).
Example 3 (2R)-1,1,1-Trifluoro-3-hydroxypmpan-2-yl (1a,5a,6a)-6-[1-(4-fluorophenyl)-1H-pyrazol-3-yl]-3azablcyclo[3.1.0]hexane-3-carboxylate (3)
Step 1. Synthesis of 1-{[({(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2yl}oxy)carbonyi]oxy}pynolidine-2,5-dione (C17).
To a solution of C1 (701 mg, 2.80 mmol) in dichloromethane (20 mL) were added triethylamine (850 mg, 8.40 mmol) and I.T-fcarbonylbisioxyJJdipyrrolidine-S.S-dions (717 mg,
2.80 mmol). The reaction mixture was stirred for 18 hours at 25 “C, then used directly in Step 4.
For subséquent synthèses described herein that utilize C17, this material was generated at the appropriate scale, and the reaction solution of C17 was used directly in the coupling reaction. Step 2. Synthesis of tert-butyl (1a,5a,6a)-6-[1-(4-fluorophenyl)~1H'pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate (C18).
To a 15 ’C solution of C6 (4.0 g. 16 mmol) in dichloromethane (300 mL) were added (4fluorophenyl)boronic acid (2.92 g, 20.9 mmol), copper(ll) acetate (4.37 g, 24.1 mmol), pyridîne (3.81 g, 48.2 mmol), and 4A molecular sieves (0.5 g). The réaction mixture was stirred for 18 hours at 30 ’C, whereupon it was washed with aqueous ammonium hydroxide solution (100 5 mL). This aqueous layer was extracted with dichloromethane (2 x 100 mL), and the combined organic layers were washed with saturated aqueous sodium chloride solution (150 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 25% ethyl acetate in petroleum ether) provided the product as a white solid. Yield: 3.3 g,
9.6 mmol, 60%. LCMS m/z 287.8 [(M - 2-methylprop-1-ene)+H]*. 1H NMR (400 MHz, CDCb) δ 10 7.73 (d, J=2.5 Hz, 1H), 7.62-7.56 (m, 2H), 7.12 (brdd, J=8.9, 8.4 Hz, 2H), 6.16 (d, J=2.4 Hz,
1H), 3.80 (d, half of AB quartet, J=11.0 Hz, 1H), 3.71 (d, half of AB quartet, J=10.9 Hz, 1H),
3.52-3.42 (m, 2H), 1.99-1.90 (m, 2H), 1.85 (dd, J=3.4, 3.4 Hz, 1 H), 1.47 (s, 9H). Step 3. Synthesis of(1a,5a,6a)-6-[1-(4-fluorOphenyl)-1iï-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane, trifluoroacetate sait (C19).
A mixture of C18 (1.0 g, 2.9 mmol) in trifluoroacetic acid (10 mL) was stirred for 30 minutes at 15 ’C, whereupon it was concentrated in vacuo. The residue was triturated with tertbutyl methyl ether (10 mL) to provide the product as a white soiid, which was used directly in the following step. LCMS m/z 243.9 [M+H]*.
Step 4. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl (1α,5α,6α)·6-[1-(420 fluorophenyl)-1H-pyrazol-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (C20).
To a 15 ’C solution of C17 [the reaction mixture from Step 1; S2.80 mmol in dichloromethane (20 mL)] was added a solution of C19 (from the previous step, S2.9 mmol) and triethylamine (566 mg, 5.59 mmol) in dichloromethane (10 mL). The reaction mixture was stined overnight at 18 ’C, whereupon it was concentrated in vacuo. Purification using silica gel chromatography (Gradient: 0% to 25% ethyl acetate in petroleum ether) provided the product as a gum. By 1H NMR analysis, this was judged to be a mixture of rotamers. Yield: 900 mg, 1.7 mmol, 61% over two steps. 1H NMR (400 MHz, CDCh) δ 7.74 (d, J*2.4 Hz, 1H), 7.62-7.57 (m, 2H), 7.29-7.24 (m. 2H), 7.13 (br dd, J=8.9, 8.3 Hz, 2H), 6.93-6.88 (m, 2H), [6.19 (d, J=2.4 Hz) and 6.16 (d, J=2.4 Hz), total 1H], 5.53-5.43 (m, 1H), 4.60-4.54 (m, 1H), 4.48 (d, half of AB quartet, J=11.7 Hz, 1 H), 3.90-3.84 (m, 1 H), 3.84-3.73 (m, 2H), [3.82 (s) and 3.79 (s), total 3H],
3.73-3.65 (m, 1H), 3.65-3.54 (m, 2H), 2.04-2.00 (m, 2H), 1.87-1.82 (m, 1H).
Step 5. Synthesis of (2R)-1,1,1-tnfluoro-3-hydroxypropan-2-yl (1a,5a,6a)-6-[1-(4-fluorophenyl)· 1H-pyrazol-3-yl]-3-azabicyclo[3.1.0]hexane-3-carboxylate (3).
Trifluoroacetic acid (10 mL) was added to a solution of C20 (890 mg, 1.7 mmol) in 35 dichloromethane (30 mL), and the reaction mixture was stirred for 4 hours at 15 ’C. It was then slowly poured into saturated aqueous sodium bicarbonate solution, and the resulting mixture was extracted with dichloromethane (3 x 50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo; purification via chromatography on silica gel
(Gradient: 0% to 50% ethyl acetate in petroleum ether) afforded the product as a white solid.
Yield: 440 mg, 1.1 mmol, 65%. LCMS m/z 399.9 [M+H]+. 1H NMR (400 MHz, CDCh) δ 7.74 (d, 7=2.4 Hz, 1H), 7.63-7.56 (m, 2H), 7.13 (dd, 7=8.7, 8.5 Hz, 2H), 6.21-6.17 (m, 1H), 5.31-5.21 (m, 1H), 4.06-3.96 (m, 1H), 3.93-3.80 (m, 3H), 3.67-3.58 (m, 2H), 2.38-2.27 (br m, 1H), 2.08-2.01 (m, 2H), 1.90-1.84 (m, 1 H).
Examples 4 and 5 (28)-1,1,1-Trif!uom-3-hydmxypropan-2-yl 4-(tetrahydro-2Yi-pyran-3-ylmethyl)-1-Oxa-4,9diazaspiro[5.5]undecane-9-carboxylate [fmm C25, DIAST-1] (4) and (28)-1,1,1-Trifluoro-3hydmxypmpan-2-yl 4-(tetrahydm-2H-pyran-3-ylmethyl)- 1-oxa-4,9-diazasplro[5.5]undecane-910 carboxylate [from C26, DIAST-2] (5)
NEt3
2)
o cf3 γ-Ν·Α·ΟΛ-ΟΡΜΒ DIAST-1 + M O CF3 <'nJ\A0PMB C°P DIAST-2 N
t? | 25 CF3COOH A C26 | cf3cooh
o cf3 L· [From C25, , DIAST-1] 0 cf3 L· J [From C26. . DIAST-2]
Step 1. Synthesis ofted-butyl 4~{[(chloroacetyI) amino]methyl]-4-hydroxypiperidino-1~carboxylato (C21).
A solution of potassium carbonate (1.32 kg, 9.55 mol) in water (11 L) was added to a solution of tert-butyl 4-(aminomethyl)-4-hydroxyplperidine-1-carboxylate (1.10 kg, 4.78 mol) in ethyl acetate (11 L). The mixture was cooled to 0 °C, and then treated in a drop-wise manner with chloroacetyl chloride (595 g, 5.27 mol). After completion of the addition, the reaction mixture was warmed to 25 *C and allowed to stir for 16 hours. The aqueous layer was extracted with ethyl acetate (3x10 L), and the combined organic layers were dried over sodium sulfate, 10 filtered, and concentrated In vacuo: trituration of the residue with tert-butyl methyl ether (10 L) afforded the product (1040 g). The filtrate from the trituration was concentrated and triturated with a mixture of tert-butyl methyl ether and petroleum ether (1:1 ; 300 mL) to provide additional product (123 g) as a white solid. Combined yield: 1.16 kg, 3.78 mol, 79%. 1H NMR (400 MHz, CDCh) δ 7.02 (brt, J=5 Hz, 1H), 4.09 (s, 2H), 3.88-3.70 (br m, 2H), 3.43-3.28 (br s, 2H), 3.20 15 (brdd, J=11,11 Hz, 2H), 2.71 (s, 1H), 1.62-1.46 (m, 4H), 1.45 (s, 9H).
Step 2. Synthesis oftert-butyl 3~oxo-1~oxa~4,9-diazaspiro[5.5]undecane-9-carboxylate (C22). This reaction was carried out in two similar batches. To a solution of C21 (540 g, 1.76 mol) in 2-propanol (20 L) was added potassium tert-butoxide (1.98 kg, 17.6 mol) at 25 *C, and the reaction mixture was stirred at 25 °C for 16 hours. After removal of solvent in vacuo, the 20 residue was partitioned between ethyl acetate (15 L) and water (20 L). The aqueous layer was extracted with ethyl acetate (2x15 L), and the combined organic layers were washed wilh saturated aqueous sodium chloride solution (15 L), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was triturated with fert-butyl methyl ether (2 L) at 25 ’C for 3 hours to afford the product as a white solid. Combined yield from the two 25 batches; 540 g, 2.00 mmol, 57%. 1H NMR (400 MHz, CDCh) δ 6.78-6.59 (br m, 1H), 4.16 (s,
2H), 3.96-3.74 (brs. 2H), 3.24 (d, J=2.6 Hz, 2H), 3.11 (brdd, J=12,12 Hz, 2H), 1.89 (brd, J=13 Hz, 2H), 1.58-1.48 (m, 2H), 1.46 (s, 9H).
Step 3. Synthesis oftert-butyi 1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxyiate (C23).
This reaction was carried out In 12 batches, as foilows. Borane-dimethyl sulfide complex 5 (10 M In dimethyl sulfide, 75 mL, 750 mmol) was added in a drop-wise manner to a solution of
C22 (50 g, 180 mmol) In tetrahydrofuran (1.5 L). The reaction mixture was heated at reflux (70 C) for 6 hours and subsequently allowed to stir at 25 'C for 10 hours. It was then quenched with methanol (500 mL), stirred for 30 minutes at 25 °C, and concentrated under reduced pressure. The resulting white solid was dissolved in methanol (1 L), treated with N,N'10 dimethylethane-1,2-diamine (65 g, 740 mmol), and heated at reflux (70 ’C) for 16 hours. The 12 reaction mixtures were combined and concentrated in vacuo to provide a light yellow oil; this was dissolved In dichloromethane (4 L), washed with aqueous ammonium chloride solution (4 x 2 L), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The residue was triturated with petroleum ether (500 mL) at 25 *C for 30 minutes to provide the product (304 15 g) as a white solid. The filtrate from the trituration was concentrated In vacuo, and the residue was triturated with petroleum ether (200 mL) at 25 C for 36 hours, affording additional product (135 g) as a white solid. Combined yield: 439 g, 1.71 mol, 77%. LCMS m/z 257.2 [M+H]*. 1H NMR (400 MHz, CDCh) δ 3.85-3.59 (m, 4H), 3.14 (br dd, J=11,11 Hz, 2H), 2.84 (dd, J=4.9, 4.6 Hz, 2H), 2.68 (s, 2H), 2.02-1.84 (brm, 2H), 1.47-1.33 (m, 2H), 1.45 (s, 9H).
Step 4. Synthesis oftert-butyl 4-(tetrahydtΌ-2l·]-pyran-3-yimethyl)-1-oxa-4,9dlazaspiro[5.5]undecane~9-carboxylate (C24).
Titanium(IV) isopropoxide (998 mg, 3.51 mmol) was added to a mixture of C23 (300 mg, 1,17 mmol) and tetrahydro-2H-pyran-3-carbaldehyde (160 mg, 1.40 mmol) In éthanol (10 mL) at 27 ‘C, and the reaction mixture was stirred at 27 *C for 15 hours. It was then cooled to 0 °C, 25 treated with sodium borohydride (88.6 mg, 2.34 mmol), and allowed to stir at 25 ‘C for 4 hours. Water (10 mL) was added slowly, and the resulting mixture was stirred at 25 *C for 30 minutes. After combination with a mixture derived from a smaller-scale reaction carried out on C23 (50 mg. 0.20 mmol), this was extracted with ethyl acetate (3 x 30 mL). The combined organic layers were dried, filtered, and concentrated in vacuo; purification via chromatography on silica gel 30 (Gradient: 0% to 5% methanol in dichloromethane) provided the product as a colorless oil.
Starting material C23 (200 mg) was also recovered, as a yellow gum. Yield: 106 mg, 0.299 mmol, 22% (51% based on recovered starting materiai). LCMS m/z 355.3 [M+H]*. 1H NMR (400 MHz, CDCh) δ 3.96-3.88 (m, 1H), 3.88-3.80 (m, 1H), 3.79-3.58 (m, 4H), 3.42-3.33 (m, 1H),
3.19-3.04 (m, 3H), 2.42-2.33 (m, 1H), 2.33-2.26 (m, 1 H), 2.26-2.19 (m, 1H), 2.15-2.01 (m, 3H), 35 1.98-1.73 (m, 5H), 1.64-1.53 (m, 2H), 1.44 (s, 9H), 1.44-1.34 (m, 2H).
Step 5. Synthesis of (2R.)· 1,1,1 •trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(tetrahydm-2Hpyran-3-ylmethyl)-1-oxa-4,9-diazaspiro[5.5]undecane-9~carboxylate, DIAST1 (C25) and(2R)18588
1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]prOpan-2-yl 4-(tetrahydro-2H-pyran-3-ylmethyl)-1-oxa4,9-diazaspiro[5.5]undecane-9-carboxylate, DIAST 2 (C26)
A solution of C24 (106 mg, 0.299 mmol) In dichloromethane (2 mL) was cooled to 0 °C and treated with trifluoroacetic acid (0.5 mL). The reaction mixture was stirred at 25 ’C for 50 minutes, whereupon it was concentrated In vacuo to provide 4-(tetrahydro-2H-pyran-3ylmethyl)-1-oxa-4,9-diazaspiro[5.5]undecane, bis-trifluoroacetic acid sait as a yellow oil (100 mg). This material was taken up in acetonitrile (5 mL) and cooled to 0 ’C. Triethylamine (151 mg, 1.49 mmol) was added, and the reaction mixture was allowed to stir at 0 ’C for a few minutes, whereupon C2 (reaction solution in acetonitrile, containing 0.49 mmol) was added drop-wise. The resulting solution was stirred at 0 °C for a few minutes, and then allowed to stir at 25 ’C for 15 hours. The reaction mixture was cooled to 0 ’C and treated in a drop-wise manner with additional C2 (reaction solution In acetonitrile, containing 0.22 mmol). The reaction mixture was again stirred for a few minutes at 0 ’C, before being allowed to stir at 25 ’C for another 15 hours. It was then concentrated in vacuo, and the resîdue was subjected to préparative thin layer chromatography on silica gel (Eluent: 1:1 petroleum ether / ethyl acetate) to afford a mixture of diastereomeric products (100 mg). The diastereomers were separated via supercritical fluid chromatography (Column: Chiral Technologies Chiralpak AD, 5 pm; Mobile phase: 1:3 éthanol / carbon dioxide). The first-eluting compound was C25, obtained as a light yellow gum. Yield: 50 mg, 94 pmol, 31%. LCMS mZr 531.1 [M+H]*. ’H NMR (400 MHz, CDCI3), characteristic peaks: S 7.28-7.23 (m, 2H, assumed; partîally obscured by solvent peak), 6.89 (d. J=8.8 Hz, 2H), 5.53-5.44 (m, 1H), 4.51 (AB quartet, downfield doublet Is broadened, Jab=11.7 Hz, ûvab=28 Hz, 2H), 3.97-3.90 (m, 1H), 3.90-3.82 (m, 2H), 3.82 (s, 3H), 3.45-3.36 (m, 1H),
3.28-3.16 (m, 2H), 3.15-3.07 (m, 1H), 2.45-2.36 (m, 1H), 2.36-2.27 (m, 1H), 2.13-2.06 (m, 2H), 2.05-1.93 (m, 2H), 1.87-1.76 (m, 2H), 1.47-1.35 (m, 2H).
The second-eluting diastereomer was C26, also obtained as a light yellow gum. Yield:
mg, 94 pmol, 31%. LCMS m/z 531.2 [M+H]*. Ή NMR (400 MHz, CDCh), characteristic peaks: δ 7.28-7.22 (m, 2H, assumed; partîally obscured by solvent peak), 6.89 (d, J=8.8 Hz, 2H), 5.54-5.43 (m, 1H), 4.51 (AB quartet, Jab=12 Hz, Avab=26 Hz, 2H), 3.97-3.90 (m, 1H), 3.903.82 (m, 2H), 3.82 (s, 3H), 3.45-3.35 (m, 1H), 3.29-3.16 (m, 2H), 3.16-3.07 (m, 1H), 2.45-2.36 (m, 1H), 2.36-2.28 (m, 1H), 2.14-2.03 (m, 2H), 2.03-1.92 (m, 2H), 1.86-1.75 (m, 2H), 1.46-1.34 (m, 2H).
Step 6. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(tetrahydm-2H-pyran-3ylmethyl)-1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate [from C25, DIAST-1] (4).
Trifluoroacetic acid (1 mL) was added in a drop-wise manner to a 0 ’C solution of C25 (50 mg, 94 pmol) In dichloromethane (4 mL), and the réaction mixture was allowed to stir at 0 ’C for 1 hour. Saturated aqueous sodium bicarbonate solution (20 mL) was added, and the mixture was extracted with dichloromethane (3x15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated In vacuo; préparative thin layer chromatography on silica gel (Eluent: 1:1 ethyl acetate / petroleum ether) provided the product as a light yellow gum. Yield: 34.5 mg, 84.0 pmol, 89%. LCMS m/z 411.2 [M+H]*. 1H NMR (400 MHz, CDCh) δ 5.31-5.18 (br m, 1H), 4.04-3.76 (m, 6H), 3.76-3.66 (m, 2H), 3.44-3.35 (m, 1H). 3.32-3.15 (m, 2H), 3.11 (br dd, J=10,10 Hz, 1H), 2.68-2.46 (br m, 1H), 2.47-2.28 (m, 2H), 2.28-
2.21 (m, 1H), 2.20-1.93 (m, 5H), 1.89-1.75 (m, 2H), 1.65-1.54 (m, 2H), 1.51-1.35 (m, 2H). Step 7. Synthesis of (2R)-1,1,1 - trifluoro-3-hydroxypropa n-2-yl 4-(tetrahydno-2H-pyran-3ylmethyl)-1-oxa-4,9-diazasplro[5.5]undecane-9-carboxylate [from C26, DIAST-2] (5)
Compound C26 was converted to the product using the method described for synthesis of 4 from C25. The product was isolated as a yellow gum. Yield: 34.0 mg, 82.8 pmol, 88%. LCMS m/z 411.1 [M+H]*. Ή NMR (400 MHz, CDCb) δ 5.30-5.19 (br m, 1 H), 4.05-3.77 (m, 6H),
3.77-3.65 (m, 2H), 3.44-3.35 (m, 1H), 3.32-3.17 (m, 2H), 3.12 (br dd, J=10,10 Hz, 1H), 2.612.20 (m, 4H), 2.20-1,94 (m, 5H), 1.90-1.75 (m, 2H), 1.64-1,53 (m, 2H, assumed; partially obscured by water peak), 1.51-1.38 (m, 2H).
Exemple 6 (2R)~1,1,1-Trifluoro-3-hydroxypropan-2-yl 4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (6)
O O=S-CI
O CF3 ^A_oh cf3cooh
CF3COOH
AoA^opmb f
• CF3COOH
Stop 1. Synthesis of tert-butyl 4-[{4-fluorophcnyl)sulfonyl]-1-oxa-4,9-diazaspiro[5 5]undecana-9carboxylate (C27).
4-Fluorobenzenesulfonyl chloride (4.18 g, 21.5 mmol) was added portion-wise to a mixture of C23 (5.0 g, 20 mmol), saturated aqueous sodium bicarbonate solution (55 mL), and dichloromethane (195 mL). The reaction mixture was stirred at room température ovemight, whereupon the aqueous layer was extracted twîce with dichloromethane, and the combined organic layers were dried over magnésium sulfate, filtered, and concentrated In vacuo. Silica gel
chromatography (Gradient: 0% to 10% methanol in dichloromethane) afforded the product as a white foam. Yield: 8.4 g, 20 mmol, quantitative. 1H NMR (400 MHz, CDCh) δ 7,79-7.73 (m, 2H),
7.28-7.22 (m, 2H, assumed; partially obscured by solvent peak), 3.8-3.66 (m, 2H), 3.79 (dd, J=5.0, 5.0 Hz, 2H), 3.19-3.08 (m, 2H), 3.08-2.89 (m, 2H), 2.89-2.67 (m, 2H), 1.96-1.82 (m, 2H), 5 1.54-1.48 (m, 2H), 1.47 (s, 9H).
Step 2. Synthesis of4-[(4-fluorophenyl)sulfonyl}-1-oxa-4,9-diazaspiro[5.5Jundecane, trifluoroacetic acid sait (C28).
Trifluoroacetic acid (15 mL) was slowly added to a solution of C27 (3.16 g, 7.62 mmol) and dichloromethane (38 mL). After the reaction mixture had stirred at room température for 2 10 hours, it was concentrated in vacuo to afford the product, which was used In the next step without further purification. LCMS m/z 315.4 [M+H]*. 1H NMR (400 MHz, CDCh) δ 7.81-7.75 (m, 2H), 7.31-7.24 (m, 2H, assumed; partially obscured by solvent peak), 3.81 (br dd, J=5.1,4.7 Hz, 2H), 3.43-3.34 (m, 2H), 3.33-3.21 (m, 2H), 3.04 (br dd, J=4.9, 4.7 Hz, 2H), 2.86 (s, 2H), 2.24 (br d, J=14.4 Hz, 2H), 1.82 (ddd, J=14.8, 13.3,4.5 Hz, 2H).
Step 3. Synthesis of (2R)· 1,1,14nfluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-((4fluorophenyl)sulfonyl(- 1-oxa-4,9-diazaspiro[5.5Jundecane-9-carboxylate (C29).
Triethylamine (5.3 mL, 38 mmol) was added to a 0 'C solution of C28 (from the previous step, £7.62 mmol) in In acetonitrile (40 mL). The reaction mixture was allowed to stir at 0 ’C for a few minutes, whereupon C2 (reaction solution in acetonitrile, containing 9.9 mmol) was added 20 drop-wise. The température was maintained at 0 ’C for a few minutes, and then the reaction mixture was allowed to stir at room température for 3 days. Solvents were removed in vacuo, and the residue was purified using silica gel chromatography (Gradient: 0% to 50% ethyl acetate in heptane) to afford the product as a white foam. Yield: 3.9 g, 6.6 mmol, 87% over 2 steps. LCMS m/z 635.5 [(M + HCOOH) - H*]. ’H NMR (400 MHz, CDCh) δ 7.79-7.73 (m, 2H), 25 7.29-7.22 (m, 4H, assumed; partially obscured by solvent peak), 6.96-6.85 (m, 2H), 5.54-5.43 (m, 1H), 4.51 (AB quartet, downfield doublet Is broadened, Jabs11.7 Hz, Avab=28 Hz, 2H), 3.95-
3.64 (m, 9H), 3.26-3.13 (m, 2H), 3.08-2.89 (m, 2H), 2.85-2.65 (m, 2H), 2.00-1.87 (m, 2H), 1.55-
1.38 (m, 2H).
Step 4. Synthesis of(2R)-1,1,1-tnfluom-3-hydmxypropan-2~yl 4-[(4-fluorophenyl)sulfonyl]-1-oxa30 4,9-diaza splm[5.5]undecane-9-carboxylate (6).
Trifluoroacetic acid (25 mL) was added drop-wise to a 0 ’C solution of C29 (3.9 g, 6.6 mmol) in dichloromethane (100 mL), and the reaction mixture was allowed to warm to room température and stir for 2 hours. It was then concentrated in vacuo; the residue was dissolved In ethyl acetate, washed sequentially with saturated aqueous sodium bicarbonate solution and 35 with saturated aqueous sodium chloride solution, dried over magnésium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (Gradient: 0% to 100% ethyl acetate in heptane) provided the product as a white foam. Yield: 2.6 g, 5.5 mmol, 83%. LCMS m/z 471.5 [M+H]*. Ή NMR (400 MHz, DMSO-de) δ 7.85-7.78 (m, 2H), 7.51 (br dd, J=8.9, 8.8
Hz, 2H), 5.30-5.16 (m, 2H), 3.78-3.60 (m, 6H), 3.20-3.02 (m, 2H), 2.94-2.82 (m, 2H), 2.81-2.69 (m, 2H), 1.89-1.75 (m, 2H), 1.57-1.38 (m, 2H).
Crystal lization of 6 (1 g) was carried out using ethyl acetate (10 mL) and hexanes (20 mL), providing the product as a white solid, melting point 132 C; this material was determined to be crystalline via powder X-ray diffraction. Yield for crystallization: 826 mg, 83%. LCMS m/z
471.4 [M+H]*. Ή NMR (400 MHz, DMSO-Λ) δ 7.85-7.78 (m, 2H), 7.51 (brdd, 7=8.8, 8.7 Hz, 2H), 5.30-5.16 (m, 2H), 3.78-3.60 (m, 6H), 3.21-3.01 (m, 2H), 2.95-2.82 (m, 2H), 2.81-2.69 (m, 2H), 1.89-1.75 (m, 2H), 1.58-1.38 (m, 2H).
Example 7 (28)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 4-(phanylsu!fonyl)-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (7)
F F>AfFO CF3 fAAoAoa^opmb
F C2
NEta
O CF3 nAoA^opmb silica gel
O CF3
AoA^opmb
O CF3
Γ cf3cooh ο=έ*ο 7 ô
Çp3
Step 1. Synthesis of 4-ted-butyl 9-{(28)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl} 1oxa-4,9-diazaspiro[5.5]undecane-4,9-dicarboxylate (C30).
Triethylamlne (9.28 g, 91.7 mmol) was added to a 0 ’C solution of fert-butyl 1-oxa-4,9diazaspiro[5.5]undecane-4-carboxylate (4.70 g, 18.3 mmol) in acetonitrile (60 mL); C2 (reaction solution in acetonitrile, containing 27.5 mmol) was then added drop-wise, and the reaction mixture was stirred at 0 *C few minutes, It was then allowed to warm to 25 °C and stir for 15 hours, whereupon it was concentrated In vacuo and purified via silica gel chromatography (Gradient: 0% to 100% dichloromethane In petroleum ether). The product (11.2 g) was isolated as a yellow oil, which by LCMS analysis was impure; this material was used without additional purification. LCMS m/z 555.1 [M+Na*].
Step 2. Synthesis of (28)-1,1,1-tnfluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 1-oxa-4,9dîazasplro[5.5Jundecane-9-carboxylate (C31).
A mixture of C30 (from the previous step, 4.5 g, £7.4 mmol) and silica gel (5.0 g) was stirred at 150 *C for 3.5 hours, whereupon it was combined with a similar reaction carried out on C30 (4.5 g, £7.4 mmol) and purified via silica gel chromatography (Gradient: 0% to 8%
methanol in dichloromethane). The product was obtained as a brown oil. Yield: 2.53 g, 5.85 mmol, 40% over 2 steps. LCMS m/z 433.2 [M+H]*. Ή NMR (400 MHz, CDCh) δ 7.25 (br d, J=8.4 Hz, 2H), 6.88 (br d, J=8.5 Hz, 2H), 5.54-5.43 (br m, 1H), 4.51 (AB quartet, Jab=1 1.7 Hz, Avab=27.5 Hz, 2H). 3.95-3.79 (m, 2H), 3.81 (s, 3H), 3.79-3.63 (m, 4H), 3.30-3.14 (m, 2H), 2.86 5 (dd, J=4.8, 4.5 Hz, 2H), 2.73-2.62 (m, 2H), 2.10-1.91 (m, 2H), 1.50-1.29 (m, 2H).
Step 3. Synthesis of(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(phenylsulfonyl)~ 1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate (C32).
Benzenesulfonyl chloride (61.3 mg, 0.347 mmol) was added to a 5 °C solution of C31 (100 mg, 0.23 mmol) in saturated aqueous sodium bicarbonate solution (2 mL) and dichloromethane (5 mL), and the reaction mixture was stirred at 5 ’C for 16 hours. The aqueous layer was extracted with dichloromethane, and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Préparative thin layer chromatography on silica gel (Eluent: 3:1 petroleum ether / ethyl acetate) provided the product as a colorless gum. Yield: 116 mg, 0.203 mmol, 88%. LCMS m/z 594.9 [M+Na*].
Step 4. Synthesis of (2R)-1,1,1-trifluoro-3~hydmxypmpan-2~yl 4-(phenylsulfonyl)-1 -oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (7).
To a solution of C32 (203 mg, 0.354 mmol) in dichloromethane (10 mL) was added trifluoroacetic acid (2 mL, 30 mmol) and the reaction mixture was stirred at 25 ’C for 10 minutes. The reaction was quenched via addition of saturated aqueous sodium bicarbonate solution to a 20 pH of -8, and the resulting mixture was extracted with dichloromethane (2 x 20 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via reversed phase HPLC (Column: Phenomenex Luna C18; Mobile phase A: 0.225% formic acid In water; Mobile phase B; acetonitrile; Gradient: 40% to 60% B) afforded the product as a white solid. Yield: 101 mg, 0.224 mmol, 63%. LCMS m/z 452.9 [M+H]*. 1H NMR 25 (400 MHz, CDCIj) δ 7.78-7.72 (m, 2H), 7.68-7.62 (m, 1 H), 7.61-7.55 (m, 2H), 5.32-5.20 (br m,
1H), 4.05-3.95 (br m, 1H), 3.95-3.8 (m, 3H), 3.79 (dd, J=5.1,4.8 Hz, 2H), 3.32-3.13 (m, 2H), 3.10-2.92 (br m, 2H), 2.90-2.72 (m, 2H), 2.34-2.22 (br m, 1H), 2.04-1.90 (m, 2H), 1.6-1.44 (m, 2H, assumed; partially obscured by water peak).
Example 8 and 9
1,1-Tnfluom-3-hydroxypropan-2-yl (3S)-3-[(phenylsulfony!)amino]-1-oxa-8azaspiro[4.5]decane-8-cait>oxylate (8) and (2R)-1,1,1'Tnfluoro-3-hydroxypropan-2-yl (3R)-3[(phenylsulfony!)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (9)
H σ
o cf3
ΑθΛ^ΟΡΜΒ
C34 «
ό
Âq^oh
HN ° »
Step 1. Synthesis of tert-butyl 3-[(phenylsulfonyl)amino}-1-oxa-8-azaspiro[4.5]decane-8· carboxylate (C33).
fert-Butyl 3-amino-1 -oxa-8-azaspiro[4.5]decane-8-carboxylate was converted to the product using the method described for synthesis of C32 from C31 in Example 7. The product was isolated as a colorie s s gum. Yield: 200 mg, 0.504 mmol, 65%. LCMS m/z 296.8 I(M-BOC)+H]*. 1H NMR (400 MHz, CDCh) δ 7.91-7.85 (m, 2H), 7.65-7.58 (m, 1H), 7.58-7.51 (m, 2H), 4.82 (br d, >8 Hz, 1H), 4.00-3.90 (m, 1 H), 3.82 (dd, >9.6, 5.7 Hz, 1H), 3.60-3.48 (m,
3H), 3.31-3.19 (m, 2H), 1.97 (dd, >13.3,7.6 Hz, 1H), 1.63-1.48 (m, 5H, assumed; partially obscured by water peak), 1,44 (s, 9H).
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxyJpropan-2-yl 3[(phcnylsu!fony!)amino]-1-oxa-8-azaspÎro[t.5]dccano-8-carboxylato (C34).
Trifluoroacetic acid (2 mL) was added to a solution of C33 (200 mg, 0.504 mmol) In dichloromethane (5 mL), and the reaction mixture was stirred at 25 *C for 1 hour. Removal of solvents in vacuo provided N-(1-oxa-8-azaspiro[4.5]dec-3-yl)benzenesulfonamide, trifluoroacetic acid sait, as a colorfess gum, LCMS m/z 297.0 [M+H]*. This matériel was dissolved In acetonitrile (5 mL), cooled to 0 *C, and treated with triethylamine (153 mg, 1,51 mmol). After this
solution had stirred at 0 °C for a few minutes, C2 (reaction solution in acetonitrile containing 0.755 mmol) was added drop-wise, and stirring was continued at 0 ’C for 30 minutes. The réaction mixture was then allowed to warm to 25 ’C and stir for 18 hours, whereupon it was concentrated under reduced pressure. Silica gel chromatography (Gradient: 1% to 34% ethyl acetate in petroleum) afforded the product as a colorless gum. Yield: 180 mg, 0.314 mmol, 62%.
LCMS m/z 595.1 [M+Na*]. ’H NMR (400 MHz, CDCh) δ 7.88 (br d, J=7 Hz, 2H), 7.65-7.60 (m,
1H), 7.59-7.52 (m, 2H), 7.23 (br d, J=8 Hz, 2H), 6.88 (br d, J=8 Hz, 2H), 5.52-5.40 (m, 1 H),
4.64-4.58 (m, 1H), 4.50 (AB quartet, Jab=11.3 Hz, ΔνΑΒ=28 Hz, 2H), 4.01-3.91 (m, 1H), 3.82 (s, 3H), 3.88-3.78 (m, 1H), 3.78-3.62 (m, 4H), 3.59-3.47 (m, 1H), 3.36-3.21 (m, 2H), 2.02-1.91 (m, 10 1H), 1.72-1.38 (m, 5H, assumed; partially obscured by water peak).
Step 3. Synthesis of(2R)-1,1,1-trifJuoro-3-hydroxypropan-2-yl 3-[(phenylsu!fonyl)amino]-1-oxa8-azaspiro[4.5]decane-8-carboxylate (C35).
Trifluoroacetic acid (2 mL) was added to a 0 ’C solution of C34 (180 mg, 0.314 mmol) in dichloromethane (8 mL) and the réaction mixture was stirred at 0 ’C for 30 minutes, whereupon 15 it was treated with saturated aqueous sodium bicarbonate solution until the pH was above 7.
The aqueous layer was extracted with ethyl acetate (5x5 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated In vacuo. Préparative thin layer chromatography on silica gel (Eluent: 1:1 petroleum ether/ethyl acetate) provided a diastereomeric mixture of the product as a colorless oil. Yield: 130 mg, 0.287 mmol, 91%.
Step 4. Isolation of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3S)-3-[(phenylsulfonyl)amlnoJ~1oxa-8-azaspiro[4.5]decane-8-carboxylate (8) and (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3R)~
3-[(phenylsulfonyl)amino]-1-oxa-8-azasplro[4.5]decane-8-carboxylate (9).
Compound C35 (130 mg. 0.287 mmol) was separated Into its component diastereomers via supercrltical fluid chromatography (Column: Chiral Technologies Chiralpak AD, 5 pm; Mobile 25 phase: 3:7 2-propanol / carbon dioxide). The first-eluting diastereomer was further purified by préparative thin layer chromatography on silica gel (Eluent: 1:1 petroleum ether / ethyl acetate) to afford 8 as a colorless gum. Yield for the séparation: 62.0 mg, 0.137 mmol, 48%. LCMS m/z
474.8 [M+Na*]. 1H NMR (400 MHz, CDCh) δ 7.91-7.85 (m, 2H), 7.66-7.59 (m, 1H), 7.59-7.52 (m, 2H), 5.30-5.18 (br m, 1H), 4.89-4.77 (br m, 1 H). 4.03-3.90 (m, 2H), 3.90-3.64 (m, 4H), 3.5830 3.50 (m, 1 H), 3.39-3.19 (m, 2H), 1.99 (dd, J=13.6,7.6 Hz, 1 H), 1.75-1.44 (m, 5H, assumed;
partially obscured by water peak).
The second-elutîng diastereomer was 9, also isolated as a colorless gum. Yield for the séparation: 67.0 mg, 0.148 mmol, 52%. LCMS m/z 475.1 [M+Na*]. 1H NMR (400 MHz, CDCb) δ 7.91-7.85 (m, 2H), 7.66-7.59 (m, 1H), 7.59-7.52 (m, 2H), 5.29-5.18 (m, 1H), 4.87-4.79 (m, 1H), 35 4.04-3.90 (m, 2H), 3.90-3.79 (m, 2H), 3.79-3.66 (m, 2H), 3.58-3.50 (m, 1H), 3.41-3.21 (m, 2H),
2.05-1.93 (m, 1H), 1.75-1.39 (m, 5H, assumed; partially obscured by water peak).
The absolute configurations Indicated for 8 and 9 were established by relation to the Xray crystal structure détermination of C48 (see Example 15) in the following mannen C48 and its enantiomer C49 were converted to samples of the general structure of 8 and 9 using the methods described in this Example. Supercritical fluid chromatography (Column: Chiral Technologies Chiralpak AD, 5 um; Mobile phase A: carbon dioxide; Mobile phase B: 2-propanol; Gradient: 5% to 60% B) provided a clear corrélation between the material derived from C48 and 9 (rétention times 7.44 and 7.45 minutes). Ltkewise, the material derived from C49 exhibited a very similar rétention time to that of 8 (6.86 and 6.87 minutes).
Example 10 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 4-[(5-cyclopropylpyridin-2-yl)oxy]piperidine-1 carboxylate (10)
1)CF3COOH
Step 1. Synthesis oftert-butyl4-[(5-cyclopropylpyridin-2-yl)oxy]piperidine-1-carboxylate (C36).
Potassium fert-butoxide (913 mg, 8.14 mmol) was added to a solution of fert-butyl 4hydroxypiperidine-1-carboxylate (983 mg, 4.88 mmol) in N,N-dimethylformamide (30 mL) and the reaction mixture was heated at 50 ’C for 2 hours. 2-Chloro-5-cyclopropylpyridine (250 mg,
1.63 mmol) was then added, and the reaction mixture was stirred at 100 ’C for 18 hours. After solvent had been removed In vacuo, the residue was diluted with water (50 mL) and extracted with ethyl acetate (3 x 50 mL); the combined organic layers were concentrated under reduced pressure. Chromatography on silica gel (Gradient: 0% to 10% ethyl acetate in petroleum ether) afforded the product as a white solid. Yield: 120 mg, 0.377 mmol, 23%. ’H NMR (400 MHz, CDCh) 5 7.97-7.93 (m, 1H), 7.3-7.21 (m, 1H, assumed; partially obscured by solvent peak), 6.62 (d, >8.4 Hz, 1H), 5.22-5.13 (m, 1H), 3.82-3.72 (m, 2H), 3.34-3.24 (m, 2H), 2.02-1.92 (m, 2H), 1.88-1.78 (m, 1 H), 1.77-1.65 (m, 2H), 1.48 (s, 9H), 0.97-0.90 (m, 2H), 0.65-0.59 (m, 2H).
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-mcthoxybonzy!)oxy]propan-2-yl 4-[(5cyc!opropy!pyridin-2-yl)oxy]piperidine-1 -carboxylate (C37).
Conversion of C36 to C37 was carried out using the method described for synthesis of C34 from C33 in Examples 8 and 9. The product was isolated as a colorless gum. Yield: 120 mg, 0.243 mmol, 64%.
’H NMR (400 MHz, CDCh) of Intermediate 5-cyclopropyl-2-(piperidin-4-yloxy)pyridine, trifluoroacetic acid sait, characteristic peaks: δ 8.07-8.03 (m, 1H), 7.79 (br d, J=8 Hz, 1H), 7.07 (d, J=9 Hz, 1H), 3.60-3.45 (m, 2H), 3.43-3.32 (m, 2H), 2.46-2.34 (m, 2H), 2.24-2.13 (m, 2H), 2.01-1.91 (m, 1H), 1,17-1.09 (m, 2H), 0.79-0.72 (m, 2H).
Compound C37: LCMS m/z 517.0 [M+Na*]. ’H NMR (400 MHz, CDCh) δ 7.97-7.92 (m, 1H), 7.30-7.22 (m, 3H, assumed; partially obscured by solvent peak), 6.89 (d. J=8.5 Hz, 2H),
6.64 (d, J=8.4 Hz, 1H), 5.55-5.44 (m, 1H), 5.24-5.15 (m, 1 H). 4.52 (AB quartet, Jab=11.5 Hz, ûvab=26.5 Hz, 2H), 3.82 (s, 3H), 3.8-3.66 (m, 4H), 3.52-3.39 (m, 2H), 2.06-1.90 (m, 2H), 1.89
1.70 (m, 3H), 0.98-0.91 (m, 2H), 0.66-0.59 (m, 2H).
Step 3. Synthesis of(2R)-1,1,1-tnfluoro-3-hydroxypropan-2-yl 4-[(5-cycloprOpylpyridin-2yl)oxy]piperidine· 1-carboxyla te (10).
Trifluoroacetic acid (5 mL) was added drop-wise to a solution of C37 (120 mg, 0.243 mmol) in dichloromethane (15 mL), and the reaction mixture was stined at 30 *C for 2 hours, whereupon It was concentrated In vacuo and diluted with ethyl acetate (20 mL). The resulting mixture was poured Into saturated aqueous sodium bicarbonate solution (20 mL) and extracted with ethyl acetate (3 x 20 mL); the combined organic layers were concentrated under reduced pressure. Préparative thin layer chromatography on silica gel (Eluent: 1:1 petroleum ether: ethyl acetate) provided the product as a colorless gum. Yield: 70 mg, 0.19 mmol, 78%. LCMS m/z 375.0 [M+H]*. Ή NMR (400 MHz, CDCh) δ 7.94 (d, J=2.1 Hz, 1H), 7.25 (dd, J=8.5, 2.4 Hz, 1H), 6.63(d, J=8.5 Hz, 1H), 5.32-5.19 (m, 2H), 4.01 (br d, halfof ABquartet, J=12 Hz, 1 H), 3.88 (dd, halfof ABX pattern, J=12,7 Hz, 1H), 3.87-3.70 (m, 2H), 3.57-3.40 (m, 2H), 2.52-2.40 (brs, 1H), 2.07-1.93 (m, 2H), 1.89-1.74 (m, 3H), 0.98-0.91 (m, 2H), 0.66-0.59 (m, 2H).
Example 11 (2R)-1,1, l-Trifluoro-3-hydmxypropan-2-yf 4-[(3-fluorOphenyl)sulfonyl]-1-oxa-4,9· diazaspiro[5.5Jundecane-9-carboxyla te (11)
O o=s-ci
H
C31
O CF3
ΑθΑ^ΟΡΜΒ
O CF3
AoA^opmb cf3cooh
O CF3
AoACoh
NaHCOj
Sfep 1. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-((3fluorOphenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undecane-9-cart)Oxylate (C38).
Conversion of C31 to the product was carried out using the method described for synthesis of C32 from C31 In Example 7, providing C38 as a colorless gum. Yield: 130 mg,
0.220 mmol, 79%. LCMS m/z 612.9 [M+Na*].
Sfep 2. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(3-fluorophenyl)sulfonyl]-1-oxa-
4,9-diazasplro[5.5]undecane-9-carboxyla te (11).
Trifluoroacetic acid (2 mL, 30 mmol) was added to a solution of C38 (190 mg, 0.322 mmol) In dichloromethane (10 mL) and the reaction mixture was stirred at 25 C for 10 minutes, whereupon it was treated with saturated aqueous sodium bicarbonate solution to a pH of -8. The mixture was extracted with dichloromethane (2 x 20 mL), and the combined organic layers 5 were dried over sodium sulfate, filtered, and concentrated in vacuo. Purification via reversed phase HPLC (Column: Phenomenex Luna C18; Mobile phase A: 0.225% formtc acid in water; Mobile phase B: acetonitrile; Gradient: 43% to 63% B) afforded the product as a white solid. Yield: 93.4 mg, 0.198 mmol, 61%. LCMS m/z 470.9 [M+H]*. 1H NMR (400 MHz, CDCh) δ 7.617.52 (m, 2H), 7.48-7.43 (m, 1H), 7.39-7.32 (m, 1 H>, 5.31-5.20 (m. 1H), 4.06-3.96 (m, 1H). 3.9510 3.83 (m, 3H), 3.80 (dd, 7=5.0, 4.9 Hz, 2H), 3.32-3.14 (m. 2H), 3.11-2.95 (m, 2H), 2.91-2.75 (m,
2H), 2.33-2.23 (m, 1H), 2.05-1.92 (m, 2H), 1.6-1.45 (m, 2H, assumed; partially obscured by water peak).
Example 12 (2R)-1,1,1-Trifluoro-3-hydroxypropan~2-yi 2-[(4-fluorophenyl)sulfonyl]-2,915 diazaspiro[5.5]undecane-9-carboxylate (12)
NaHCO3
O
:i A ~ çp >Vo 039
CF3COOH
Ο=έ=Ο C43
O CF3
ΑθΑ^ΟΗ
Step 1. Synthesis of 9-benzyl2-tert-butyl 2,9-diazaspiro[5.5]undecane-2,9-dicarboxylate (C39).
Saturated aqueous sodium bicarbonate solution (5 mL) and benzyl chloroformate (161 mg, 0.944 mmol) were added to a 0 ’C solution of tert-butyl 2,9-diazaspiro[5.5]undecane-2carboxylate (200 mg, 0.786 mmol) in ethyl acetate (5 mL), and the reaction mixture was stirred for 18 hours at 30 ’C. The aqueous layer was extracted with ethyl acetate (2x5 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo; silica gel chromatography (Gradient: 0% to 20% ethyl acetate in petroleum ether) provided the product as an oil. Yield: 235 mg, 0.605 mmol, 77%. Ή NMR (400 MHz, CDCh) δ 7.41-7.29 (m, 5H), 5.13 (s, 2H), 3.73-3.60 (m, 2H), 3.48-3.19 (m, 6H), 1.60-1.50 (m, 2H), 1.50-1.28 (m, 6H), 1.45 (m, 9H).
Step 2. Synthesis of benzyl 2,9-diazaspiro[5.5]undecane-9-carboxylate (C40).
Trifluoroacetic acid (3 mL) was added to a solution of C39 (235 mg, 0.605 mmol) in dichloromethane (5 mL) and the reaction mixture was stirred for 30 minutes at room température. After removal of solvents in vacuo, the residue was taken up In aqueous sodium bicarbonate solution (20 mL) and extracted with dichloromethane (3 x 20 mL). The combined 15 organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the product as a gum. Yield: 116 mg, 0.402 mmol, 66%. LCMS m/z 289.1 [M+H]+.
Step 3. Synthesis of benzyl 2-{(4-fluorophenyl)sulfonyl]-2,9-diazaspiro[5.5]undecane-9carboxylate (C41).
4-Fluorobenzenesulfonyl chloride (117 mg, 0.601 mmol) was added to a solution of C40 20 (116 mg, 0.402 mmol) in pyridine (2 mL) and the reaction mixture was stirred for 18 hours at 30 ’C, whereupon It was concentrated in vacuo. The residue was partitioned between dichloromethane (20 mL) and saturated aqueous sodium bicarbonate solution (20 mL), and the organic layer was concentrated under reduced pressure. Silica gel chromatography (Gradient: 0% to 25% ethyl acetate in petroleum ether) provided the product as a gum. Yield: 140 mg, 25 0.314 mmol, 78%. LCMS m/z 446.9 [M+H]*. ’H NMR (400 MHz, CDCh) δ 7.81-7.73 (m, 2H),
7.41-7.29 (m, 5H). 7.22 (dd, J=8.8, 8.4 Hz, 2H), 5.14 (s, 2H), 3.64-3,54 (m, 2H), 3.44-3.32 (m, 2H), 3.22-3.04 (m, 1 H). 3.04-2,80 (m, 2H), 2.80-2.60 (m, 1 H). 1.77-1.65 (m, 2H), 1.65-1.5 (m, 2H, assumed; obscured by water peak), 1.44 (ddd, J=14, 9,4 Hz, 2H), 1.39-1.29 (m, 2H). Step 4. Synthesis of 2-{(4-fluorophenyl)sulfonylJ-2,9-diazaspiro[5.5]undecane (C42).
To a solution of C41 (60.0 mg, 0.134 mmol) in tetrahydrofuran (10 mL) was added 10% palladium on carbon (14.3 mg, 13.4 pmol), and the mixture was stirred under a hydrogen atmosphère (45 psi) for 18 hours at 50 ’C. After filtration of the reaction mixture, the filter cake was washed with methanol (20 mL); the combined filtrâtes were concentrated in vacuo to afford the product as a coloriess gum. Yield: 42.0 mg, 0.134 mmol, 100%. LCMS m/z 312.9 [M+H]*.
Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 2-((4fluorophenyl)sulfonyl]-2,9-diazaspiro[5.5]undecane-9-carboxylate (C43).
Conversion of C42 to the product was effected using the method described for synthesis of C30 In Example 7. In this case, purification was carried out via préparative thin layer chromatography on silica ge! (Eluent: 3:1 petroleum ether / ethyl acetate) to afford the product as a gum. Yield: 55 mg, 93 pmol, 35%. LCMS m/z 611.0 [M+Na*].
Step 6. Synthesis of(2R)-1,1,1-tnfluoro-3-hydroxypropan-2-yl 2-[(4~fluorophenyl)sutfonyl]-2,9~ diazaspiro[5.5]undecane~9~carboxy!ate (12).
Conversion of C43 to the product was carried out using the method described for synthesis of 11 from C38 in Example 11, except that the reaction was carried out at 0 “C. Purification was effected via préparative thin layer chromatography on silica gel (Eluent: 9:1 dichloromethane / methanol) to provide the product as a white solid. Yield: 13 mg, 28 pmol, 30%. LCMS m/z 491.1 [M+Na*]. 1H NMR (400 MHz, CDC!3) δ 7.77 (br dd, >8.5, 5.0 Hz, 2H),
7.23 (dd, >8.5, 8.3 Hz, 2H), 5.32-5.20 (m, 1H), 4.05-3.95 (m, 1H), 3.92-3.81 (m, 1H), 3.69-3.53 (m, 2H), 3.50-3.31 (m, 2H), 3.16-3.02 (m, 1H), 3.01-2.84 (m, 2H), 2.81-2.69 (m, 1H). 1.80-1.54 (m, 5H), 1.54-1.42 (m, 2H), 1.41-1.31 (m, 2H).
Example 13 (2R)-1,1,1-Tnfluom-3-hydroxypropan-2-yl (3aR,6aS)-5-[(3,4difluorophanyl)sulfonyl]hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxy!ate (13)
O O=S-CI
HN
Step 1. Synthesis ofted-butyl (3aR,6aS)-5-[(3,4-difJuorophenyl)sulfonyl]hexahydropyrrOlo[3,4· c]pyrrole-2(1H)-carboxylate (C44).
ferf-Butyl (3aR,6aS)-hexahydropyrrolo[3,4-c]pyiTo!e-2(1H)-carboxylate was converted to C44 using the method described for synthesis of C32 from C31 in Example 7. The product was obtained as a white solid. Yield: 100 mg, 0.257 mmol, 68%. LCMS m/z 410.9 [M+Na*]. 1H NMR (400 MHz, CDCh) δ 7.68 (ddd, >9, 7, 2 Hz, 1H), 7.64-7.59 (m, 1H), 7.36 (ddd, >9, 9, 7 Hz, 1H), 3.57-3.48 (m, 2H), 3.48-3.39 (m, 2H), 3.20-2.98 (m, 4H), 2.89-2.80 (m, 2H), 1.44 (s, 9H). Step 2. Synthesis of (2R)-1,1,1-trifJuorO-3-[(4-methoxybenzyl)oxy]propan-2-yl (3aR,6aS)-5-[(3,4difluorophenyl)su!fony!]hexahydropytTolo[3,4-c]pyrTole-2(1R)-carboxylate (C45).
Conversion of C44 to C45 was effected using the method described for synthesis of C34 from C33 in Examples 8 and 9.1H NMR (400 MHz, CD3OD) of intermediate (3a/?,6aS)-2-[(3,4difluorophenyl)sulfonyl]octahydropyrrolo[3,4-c]pyrrole, trifluoroacetic acid sait, δ 7.80 (ddd, J=9.7, 7.3, 2.2 Hz, 1H). 7.72-7.67 (m, 1H), 7.58 (ddd, J=10.0, 8.7, 7.5 Hz, 1H), 3.60-3.53 (m,
2H), 3.38-3.33 (m, 2H), 3.13-3.07 (m, 2H), 3.07-2.96 (m, 4H). In this case, purification was camed out via préparative thin layer chromatography on silica gel (Eluent: 2:1 petroleum ether / ethyl acetate) to afford C45 as a colorless gum. By Ή NMR analysis, this was judged to be a mixture of rotamers. Yield: 100 mg, 0.18 mmol, 69%. LCMS m/z 587.0 [M+Na*]. Ή NMR (400 MHz, CDCb) δ 7.71-7.64 (m, 1H), 7.64-7.57 (m, 1H), 7.39-7.31 (m, 1H). 7.28-7.20 (m, 2H, assumed; partially obscured by solvent peak), 6.94-6.85 (m, 2H), 5.47-5.37 (m, 1H), 4.58-4.41 (m, 2H), [3.83 (s) and 3.81 (s), total 3H], 3.77-3.55 (m, 4H), 3.55-3.35 (m, 2H), 3.29-3.05 (m, 4H), 2.95-2.84 (m, 2H).
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydmxypmpan-2-yl (3aR,6aS)-5-[(3,4difluomphenyl)sulfon^]hexahydropyrrolo[3,4-c]pyrrole-2(1H)-carboxylate (13).
Conversion of C45 to 13 was carried out using the method described for synthesis of
C35 from C34 in Examples 8 and 9. The product was Isolated as a colorless oil. Yield: 40 mg, 90 pmol, 50%. LCMS m/z 445.0 [M+H]*. ’H NMR (400 MHz, CDCb) δ 7.68 (br dd, J=9.0, 7.3 Hz, 1H), 7.64-7.58 (m, 1H), 7.42-7.31 (m, 1H), 5.29-5.18 (m, 1H), 4.03-3.93 (m, 1H), 3.90-3.79 (m, 1H), 3.74-3.58 (m. 2H), 3.52-3.42 (m, 1H), 3.42-3.07 (m, 5H), 2.99-2.84 (m. 2H).
Example 14 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-y! 4-(5-fluoropyhdin-2-yl)-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (14)
Step 1. Synthesis of tert-butyl 4-(5-fluoropyridin-2-yl)-1-oxa-4,9-diazaspiro[5.5]undocane-9carboxylate (C46).
A mixture of C23 (100 mg, 0.39 mmol), 2-chloro-5-fluoropyridine (103 mg, 0.783 mmol), 5 [1,3-bis(2,6-diisopropylphenyl)imidazol-2-ylidene](3-chloropyridyl)palladium(ll) dichloride (26.6 mg, 39.1 Mmol), and césium carbonate (381 mg. 1.17 mmol) in toluene (10 mL) was heated at 120 *C for 3 days. The reaction mixture was then filtered and the filtrate was concentrated in vacuo; silica gel chromatography (Gradient: 0% to 10% methanol In dichloromethane) afforded the product as a brown gum. Yield: 135 mg, 0.384 mmol, 98%. LCMS m/z 352.3 [M+H]*. ’H 10 NMR (400 MHz, CDCh) δ 8.03 (d, >3.0 Hz, 1H), 7.26 (ddd, >9.2, 7.7, 3.1 Hz, 1H), 6.57 (dd, >9.3, 3.3 Hz, 1H), 3.83 (dd, >6.0,4.1 Hz, 2H), 3.8-3.65 (m, 2H), 3.42 (dd, >5.4, 4.8 Hz, 2H), 3.33 (s, 2H), 3.19 (brdd, >12, 12 Hz, 2H), 1.91 (brd. >13 Hz, 2H), 1.56-1.45 (m, 2H), 1.46 (s, 9H).
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(5-fluoropyridin15 2-yl)-1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate (C47)
Conversion of C46 to C47 was carried out using the method described for synthesis of C34 from C33 in Examples 8 and 9. LCMS of Intermediate 4-(5-fluoropyridin-2-yl)-1-oxa-4,9diazaspiro[5.5]undecane, bis(trifluoroacetic acid) sait, m/z 252.t [M+H]*. In this case, purification was carried out using préparative thin layer chromatography (Eluent: 3:1 petroleum 20 ether / ethyl acetate) to afford C47 as a light yellow gum. Yield: 70 mg, 0.13 mmol, 68%. LCMS m/z 528.2 [M+H]*.
100
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(5-fluoropyridin-2-yl)-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (14).
Trifluoroacetic acid (1 mL) was added to a 0 ’C solution of C47 (70 mg, 0.13 mmol) In dichloromethane (5 mL), and the reaction mixture was stirred at 25 'C for 1 hour. Solvents were removed in vacuo, and the residue was subjected to préparative thin layer chromatography on silica gel (Eluent: 2:3 petroleum ether / ethyl acetate). Further purification using reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.225% formic acid In water; Mobile phase B: 0.225% formic acid in acetonitrile; Gradient: 38% to 58% B) provided the product as a coloriess gum. Yield: 33.4 mg, 82.0 pmol, 63%. LCMS m/z 408.1 [M+H]*. 1H NMR (400 MHz, CDCh) δ 8.04 (d, 3=2.8 Hz, 1H), 7.31-7.23 (m, 1H, assumed; partially obscured by solvent peak), 6.59 (dd, 3=9.2, 3.1 Hz, 1H), 5.32-5.20 (m, 1H), 4.06-3.77 (m, 6H), 3.49-3.39 (m, 2H), 3.39-3.19 (m, 4H), 2.68-2.38 (br s. 1H), 2.08-1.92 (m, 2H), 1.62-1.48 (m, 2H).
Example 15 (2R)-1,1,1-Trifluoro-3-hydroxyproparh2-yl (3R)-3-[methyl(phenylsulfonyl)amino]-1-Oxa-8~ azaspiro[4.5]decane-8-carboxylate (15)
O
OS-CI ô
NaHCO3
CF3COOH
1)CF3COOH,_ FO CF3 ΟΛΟΛ^ΟΡΜΒ
C2 NEt3 O CF3
ΑθΑ^ΟΡΜΒ
Step 1. Synthesis ofted-butyl (3R)-3-[(phenylsulfonyl)amino]-1oxa-8-azaspiro[4.5]decane-8carboxylate (C48) and tert-butyl (3S)-3-[(phenylsulfonyl)amino]~1-oxa-8-azaspiro[4.5]decane-8carboxylate (C49).
101
Reaction of fert-butyl 3-amino-1-oxa-8-azaspiro[4.5]decane-8-carboxylate with benzenesulfonyl chloride was carried out as described for synthesis of C32 from C31 in Example 7. The racemic product was purified using silica gel chromatography (Gradient: 20% to 50% ethyl acetate in heptane) to afford a white solid (2.88 g), which was then separated into its 5 component enantiomers via supercritical fluid chromatography [Column: Phenomenex Lux Cellulose-3, 5 pm; Eluent: 7.5% (1:1 methanol / acetonitrile) in carbon dioxide]. The first-eluting product. obtained as a tacky white solid that exhibited a négative (-) rotation, was designated as C48. Yield: 1.35 g, 3.40 mmol, 45%. LCMS m/z 395.5 [M-H*]. 1H NMR (400 MHz, CDCh) δ 7.90-7.86 (m, 2H), 7.64-7.59 (m, 1H), 7.57-7.52 (m, 2H). 4.81 (d, J-7.9 Hz, 1H), 4.00-3.91 (m, 10 1H), 3.81 (dd. J=9.7, 5.7 Hz, 1H), 3.59-3.48 (m, 3H), 3.30-3.19 (m, 2H), 1.97 (dd, J-13.4, 7.7
Hz, 1H), 1.67-1.49 (m, 4H), 1.48-1.38 (m, 1H), 1.44 (s, 9H).
The second-eluting product, obtained as a tacky white solid that exhibited a positive (+) rotation, was designated as C49. Yield: 1.15 g, 2.90 mmol, 38%. LCMS m/z 395.5 [M-H*]. 1H NMR (400 MHz, CDCb) δ 7.90-7.86 (m, 2H), 7.64-7.59 (m, 1H), 7.57-7.52 (m, 2H), 4.79 (d, 15 J=8.0 Hz, 1H), 4.00-3.91 (m, 1H), 3.81 (dd, J=9.7, 5.7 Hz, 1H), 3.59-3.48 (m, 3H), 3.30-3.19 (m,
2H), 1.97 (dd, J=13.4, 7.7 Hz, 1 H), 1.67-1.49 (m, 4H), 1.47-1.38 (m, 1 H), 1.44 (s, 9H).
The absolute configurations shown were established as follows: a portion of this batch of C48 was recrystallized from dichloromethane / fert-butyl methyl ether, and its absolute configuration was determined via single crystal X-ray structure détermination:
Single-crystal X-ray structural détermination of C48
Data collection was performed on a Bruker APEX diffractometer at room température. Data collection consisted of oméga and phi scans.
The structure was solved by direct methods using SHELX software suite in the space group P2i2i2i. The structure was subsequently refined by the full-matrix least squares method. 25 Ail non-hydrogen atoms were found and refined using anisotropic displacement parameters.
The hydrogen atom located on nitrogen was found from the Fourier différence map and refined with distances restrained. The remaining hydrogen atoms were placed in calculated positions and were allowed to ride on their carrier atoms. The final refinement included isotropie displacement parameters for ail hydrogen atoms.
Analysis of the absolute structure using likelihood methods (Hooft, 2008) was performed using PLATON (Spek, 2010). The results indicate that the absolute structure has been correctly assigned. The method calculâtes that the probability that the structure is correct is 100.0. The Hooft parameter is reported as 0.015 with an esd of 0.09.
The final R-index was 4.2%. A final différence Fourier revealed no missing or misplaced 35 électron density.
Pertinent crystal, data collection and refinement information is summarized in Table 1. Atomic coordinates, bond lengths, bond angles, and displacement parameters are listed in Tables 2-5.
102 >
Software and References
SHELXTL, Version 5.1, Broker AXS, 1997.
PLATON, A. L. Spek, J. Appl. Cryst. 2003,36, 7-13.
MERCURY, C. F. Macrae, P. R. Edington, P. McCabe, E. Pidcock, G. P. Shields, R. Taylor, 5 M. Towler, and J. van de Streek, J. Appl. Cryst. 2006,39,453-457.
OLEX2,0. V. Dolomanov, L. J. Bourhîs, R. J. Gildea, J. A. K. Howard, and H. Puschmann, J. Appl. Cryst. 2009, 42, 339-341.
R. W. W. Hooft, L. H. Straver, and A. L. Spek, J. Appl. Cryst. 2008, 41, 96-103.
H. D. Flack, Acta Cryst. 1983, A39,867-881.
Table 1. Crystal data and structure refînement for C48.
15 Empirical formula C19H2SN2O5S
Formula weight 396.50
Température 276(2) K
Wavelength 1.54178 A
Crystal system Orthorhombîc
20 Space group P2i2i2i
Unit cell dimensions a = 9.79150(10) A a = 90' b= 11.11580(10) A β = 90' c=18.6694(2) A y = 90'
Volume 2031.98(4) A3
25 Z 4
Density (calculated) 1.296 Mg/m3
Absorption coefficient 1.686 mm-'
F(000) 848
Crystal slze 0.260x0.180x0.140 mm3
30 Thêta range for data collection 4.630 to 68.568’
Index ranges -11<=h<=11,-13<=k<=13, -20<=l<=22
Reflections collected 9404
Independent reflections 3633 [R(int) = 0.024η
35 Completeness to thêta = 70.31e 99.3 %
Absorption correction None
Refînement method Full-matrix least-squares on F2
Data / restraints / parameters 3633/1 /251
Goodness-of-fit on F2
Final R Indices [l>2sigma(t)J R Indices (all data) Absolute structure parameter 5 Extinction coefficient
Largest diff. peak and hole
103
1.067
R1 = 0.0418, wR2 = 0.1074
R1 = 0.0441, wR2 = 0.1098 0.017(9) n/a
0.428 and -0.457 e.A’3
Table 2. Atomic coordinates (x 10*) and équivalent isotropie displacement parameters (A2 x 103) forC48. U(eq) Is defined asone-third ofthetrace ofthe orthogonalized Ufitensor.
x y z U(eq)
S(1) -3733(1) 10920(1) 849(1) 53(1)
15 N(1) -3045(3) 9602(2) 839(2) 59(1)
N(2) 3033(2) 7292(2) 1366(2) 52(1)
0(1) -5113(3) 10761(2) 1075(1) 74(1)
0(2) -2848(3) 11724(2) 1218(1) 68(1)
0(3) 29(3) 8787(2) 1780(1) 68(1)
20 0(4) 5295(2) 7383(2) 1100(1) 53(1)
0(5) 4386(2) 5806(2) 1709(1) 55(1)
C(1) -4868(3) 11071(3) -483(2) 63(1)
C(2) -4920(4) 11465(4) -1195(2) 76(1)
C(3) -3910(5) 12188(4) -1452(2) 77(1)
25 C(4) -2853(5) 12532(4) -1029(2) 80(1)
C(5) -2775(3) 12136(3) -315(2) 64(1)
0(6) -3796(3) 11406(2) -54(2) 49(1)
C(7) -1575(3) 9468(3) 927(2) 49(1)
C(8) -1069(4) 9583(4) 1697(2) 77(1)
30 C(9) 248(3) 8100(3) 1135(2) 48(1)
C(10) -1087(3) 8216(3) 724(2) 51(1)
C(11) 601(3) 6821(3) 1356(2) 62(1)
0(12) 1914(4) 6735(3) 1772(2) 67(1)
C(13) 2776(3) 8526(3) 1137(2) 55(1)
35 C(14) 1463(3) 8609(3) 722(2) 49(1)
C(15) 4329(3) 6873(2) 1372(2) 46(1)
C(16) 5650(3) 5100(3) 1749(2) 50(1)
C(17) 6713(4) 5783(4) 2169(2) 69(1)
6126(5)
0(19) 5191(4)
104
4758(4) 1005(2) 82(1)
3991(3) 2158(2) 62(1)
Table 3. Bond lengths [A] and angles [’] for C48.
S(1 )-0(2) 1.423(3)
10 S(1)-O(1) 1.426(2)
S(1)-N(1) 1.613(2)
S(1)-C(6) 1.772(3)
N(1)-C(7) 1.456(4)
N(2)-C(15) 1.353(4)
15 N(2)-C(13) 1.459(4)
N(2)-C(12) 1.468(4)
O(3)-C(8) 1.400(4)
O(3)-C(9) 1.441(4)
O(4)-C(15) 1.214(4)
20 0(5)-0(15) 1.344(3)
0(5)-0(16) 1.467(3)
C(1)-C(6) 1.372(5)
C(1)-C(2) 1.400(5)
C(2)-C(3) 1.362(6)
25 C(3)-C(4) 1.358(6)
C(4)-C(5) 1.405(5)
C(5)-C(6) 1.376(4)
0(7)-0(10) 1.520(4)
C(7)-C(8) 1.525(5)
30 C(9)-C(11) 1.520(4)
C(9)-C(10) 1.521(4)
C(9)-C(14) 1.526(4)
C(11)-0(12) 1.506(5)
C(13)-0(14) 1.503(4)
35 C(16)-C(17) 1.508(5)
C(16)-C(18) 1.514(5)
C(16)-0(19) 1.518(4)
105
O(2)-S( 1)-0(1) 120.73(17)
O(2)-S(1)-N(1) 108.79(15)
O(1)-S(1)-N(1) 106.64(15)
O(2)-S(1)-C(6) 106.86(14)
5 O(1)-S(1)-C(6) 106.70(15)
N(1)-S(1)-C(6) 106.29(15)
C(7)-N(1)-S(1) 120.3(2)
C(15)-N(2)-C<13) 119.2(2)
C(15)-N(2)-C<12) 123.4(2)
10 C(13)-N(2)-C(12) 114.8(3)
C(8)-O(3)-C(9) 110.9(2)
C(15)-O(5)-C(16) 122.1(2)
C(6)-C(1)-C(2) 119.8(3)
C(3)-C(2)-C(1) 119.6(4)
15 C(4)-C(3)-C(2) 120.9(4)
C(3)-C(4)-C(5) 120.4(4)
C(6)-C(5)-C(4) 118.7(3)
C(1)-C(6)-C(5) 120.6(3)
C(1)-C(6)-S(1) 119.9(2)
20 C(5)-C(6)-S(1) 119.4(3)
N(1)-C(7)-C(10) 112.1(3)
N(1)-C(7)-C(8) 114.8(3)
C(10)-C(7)-C(8) 102.1(3)
O(3)-C(8)-C(7) 107.5(3)
25 O(3)-C(9)-C(11) 107.7(3)
O(3)-C(9)-C(10) 104.4(2)
C(11)-C(9)-C<10) 114.3(3)
O(3)-C(9)-C(14) 109.9(3)
C(11)-C(9)-C(14) 107.9(2)
30 C{10)-C(9)-C{14) 112.6(2)
C(7)-C(10)-C{9) 102.8(2)
C(12)-C(11)-C{9) 113.1(3)
N(2)-C(12)-C{11) 110.1(3)
N(2)-C(13)-C(14) 110.9(3)
35 C(13)-C(14)-C{9) 112.6(2)
0(4)-0(15)-0(5) 125.2(3)
O(4)-C(15)-N(2) 124.5(3)
O(5)-C(15)-N(2) 110.3(2)
106
0(5)-0(16)-0(17)
109.8(3)
0(5)-0(16)-0(18) 110.3(3)
C(17)-0(16)-0(18) 113.0(3)
0(5)-0(16)-0(19) 102.1(2)
0(17)-0(16)-0(19) 110.6(3)
0(18)-0(16)-0(19) 110.4(3)
Symmetry transformations used to generate équivalent atoms.
Table 4. Anisotropic displacement parameters (A2 x 103) for C48. The anisotroplc displacement
factor exponent takes the form: -2ττ22 a*2Un + . „ + 2hka* b* U12].
U11 U22 u U23 U13 U12
S(1) 48(1) 42(1) 69(1) 2(1) 10(1) 8(1)
N(1) 44(1) 42(1) 91(2) 9(1) 4(1) 3(1)
N(2) 41(1) 49(1) 67(2) 17(1) 2(1) 2(1)
0(1) 57(1) 69(1) 95(2) 19(1) 28(1) 18(1)
0(2) 80(2) 52(1) 70(1) -7(1) -6(1) 9(1)
0(3) 66(2) 88(2) 49(1) -8(1) -5(1) 24(1)
0(4) 43(1) 49(1) 68(1) 7(1) 4(1) 0(1)
0(5) 46(1) 46(1) 73(1) 16(1) KD 4(1)
0(1) 45(2) 51(2) 92(2) 0(2) -4(2) -4(1)
0(2) 66(2) 78(2) 84(2) -6(2) -20(2) 2(2)
0(3) 85(3) 77(2) 69(2) 6(2) -1(2) 2(2)
0(4) 77(2) 83(3) 81(2) 12(2) 15(2) -22(2)
0(5) 53(2) 65(2) 75(2) 1(2) 2(2) -18(2)
0(6) 40(1) 36(1) 70(2) -2(1) 5(1) 4(1)
0(7) 42(1) 44(1) 60(2) 2(1) 4(1) 4(1)
0(8) 78(2) 83(2) 70(2) -22(2) -9(2) 27(2)
0(9) 47(2) 49(2) 48(2) -KD 3(1) 6(1)
0(10) 46(1) 49(1) 57(2) -5(1) KD 7(1)
0(11) 44(2) 54(2) 91(2) 21(2) 9(2) KD
0(12) 50(2) 69(2) 83(2) 35(2) 10(2) 9(2)
0(13) 48(2) 48(2) 68(2) 10(1) -2(1) 0(1)
0(14) 51(2) 45(1) 51(2) 5(1) 1(1) 5(1)
0(15) 44(1) 43(1) 50(1) 2(1) -KD 2(1)
0(16) 51(2) 51(2) 48(2) 5(1) KD 13(1)
0(17) 56(2) 80(2) 70(2) 17(2) -7(2) -6(2)
120(4) 71(2)
C(19) 71(2) 51(2)
107
56(2) 4(2)
64(2) 12(1)
14(2)
-4(2)
37(2)
10(2)
Table 5. Hydrogen coordinates (x 104) and
Isotropie displacement parameters (A2x 10’) for C48.
X y z U(eq)
10 H(1X) -3660(30) 8980(20) 932(17) 57(9)
H(1) -5558 10584 -302 75
H(2) -5639 11234 -1490 91
H(3) -3946 12450 -1925 92
H(4) -2177 13033 -1212 96
15 H(5) -2047 12362 -25 77
H(7) -1107 10063 628 59
H(8A) -776 10401 1791 92
H(8B) -1794 9380 2029 92
H(10A) -938 8151 212 61
20 H(10B) -1738 7606 872 61
H(11A) -137 6501 1645 75
H(11B) 674 6326 929 75
H(12A) 1811 7141 2229 81
H(12B) 2127 5898 1865 81
25 H(13A) 3526 8801 840 66
H(13B) 2726 9045 1554 68
H(14A) 1562 8173 275 59
H(14B) 1285 9446 607 59
H(17A) 7038 6448 1888 103
30 H(17B) 7462 5258 2281 103
H(17C) 6316 6080 2605 103
H(18A) 5376 4423 741 124
H(18B) 6844 4173 1040 124
H(18C) 6460 5461 763 124
35 H(19A) 4803 4229 2609 93
H(19B) 5962 3476 2242 93
H(19C) 4519 3565 1883 93
108
Step 2. Synthesis of tert-butyl (3R)-3-[methyl(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5Jdecane-8-carboxylate (C50).
To a solution of C48 (1.5 g, 3.8 mmol) in Λ/,Μ-dimethylformamide at 0 ’C was added sodium hydride (60% dispersion In minerai oil; 227 mg, 5.67 mmol). The reaction mixture was stirred at room température for 30 minutes, whereupon lodomethane (1.61 g. 11.3 mmol) was added, and stirring was continued for 1 hour. Saturated aqueous ammonium chloride solution was added, and the aqueous layer was extracted three times with ethyl acetate. The combined organic layers were dried over magnésium sulfate, filtered, and concentrated in vacuo to provide the product. Yield: 1.53 g, 3.73 mmol, 98%. ’H NMR (400 MHz, CDCh) δ 7.80-7.76 (m, 2H), 7.63-7.58 (m, 1H), 7.56-7.50 (m, 2H), 4.73-4.64 (m, 1H), 3.78 (dd, J=10.2,7.4 Hz, 1H), 3.64-3.51 (m, 2H), 3.55 (dd, >10.2, 4.9 Hz, 1H), 3.27-3.13 (m, 2H), 2.76 (s, 3H), 1.87 (dd, >13.5, 9.1 Hz, 1H), 1.63-1.54 (m. 3H), 1.44 (dd, >13.5, 6.8 Hz, 1H), 1.43 (s, 9H), 1.37 (br ddd, >13,10,4 Hz, 1H).
Step 3. Synthesis of(2R)~1,1,1'-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl (3R)-3[methyl(phenylsuifonyi)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C51).
Conversion of C50 to C51 was carried out using the method described for synthesis of C34 from C33 In Exampies 8 and 9. Purification in this case was effected via silica gel chromatography (Gradient: 0% to 60% ethyl acetate in heptane) to afford the product as a 20 colorless oil. Yield: 1.7 g, 2.9 mmol, 77%. LCMS m/z 609.4 [M+Na*]. ’H NMR (400 MHz, CDCh) δ 7.82-7.78 (m, 2H), 7.64-7.59 (m, 1H), 7.57-7.52 (m, 2H), 7.23 (br d, >8.7 Hz, 2H), 6.87 (br d, >8.6 Hz, 2H), 5.52-5.40 (m, 1H), 4.75-4.63 (m, 1H), 4.49 (AB quartet, upfield doublet is broadened, Jab=11.7 Hz, Avab=28.4 Hz, 2H), 3.85-3.62 (m, 5H), 3.81 (s, 3H), 3.62-3.52 (m, 1H), 3.34-3.17 (m, 2H), 2.77 (s, 3H), 1.85 (dd, >13.5,9.1 Hz, 1H), 1.71 -1.53 (m, 3H), 1.46 (dd, 25 >13.5, 6.9 Hz, 1H), 1.38 (ddd, >13.5,11.2,4.4 Hz, 1 H).
Step 4. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3R)-3[methyl(phenylsulfonyi)aminoJ~1oxa-8-azaspiro[4.5]decane-8-carboxylate (15).
Trifluoroacetic acid (10.8 mL) was added drop-wise to a 0 ’C solution of C51 (1.7 g, 2.9 mmol) in dichloromethane (30 mL) and the reaction mixture was stirred for 1.5 hours at room 30 température. After removal of solvents in vacuo, the residue was dissolved in ethyl acetate and washed with saturated aqueous sodium bicarbonate solution. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were dried over magnésium sulfate, filtered, and concentrated under reduced pressure. Silica gei chromatography (Gradient; 0% to 80% ethyl acetate in heptane) provided the product as a white solid. Yield: 1.06 g, 2.27 mmol, 78%.
LCMS m/z 467.4 [M+HJ*. ’H NMR (400 MHz, CDCh) δ 7.81-7.77 (m, 2H), 7.64-7.59 (m, 1H), 7.57-7.51 (m, 2H), 5.28-5.18 (m, 1 H), 4.74-4.65 (m, 1 H), 3.98 (dd, half of ABX pattern, >12.5, 3.3 Hz, 1H), 3.89-3.69 (m, 3H), 3.80 (dd, >10.3, 7.4 Hz, 1H), 3.62-3.54 (m, 1H), 3.38-3.19 (m,
109
2H), 2.77 (s, 3H), 2.4-2.0 (v br s, 1 H), 1.94-1.81 (m, 1 H), 1.72-1.59 (m. 3H), 1.48 (br dd, J=13, 6 Hz, 1H), 1.45-1.34 (m, 1H).
Example 16 (2R)-1,1,1-TrifJuoro-3-hydroxypropan-2-yl 4-hydroxy~4~ {[(phenylsulfonyl)amino]methyl}piperidine-1-carboxylate (16)
O
O’S-CI
H2N
Step 1. Synthesis of tert-butyl 4-hydroxy-4-{[(phenylsulfonyl)amino]methy!}pipendine~1 carboxylate (C52).
tert-Butyl 4-(aminomethyl)-4-hydroxypiperidine-1-carboxylate was converted to C52 using the method described for synthesis of C32 from C31 in Example 7. Purification via préparative thln layer chromatography (Eluent: 10:1 dichloromethane / methanol) afforded the product as a coloriess gum. Yield: 127 mg, 0.343 mmol, 79%. LCMS m/z 393.0 [M+Na*]. 1H NMR (400 MHz, CDCh) δ 7.90-7.84 (m, 2H), 7.64-7.58 (m, 1H), 7.57-7.51 (m, 2H), 5.10 (br t, J=6.6 Hz. 1H), 3.83-3.70 (m, 2H), 3.17 (br dd, J-12,11 Hz, 2H), 2.92 (br d, J=6 Hz, 2H), 2.16 (brs, 1H), 1.63-1.54(m,2H), 1.53-1.45 (m,2H), 1.45 (s, 9H).
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-hydroxy-4{[(phenylsulfonyl)amino]methyl}plperidine-1-cartjoxylate (C53).
Conversion of C52 to C53 was carried out using the method described for synthesis of C34 from C33 in Examples 6 and 9. Purification In this case was effected via préparative thin layer chromatography on silica gel (Eluent: 1:1 ethyl acetate / petroleum ether) to afford the product as a coloriess gum. Yield: 60 mg, 0.11 mmol, 58% over 3 steps. LCMS m/z 569.1 [M+Na*].
Step 3. Synthesis of (2R)-1,1,1-tnnuoro-3-hydroxypropan-2-yl 4-hydroxy-4{[(phenylsulfonyl)amino]methyl}piperidine-1-cartjoxylate (16).
Conversion of C53 to 16 was carried out using the method described for synthesis of 1 from C12 in Example 1. Purification via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.1% aqueous hydrochloric acid; Mobile phase B: acetonitrile; Gradient: 28% to 48% B) afforded the product as a white solid. Yield: 23 mg, 54 pmol, 49%. LCMS m/z
110
449.0 [M+Na*]. 1H NMR (400 MHz, CDCh), characteristic peaks: δ 7.85 (br d, 7=7 Hz, 2H), 7.59 (br dd, half of ABX pattern, 7=7, 7 Hz, 1H), 7.53 (br dd, half of ABX pattern, 7=7,7 Hz, 2H),
5.87-5.69 (m, 1H), 5.33-5.20 (m, 1 H), 4.02-3.91 (m, 1H), 3.92-3.74 (m, 3H), 3.39-3.16 (m, 2H),
1.74-1.38 (m, 4H).
Example 17 (2R)-1,1,1-Trifluoro~3-hydroxypropan-2-yl 4-(4-fluorobenzyl)-3-oxo-1-oxa-4,9diazaspiro[5.5}undecane~9-carboxylate (17)
CF3COOH
O CF3
ΑθΑ^,ΟΡΜΒ
C55
Step 1. Synthesis oftert-butyl 4-(4-fluorobenzyl)-3-oxo-1-oxa-4,9-diazaspiro[5.5]undecane-910 carboxylate (C54).
A mixture of C22 (100 mg, 0.370 mmol), 1-(bromomethyl)-4-fluorobenzene (119 mg, 0.629 mmol), and césium carbonate (241 mg, 0.740 mmol) in /V,N-dimethylformamide (2 mL) was stirred at 100 ’C for 64 hours. The reaction mixture was then filtered and concentrated in vacuo; the residue was purified by préparative thin layer chromatography on silica gel (Eluent:
1:1 petroleum ether/ethyl acetate), affording the product as a colorless gum. Yield: 42 mg, 0.11 mmol, 30%. Ή NMR (400 MHz, CDCh) δ 7.28-7.21 (m, 2H, assumed; partially obscured by solvent peak), 7.04 (br dd, 7=8.7, 8.5 Hz, 2H), 4.57 (s, 2H), 4.23 (s, 2H), 3.82-3.67 (m, 2H), 3.14-3.03 (m, 2H), 3.07 (s, 2H), 1.84-1.74 (m, 2H), 1.44 (s, 9H), 1.42-1.32 (m, 2H).
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(4-fluorobenzyl)20 3-oxo-1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate (CSS).
Conversion of C54 to C55 was carried out using the method described for synthesis of C34 from C33 in Examples 8 and 9. Purification was effected via préparative thin layer chromatography on silica gel (Eluent: 1:1 ethyl acetate / petroleum ether) to provide the product as a colorless gum. Yield: 48 mg, 87 pmol, 78% over 2 steps. LCMS m/z 577.3 [M+Na*]. 1H
NMR (400 MHz, CDCh) δ 7.27-7.19 (m, 4H, assumed; partially obscured by solvent peak), 7.04 (brdd, 7=8.7, 8.4 Hz, 2H), 6.92-6.82 (m, 2H), 5.50-5.40 (m, 1H), 4.64-4.40 (m, 4H), 4.22 (s, 2H),
111
3.96-3.78 (m, 2Η), 3.81 (s, 3Η), 3.78-3.63 (m, 2H), 3.24-2.97 (m, 4H), 1.91-1.73 (m, 2H), 1.441.28 (m, 2H).
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(4-fluoroben2yl)-3-oxo-1-oxa-
4,9-diazaspiro[5.5]undecane-9-carboxylate (17).
Conversion of C55 to 17 was carried out using the method described for synthesis of C35 from C34 in Examples 8 and 9. Purification via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.225% formic acid in water; Mobile phase B: 0.225% formic acid in acetonitrile; Gradient: 30% to 50% B) afforded the product as a colorless gum. Yield: 15.4 mg, 35.4 pmol, 41%. LCMS m/z 435.0 [M+H]*. Ή NMR (400 MHz, CDCh) δ 7.28-
7.21 (m, 2H, assumed; partially obscured by solvent peak), 7.05 (br dd, J=8.5, 8.5 Hz, 2H),
5.29-5.18 (m, 1H), 4.66-4.49 (m, 2H), 4.23 (s, 2H), 4.02-3.95 (m, 1H), 3.93-3.79 (m, 3H), 3.283.11 (m, 2H), 3.09 (s, 2H), 1.93-1.80 (m, 2H), 1.46-1.35 (m, 2H).
Ex ample 18 (2R)-1,1,1-TrifIuoro-3-hydroxypropan-2-yl 2-ethyl-4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (18)
O
F
3)CF3COOH
Step 1. Synthesis ofted-butyl 4-({[(4-fluomphenyl)sulfonyl]amino}methyl)-4-hydmxypiperidine~1· carboxylate (C55).
4-Fiuoroben?enesulfonyl chloride (2.21 g, 11.4 mmol) was added portion-wise to a 0 ’C solution of tert-butyl 4-(aminomethyl)-4-hydroxyplperidine-1-carboxyiate (2.95 g, 12.8 mmol) and triethylamine (4.7 mL, 33.7 mmol) In dichloromethane (150 mL) and the reaction mixture was allowed to warm to room température and stir for 1 hour. It was then diluted with dichloromethane (100 mL) and washed sequentiafly with water (200 mL) and with saturated aqueous sodium chloride solution (200 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was dissolved in ethyl acetate (20 mL); addition of heptane
112
(100 mL) caused a solid to precipitate. Solvents were evaporated off to afford the product as a white solid. Yield: 4.3 g, 11.1 mmol, 97%. LCMS m/z 387.4 [M-H*J. Ή NMR (500 MHz, CDCb) δ 7.91-7.86 (m, 2H), 7.22 (br dd, J=8.6, 8.5 Hz. 2H), 5.2-4.9 (v br s, 1 H), 3.87-3.67 (m, 2H), 3.24-3.09 (m, 2H), 2.92 (s, 2H), 2.12-1.94 (brs, 1H), 1.63-1.55 (m, 2H), 1.53-1.45 (m, 2H), 1.45 5 (s, 9H).
Step 2. Synthesis oftert~buty/2-ethyl-4-f(4-fluowpheny/)sulfony/J~1-oxa-4,9~ diazaspiro[5.5]undecane-9-carboxylate (C57).
1,2-Dibromobutane (0.14 mL, 1.2 mmol) was added to a solution of C56 (150 mg, 0.386 mmol) in /V,N-dimethylformamide (2 mL). Potassium carbonate (330 mg, 2.4 mmol) was added, 10 and the reaction mixture was heated at 100 ’C for 1 hour. It was then cooled to room température and treated with additional 1,2-dibromobutane (0.14 mL, 1.2 mmol), followed by potassium carbonate (330 mg, 2.4 mmol). The reaction température was increased to 110 ’C for 1 hour, whereupon the reaction mixture was partitioned between ethyl acetate (50 mL) and water (50 mL). The organic layer was washed sequentially with water (50 mL) and with 15 saturated aqueous sodium chloride solution (50 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 60% ethyl acetate in heptane) provided the product as a colorless, viscous oil. Yield: 115 mg, 0.260 mmol, 67%. LCMS m/z 465.5 [M+Na*].
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypmpan-2-yl 2-ethyl~4-[(420 f!uorophenyl)sulfonyl]-1-oxa-4,9-diazasplro[5.5]undecane~9-carboxylate (18).
Trifluoroacetic acid (0.40 mL, 5.2 mmol) was added to a solution of C57 (115 mg, 0.260 mmol in dichloromethane (5 mL) and the reaction mixture was allowed to stirfor 1 hour at room température, whereupon It was concentrated in vacuo and mixed with dichloromethane (5 mL) and triethylamine (1.5 mL, 11 mmol). In a separate flask, a solution of C1 (65.0 mg, 0.260 25 mmol) in tetrahydrofuran (2 mL) was treated sequentially with bis(pentafluorophenyl) carbonate (102 mg, 0.259 mmol) and triethylamine (1.8 mL, 13 mmol), and this reaction was allowed to stir at room température for 1 hour. The solution containing the deprotected C57 was added to the carbonate reaction mixture, and stirring was continued for 2 hours at room température. The reaction mixture was then partitioned between ethyl acetate (100 mL) and saturated aqueous 30 sodium bicarbonate solution (60 mL), and the organic layer was washed with aqueous sodium hydrogen sulfate solution (1 M, 60 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure. The resulting viscous oil was taken up in dichloromethane (5 mL); trifluoroacetic acid (2 mL) was added at room température while the reaction mixture was stirred. The reaction mixture was allowed to stir for an additional 30 minutes, whereupon it was 35 concentrated in vacuo; the residue was dissolved In dichloromethane (5 mL) and concentrated once more. Purification was carried out via reversed phase HPLC (Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v): Gradient: 35% to 55% B) to provide the product. Yield:
113 ►
12.3 mg, 24.7 pmol, 10%. LCMS m/z 499.2 [M+H]*. Rétention time: 2.79 minutes [Analytical HPLC column: Waters Atlantis dC18,4.6 x 50 mm, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid In water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 5.0% to 95% B, linear over 4.0 minutes; Flow rate: 2 mL/minute].
Examples 19, 20 and 21
1,1,1,3t3-Pentafluoro-4-hydroxybutan-2-yl 4-[(4-fJuorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate (19); 1,1,1,3,3-PentafJuoro-4-hydroxybutan-2-yl4-((4fiuorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5Jundecane-9-carboxylate, ENT-1 (20); and
1, 1, 1,3,3-Pentafluoro-4-hydmxybutan-2-yl 4-[(4-fluomphenyl)sulfonyl]-1-oxa-4,9- diazaspifO[5.5]undecane-9-carboxylate. ENT-2(21)
C60
FF F
Step 1. Synthesis of 2,2,4,4,4-pentaiïuombutane-1,3-dfoi (CSS).
n-Butyllithium (2.5 M solution in hexanes; 23.9 mL, 59.8 mmol) was added drop-wise to a -78 ’C solution of 1,1,1,3,3,3-hexafluoropropan-2-ol (4.90 g, 29.2 mmol) in tetrahydrofuran
114 (40 mL). The reaction mixture was stirred for 10 minutes at -78 ’C, then allowed to warm to 0
C and stir for 1 hour. Paraformaldéhyde (8.7 g, 0.29 mol) was added In a portlon-wise manner, and the reaction mixture was stirred at room température ovemight. Water (50 mL) was added, followed by sodium borohydride (3.7 g, 98 mmol) {Caution: exothermic reaction, accompanied bygas évolution!}; in the course of the addition, the reaction mixture was cooled in an ice bath to control the reaction. Upon completion of the addition, stirring was continued ovemight at room température, whereupon the reaction was quenched via addition of 1 M aqueous hydrochlorlc acid {Caution: gas évolution}. The resulting mixture was extracted with ethyl acetate, and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the product as a yellow-brown oil. Yield: 4.5 g, 25 mmol, 86%. 1H NMR (400 MHz, CDCh) δ 4.50-4.38 (m, 1H), 4.14-4.02 (m, 1 H), 4.00-3.89 (m, 1H).
Step 2. Synthesis of 4-{[tert-butyl(dimethyl)silyl]oxy}-1,1,1,3,3-pentafluorobutan-2-ol (C59). N.N-Dimethylformamide (5 mL) was added to a 0 ’C solution of C58 (6.30 g, 35.0 mmol) and 1H-imidazole (2.62 g, 38.5 mmol) in dichloromethane (60 mL). fert-Butyl(dimethyl)silyl chloride (5.27 g, 35.0 mmol) was then introduced portion-wise, and the reaction mixture was allowed to warm to room température and stir for 4 days. Saturated aqueous ammonium chloride solution (100 mL) was added, and the aqueous layer was extracted with dichloromethane (2 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated In vacuo; silica gel chromatography (Eluent: 5% ethyl acetate in petroleum ether) afforded the product as a yellow oil. Yield: 3.0 g, 10 mmol, 29%. 1H NMR (400 MHz, CDCh) δ 4.44-4.31 (m, 1 H), 4.13-4.01 (m, 1H), 3.95-3.85 (m, 1H), 3.57-3.46 (m, 1H), 0.92 (s,9H), 0.13 (s, 6H).
Step 3. Synthesis of 4-{[tert-butyl(dimethyl)silyl]oxy}~1,1,1,3,3-pentafluorobutan-2-yl pentafluorophenyl carbonate (C60).
Bis(pentafluorophenyl) carbonate (158 mg, 0.401 mmol) was added to a 0 ’C solution of C59 (118 mg, 0.401 mmol) in acetonitrile (4 mL). Triethylamine (122 mg, 1.20 mmol) was added drop-wise to the reaction mixture, which was stirred briefly in the ice bath, and then allowed to warm to 25 ’C and stir for 2 hours. The réaction solution of C60 was used directly in the following step.
Step 4. Synthesis of 4-{[tert-butyl(dimethyl)silyl]oxy}-1,1,1,3,3-pentafluorobutan-2-yl 4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate (C61).
Triethylamine (118 mg, 1.17 mmol) was added to a 0 ’C solution of C28 (100 mg, 0.233 mmol) in acetonitrile (5 mL). After a few minutes, C60 (reaction solution from the previous step; 0.401 mmol) was added drop-wise to the 0 ’C mixture, which was stirred in the ice bath for several minutes, stirred at 28 ’C for 20 hours, and then cooled to 0 ’C. A second batch of C60 (using the same scale and method as step 3 above; 0.401 mmol) was prepared and added to the 0 ’C reaction mixture, which was allowed to warm to room température and stir ovemight. After removal of volatiles in vacuo, the residue was purified using préparative thin layer
115
chromatography on silica gel (Eluent: 3:1 petroleum ether / ethyl acetate) to afford the product as a white solid. Yield: 120 mg, 0.189 mmol, 81%. By Ή NMR analysis, this material was judged to be a mixture of rotamers. ’H NMR (400 MHz, CD3OD) δ 7.87-7.80 (m, 2H), 7,36 (br dd, >8.7, 8.7 Hz, 2H), 5.84-5.70 (m, 1H), 4.63-4.54 (m, 1H), 3.97-3.77 (m, 5H), 3.3-3.15 (m,
2H, assumed; partially obscured by solvent peak), 3.02-2.95 (m, 2H), 2.86-2.78 (m, 2H), 2.061.94 (m, 2H), 1.61-1.45 (m, 2H), [0.93 (s) and 0.90 (s), total 9H], [0.12 (s), 0.10 (s), and 0.08 (s), total 6H],
Step 5. Synthesis of 1,1,1,3,3-pentafluoro-4-hydroxybutan-2~yl 4-[(4-fluorophenyl)sulfonyl]-1· oxa-4,9-diazaspim[5.5Jundecane-9-carboxylate (19).
Trifluoroacetic acid (4 mL) and water (1 mL) were added drop-wise to a 0 C solution of
C61 (120 mg, 0.189 mmol) in dichloromethane (6 mL) and the reaction mixture was stirred at 28 *C for 3 hours. It was then concentrated in vacuo and partitioned between ethyl acetate (50 mL) and saturated aqueous sodium bicarbonate solution (50 mL); the organic layer was washed with saturated aqueous sodium bicarbonate solution (3 x 20 mL), dried over sodium sulfate, filtered, 15 and concentrated in vacuo. Préparative thin layer chromatography on silica gel (Eluent: 1:1 petroleum ether/ethyl acetate) provided the product as a coloriess gum. Yield: 73 mg, 0.14 mmol, 74%. LCMS m/z 521.1 [M+H]+. ’H NMR (400 MHz, CD3OD) δ 7.84 (brdd, >8.8, 5.0 Hz, 2H), 7.37 (br dd, >8.8, 8.7 Hz, 2H), 5.86-5.73 (m, 1H), 3.91-3.72 (m, 6H), 3.3-3.17 (m, 2H, assumed; partially obscured by solvent peak), 3.01-2.94 (m, 2H), 2.86-2.76 (m, 2H), 2.02-1.92 20 (m, 2H), 1.60-1.47 (m,2H).
Step 6. Isolation of 1,1,1,3,3~pentafluoro-4-hydroxybutan-2-yl4-[(4-fluomphenyl)sulfonylJ-1-oxa-
4,9-dlazasplm[5.5]undecane-9-carboxylate, ENT-1 (20), and 1,1,1,3,3-pentafluom-4hydroxybutan-2-yl 4-[(4~fluomphenyl)sulfonyl]- 1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate, ENT-2 (21).
The racemate 19 was separated into its component enantiomers via supercritical fluid chromatography [Column: Chiral Technologies Chiralcel OD, 3 pm; Gradient: 5% to 40% (2propanol containing 0.05% diethylamine) in carbon dioxidej. The first-eluting enantiomer was 20, obtained as a coloriess gum. Yield: 19.9 mg, 38.2 pmol, 27% for the séparation. LCMS m/z
521.2 [M+H]*. ’H NMR (400 MHz, CD3OD) δ 7.84 (br dd, >8.7, 5.1 Hz, 2H), 7.37 (br dd, >8.8,
8.7 Hz, 2H), 5.86-5.73 (m, 1H), 3.91-3.72 (m, 6H), 3.3-3.17 (m, 2H, assumed; partially obscured by solvent peak), 3.01-2.94 (m, 2H), 2.86-2.76 (m, 2H), 2.03-1.92 (m, 2H), 1.61-1.47 (m, 2H). Rétention time via supercritical fluid chromatography: 3.97 minutes (Column: Chiral Technologies Chiralcel OD-3,4.6 mm x 150 mm I.D., 3 pm; Mobile phase A: carbon dioxide; Mobile phase B: 2-propanol containing 0.05% diethylamine; Gradient: 5% to 40% B; Flow rate:
2.5 mL/minute).
The second-eluting enantiomer was 21, also isolated as a coloriess gum. Yield: 19.6 mg,
37.6 pmol, 27% for the séparation. LCMS m/z 521.2 [M+H]*. ’H NMR (400 MHz, CD3OD) δ
7.87-7.81 (m, 2H), 7.41-7.33 (m, 2H), 5.85-5.73 (m, 1H), 3.91-3.72 (m. 6H), 3.3-3.17 (m, 2H,
116 assumed; partially obscured by solvent peak), 3.01-2.94 (m, 2H), 2.86-2.76 (m, 2H), 2.03-1.92 (m, 2H), 1.60-1.47 (m, 2H). Rétention time via supercritical fluid chromatography: 4.38 min (Same analytical conditions as those described for 20).
Example 22 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 4-(morpholin-4-ylsulfonyl)-1-oxa-4,9· diazaspiro[5.5]undecane-9-carboxylate (22)
O
II
O=S-CI I
NaHCO3
O CF3
ΑοΛ<οη
O=S=O
O CF3
AqA^opmb
CF3COOH
o=s=o
C63
Step 1. Synthesis of tert-butyl 4-(morpholin-4-ylsulfonyl)-1-Oxa-4,9-diazaspiro[5.5]undecane-9carboxylate (C62).
Réaction of C23 with morpholine-4-sulfonyl chloride was carried out using the method described for synthesis of C32 from C31 in Example 7, providing the product as a colorless gum. Yield: 100 mg, 0.247 mmol, 63%. LCMS m/z 428.2 [M+Na*].1H NMR (400 MHz, CDCh) δ 3.81-3.70 (m, 8H), 3.29-3.21 (m, 6H), 3.15 (brdd, >12,12 Hz, 2H), 3.06 (s, 2H), 1.95-1.86 (m. 2H), 1.52-1.41 (m, 2H), 1.46 (s, 9H).
Step 2. Synthesis of (2R)-f, 1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(morpholin-4ylsulfonyl)- 1-oxa-4,9-diazasplm[5.5]undecane-9-carboxyla te (C63).
Conversion of C62 to C63 was carried out using the method described for synthesis of C34 from C33 In Examples 8 and 9. LCMS of intermediate 4-(morpholin-4-ylsulfonyl)-1-oxa-4,9diazaspiro[5.5]undecane, trifluoroacetic acid sait: m/z 306.0 [M+H]’. In this case, purification was carried out using préparative thln layer chromatography (Eluent: 1:1 petroleum ether/ethyl acetate) to afford C63 as a colorless gum. Yield: 90.0 mg, 0.155 mmol, 65%. LCMS m/z 603.9 [M+Na*].’H NMR (400 MHz, CDCh) δ 7.23 (d, >8.5 Hz, 2H), 6.87 (d, >8.4 Hz, 2H), 5.52-5.41 (m, 1H), 4.50 (AB quartet, JAB=11.7 Hz, ΔνΑΒ=28.2 Hz, 2H), 3.95-3.80 (m, 2H), 3.80 (s, 3H),
3.78-3.64 (m, 8H), 3.28-3.16 (m, 8H), 3.07-3.00 (m, 2H), 1.99-1.90 (m, 2H), 1.50-1,40 (m, 2H).
117
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydfOxypropan-2-yl 4-(morpholin-4-ylsulfonyl)~1-oxa-
4,9-diazaspiro[5.5]undecane-9-carboxylate (22).
Conversion of C63 to 22 was carried out using the method described for synthesis of C35 from C34 in Examples 8 and 9. Purification via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.225% formic acid in water; Mobile phase B: 0.225% formic acid in acetonitrile; Gradient: 25% to 45% B) afforded the product as a colorless gum.
Yield: 33.4 mg, 72.3 pmol, 47%. LCMS m/z 462.1 [M+H]*. 1H NMR (400 MHz, CDCIj) δ 5.32-
5.21 (m, 1H), 4.06-3.96 (m, 1H), 3.96-3.82 (m, 3H), 3.82-3.69 (m, 6H), 3.34-3.18 (m, 8H). 3.07 (s, 2H), 2.34-2.21 (m, 1H), 2.06-1.95 (m, 2H), 1.6-1.42 (m, 2H, assumed; partially obscured by water peak).
Example 23 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 3-(4-fluombenzyl)-3,8-diazabicyclo[3.2.1]octane-8carboxylate (23)
O CF3 λΑοη
CF3COOH
O CF3
AqA^opmb
C65
Step 1. Synthesis of fed-butyl 3-(4-fluorobenzyl)-3,8-diazabicyclo[3.2.1]octane-8-cart)Oxylate (C64).
A solution of 1-(bromomethyl)-4-fluorobenzene (134 mg, 0.709 mmol) in acetonitrile (3 ml) was slowly added to a room température mixture of tert-butyl 3,8-diazablcyclo[3.2.1]octane8-carboxylate (150 mg, 0.706 mmol) and potassium carbonate (293 mg, 2.12 mmol) in acetonitrile (12 mL) and the réaction mixture was stirred at 25 °C for 16 hours. It was then filtered, and the filtrate was concentrated In vacuo', silica gel chromatography (Gradient: 0% to 20% ethyl acetate in petroleum ether) afforded the product as a colorless gum. Yield: 226 mg, 0.705 mmol, quantitative. 1H NMR (400 MHz, CDCI3) δ 7.30-7.24 (m, 2H), 6.99 (br dd, J=8.8,
118
1.93-1.78 (m, 4H), 1.47 (s, 9H).
Sfep 2. Synthesis of (2R)-1,1,1-trif1uoro~3-[(4-methoxybenzyl)oxy]propan-2-yl3-(4-fluorobenzyl) 3,8-diazablcyclo[3.2.1]octane-8-carboxylate (C65).
Conversion of C64 to C65 was carried out using the method described for synthesis of C34 from C33 in Examples 8 and 9. LCMS of intermediate 3-(4-fiuorobenzyl)-3,8diazabicyclo[3.2.1]octane, bis(trifluoroacetic acid) sait: m/z 221.1 [M+H]*. In this case, purification was carried out via silica gel chromatography (Gradient: 0% to 5% methanol in dichloromethane) to afford C65 as a color! es s gum. Yieid: 150 mg, 0.302 mmol, 88% over 2 steps. LCMS m/z 497.2 [M+H]*.
Sfep 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-(4-fluorobenzyl)-3,8diazabicyclo[3.2.1]octane-8-carboxylate (23).
Conversion of C65 to 23 was carried out using the method described for synthesis of 7 from C32 in Example 7. In this case, purification was effected via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.225% formic acid in water; Mobile phase B: 0.225% formic acid in acetonitrile; Gradient: 10% to 30% B) to provide the product as a colortess gum. Yield: 75 mg, 0.199 mmol, 66%. LCMS m/z 377.0 [M+H]*. Ή NMR (400 MHz,
CDCIj) δ 7.31-7.23 (m, 2H, assumed; partially obscured by solvent peak), 7.01 (br dd, >8.8,
8.7 Hz, 2H), 5.33-5.22 (m, 1 H), 4.31-4.21 (m, 2H), 4.06-3.96 (m, 1 H), 3.93-3.83 (m, 1 H). 3.493.43 (m, 2H), 2.69-2.61 (m, 2H), 2.38-2.20 (m, 3H), 2.00-1.84 (m, 4H).
Example 24 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 4-hydroxy-4~([methyi(phenyisu!fonyi) amino]methyl)piperidlne~1-carboxylate(24) o
O=S-CI
O
A o cf3 qAqÂ^OPMB
C2 NEt3
1)CF3COOH
CFaCOOH
AqA^opmb
Sfep 1. Synthesis of tert-butyl 4-hydroxy-4-[(methylamino)methyl]piperidine-1-carboxylate (C66).
Methylamine (2 M solution in tetrahydrofuran; 0.245 mL, 0.490 mmol) was added to a solution of tert-butyl 1-oxa-6-azaspiro[2.5]octane-6-carboxylate (95 mg, 0.44 mmol) in éthanol (2
119
mL) and the reaction mixture was heated to 80 ’C for 20 hours. Concentration in vacuo provided the product as an oi! (105 mg); this material was used in the following step without additional purification.
Step 2. Synthesis oftert-butyl 4-hydroxy-4f[methyl(phenylsulfonyl)amino]methyl}pipehdine-15 carboxylate (C67).
To a solution of C66 (from the previous step; 105 mg, £0.44 mmol) in acetonitrile (2 mL) were added benzenesulfonyl chloride (0.110 mL, 0.862 mmol) and potassium carbonate (119 mg, 0.861 mmol). The reaction mixture was stirred at 25 ’C for 3 hours, whereupon it was concentrated in vacuo; silica gel chromatography (Eluent: ethyl acetate) afforded the product as a gum. Yield: 115 mg, 0.299 mmol, 68% over 2 steps. Ή NMR (400 MHz, CDCh) 8 7.82-7,77 (m, 2H), 7.65-7.59 (m, 1H), 7.58-7.52 (m, 2H), 3.95-3.81 (m, 2H). 3.24-3.10 (m, 2H), 3.04-2.91 (m, 2H), 2.90 (s, 3H), 1.70-1.61 (m, 2H), 1.56-1.46 (m, 2H), 1.45 (s, 9H).
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3~[(4-methoxybenzyl)oxy]propan-2-yl 4-hydroxy-4{[methyl(phenylsulfonyl)amino]methyl}piperidine-1-carboxylate (C68).
Conversion of C67 to C68 was carried out using the method described for synthesis of
C34 from C33 In Examples 8 and 9. LCMS of Intermediate N-[(4-hydroxyplperidin-4-yl)methyl]N-methylbenzenesulfonamide, trifluoroacetic acid sait: m/z 285.0 [M+H]*. In this case, purification was carried out via silica gel chromatography (Gradient: 40% to 60% ethyl acetate In petroleum ether), affording C68 as a colorless gum. By 1H NMR analysis, this was judged to be a mixture of diastereomers. Yield: 130 mg, 0.232 mmol, 78% over 2 steps. LCMS m/z 583.1 [M+Na*]. ’H NMR (400 MHz, CDCh) 8 7.81 (br d, J=8 Hz, 2H), 7.67-7.62 (m, 1H), 7.61-7.54 (m, 2H), 7.25 (d, J=8.5 Hz, 2H), 6.92-6.84 (m, 2H), 5.55-5.43 (m, 1H), 4.51 (AB quartet, upfield doublet is broadened, Jab=1 1.7 Hz, ΔνΑΒ“29 Hz, 2H), 4.07-3.90 (m, 2H), 3.85-3.65 (m, 2H), [3.82 (s) and 3.77 (s), total 3H], 3.37-3.22 (m, 2H), 3.01-2.79 (m, 2H), [2.91 (s) and 2.87 (s), total 3H], 1.75-1.64 (m, 2H), 1.55-1.43 (m, 2H).
Step 4. Synthesis of(2R)-1,1,1-tnfluoro-3-hydroxypropan-2-yl 4-hydroxy-4{[methyl(phenylsulfonyl)amino]methyl}plperidine-1-carboxylate (24).
Trifluoroacetic acid (1.2 mL, 16 mmol) was added drop-wise to a 0 ’C solution of C68 (130 mg, 0.232 mmol) in acetonitrile (5 mL). The reaction mixture was stirred at room température for 30 minutes, whereupon saturated aqueous sodium bicarbonate solution was added until the mixture reached a pH of approximately 8. The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo to provide an off-white solid; purification via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.225% formic acid in water;
Mobile phase B: acetonitrile; Gradient: 30% to 50% B) afforded the product. Yield: 51.6 mg,
0.117 mmol, 50%. LCMS m/z 463.1 [M+Na*]. 1H NMR (400 MHz, CDCh) 8 7.83-7.77 (m, 2H),
7.67-7.61 (m, 1H), 7.60-7.53 (m, 2H), 5.31-5.21 (m, 1H), 4.05-3.92 (m, 3H), 3.90-3.81 (m, 1H), 3.41-3.22 (m, 2H), 3.04-2.93 (m, 2H), 2.90 (s, 3H), 2.84-2.74 (brs, 1H), 1.77-1.67 (m, 2H), 1.64-
1.46 (m, 2H).
120
Example 25 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yi 4-(4-fiuorobenzyl)piperazine-1-carboxylate (25)
Br λ
9f3
Sfep 1. Synthesis of tert-butyl 4-(4-fJuorobenzyl)piperazine-1-carboxylate (C69).
To a 30 ’C solution of tert-butyl piperazine-1-carboxylate (200 mg, 1,07 mmol) and potassium carbonate (445 mg, 3.22 mmol) in acetonitrile (8 mL) was added a solution of 1(bromomethyl)-4-fluorobenzene (203 mg, 1.07 mmol) in acetonitrile (2 mL), In a drop-wise manner. The reaction mixture was stirred for 16 hours at 30 ‘C, whereupon it was concentrated ln vacuo and purified via chromatography on silica gel (Gradient: 0% to 20% ethyl acetate In petroleum ether) to afford the product as a colorless gum. Yield: 250 mg, 0.849 mmol, 79%. Ή NMR (400 MHz, CDCh) δ 7.28 (br dd, 4=8.2, 5.5 Hz, 2H), 7.01 (br dd, 4=8.8, 8.7 Hz, 2H), 3.47 (s, 2H), 3.43 (br dd, 4=5, 5 Hz, 4H), 2.37 (brdd, 4=5, 5 Hz, 4H), 1.46 (s, 9H).
Sfep 2. Synthesis of (2R)-1,1,1-tnfluoro-3-[(4-methoxybanzyl)oxy]propan-2-yl 4-(4fluorobenzyl)piperazine-1 -carboxylate (C70).
Conversion of C69 to C70 was carried out using the method described for synthesis of C34 from C33 In Examples 8 and 9. In this case, purification was carried out using préparative thin layerchromatography (Eluent: 3:1 petroleum ether/ethyl acetate) toafford the product as a colorless gum. Yield: 71 mg, 0.15 mmol, 74% over 2 steps. LCMS m/z 471.2 [M+H]*. Ή NMR (400 MHz, CDCh) δ7.31-7.22 (m, 4H), 7.01 (brdd, 4=8.8, 8.7 Hz, 2H), 6.88 (brd, 4=8.8 Hz, 2H), 5.53-5.43 (m, 1H), 4.51 (AB quartet, 4ab=1 1.7 Hz, ΔνΑΒ=27.9 Hz, 2H), 3.81 (s, 3H), 3.76 (dd, halfof ABX pattern, 4=11.1,4.0 Hz, 1H), 3.69 (dd, halfof ABX pattern, J=11.2,7.0 Hz, 1H), 3.60-3.45 (m, 4H), 3.50 (s, 2H), 2.51-2.36 (m, 4H).
Sfep 3. Synthesis of (2R)-1,1,1-tnfluoro-3-hydroxypropan-2-yl 4-(4-fluorobenzyl)piperazine-1carboxylate (25).
121
Trifluoroacetic acid (1 mL) was added to a 0 *C solution of C70 (61 mg, 0.13 mmol) In dichloromethane (4 mL). The réaction mixture was stirred at 25 C for 1 hour, whereupon it was basified to pH 7 via addition of saturated aqueous sodium bicarbonate solution, and extracted with dichloromethane (2x10 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Préparative thin layer chromatography (Eluent: 10:1 dichloromethane / methanol) provided the product as a colorless gum. Yield: 24.2 mg, 69.1 pmol, 53%. LCMS m/z 351.1 [M+H]*. Ή NMR (400 MHz, CDCh) δ 7.28 (br dd, J=8.2, 5.6 Hz, 2H), 7.02 (brdd, J=8.7, 8.7 Hz, 2H), 5.30-5.20 (m, 1H), 4.00 (br dd, half of ABX pattern, J=12, 3 Hz, 1H), 3.86 (dd, half of ABX pattern, J=12.4, 6.8 Hz, 1H), 3.63-3.43 (m, 4H), 3.49 (s, 2H),
2.52-2.34 (m, 4H).
Example 26 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yt 4-(isoquinolin-1-yloxy)piperidine-1-carboxylate, trifluoroacetic acid sait (26)
1) f-BuOK
2) CF3COOH
3) F FO CF3 qAqA^OPMB
F C2 NEt3
O CF3
ΑθΑ^ΟΗ • CF3COOH
4) CF3COOH
A solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (30.2 mg, 0.15 mmol) in N.Ndimethylformamide (0.5 mL) was added to 1-chloroisoquinolîne (24.5 mg, 0.15 mmol) in a reaction vial. Potassium tert-butoxide (1 M solution In tetrahydrofuran; 0.45 mL, 0.45 mmol) was added, and the reaction mixture was shaken at 60 ’C for 18 hours, then at 100 ’C for 1 hour. It was then partîtioned between half-saturated aqueous sodium bicarbonate solution (1.5 mL) and ethyl acetate (2.4 mL) and subjected to vortexing, followed by centrifugation to break up an émulsion. The organic layer was eluted through a solid phase extraction cartridge (6 mL) charged with sodium sulfate (-1 g); this extraction procedure was repeated twice, and the combined eluents were concentrated in vacuo. A mixture of trifluoroacetic acid and 1,2dichloroethane (1:1,1 mL) was added, and the reaction mixture was shaken at room température for 2.5 hours, whereupon it was concentrated in vacuo and dissolved in 1,2dichloroethane (2.4 mL) with vortexing. This material was loaded onto an SCX (strong cation exchanger) solid phase extraction cartridge (Silicycle, 6 mL, 1 g); the vial was rinsed with a mixture of methanol and 1,2-dichloroethane (1:1; 2 x 2.4 mL). The cartridge was eluted with methanol (5 mL), followed by a solution of triethylamîne in methanol (1 M, 7.5 mL) to elute the deprotected intermediate. Fractions containing the desired material were concentrated in vacuo,
122
and the residue was azeotroped with toluene (2 x 1 mL) to remove trace methanol. The residue was dissolved in dichloromethane (0.5 mL).
A crude solution of C2 was prepared separately, as follows: Bis(pentafluorophenyl) carbonate (1.89 g, 4.80 mmol) and triethylamine (13.4 ml, 96.1 mmol) were added to a stirring solution of C1 (1.23 g, 4.91 mmol) in tetrahydrofuran (15 mL). Sufficient tetrahydrofuran was added to bring the total volume to 32 mL, and the reaction mixture was stirred at room température for 1 hour. A portion of this crude C2 solution (1.0 mL, 0.15 mmol of C2 and 3 mmol of triethylamine) was added to the deprotected amine solution prepared above, and the reaction mixture was shaken at room température overnight. It was then partitioned between half-saturated aqueous sodium bicarbonate solution (1.5 mL) and ethyl acetate (2.4 mL) and subjected to vortexing. The organic layer was eluted through a solid phase extraction cartridge (6 mL) charged with sodium sulfate (~1 g); this extraction procedure was repeated twice, and the combined eluents were concentrated in vacuo. This material was treated with a mixture of trifluoroacetic acid and 1,2-dichloroethane (1:1,1 mL) and shaken at room température for 1 hour, whereupon it was concentrated in vacuo and purified using reversed phase HPLC (Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid In acetonitrile (v/v); Gradient: 20% to 100% B) to afford the product. Yield: 2.5 mg, 6.5 pmol, 4%. LCMS m/z 385.1 [M+H]+. Rétention time 3.01 minutes [Analytical HPLC conditions - Column: Waters Atlantis dC18,4.6 x 50 mm, 5 pm;
Mobile phase A: 0.05% trifluoroacetic add in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid In acetonitrile (v/v); Gradient: 5.0% to 95% B, linear over 4.0 minutes; Flow rate: 2 mL/minute].
Example 27 (2R)-1,1,1-Trifluoro-3-hydrOxyprOpan-2-yl 3-(pyridin-2-ylamino)-1-oxa-8~azaspiro[4.5]decane-825 carboxylate (27)
123
HN
Step 1. Synthesis ofted-butyl 3~{[(prop-2-en-1-yloxy)carbonyl]amino}-1-oxa-85 azaspiro[4.5]decane-8-carboxylate (C71).
Prop-2-en-1-yl carbonochloridate (9.87 g, 81.9 mmol) was added to a 0 ’C solution of tert-butyl 3-amino-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (14.0 g, 54.6 mmol) in saturated aqueous sodium bicarbonate solution (400 mL) and tetrahydrofuran (100 mL). The réaction mixture was stirred at 22 ’C for 16 hours, whereupon it was filtered and the fîlter cake was 10 washed with ethyl acetate. The aqueous layer from the combined filtrâtes was extracted with ethyl acetate (2 x 200 mL), and the combined organic layers were washed with saturated ammonium chloride solution (3 x 100 mL), dried over sodium sulfate, filtered, and concentrated In vacuo to provide the product as a yellow oil, which solidified upon standing at room température. Yield: 18.3 g, 53.8 mmol. 98%. 1H NMR (400 MHz, CDCb) δ 5.98-5.85 (m, 1H), 15 5.31-5.27 (m, 1H), 5.26-5.20 (m, 1 H). 4.95-4.86 (m. 1H), 4.56 (br d. J-4.6 Hz. 2H), 4.38-4.28 (m, 1H), 4.00 (dd, J=9.5, 5.6 Hz, 1H), 3.67 (brdd, J=9.7,4.0 Hz, 1H), 3.66-3.52 (m, 2H), 3.37-
3.24 (m, 2H), 2.13 (dd, J=13.3, 7.6 Hz, 1H), 1.72-1.49 (m, 5H, assumed; partially obscured by water peak), 1.46 (s, 9H).
Step 2. Synthesis of (2R)-1,1,1-tnfluoro-3-[(4-methoxybenzyl)oxy]prcpan-2-yl 3-{[(prop-2-en-1~ 20 yloxy)carbonyl]amino}-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C72).
124
Conversion of C71 to C72 was effected using the method described for synthesis of C34 from C33 in Examples 8 and 9. The product was isolated as a light yellow oil. Yield: 12.6 g, 24.2 mmol, 89% over 2 steps. LCMS m/z 539.1 [M+Na*]. ’H NMR (400 MHz, CDCh) δ 7.24 (br d, >8.5 Hz, 2H), 6.88 (br d, >8.7 Hz, 2H), 5.98-5.85 (m, 1H), 5.53-5.41 (m, 1H), 5.35-5.26 (m,
1H), 5.26-5.19 (m, 1H), 5.00-4.89 (m, 1H), 4.62-4.50 (m, 3H), 4.46 (d, half of AB quartet, >11.7
Hz, 1H), 4.38-4.26 (m, 1H), 4.04-3.96 (m, 1H), 3.85-3.62 (m, 4H), 3.81 (s, 3H), 3.41-3.25 (m, 2H), 2.19-2.06 (m, 1 H), 1.78-1.46 (m, 5H, assumed; partially obscured by water peak).
Step 3. Synthesis of(2R)~1,1,1-tnfluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3-amino-1-oxa-8· azaspiro[4.5]decane-8-carboxylate (C73).
Palladium(ll) acetate (520 mg, 2.32 mmol) was added to a solution of C72 (12.6 g, 24.2 mmol), 1,3-dimethylpyrimidine-2,4,6(1H,3H,5H)-trione (7.62 g, 48.8 mmol), and triphenylphosphine (1.92 g, 7.32 mmol) in dichloromethane (100 mL). The reaction mixture was heated to 35 ’C for 5 hours, whereupon it was concentrated in vacuo and purified via silica gel chromatography (Gradient: 0% to 100% ethyl acetate in petroleum, followed by a gradient of 0% 15 to 10% methanol in dichloromethane) to afford the product as an orange solid. Yield: 9.40 g,
21,7 mmol, 90%. ’H NMR (400 MHz, CDCh) δ 7.24 (br d, >8.5 Hz, 2H), 6.87 (br d, >8.5 Hz, 2H), 5.53-5.41 (m, 1H), 4.50 (AB quartet, Jab=11.7 Hz, Avab=26.7 Hz, 2H), 4.02-3.94 (m, 1H),
3.87-3.62 (m, 4H), 3.80 (s, 3H), 3.42-3.17 (m, 4H), 2.18-2.05 (m, 1H), 1.86-1.59 (m, 4H), 1.55-
1.46 (m, 1H).
Step 4. Synthesis of (2R)-1,1,1-tnfJuoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3-(pyridin-2~ ylamino)- 1-oxa-8-azaspiro[4.5]decane-8-carboxyiate (C74).
A mixture of C73 (100 mg, 0.231 mmol), 2-chloropyridine (52.5 mg, 0.462 mmol), (1,3bÎs(2,6-diisopropylphenyl)imldazol-2-ylidene](3-chloropyridyl)palladium(ll) dichlorlde (15.8 mg,
23.2 pmol), and césium carbonate (226 mg, 0.694 mmol) in toluene (9 mL) was heated at 130 25 ’C for 18 hours. The reaction mixture was filtered, concentrated in vacuo, and subjected to préparative thin layer chromatography (Eluent: ethyl acetate), followed by a second préparative thin layer chromatographie purification (Eluent: (1:1 ethyl acetate / petroleum ether) containing 0.5% ammonium hydroxide] to provide the product as a light yellow gum. Yield: 36 mg, 71 pmol, 31%. LCMS m/z 532.2 (M+Na*]. ’H NMR (400 MHz, CDCh) δ 8.10 (d. J=4 Hz, 1H). 7.43 (dd, 30 >8, 8 Hz, 1 H), 7.24 (d, >8.4 Hz, 2H), 6.88 (br d, >8 Hz, 2H), 6.64-6.59 (m, 1 H), 6.38 (d, >8
Hz, 1H), 5.53-5.43 (m, 1H), 4.64-4.58 (m, 1H), 4.55 (d, half of AB quartet, >12 Hz, 1H), 4.514.40 (m, 2H), 4.19-4.12 (m, 1H), 3.81 (s, 3H), 3.8-3.65 (m, 4H). 3.44-3.31 (m, 2H), 2.27-2.15 (m, 1H), 1.85-1.51 (m, 5H, assumed; partially obscured by water peak).
Step 5. Synthesis of (2R)-1,1,1-tnfJuoro-3-hydroxypropan-2-y/ 3-(pyridin-2-ylamino)~1-oxa-835 azaspiro[4.5]decane-8-carboxylate (27).
Trifluoroacetic acid (1 mL) was added to a 0 ’C solution of C74 (18 mg, 35 pmol) In dichloromethane (2 mL). The reaction mixture was stirred for 45 minutes, whereupon It was treated with aqueous sodium bicarbonate solution (10 mL) and extracted with dichloromethane
125
(3x15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo; purification via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: 0.225% formic acid In water; Mobile phase B: acetonltrile; Gradient: 8% to 28% B) afforded the product as a white solid. Yield: 10.0 mg, 25.7 pmol, 73%. LCMS m/z 389.9 [M+H]*. Ή NMR (400 MHz, CD3OD), characteristic peaks: δ 7.93 (br d, 7=5 Hz, 1 H), 7.42 (br dd, 7=8, 7 Hz, 1H), 6.59-6.52 (m, 2H), 5.33-5.24 (m, 1H), 4.49-4.40 (m, 1 H), 4.14 (dd, 7=9, 6 Hz, 1H), 3.91-3.83 (m, 1H), 3.81-3.67 (m, 4H), 2.26 (dd, 7=13,8 Hz, 1 H).
Example 28 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 4-(4-fluombenzyl)-1-oxa-3-thia-4,910 diazaspiro[5.5]undecane-9-carboxylate 3,3-dioxide (28)
O CF3
ΑθΛ^,ΟΡΜΒ
1)CF3COOH
C78
F \ \ CF3COOH
2) î ΡΥΎΡ° CF3 fAsAoaoA^opmb F C2 NEt3 O CF3
XaLah
Step 1. Synthesis oftert-butyl 4-({[(chloromethyl)sulfonyl]amino}methyl)-4-hydroxypipendine-1· carboxylate (C75).
Pyridine (3.0 mL, 37 mmol) was added to a solution of tert-buty14-(amînomethyl)-4hydroxypiperidine-1-carboxylate (2 g, 8.7 mmol) In dichloromethane (40 mL). and the reaction mixture was cooled to 0 ’C. A solution of chloromethanesulfonyl chloride (0.930 mL, 10.2 mmol) In dichloromethane (40 mL) was then added drop-wise over 25 minutes, and the reaction mixture was allowed to stîr at 0 ’C for 5 minutes before being warmed to room température and
126
stirred for 2 days. After solvents had been removed in vacuo, the residue was partitioned between dichloromethane and saturated aqueous ammonium chloride solution. The aqueous layer was extracted with dichloromethane, and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography 5 (Eluents: 50%, then 75%, then 90% ethyl acetate in heptane) provided the product as a tacky yellow solid. Yield: 851 mg, 2.48 mmol, 28%. LCMS m/z 341.5 [M-H*]. 1H NMR (400 MHz, CDCh) δ 5.28 (br t, J-6.2 Hz, 1H), 4.58 (s, 2H), 3.83 (br ddd, J=13.6,4,4 Hz, 2H), 3.24-3.15 (m, 4H), 1.69-1.61 (m, 2H), 1.56 (ddd, J=13.5,11.1, 4.7 Hz, 2H), 1.46 (s, 9H).
Step 2. Synthesis of tert-butyl 1-oxa-3-thia-4,9-diazaspiro[5.5]undecane-9-carboxylate 3,310 dioxide (C76).
A solution of C75 (360 mg, 1.05 mmol) in tetrahydrofuran (7 mL) was cooled to 0 *C and treated with sodium hydride (60% suspension in minerai oil; 109 mg, 2.72 mmol). After the reaction mixture had been stirred for two days at room température, more sodium hydride (60% suspension in minerai oil; 109 mg, 2.72 mmol) was added, and stirring was continued for 2 days 15 at room température. Saturated aqueous ammonium chloride solution was added, and the mixture was diluted with ethyl acetate; the aqueous layer was extracted with ethyl acetate, and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo. Chromatography on silica gel (Gradient: 25% to 50% ethyl acetate in heptane) afforded the product as a white solid. Yield: 20 430 mg, assumed quantitative. GCMS m/z 306.1 [M*J. 1H NMR (400 MHz, CDCI3) δ 4.67 (s,
2H), 4.63 (br t, J=7 Hz, 1H), 3.99-3.81 (m, 2H), 3.45 (br d, J-7 Hz, 2H), 3.06 (br dd, J=12,11 Hz, 2H), 2.08-1.92 (m, 2H). 1.49 (ddd, J=14.0,11.8, 4.7 Hz, 2H), 1.47 (s, 9H).
Step 3. Synthesis of tert-butyl 4-(4-fluorobenzyl)-1-oxa-3-thia-4,9-diazaspiro[5.5]undecane-9carboxylate 3,3-dioxide (C77).
A mixture of C76 (100 mg, 0.326 mmol), sodium iodide (74 mg, 0.49 mmol), césium carbonate (319 mg, 0.979 mmol), and acetonitrile (3 mL) was treated with 1-(bromomethyl)-4fluorobenzene (63 pL, 0.51 mmol) and stirred at room température ovemight. The reaction mixture was then filtered through diatomaceous earth, and the filter pad was rinsed with acetonitrile. The combined filtrâtes were concentrated in vacuo, and the residue was purified twice via silica gel chromatography (#1 - Gradient: 10% to 33% ethyl acetate in heptane; #2 dichloromethane as eluent, followed by a gradient of 5% to 33% ethyl acetate in heptane) to afford the product as a white solid. Yield: 128 mg, 0.309 mmol, 95%. 1H NMR (400 MHz, CDCIj) δ 7.32-7.27 (m, 2H). 7.07 (br dd, J=8.6, 8.6 Hz, 2H), 4.68 (s, 2H), 4.27-4.17 (br s, 2H), 3.74-3.59 (m, 2H), 3.14-2.99 (m, 4H), 2.04-1.88 (m, 2H), 1.43 (s, 9H), 1.33 (ddd, J=14.1,11.3, 4.6 Hz,
2H).
Step 4. Synthesis of(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 4-(4-fluombenzyl)1-oxa-3-thia-4,9-diazaspiro[5.5]undecane-9-carboxylate 3,3-dioxide (C78).
127
Conversion of C77 to C78 was effected using the method described for synthesis of C34 from C33 in Examples 8 and 9. ’H NMR (400 MHz, CD3OD) of intermediate 4-(4-fluorobenzyl)1-oxa-3-thia-4,9-diazaspiro[5.5]undecane 3,3-dioxide, trifluoroacetïc acid sait, δ 7.44-7.38 (m, 2H), 7.11 (brdd, >8.8, 8.8 Hz, 2H), 4.82 (s, 2H), 4.26 (brs. 2H), 3.24-3.17 (m, 2H), 3.23 (s.
2H), 3.17-3.08 (m, 2H), 2.34-2.26 (m, 2H), 1.58 (ddd, >15,13, 5 Hz, 2H); LCMS m/z 315.3 [M+H]*. In this case, purification was effected via chromatography on silica gel (Eluents: 10%, then 25%, then 50% ethyl acetate in heptane), affording C78 as a tacky white solid. Yield: 156 mg, 0.264 mmol, 85%. LCMS m/z 613.1 [M+Na*] ’H NMR (400 MHz, CDCb) δ 7.29 (br dd, >8.6, 5.3 Hz, 2H), 7.26-7.16 (br m, 2H), 7.07 (br dd, >8.6, 8.6,2H), 6.91-6.81 (br m, 2H), 5.49-5.38 (m, 1H), 4.73-4.63 (m, 2H), 4.55-4.39 (m, 2H), 4.33-4.14 (m, 2H), 3.88-3.7 (m, 2H), 3.81 (s, 3H), 3.73 (dd, half of ABX pattern, >11.1,3.8 Hz, 1 H), 3.65 (dd, half of ABX pattern, >11.1, 7.2 Hz, 1H), 3.22-2.99 (m, 4H), 2.12-1.91 (m, 2H), 1.40-1.23 (m, 2H).
Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(4-fluorobenzyl)-1-oxa-3-thla-
4,9-diazaspiro[5.5]undecane-9-carboxylate 3,3-dioxide (28).
Trifluoroacetic acid (1 mL) was added portion-wise to a 0 *C solution of C78 (151 mg, 0.256 mmol) in dichloromethane (4 mL). The reaction mixture was stirred for 1 hour at room température, whereupon it was concentrated In vacuo, and the resïdue was partitioned between saturated aqueous sodium bicarbonate solution and ethyl acetate. The organic layer was dried over sodium sulfate, filtered, concentrated under reduced pressure, and chromatographed on silica gel (Eluents: 10%, then 25%, then 50% ethyl acetate in heptane) to afford the product as a tacky white solid. Yield: 109 mg, 0.232 mmol, 91%. LCMS m/z 471.4 [M+H]*. ’H NMR (400
MHz, CDCI3) δ 7.30 (brdd, >8.5, 5.4 Hz, 2H), 7.08 (brdd, >8.6, 8.5 Hz, 2H), 5.27-5.17 (m, 1H), 4.74-4.63 (m, 2H), 4.34-4.13 (m, 2H), 3.98 (dd, half of ABX pattern, >12.5, 3.3 Hz, 1H), 3.92-3.73 (m, 3H), 3.27-3.01 (m, 4H), 2.15-1.96 (m, 2H), 1.43-1.3 (m, 2H).
Example 29 (2R)-1,1,1-Trlfluoro-3-hydroxypropan-2-yl 4-[(4-fluorophenyl)sulfonyl]-3-hydroxy-1-oxa-4,9diazasplro[5.5Jundecane-9-carboxylate (29)
MicroCyp® Reaction Buffer mix (Codexis; 519.0 mg) was mixed with deionized water (28.1 mL) to provide a buffer solution containing NADP*, glucose, glucose dehydrogenase, and
128
potassium phosphate. Compound 6 (6.0 mg, 13 pmol) was dissolved in a mixture of dimethyl sulfoxide (0.72 mL) and the buffer solution (0.24 mL).
MCYP-P1.2-B12 (Codexis; 6.8 mg, 0.72 nmol/mg) was treated with the buffer solution prepared above (27.4 mL), followed by the solution of 6 prepared above. The reaction mixture 5 was divided in half (14.2 mL each) and transferred into two 25 mL glass vials; the reaction mixtures were left open to the atmosphère and shaken on an orbital shaker (30 °C, 225 rpm) for 24 hours. The combined reaction mixtures contained:
[MCYP-P1.2-B12] = 0.24 mg/mL (0.17 μΜ. 6.8 mg, 4.89 nmol) [6] = 0.21 mg/mL (0.44 mM, 6.0 mg, 13 pmol)
2.5% dimethyl sulfoxide [NADP*] = 0.75 mg/mL (0.99 mM, 21.5 mg, 28.1 pmol) [Glucose] = 3.55 mg/mL (19,7 mM, 100.8 mg, 559.7 pmol) [Glucose dehydrogenase] = 0.39 mg/mL (11.2 mg)
0.1 M potassium phosphate buffer, pH 8.0
After 24 hours, the crude reaction mixtures were combined and purified via reversed phase HPLC (Column: Phenomenex Luna (2) C18, 5 pm; Mobile phase A: 0.1% formic acid In water; Mobile phase B: 0.1% formic acid in acetonitrile; Gradient: 50% to 100% B) to afford the product as a solid (3.0 mg), presumed to be a mixture of diastereomers. 1 -Dimensional and 2dimensional NMR spectroscopic studies established the regiochemïstry of oxidation as shown 20 for 29. The 1H NMR Indicated that some impurities were présent; peaks belonging to the product were Identified via 2D NMR. Yield, corrected by quantitative NMR: 1.6 mg, 3.3 pmol, 25%. LCMS m/z 469.2 [(M - H2O)+H]* and 509.1 [M+Na*]. 1H NMR (500 MHz, DMSO-de), characteristlc peaks: δ 7.93-7.88 (m, 2H), 7.42 (br dd, J=8.9,8.8 Hz, 2H), 5.25-5.17 (m, 1H), 5.17 (br s, 1H), 3.83-3.78 (m, 1H), 3.70-3.53 (m. 5H), 3.26-3.13 (m, 2H), 3.19 (d, J=12.0 Hz, 25 1 H), 2.79 (d, J=12.0 Hz, 1H), 1.53-1.41 (m, 2H).
Example 30 (2R)-3,3,3-Trifluoro-2-[({(3R)-3-[methyl(pheny!sulfony!)aminoJ- 1-oxa-8-azaspiro[4.5Jdec-8yl}carbonyl)oxy]propyl phosphate, disodium sait (30)
129
Step 1. Synthesis of (2R)-1,1,1-tnfluoro-3-(phosphonooxy)propan-2-yl (3R)~3~ [methyl(phenylsulfonyl)amlno]~ 1-oxa-8-azasplro[4.5]decane-8-carboxylate (C79).
Diphosphoryl tetrachloride (98%, 850 pL, 6.02 mmol) was added to a 0 ’C solution of 15 5 (560 mg, 1.20 mmol) In acetonitrile (7.5 mL), and the reaction mixture was stirred at 0 ’C for 3 hours, whereupon it was poured into ice. After it had been stirred at room température for 1.75 hours, the resulting mixture was concentrated in vacuo to remove acetonitrile. The aqueous residue was extracted 4 times with ethyl acetate, and the combined organic layers were dried over magnésium sulfate, filtered and concentrated under reduced pressure. The resulting clear 10 oil was treated with diethyl ether and again concentrated in vacuo; this diethyl ether treatment was repeated, affordïng the product as a white solid. Yield: 510 mg, 0.933 mmol, 78%. LCMS m/Z 547.2 [M+H]*. Ή NMR (400 MHz, CD3OD) δ 7.85-7.80 {m, 2H), 7.70-7.65 {m, 1H), 7.637.57 (m, 2H), 5.53-5.43 (m, 1H). 4.75-4.64 (m, 1H), 4.30-4.16 (m, 2H), 3.80 (dd, J=10.0, 7.4 Hz, 1H), 3.77-3.63 (m, 2H), 3.55 {dd, J=rf0.1, 5.0 Hz, 1H), 3.38-3.18 (m, 2H, assumed; partîally obscured by solvent peak), 2.76 (s, 3H), 1.91 (brdd, J=13.3, 9.3 Hz, 1H), 1.78-1.57 {m, 3H), 1.51 (dd, J=13.5, 6.8 Hz, 1H), 1.48-1.37 (m, 1H).
Step 2. Synthesis of (2R)-3,3,3-trifluoro-2-[(((3R)~3~[methy!(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]dec-B-yl}cart>onyl)oxy]propyl phosphate, disodium sait (30).
To a solution of C79 (820 mg, 1.50 mmol) in éthanol (9 mL) was added aqueous sodium 20 hydroxide solution (1 M; 2.9 mL, 2.9 mmol) and the reaction mixture was stirred at room température for 3 hours Ethanol (10 mL) was added, and the mixture was concentrated in vacuo; this éthanol treatment was repeated three times; the resulting solid was washed with éthanol and collected via filtration, affording the product as a white solid. Yield: 660 mg, 1.12 mmol, 75%. LCMS m/Z 547.2 [M+H]*. 1H NMR (400 MHz, D2O) δ 7.85-7.80 (m, 2H), 7.75-7.69 25 (m, 1H), 7.65-7.60 (m, 2H), 5.46-5.36 (m, 1H), 4.78-4.65 (m, 1H, assumed; partîally obscured by solvent peak), 4.15-4.08 (m, 1H), 4.07-3.99 (m, 1H), 3.85 (dd, >10, 8 Hz, 1H), 3.63-3.25 (m,
130
5H), 2.76 (s, 3H), 1.94 (dd, >13.6,9.3 Hz, 1H), 1.79-1.57 (m, 3H), 1.57-1.40 (m, 1H), 1.49 (dd, >13.7, 6.7 Hz, 1H).
Example 31 (2R)-3,3,3- Trifluoro-2-[({(3R)-3-[(phenylsulfonyl)amÎno]- 1-oxa-8-azaspiro[4.5]dec-8~ yl}carbonyl)oxyjpropyl phosphate, disodium sait (31)
CF,COOH
Hd 0H
-à O’Na* Na*
Step 1. Synthesis of dibenzyl (2R)-3,3,3-trifluoro-2-hydroxypropyl phosphate (C80).
(2R)-2-(Trifluoromethyl)oxirane (14.85 g, 132.5 mmol) was added to dibenzyl hydrogen phosphate (99%, 10.8 g, 38.4 mmol) In an amber bottle, and the thîck slurry was heated In a 65 °C oil bath for 25 hours. Excess (2R)-2-(trifluoromethyl)oxirane was removed via concentration in vacuo. The resulting oil was diluted with dichloromethane (10 mL) and subjected to silica gel chromatography (Eluents: 5%, then 10%, then 15%, then 20% ethyl acetate in dichloromethane)
131
to afford a pale yellow oil, which was treated with heptane (90 mL) and vîgorously stirred. The resulting solids were allowed to granulate for 1.5 hours, whereupon they were coilected via filtration and washed with heptane (38 mL), affording the product as a white solid. Yield: 9.11 g,
23.3 mmol, 61%. Melting point: -45 ‘C by différentiel scanning calorimetry. Ή NMR (400 MHz, 5 CDjCN) δ 7.42-7.34 (m, 10H), 5.06 (d, 4=8.3 Hz, 4H), 4.27-4.14 (m, 2H), 4.14-4.05 (m, 1H).
Step 2. Synthesis ofN-[(3R)-1-oxa-8-azaspimf4.5Jdec-3-y/]benzenesu/fonam/de, trifluoroacetic acid sait (C81).
Conversion of C48 (1.3 g, 3.3 mmol) to C81 was carried out using the method described for synthesis of C10 from C9 In Example 1. The product was obtained as a colorless oil, and 10 taken on without additional purification. The ’H NMR Indicated that the product was Impure.
Yield: 2.17 g, assumed quantitative. LCMS m/z 297,2 [M+H]+. ’H NMR (400 MHz, CDCI3), product peaks only: δ 7.89-7.85 (m, 2H), 7.68-7.62 (m, 1H), 7.60-7.54 (m, 2H), 3.98-3.91 (m, 1H), 3.88 (dd, halfof ABX pattern, 4=9.7, 5.6 Hz, 1H), 3.62 (brdd, 4=9.8,4.4 Hz, 1H), 3.38-3.24 (m, 4H), 2.05 (dd, 4=13.6, 7.3 Hz, 1H), 1.99-1.88 (m, 2H), 1.88-1.81 (m, 1H), 1.81-1.71 (m, 2H). 15 Sfep 3. Synthesis of (2R)-3-([bis(benzyloxy)phosphoryl]oxy}-1,1,1-trifluoropropan-2-yl (3R)-3[(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-cart)oxy!ate (C82).
Compound C80 (1.90 g, 4.87 mmol) was added to a solution of Ι,Γ-carbonyldiimîdazole (790 mg, 4.87 mmol) in acetonitrile (23 mL). The réaction mixture was allowed to stlr for 1.5 hours at room température, whereupon a solution of C81 (from the previous step, 2.00 g) In 20 acetonitrile (2 mL) was added in a drop-wise manner over 1 minute. After the reaction mixture had been stirred for an additional 5 hours at room température, it was partitioned between ethyl acetate (250 mL) and water (250 mL). The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo; silica gel chromatography (Gradient: 30% to 80% ethyl acetate In heptane) provided the product as a colorless oil. Yield: 2.02 g. 2.83 mmol, 66% over 2 steps.
LCMS m/z 713.1 [M+H]*. 1H NMR (400 MHz, CDCh) δ 7.88 (br d, 4=8 Hz, 2H), 7.64-7.58 (m, 1H), 7.57-7.51 (m, 2H), 7.40-7.30 (m, 10H), 5.46-5.36 (m, 1H), 5.09-4.96 (m, 4H). 4.73-4.62 (m, 1H), 4.28-4.16 (m, 2H), 3.99-3.86 (m, 1H), 3.85-3.60 (m, 3H), 3.56-3.45 (m, 1H), 3.31-3.14 (m, 2H), 1.99-1.83 (m, 1H), 1.67-1.45 (m,4H), 1.44-1.3 (m, 1H).
Sfep 4. Synthesis of(2R)-1,1,1-trifluoro-3-(phosphonooxy)propan-2-yl (3R)-330 [(phenylsulfonyl)amino)-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C83).
A solution of C82 (1.80 g, 2.53 mmol) In methanol (50 mL) was treated with 10% palladium on carbon (180 mg) and hydrogenated at 25 psi using a Parr reactorfor 4 hours at room température. The reaction mixture was filtered through diatomaceous earth, and the filtrate was concentrated in vacuo to provide an oil, which was taken up In methanol (20 mL) 35 and again concentrated under reduced pressure. The product was obtained as a brittle foam.
Yield: 1.14 g, 2.14 mmol, 85%. LCMS m/z 533.0 [M+H]*. ’H NMR (400 MHz, CDCh) δ 7.87 (br d, 4=8 Hz, 2H), 7.63-7.57 (m, 1H), 7.56-7.49 (m, 2H), 5.53-5.41 (m, 1H), 4.39-4.15 (m, 2H), 3.98-3.18 (m, 7H), 2.06-1.92 (m, 1 H), 1.88-1.43 (m, 5H).
132
Step 5. Synthesis of (2R)-3,3,3-trifluoro-2-[(((3R)-3-[(phenylsu!fonyl)amino]-1-oxa-8· azaspiro[4.5]dec-8-yl}carbonyl)oxy]propyl phosphate, disodium sait (31).
Sodium tert-butoxide (2 M solution in tetrahydrofuran, 1.98 mL, 3.96 mmol) was added drop-wise over 5 minutes to a 0 *C solution of C83 (1.08 g, 2.03 mmol) in acetonitrile (20 mL), and the reaction mixture was allowed to warm to room température and stir for 2 hours. The resulting solid was collected on a Teflon filter, affording the product as a white solid. Yield: 1.02 g, 1.77 mmol, 87%. LCMS m/z 532.9 [M+H]*. Ή NMR (400 MHz, D2O) δ 7.90 (br d, J=8 Hz, 2H), 7.77-7.71 (m, 1H), 7.66 (br dd, J=8, 8 Hz, 2H), 5.47-5.37 (m, 1 H), 4.14-4.05 (m, 1H), 4.023.86 (m, 3H), 3.68-3.31 (m, 5H), 2.08-1.97 (m, 1H), 1.80-1.49 (m, 5H).
Example 32 (2R)-3,3,3-Trifluoro-2-[((4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl)carbonyl)oxy]propylphosphate, disodium sait (32)
O CF3
AACoh
O ÇFa
t-BuONa
Step 1. Synthesis of (2R)-1,1,1-trifluoro-3-(phosphonooxy)pmpan-2-yl 4-((4fluorophenyl)sulfonyl]-l-oxa-4,9-diazaspiro(5.5]undecane-9-carboxylate (C84).
4-Methylmorpholine (14.5 mL, 132 mmol) was added to a solution of 6 (12.3 g, 26.1 mmol) in acetonitrile (750 mL) and the reaction mixture was cooled to -10 eC in an ice-salt bath. Phosphores oxychloride (2.9 mL, 31 mmol) was added over 1 minute with vîgorous stirring, and the reaction mixture was allowed to stir at -10 °C for one hour, whereupon it was poured Into ice water (500 mL) and stirred for 1.5 hours to ensure complété quench of excess reagent. After concentration ofthe mixture to approximately one-half its original volume, the remaining liquid was extracted with ethyl acetate (1 L), and the organic layer was washed sequentially with aqueous hydrochloric acid (1 M; 3 x 300 mL) and with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated In vacuo to afford the product as a brittle foam (15.0 g) containing some ethyl acetate by 1H NMR analysis. Yield, corrected for
133
ethyl acetate: 14.2 g, 25.8 mmol, 99%. LCMS m/z 550.9 [M+H]*. 1H NMR (500 MHz, CDCb), characteristlc peaks: δ 7.81-7.73 (m, 2H), 5.57-5.48 (m, 1 H>, 4.45-4.34 (m, 1H), 4.32-4.20 (m, 1H), 3.97-3.74 (m, 4H), 3.35-3.11 (m, 2H), 3.06-2.89 (m, 2H), 2.89-2.72 (m, 2H), 2.03-1.87 (m, 2H), 1.68-1.46 (m, 2H).
Step 2. Synth esis of (2R)-3,3,3-trifluoro-2-[((4-[(4-fluorophenyl) sulfonyl]- 1-oxa-4,9diazaspiro[5.5]undec-9-yl}carbonyl)oxyJpropyl phosphate, disodium sait (32).
A stirring solution of C84 (20.0 g, 36.3 mmol) in water (1.2 L) was treated with solid sodium bicarbonate until the pH of the mixture was approximately 7. The mixture was washed with ethyl acetate (500 mL), and the aqueous layer was acidified to pH 1.5 - 2 via portion-wise 10 addition of concentrated hydrochloric acid. It was then extracted with ethyl acetate (1.5 L); the organic layer was washed with saturated aqueous sodium chloride solution (100 mL), dried over sodium sulfate, filtered, and concentrated In vacuo to provide a white solid (20 g). This material was dissolved in acetonitrile (600 mL), cooled to 0 ’C, and treated in a drop-wise manner over 5 minutes with a solution of sodium fert-butoxide in tetrahydrofuran (2 M; 35.4 mL, 70.9 mmol).
After the reaction had stirred for one hour at 0 ’C, it was concentrated under reduced pressure to afford a solid (21.4 g). This material was mixed with éthanol (30 mL) and stirred at room température for 30 minutes, whereupon the solid was collected via filtration to provide the product as a solid (21.3 g) that contained some solvents via 1H NMR analysis. Yield, corrected for solvents: 20.8 g, 35.0 mmol, 96%. LCMS m/z 551.3 [M+H]*. 1H NMR (500 MHz, D2O) δ 7.8720 7.81 (m, 2H), 7.37 (dd, >8.9, 8.7 Hz, 2H), 5.46-5.39 (m, 1H), 4.12-4.05 (m, 1H), 4.01-3.94 (m,
1H), 3.93-3.8 (m, 1H), 3.83 (dd, >5.0, 4.8 Hz, 2H), 3.78-3.65 (m, 1H), 3.34-3.13 (m, 2H), 3.102.98 (m, 2H), 2.97-2.85 (m, 2H), 1.99-1.81 (m, 2H), 1.75-1.51 (m, 2H).
Altemate Synthesis ofExampie 32 (2R)-3,3,3-Trifluoro-2-[({4-[(4-f!uorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-925 yl}carbonyl)oxy]propyl phosphate, disodium sait (32)
Br^Br
F
134
NaOH
Step 1. Synthesis of 4-[(4-fluomphenyf)su/fony/J-1-oxa-4,9-d/azaspifO[5.5Jundecane, paratoluenesulfonic acid sait (C85).
Potassium carbonate (24.0 g, 174 mmol) was added to a solution of tert-butyl 4(aminomethyl)-4-hydroxypiperldîne-1-carboxylate (5.00 g, 21.7 mmol) ln acetonitrile (35 mL), and the reaction mixture was allowed to stir for 5 minutes. A solution of 4-fluorobenzenesulfonyl chloride (4.31 g, 22.1 mmol) in acetonitrile (15 mL) was slowly added overfive minutes, and the resulting suspension was stirred at 25 ’C ; after 1 hour, 1,2-dibromoethane (7.50 mL, 87.0 mmol) was added, and the reaction mixture was heated at 80 'C for 27 hours, whereupon it was cooled to 25 ’C and filtered. The reaction flask was rinsed with acetonitrile (2x18 mL), and the combined filtrâtes were concentrated under reduced pressure and diluted with ethyl acetate (72 mL). para-Toluenesulfonic acid monohydrate (8.38 g, 44.0 mmol) was added in one portion, and the reaction mixture was stirred at room température for 10 minutes, until a solution was obtained. It was then heated at 50 ’C for 1.5 hours, at which point it was cooled to 25 ’C and stirred for 2 hours to granulate the precipitate. This material was collected via filtration and rinsed with ethyl acetate, affording the product as a white solid. Yield: 7.26 g, 14.9 mmol, 69%. ’H NMR (600 MHz, CD3OD) δ 7.84 (br dd, J=8, 5 Hz, 2H), 7.71 (br d, J=7.9 Hz, 2H), 7.38 (br dd, J=8.5, 8.5 Hz. 2H), 7.24 (br d, J=7.9 Hz, 2H), 3.81 (dd, J=5.0,4.7 Hz, 2H), 3.26-3.20 (m, 2H),
3.19-3.12 (m, 2H), 3.03-2.98 (m, 2H), 2.86 (brs, 2H), 2.37 (s, 3H), 2.20 (brd, J=14.4 Hz, 2H),
1.74-1.67 (m, 2H).
Step 2. Synthesis of (2R)-3-{[bis(benzyloxy)phosphofyl]oxy}-1,1,1-trifluoropropan-2-yl 4-((4fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro(5.5]undecane-9-carboxylate (C86).
135
A solution of C80 (28.0 g, 71.7 mmol) in acetonitrile (75 mL) was added over 15 minutes to a mixture of 1,T-carbonyldiimidazole (97%, 12.6 g, 77.7 mmol) In acetonitrile (93 mL). The C80 solution was rinsed in with acetonitrile (5 mL) and the reaction mixture was allowed to stir at room température for 30 minutes. Compound C85 (37.0 g, 76.0 mmol) was added In one portion, and stirring was continued at room température for 6 hours, whereupon the reaction mixture was concentrated in vacuo. The residue was mixed with ethyl acetate (520 mL), and the mixture was washed twice with water (2 x 260 mL), then concentrated under reduced pressure. The residue was dissolved in a mixture of ethyl acetate and heptane (1:1,206 mL) and eluted through a pad of silica gel (150 g) using a mixture of ethyl acetate and heptane (1:1,1.3 L).
Fractions containing the product were combined and concentrated under reduced pressure to provide the product. Yield: 42.1 g, 57.6 mmol, 80%. Ή NMR (600 MHz, CD3CN) 8 7.80-7.74 (m, 2H), 7.44-7.34 (m, 10H), 7.34 (dd, J=8.8, 8.7 Hz, 2H), 5.52-5.46 (m, 1H), 5.09-4.99 (m, 4H), 4.35-4.21 (m, 2H), 3.77-3.67 (m, 4H), 3.16-3.02 (m, 2H), 2.96-2.86 (m, 2H), 2.79-2.63 (m, 2H), 1.86-1.72 (m, 2H), 1.51-1.26 (m, 2H).
Step 3. Synthesis of (2RJ-1,1,1-trifluoro-3-(phosphonooxy)propan-2-yl 4-[(4fJuorophenyl)sulfonyl]- 1-oxa~4,9-diazaspiro[5.5]undecane-9-carboxyla te (C84).
A solution of C86 (2.0 grams, 2.7 mmol) in tetrahydrofuran (26 mL) was added to 5% palladium on carbon (Evonik Noblyst P1142; 40 mg) in a Biotage Atlantis reactor. Additional tetrahydrofuran (4.0 mL) was used to rinse the vessel containing starting material; this was 20 added to the reaction mixture. The reactor was purged three times with nitrogen while the reaction mixture was stirred, and then three times with hydrogen without stirring. The hydrogen pressure was brought to 5 pslg at 25 ’C, and then to 15 pslg. The agitation was increased to 1200 rpm for 4 hours, whereupon the reactor was purged three times with nitrogen, and the réaction mixture was filtered. The filter cake was rinsed with tetrahydrofuran (20 mL), the 25 combined filtrâtes were concentrated in vacuo, and the residue was dissolved in tert-butyl methyl ether (300 mL) and concentrated again. This dissolution/concentration was repeated, affording the product as a white foam. Yield: 1.35 g, 2.45 mmol, 91%.
Step 4. Synthesis of(2R)-3,3,3-trifluoro-2-[({4-[(4-fIuorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5JundeC‘9-yl}caitonyl)oxy]propylphosphate, disodium sait (32)
Aqueous sodium hydroxide solution (1 M, 12.0 mL, 12.0 mmol) was added drop-wise over 1 minute to a solution of C84 (97%, 3.50 g, 6.17 mmol) In éthanol (35.0 mL). The reaction mixture was stirred at room température for 1.5 hours; éthanol (120 mL) was added, and stirring was continued for 30 minutes, whereupon the reaction mixture was filtered. The filter cake was washed with éthanol (25 mL) to provide the product as a white solid. Yield: 2.88 g, 4.84 mmol,
78%. Ή NMR (600 MHz, D2O) 8 7.85 (br dd, J=7, 5 Hz, 2H), 7.38 (br dd, J=9,8 Hz, 2H), 5.47-
5.39 (m, 1H), 4.12-4.06 (m, 1H), 4.01-3.95 (m, 1H), 3.94-3.66 (m, 2H), 3.84 (brdd, J=5, 4 Hz, 2H), 3.35-3.15 (m, 2H), 3.11-3.00 (m, 2H), 2.98-2.86 (m, 2H), 2.00-1.82 (m, 2H), 1.76-1.52 (m, 2H).
136
Example 33 (2R)-3,3,3-Tnf]uoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, (bls)-L-lysine sait (33)
A solution of L-lysine (3.63 g, 24.8 mmol) In water (14 mL) was added to a solution of
C84 (7.00 g, 12.7 mmol) In methanol (56 mL). The lysine solution was rinsed In with water (3 mL), and the reaction mixture was stirred at room température. Methanol (280 mL) was added to improve stirring of the slurry, and stirring was continued at room température for 1 hour. The reaction mixture was heated to 40 ’C and stirred for 30 minutes, then cooled to 0 ’C to 5 ’C with stirring. After being held at 0 ’C for 30 minutes, It was warmed to room température and stirred for 30 minutes, whereupon it was filtered through a Büchner funnel. The collected material was washed with methanol (140 mL) to afford a white solid (9.44 g). The buik of this material (8.44 g) was slurried in methanol (140 mL) and stirred at room température for 4 hours, whereupon it was filtered through a Büchner funnel, providing the product as a white solid. Yield: 8.24 g, 9.77 mmol, 86% (corrected for material that was removed prior to reslurry). LCMS m/z 551.2 [M+H]+. 1H NMR (400 MHz, D2O) δ 7.88-7.81 (m, 2H), 7.38 (br dd, J=8.8, 8.8 Hz, 2H), 5.48-5.38 (m, 1H), 4.13-4.05 (m, 1 H), 4.03-3.94 (m, 1H), 3.94-3.8 (m, 1H), 3.84 (br dd, J=5.0,4.9 Hz, 2H),
3.79-3.64 (m, 1H), 3.71 (dd, J-6.2, 6.0 Hz, 2H), 3.36-3.13 (m, 2H), 3.10-3.02 (m, 2H), 2.99 (dd, J=7.7, 7.5 Hz, 4H), 2.95-2.86 (m, 2H), 2.01-1.81 (m, 6H), 1.76-1.54 (m, 6H), 1.54-1.34 (m, 4H).
Example 34 (2R)-3,3,3-Trifluoro-2-[({4-[(3-!luoropheny1)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, disodium sait (34)
137
NaOH
Step 1. Synthesis of(2R)-1,1,1-trifluom-3-(phosphonooxy)propan-2-yl 4-((3fluorophenyl)sulfonyl]- 1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate (C87).
Diphosphoryl tetrachloride (2.63 mL, 19.0 mmol) was added drop-wise over 5 minutes to 5 a 0 'C solution of 11 (1.74 g, 3.70 mmol) In acetonitrile (20 mL), and the réaction mixture was stirred at 0 °C for 3 hours, whereupon it was poured into Ice (20 g) and stirred at room température for 1.75 hours. The reaction mixture was concentrated in vacuo, and the aqueous residue was partitioned between ethyl acetate (50 mL) and aqueous hydrochloric acid (1 M; 10 mL); the organic layer was washed sequentially with aqueous hydrochloric acid (1 M; 10 mL) 10 and saturated aqueous sodium chloride solution (2x10 mL), then dried over sodium sulfate, filtered, and concentrated in vacuo. The resulting thick oil was taken up in water (75 mL), basified via addition of saturated aqueous sodium bicarbonate solution and solid sodium bicarbonate, and washed with ethyl acetate (50 mL). The pH of the aqueous layer was then adjusted to -2 using concentrated hydrochloric acid, and the product was extracted with ethyl 15 acetate (2 x 50 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was mixed with ethyl acetate and filtered through a 0.45 pm membrane filter; the filtrate was concentrated under reduced pressure to provide the product as a white solid. Yield: 1.36 g, 2.47 mmol, 67%. LCMS m/z 551.1 [M+H]*. 1H NMR (400 MHz, DMSO-de) δ 7.76-7.70 (m. 1H), 7.65-7.55 (m, 3H), 5.50-5.40 (m, 1H), 4.13-3.98 (m, 2H), 3.7720 3.63 (m, 4H), 3.21-3.02 (m, 2H), 2.98-2.88 (m, 2H), 2.84-2.73 (m, 2H), 1.88-1.71 (m, 2H), 1.621.38 (m, 2H).
Sfep 2. Synthesis of (2R)-3,3,3-trifluoro-2-[((4-[(3-fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undec-9-yl}carbonyl)oxy]propylphosphate, disodium sait (34).
Aqueous sodium hydroxide solution (1 M, 4.78 mL, 4.78 mmol) was added drop-wise 25 over 3 minutes to a solution of C87 (1.35 g, 2.45 mmol) in éthanol (15 mL), and the reaction mixture was allowed to stîr at room température for 1 hour. Ethanol (50 mL) was then added to the suspension, which was allowed to stir for 5 minutes before being filtered. The filter cake was
138
1)SOCI2 o CF3
ΑθΑ^ΟΡΜΒ
2)
C31
H________ r
3) CF3COOH
rinsed with éthanol (10 mL) to afford the product as a white solid. Yield: 1.01 g, 1.70 mmol, 69%. LCMS m/z 551.1 [M+H]*. ’H NMR (400 MHz, D2O) δ 7.67 (ddd, half of ABXY pattern, 7=8.0, 7.8, 5.2 Hz, 1H), 7.62 (ddd, half of ABXY pattern, 7=7.8,1.4,1.3 Hz, 1H), 7.60-7.56 (m, 1H), 7.49 (dddd, 7=8.7, 8.0, 2.5, 1 Hz, 1H), 5.47-5.38 (m, 1H), 4.13-4.05 (m, 1H), 4.02-3.93 (m,
1H), 3.93-3.64 (m, 2H), 3.84 (dd, 7=5.2,4.8 Hz, 2H), 3.36-3.13 (m, 2H), 3.13-3.01 (m, 2H), 3.012.87 (m, 2H), 2.02-1.81 (m, 2H), 1.77-1.50 (m, 2H).
Method A
Method A describes a spécifie synthetic method for préparations of certain exemplar compounds of the Invention.
Synthesis of(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(R70-sulfonyl)-1-oxa-4,9diaza$piro[5.5]undecane-9-carboxylate analogues (MA-2) via sulfonylation of C31 followedby de protection
O
O=s—OH I R70
MA-1
A solution of the sulfbnic acid MA-1 (0.15 mmol) In ΛΙ,Ν-dimethylformamide (0.15 mL) 15 was treated with thionyl chloride (0.12 ml, 1.6 mmol), and the reaction mixture was heated with shaking at 50 *C for 16 hours. Volatiles were removed using a Genevac evaporator; 1,2dichloroethane (2 mL) was added, and the mixture was concentrated again. A solution of C31 (25.9 mg, 60.0 mmol) in 1,2-dichloroethane (0.5 mL) was added to the crude sulfonyl chloride, followed by Ν,/V-diisopropylethylamine (0.225 mL, 1.29 mmol), and the réaction mixture was 20 shaken ovemight at room température. It was then partitioned between half-saturated aqueous sodium bicarbonate solution (1.5 mL) and ethyl acetate (2.4 mL) and subjected to vortexîng. The organic layer was eluted through a solid phase extraction cartridge (6 mL) charged with sodium sulfate (-1 g); this extraction procedure was repeated twice, and the combined eluents were concentrated In vacuo. A mixture of trifluoroacetic acid and 1,2-dichloroethane (1:1, 1 mL) 25 was added, and the reaction mixture was shaken at room température for 2 hours, whereupon it was concentrated in vacuo and subjected to purification via reverscd phase HPLC (Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 5% to 100% B).
Table 6. Method of synthesis, structure, and physicochemical properties for Examples 35 - 91.
Exam Method Structure ’H NMR (400 MHz, CDCI3) δ; Mass
139
pie Numb er of Synthesi s; Noncommerc ial starting materials spectrum, observed ion m/z [M+H]* or HPLC rétention time; Mass spectrum m/z [M+H]* (unless otherwise indicated)
35 Footnote s1,2 0 CF3 <'νΛοΛ-οη N^N ό 0 8.25 (d, >5.1 Hz, 1H), 6.41 (d, >5.0 Hz, 1H), 5.33-5.22 (m, 1H), 4.34-4.19 (m, 2H), 4.06-3.99 (m, 1H), 3.89 (brdd, half of ABX pattern, >12,7 Hz, 1H), 3.83-3.74 (m, 8H), 3.08-2.88 (m, 2H), 2.68 (tt, >11.5, 3.6 Hz, 1H), 1.99-1.88 (m. 2H), 1.84-1.69 (m, 2H); 405.1
36 C1934 o ÇF3 CF3 n-n 0 F By 1H NMR analysis, this was judged to be a mixture of rotamers. ’H NMR (400 MHz, CDaOD) δ 8.03 (d, >2.5 Hz. 1H), 7.72-7.66 (m, 2H), 7.20 (br dd, >8.9,8.5 Hz, 2H), [6.27 (d, >2.5 Hz) and 6.26 (d, >2.5 Hz), total 1 H]t 5.50-5.40 (m, 1H), 4.43-4.33 (m, 1H), 3.83-3.76 (m, 2H), 3.63-3.55 (m, 2H). 2.08-2.01 (m, 2H), [1.77 (dd, >3.6, 3.5 Hz) and 1.72 (dd, >3.4, 3.4 Hz), total 1H]; 468.0
37 Example 3S; C6, C17 0 CF3 Η^ιΛο^θπ fT'h N-N b By Ή NMR analysis, this was judged to be a mixture of rotamers. 1H NMR (400 MHz, CDjOD) δ 8.51 (br d, >5 Hz, 1H), 7.79 (ddd, >7.8, 7.8,1.5 Hz, 1H), 7.64 (d, >2.3 Hz, 1H), 7.33 (ddd, >7.3,5.0, 0.6 Hz, 1H), 7.03-6.98 (m, 1H), [6.11 (d, >2.3 Hz) and 6.10 (d, >2.4 Hz), total 1H], 5.37 (s, 2H), 5.32-5.22 (m, 1H), 3.90-3.71 (m, 4H), 3 653.52 (m, 2H), 1.98-1.91 (m, 2H), [1.76 (dd, >3.5, 3.4 Hz) and 1.70 (dd, >3.6, 3.4 Hz), total 1H]; 396.9
140
38 Example 6e7; Cl 3 0 cf3 <·νλ<Α- n-n 0 ’H NMR (400 MHz, CD3OD) δ 7.58 (d, J=2.4 Hz, 1H), 6.13 (d, J=2.4 Hz, 1H), 5.35-5.26 (m, 1H), 4.38-4.28 (m, 1H), 4.27-4.14 (m, 2H), 4.09-4.01 (m, 2H), 3.88 (br dd, half of ABX pattern, J=12, 4 Hz, 1H), 3.79 (dd, half of ABX pattern, J=12.4, 6.9 Hz, 1H), 3.603.51 (m, 2H), 3.13-2.94 (m, 2H), 2.92-2.83 (m, 1H), 2.10-1.87 (m, 6H), 1.76-1.53 (m, 2H); 391.9
39 Example s 19,20 and 21; C19, C59 o cf3 h^nAtSHoh N-N 0 F By ’H NMR analysis, this was judged to be a mixture of rotamers. ’H NMR (600 MHz, DMSO-di) δ 8.33-8.30 (m, 1H), 7.81-7.76 (m, 2H), 7.30 (br dd, J=8.9, 8.8 Hz, 2H), [6.33 (d, J=2.4 Hz) and 6.31 (d, J=2.4 Hz), total 1 H], 5.88-5.83 (m, 1H), 5.82-5.73 (m, 1H), 3.83-3.56 (m, 6H), 2.06-1.98 (m, 2H), [1.82 (dd, J=3.4, 3.4 Hz) and 1.76 (dd, J=3.4, 3.3 Hz), total 1 H]; 450.2
40 Example 13; C17 0 CFa F<r% ’H NMR (400 MHz, CDjOD) δ 7.94-7.87 (m, 2H), 7.40-7.32 (m, 2H), 5.29-5.18 (m, 1H), 3.90-3.81 (m, 1H), 3.79-3.69 (m, 1H), 3.683.51 (m, 2H), 3.49-3.25 (m, 3H, assumed; partially obscured by solvent peak), 3.21- 3.00 (m, 3H), 2.96-2.82 (m, 2H); 426.9
41 Example 6e; C2 0 cf3 γΛοΛ-οη œ ‘o.. 7,32-7.21 (m, 2H, assumed; partially obscured by solvent peak), 7.05-6.95 (m, 2H), 5.27-5.16 (m, 1H), 4.03-3.93 (m, 1H), 3.90-3.80 (m, 1H), 3.55-3.35 (m, 2H). 3.40 (br s, 2H), 3.35-3.16 (m, 2H), 2.51-2.32 (m, 3H), 2.21-2.06 (m, 2H), 1.71-1.41 (m, 6H, assumed; partially obscured by water peak), 1.41-1.30 (m, 2H); 419.2
141
42 Example 23; C23, C2 C°' Sr 0 CF3 -ΆεΑ-θπ aF 7.31-7.23 (m, 2H, assumed; partially obscured by solvent peak), 7.01 (br dd, J=8.8, 8.5 Hz, 2H), 5.28-5.18 (m, 1H), 3.99 (br dd, half of ABX pattern, J=12, 3 Hz, 1H), 3.91-3.77 (m, 3H), 3.77-3.70 (m, 2H), 3.463.36 (m, 2H), 3.33-3.15 (m, 2H), 2.48-2.38 (m, 2H), 2.25-2.15 (m, 2H), 2.13-1.97 (m, 2H), 1.48-1.36 (m, 2H); 421.1
43 Example 139; C23, C2 d Sr 0 CF3 AtA-0H x> •HCOOH Ck 7.31-7.23 (m, 2H, assumed; partially obscured by solvent peak), 7.01 (br dd, J=8.8, 8.5 Hz, 2H), 5.27-5.18 (m, 1H), 4.023.95 (m, 1H), 3.90-3.69 (m, 5H), 3.36-3.13 (m, 3H), 2.69-2.55 (m, 1H), 2.40-2.05 (m, 5H), 1.98-1.86 (m, 1H), 1.45-1.3 (m, 2H), 1.31 (d, J=6.5 Hz, 3H); 435.2
44 Example 1310 c A 0 CF3 —. —-OH DIAST-1 aF 7.26 (br dd, J=8.5, 5.6 Hz, 2H), 7.00 (br dd, J=8.8, 8.7 Hz, 2H), 5.28-5.17 (m, 1H), 4.03- 3.94 (m, 1H), 3.90-3.67 (m, 5H), 3.33-3.14 (m, 3H), 2.65-2.44 (m, 2H), 2.36-2.27 (m, 1H), 2.24-2.05 (m, 3H), 2.02-1.84 (m, 1H), 1.44-1.32 (m, 2H), 1.29 (d, J=6.5 Hz, 3H); 435.2
45 Example 1310 c0' A 0 CF3 A'N'^‘Ox*x*zOH DIAST-2 aF 7.30-7.22 (m, 2H), 7.00 (br dd, J=8.7, 8.7 Hz, 2H), 5.27-5.17 (m, 1H), 4.02-3.94 (m, 1H), 3.89-3.67 (m, 5H), 3.32-3.12 (m, 3H), 2.68-2.53 (m, 1H), 2.36-2.25 (m, 1H), 2.242.05 (m, 3H), 1.98-1.63 (m, 2H). 1.44-1.32 (m, 2H), 1.28 (d, J=6.6 Hz, 3H); 435.2
46 Example 24”; C2 o cf3 ’H NMR (400 MHz, CD3OD) δ 7.42-7.33 (m, 2H), 7.02 (br dd, J=8.5, 8.5 Hz, 2H), 5.335.22 (m, 1H), 4.01-3.91 (m, 2H), 3.91-3.72 (m, 6H), 3.3-3.12 (m, 4H, assumed; partially obscured by solvent peak), 2.83-2.71 (m, 1H), 2.55 (s, 2H), 1.78-1.69 (m, 2H), 1.671.43 (m, 6H); 479.1
142
47 Example 13; C2 0 cf3 hnJ3 ’H NMR (400 MHz, CD3OD) δ 7.95-7.88 (m, 2H), 7.32 (br dd, J=8.8,8.8 Hz, 2H), 5.33- 5.24 (m, 1 H), 4.21-4.08 (m, 2H), 3.88 (br dd, haîf of ABX pattern, J=12, 4 Hz, 1H), 3.78 (dd, half of ABX pattern, J=12.3,6.7 Hz, 1H), 2.95-2.76 (m, 2H), 2.77 (d, J=6.5 Hz, 2H), 1.79-1.61 (m, 3H), 1.24-1.01 (m, 2H); 429.0
48 Example 14; C2 o cf3 *V'nAoA-oh r NYNA ï By Ή NMR analysis, this was judged to be a mixture of rotamers. [8.01 (d, J=6.0 Hz) and 7.96 (d, J=6.0 Hz), total 1 H], 6.35-6.23 (m, 2H), 5.32-5.21 (m, 1H), 4.03-3.64 (m, 7H), 3.61-3.38 (m, 3H), 3.18-3.07 (m, 2H), 1.93-1.82 (m, 1H), 1.22-1.11 (m, 2H), 0.920.83 (m, 2H); 386.0
49 Example 13; C23, C2 o cf3 _ |^nAo^oh c°p N O=S=O 0 5.32-5.20 (m, 1H), 4.14-4.04 (m, 2H), 4.043.95 (m, 1H), 3.95-3.80 (m, 3H), 3.76 (br dd, J=4.9, 4.8 Hz, 2H), 3.45-3.32 (m, 4H), 3.323.10 (m, 5H), 2.53-2.34 (m, 1H), 2.05-1.80 (m, 6H), 1.60-1.43 (m, 2H); 461.1
50 Example 23”; C2 o cf3 F 7.31-7.23 (m, 2H. assumed; partîally obscured by solvent peak), 7.00 (br dd, J-8.5, 8.5 Hz, 2H), 5.27-5,18 (m, 1H), 4.183.95 (m, 5H), 3.90-3.81 (m, 1H), 3.60 (s, 2H), 3.35-3.26 (m, 2H), 2.87-2.59 (m, 3H), 2.47-2.38 (m, 1H), 2.35 (d, A6.8 Hz, 2H), 1.84-1.73 (m, 2H), 1.70-1.46 (m, 4H, assumed; partîally obscured by solvent peak), 1.09-0.92 (m, 2H); 463.2
51 Example 13; C23, C2 o cf3 n <'νΛοΛ-c o=s=o φ. F 7.63-7.53 (m, 2H), 7.18 (dd, J=9.0, 8.7 Hz, 1 H), 5.31-5.21 (m, 1H). 4.0G-3.96 (m, 1 H), 3.95-3.82 (m, 3H), 3.80 (br dd, J=5.0, 5.0 Hz, 2H), 3.32-3.13 (m, 2H), 3.09-2.91 (m, 2H), 2.89-2.71 (m, 2H), 2.43-2.24 (m, 1H), 2.37 (br s, 3H), 2.05-1.91 (m, 2H), 1.69-1.43 (m, 2H, assumed; partîally obscured by
143
water peak); 485.1
52 Example 13; C23, C2 0 cf3 rNA0+-0H C o=s=o ¢1., F 7.59 (ddd, J-9.2,7.2, 2.1 Hz, 1H), 7.56-7.50 (m, 1H), 7.38 (ddd, J=9.0, 8.8,7.4 Hz, 1H), 5.32- 5.21 (m, 1H), 4.05-3.97 (m, 1H), 3.963.83 (m, 3H), 3.81 (dd, J=4.9,4.9 Hz, 2H), 3.32- 3.13 (m, 2H), 3.10-2.95 (m, 2H), 2.892.75 (m, 2H), 2.5-2.2 (brm, 1H), 2.05-1.92 (m, 2H), 1.6-1.43 (m, 2H); 489.1
53 Example 13; C2 0 ÇF3 <nAoA-'oh V cf3 8.42 (br s, 1 H), 7.79 (br d, A8.8 Hz, 1 H), 6.81 (d, J=8.8 Hz, 1H), 5.41-5.32 (m, 1H), 5.32-5.23 (m, 1H). 4.06-3.98 (m, 1H), 3.933.72 (m, 3H), 3.59-3.40 (m, 2H), 2.55-2.32 (m, 1H), 2.12-1.95 (m, 2H), 1.92-1.75 (m, 2H); 403.0
54 Method A o cf3 pA^0H _ A · CF3COOH o=s=o \> 1.65 minutes13; 488.2
55 Method A 0 CF3 ρΑοΑ,θπ · CF3C00H o=s=o r^N U 1.65 minutes13; 482.1
144
56 Method A O cf3 cP n A n · CF,COOH o=s=o 3 1.63 minutes13; 485.1
57 Example 10; C2 0 CF3 Pn 9 7.97-7.92 (m, 1H), 7.44-7.39 (m, 1H), 6.65 (d, >8.0 Hz, 1H), 5.32-5.20 (m, 2H), 4.02 (dd, half of ABX pattern, >12.4, 3.0 Hz, 1H), 3.89 (dd, half of ABX pattern, >12.5, 6.8 Hz, 1H), 3.88-3.72 (m, 2H), 3.57-3.41 (m, 2H), 2.25 (s, 3H), 2.07-1.96 (m, 2H), 1.94-1.73 (m, 2H, assumed; partially obscured by water peak); 348.9
58 Example 10; C2 0 ÇF3 <A> tpN . Ύ 8.46 (s, 1H), 7.57 (d. >2.0 Hz, 1 H), 6.55 (d, >2.0 Hz, 1H). 5.32-5.22 (m, 2H), 4.07 (s, 3H), 4.05-3.98 (m, 1H), 3.93-3.73 (m, 3H), 3.63-3.45 (m, 2H), 2.32 (s, 3H), 2.10-1.87 (m, 4H); 430.1
59 Example 13; C23, C2 0 CF3 n |^NAcA^0H cP n ο=έ=ο 7.72 (d, >1.5 Hz, 1 H), 7.52 (dd, half of ABX pattern, >8.0,1.6 Hz, 1H), 7.43 (d, half of AB quartet, >8.3 Hz, 1H), 5.31-5.21 (m, 1H), 4.06-3.96 (m, 1H), 3.95-3.82 (m, 3H), 3.80 (dd, >5.0, 4.8 Hz, 2H), 3.32-3.13 (m, 2H), 3.10-2.94 (m, 2H), 2.89-2.73 (m, 2H), 2.48 (s, 3H), 2.39-2.26 (m, 1H), 2.05-1.92 (m, 2H), 1.6-1.44 (m, 2H, assumed; partially obscured by water peak); 523.1 [M+Na*]
60 Example 13; C23, C2 X O O^° 7.82 (dd, >6.6, 2.3 Hz, 1H), 7.65 (ddd, >8.6,4.3, 2.3 Hz, 1H), 7.34 (dJ, >8.5, 8.4 Hz, 1H), 5.31-5.21 (m, 1H), 4.05-3.97 (m, 1H), 3.96-3.83 (m, 3H), 3.81 (dd, >5.0,4.9 Hz, 2H), 3.31-3.13 (m, 2H), 3.11-2.96 (m, 2H), 2.90-2.75 (m, 2H), 2.35-2.23 (m, 1 H), 2.04-1.93 (m, 2H), 1.6-1.45 (m, 2H,
145
assumed; partially obscured by water peak); 527.1 [M+Na*J
61 Example 10; C2 O cf3 t 1H NMR (400 MHz, CDjOD) δ 7.73 (br s, 1 H), 7.34 (br s, 1 H), 5.36-5.27 (m, 1 H), 5.27-5.19 (m, 1H), 3.89 (dd, half of ABX pattern, >12.4,3.8 Hz, 1H), 3.84-3.69 (m, 2H), 3.79 (dd, half of ABX pattern, >12.4, 6.6 Hz, 1H), 3.60-3.44 (m, 2H), 2.20 (s, 3H), 2.16 (s, 3H), 2.08-1.92 (m, 2H), 1.86-1.68 (m. 2H), 363.0
62 Example 1314; C2 0 CF3 <'N'S'o+-'OH 7.84-7.77 (m, 2H), 7.23 (dd, >8.7, 8.5 Hz, 2H), 5.31-5.20 (m, 1H), 4.28-4.12 (m, 2H), 4.05-3.97 (m, 1H), 3.88 (dd, half of ABX pattern, >12.5, 7 Hz, 1H), 2.96-2.78 (m, 4H), 2.76 (s, 3H), 1.88-1.75 (m, 3H), 1.301.15 (m,2H); 443.1
63 Example 10; C2 O cf3 Âf01 cf3 8.33-8.30 (m, 1 H), 7.87 (d, >2.3 Hz, 1H), 5.50-5.41 (m, 1H), 5.33-5.24 (m, 1H), 4.073.98 (m, 1H), 3.94-3.85 (m, 1 H). 3.83-3.54 (m, 4H), 2.38-2.27 (m, 1H), 2.09-1.87 (m, 3H); 437.0
64 Example 1O1S; C2 o cf3 jQnAo^0H XN 8.21 (d, >5.4 Hz, 1H), 7.54 (d, >1.9 Hz, 1H), 6.95 (dd, >5.3,1.5 Hz, 1H), 6.80-6.78 (m, 1H), 6.41 (d, >2.0 Hz, 1H), 5.39-5.31 (m, 1H), 5.31-5.24 (m, 1 H), 4.03 (dd, half of ABX pattern, >12,3 Hz, 1H), 3.97 (s, 3H), 3.92-3.76 (m, 2H), 3.90 (dd, >13, 7 Hz, 1H), 3.59-3.42 (m, 2H), 2.12-1.98 (m, 2H), 1.92-1.79 (m, 2H); 415.1
146
65 Example 10; C2 o cf3 ρ,Αθ^,ΟΗ r n i 7.65 (dd. >7.9, 7.8 Hz, 1H), 7.49 (d, >1.6 Hz, 1H), 7.20 (d, >7.4 Hz, 1H), 6.70 (d, >8.3 Hz, 1H), 6.57 (d, >1.8 Hz, 1H), 5.37- 5.24 (m, 2H), 4.21 (s, 3H), 4.06-3.97 (m, 1H), 3.93-3.69 (m, 3H), 3.62-3.45 (m. 2H), 2.72-2.64 (m, 1H), 2.09-1.96 (m, 2H), 1.95- 1.83 (m, 2H); 415.1
66 C3117 0 cf3 pAo^OH o-s=o Ôy 3.22 minutes18; 495
67 Example 7”; C31 o cf3 pAoH V o=s=o hn. ό 7.42-7.30 (m, 5H), 5.30-5.20 (m, 1H), 4.584.49 (m, 1H), 4.24 (d, >5.9 Hz, 2H), 4.043.95 (m, 1H), 3.92-3.77 (m, 3H), 3.71 (dd, >4.9, 4.8 Hz, 2H), 3.31-3.09 (m, 4H), 2.95 (br s, 2H), 2.44-2.30 (m, 1H), 1.98-1.87 (m, 2H), 1.53-1.36 (m, 2H); 481.9
68 Example 7M; C31 o cf3 ηρΛθΛ^ΟΗ Y o=s=o ô II 1H NMR (400 MHz, CDjOD) 6 7.73 (br AB quartet, Jab=8.5 Hz, ûvab=22.4 Hz, 4H), 5.33-5.24 (m, 1H), 3.92-3.73 (m, 7H), 3.33.14 (m, 2H), 3.05-2.93 (m, 2H), 2.89-2.76 (m, 2H), 2.02-1.88 (m, 2H), 1.64-1.45 (m. 2H); 477.5
69 Example 3; C6, C17 0 CF3 on 0 By 1H NMR analysis, this was judged to be a mixture of rotamers. 8.39 (d, >2.7 Hz, 1H). 7.90-7.85 (m, 1H), 7.71 (d, >2.3 Hz, 1H), 6.83 (d, >8.9 Hz, 1H), [6.20 (d, >2.3 Hz) and 6.19 (d, >2.3 Hz), total 1H], 5.31- 5.21 (m, 1H), 4.04-3.98 (m, 1H), 3.97 (s, 3H), 3.92-3.81 (m, 3H), 3.66-3.58 (m, 2H), 2.07-2.00 (m, 2H), 1.90-1.84 (m, 1 H); 413.0
147
70 Example 182’·22; C2 □ cf3 y DIAST-1 O=S=O Ψ F By ’H NMR analysis, this was judged to be a mixture of rotamers. 7.84-7.76 (m, 2H), 7.20 (br dd, >8.5, 8.5 Hz, 2H), 5.31-5.21 (m, 1 H), 4.04-3.63 (m, 7H). 3.54-3.45 (m, 1H), 3.31-2.96 (m, 3H), 2.59-2.47 (m, 1H), 1.62-1.23 (m, 4H), 1.04-0.91 (m, 3H); 485.0
71 Example 1821·22; C2 0 cf3 I^nÂt^0H ¢9 DIAST-2 O=S=O Ψ F By ’H NMR analysis, this was judged to be a mixture of rotamers. 7.84-7.76 (m, 2H), 7.21 (brdd, >8.8, 8.3 Hz, 2H), 5.31-5.19 (m, 1H), 4.04-3.94 (m, 1H), 3.94-3.63 (m, 6H), 3.55-3.45 (m, 1H), 3.32-2.99 (m, 3H), 2.60-2.43 (m, 2H), 1.62-1.50 (m, 1H), 1.481.34 (m, 2H), 1.03-0.91 (m, 3H); 485.0
72 Example 1323·24; C2 0 cf3 <'νΛοΛ-οη 0=<0jv> b F ’H NMR (600 MHz, DMSO-de) δ 7.32 (brdd, >8.4, 5.7 Hz, 2H), 7.19 (brdd, >9.0, 8.8 Hz, 2H), 5.26-5.17 (m, 2H), 4.35 (s, 2H), 3.77-3.71 (m, 1H), 3.71-3.61 (m, 3H), 3.383.24 (m, 2H, assumed; obscured by water peak), 3.24-3.20 (m, 2H), 1.86-1.63 (m, 4H); 421.2
73 C3125 0 cf3 <·νΛ0Λ-'r°'x> o=s=o 3.20 minutes28; 485
74 Example 717; C31 0 cf3 c°p N O=S=O hSl Ό 8.59-8.54 (m, 1 H). 7.75-7.69 (m, 1H), 7.307.23 (m, 2H, assumed; partially obscured by solvent peak), 5.75 (br t, >5 Hz, 1H), 5.295.20 (m, 1H), 4.37 (d, >5.0 Hz, 2H), 4.043.96 (m, 1H), 3.91-3.77 (m, 3H), 3.74-3.69 (m, 2H), 3.30-3.11 (m, 4H), 3.04-2.98 (m, 2H), 1.98-1.89 (m, 2H), 1.52-1.33 (m, 2H); 483.0
148
75 Example 26; C2 0 cf3 jQnÂP^0H n\ Y OH 1.72 minutes13; 365.1
76 Example 27; C31 o cf3 AA™ cP N N^N Y 8.15 (s, 2H), 5.31-5.19 (m, 1H), 4.04-3.94 (m. 1H), 3.91-3.72 (m, 7H), 3.72-3.62 (m, 2H), 3.40-3.22 (m, 2H), 2.56-2.37 (br s, 1H), 2.13 (s. 3H), 1.96-1.83 (m, 2H), 1.63-1.48 (m, 2H); 405.0
77 C3117 o cf3 AA™ t°p N o=s=o À 3.26 minutes28; 481
78 C3117 0 cf3 rNA<A-OH C Sr* o=s=o Φ Ύ 3.30 minutes28; 511
79 Example 68; C31 0 cf3 AtA™ cP N O=S=O <x ’H NMR (400 MHz, CD3OD) δ 7.83 (br s, 1H), 7.80-7.75 (m, 2H), 7.62 (dd, J=7.9. 7.7 Hz, 1H), 5.33-5.24 (m, 1H), 3.92-3.76 (m, 6H), 3.75 (s, 1H), 3.29-3.14 (m, 2H), 3.052.94 (m, 2H), 2.SC-2.77 (m, 2H), 2.01-1.89 (m, 2H), 1.65-1.46 (m, 2H); 477.5
149
βο Example 1328; C2 0 cf3 ι^ν^ο'Λ^οη n-n Ή NMR (400 MHz, CDjOD) δ 8.14 (br s, 1H), 7.63 (AB quartet, 4as=7.7 Hz, ΔνΑΒ=66.8 Hz, 4H), 6.40 (br s, 1H), 5.365.27 (m, 1H), 4.28-4.15 (m, 2H), 3.89 (dd, halfof ABX pattern, 4=12.4, 3.7 Hz, 1H), 3.79 (dd, halfof ABXpattern, 4=12.3, 6.9 Hz, 1H), 3.53 (s, 1H), 3.18-2.93 (m, 3H), 2.08-1.96 (m, 2H), 1.85-1.61 (m. 2H); 408.2
81 Example 3; C6. C17 o cf3 N-N ? 7.84-7.80 (m, 1H), 7.58 (br AB quartet, 4ab=8.3 Hz, ΔνΑΒ=21.3 Hz, 4H), 6.24-6.19 (m, 1H), 5.32-5.21 (m, 1H), 4.05-3.97 (m, 1H), 3.93-3.81 (m, 3H), 3.68-3.58 (m, 2H), 3.12 (s, 1H), 2.09-2.02 (m, 2H), 1.90-1.84 (m, 1H); 406.0
82 Example 3»; C6 O çf3 H^nA/^OH N-N 7.82 (d, 4=2.3 Hz, 1H), 7.58 (br AB quartet, 4ab=8.6 Hz, ΔνΑΒ=21.2 Hz, 4H), 6.24-6.19 (m, 1H), 5.32-5.21 (m, 1H), 4.05-3.97 (m, 1H), 3.93-3.81 (m, 3H), 3.69-3.57 (m, 2H), 3.12 (s, 1H), 2.09-2.02 (m, 2H), 1.90-1.84 (m, 1H); 406.0
83 Example 6; C23, C2 o cf3 ΛνΛΑ011 cP N o=s=o 0, Ή NMR (400 MHz, CD3OD) δ 7.81-7.77 (m, 1H), 7.74-7.69 (m, 2H), 7.63 (dd, component of ABC system, 4=7.8, 7.8 Hz, 1H), 5.33-5.25 (m, 1H), 3.92-3.74 (m, 6H), 3.3-3.12 (m, 2H, assumed; partially obscured by solvent peak), 3.04-2.97 (m, 2H), 2.88-2.81 (m, 2H), 2.00-1.90 (m, 2H), 1.64-1.46 (m, 2H); 486.9
84 C312S o cf3 n (ΛοΛ-οη cP N o=s=o <y 3.00 minutes28; 471
150
85 030,31 0 CF3 « C nh4*u N ο=έ=ο $ Presumed to be a mixture of diastereomers around the phosphorus atom; 1H NMR (400 MHz, CDjOD) δ 7.88-7.81 (m, 2H), 7.37 (br d, 7=9.0, 8.6 Hz, 2H), 5.53-5.43 (m, 1H), 4.20-4.12 (m, 1 H), 4.12-4.03 (m, 1 H), 3.943.76 (m, 2H), 3.79 (dd, 7=5.1,4.9 Hz, 2H), 3.62-3.52 (m, 3H), 3.29-3.12 (m, 2H), 3.062.90 (m, 2H), 2.90-2.73 (m, 2H), 2.02-1.83 (m, 2H), 1.67-1.44 (m, 2H); 565.3
86 030,31 r Ao^o. £ c°P 0 N O=S=O 1H NMR (400 MHz, CDsOD), characteristic peaks: δ 7.87-7.81 (m, 2H), 7.37 (dd, 7=8.7, 8.7 Hz, 2H), 5.59-5.50 (m, 1H), 4.48-4.31 (m, 2H), 3.93-3.73 (m, 8H), 3.3-3.15 (m, 2H), 3.04-2.92 (m, 2H), 2.87-2.76 (m, 2H), 2.01-1.91 (m, 2H), 1.64-1.47 (m, 2H); 579.3
87 030,32 οΓΛΑΡ° £p »ηΓ ο=έ=ο φ F Presumed to be a mixture of diastereomers around the phosphorus atom; Ή NMR (400 MHz, CD3OD) δ 7.87-7.81 (m, 2H), 7.37 (dd, 7=8.8, 8.7 Hz, 2H), 5.52-5.43 (m, 1H), 4.204.13 (m, 1 H), 4.11 -4.03 (m, 1 H), 3.97-3.82 (m, 4H), 3.79 (dd, 7=5.1,4.8 Hz, 2H), 3.33.12 (m, 2H), 3.06-2.89 (m, 2H), 2.89-2.74 (m, 2H), 2.02-1.84 (m, 2H), 1.66-1.44 (m, 2H), 1.31-1.20 (m, 3H); 579.2
88 030.32 o CF3 λ khT / ο=έ=ο 1H NMR (400 MHz, CD3OD) δ 7.87-7.80 (m, 2H), 7.37 (dd, 7=8.7, 8.6 Hz, 2H), 5.58-5.49 (m, 1H), 4.44-4.28 (m, 2H), 4.21-4.07 (m, 4H), 3.93-3.8 (m, 2H), 3.80 (dd, 7=5.1, 4.8 Hz, 2H), 3.3-3.15 (m, 2H), 3.03-2.93 (m, 2H), 2.87-2.75 (m, 2H), 2.01-1.91 (m, 2H), 1.63-1.47 (m, 2H), 1.40-1.27 (m, 6H), 607.2
151
89 Example 85“; 6 O CF3 c P V ο=έ=ο 9 Presumed to be a mixture of diastereomers around the phosphorus atom; ’H NMR (400 MHz, CDsOD) δ 7.87-7.81 (m, 2H), 7.37 (dd, >8.8, 8.7 Hz, 2H), 5.56-5.46 (m, 1H), 4.284.20 (m, 1H), 4.20-4.07 (m, 3H), 3.93-3.82 (m, 2H), 3.80 (dd, >5.0,4.9 Hz, 2H), 3.433.34 (m, 2H), 3.3-3.14 (m, 2H), 3.00-2.90 (m, 8H), 2.85-2.79 (m, 2H), 2.01-1.89 (m, 2H), 1.68-1.44 (m, 2H); 622.5
90 Example 85”; 6 O ÇF3 λ ι^νλο^ο'ρ: cP V ’ N O-S=O Z|S Φ F Presumed to be a mixture of diastereomers around the phosphorus atom; 1H NMR (400 MHz, CDaOD) δ 7.87-7.81 (m, 2H), 7.37 (dd, >8.7, 8.7 Hz, 2H), 5.55-5.46 (m, 1H), 4.334.20 (m, 3H), 4.15-4.07 (m, 1H), 3.93-3.82 (m, 2H), 3.80 (dd, >5.2, 4.7 Hz, 2H), 3.693.60 (m, 2H), 3.28-3.17 (m, 11 H), 3.01-2.95 (m, 2H), 2.86-2.78 (m, 2H), 2.01-1.90 (m, 2H), 1.67-1.44 (m,2H); 636.2
91 Example 30; 7 O CF3 _ Na* N o=é=o ô 1H NMR (400 MHz, D2O) δ 7.82-7.77 (m, 2H), 7.77-7.72 (m, 1H), 7.68-7.62 (m, 2H), 5.47-5.37 (m, 1 H), 4.12-4.04 (m, 1H), 4.013.93 (m, 1H), 3.93-3.63 (m, 2H), 3.83 (dd, >5.1,4.9 Hz, 2H), 3.36-3.12 (m, 2H), 3.112.98 (m, 2H), 2.97-2.84 (m, 2H), 2.00-1.80 (m, 2H), 1.76-1.49 (m, 2H); 533.1
1. 3,3,3-Trifluoropropane-1,2-diol was converted to S-fltert-butyltdimethyOsilylloxyJ-I.IJtrifluoropropan-2-oi using the method described for synthesis of C59 from C58 In Examples 19, 20 and 21.
2. Reaction of 4-[4-(plperidin-4-yl)pyrimidin-2-yl]morpholine, hydrochloride sait with bis(trichloromethyl) carbonate and 3-{[tert-butyl(dimethyl)silyl]oxy}-1,1,1-trifluoropropan-2-ol (see footnote 1) in the presence of Μ,Μ-diisopropylethylamine afforded 3-{[fertbutyl(dimethyl)silyl]oxy}-1,1,1-trifluoropropan-2-yl4-[2-(morpholin-4-yl)pyrimidin-4-yl]piperidine1-carboxylate. This material was desilylated via treatment with acetic acid in a mixture of water and tetrahydrofuran to afford Example 35.
3. Examination of coupling constants In the NMR spectra of re/-(2S,3R)-1,1,1,4,4,4hexafluorobutane-2,3-diol and re/-(2f?,3/?)-1,1,1,4,4,4-hexafluorobutane-2,3-dio! allowed tentative assignaient of the starting material used for Example 36 as the re/-(2S.3R) Isomer.
152
4. Reaction of C19 with bls(trlchloromethyl) carbonate and rel-(2S,3R)-1,1,1,4,4,4hexafluorobutane-2,3-diol in the presence of /V,N-diisopropylethylamine afforded Example 36.
5. Alkylation of C6 with 2-(chloromethyl)pyridine In the presence of potassium hydroxide In acetone provided the requisite tert-butyl (1a,5a,6a)-6-[1-(pyridin-2-ylmethyl)-1H-pyrazol-3-yl]-3- azabicyclo[3.1.0]hexane-3-carboxylate.
6. Reaction of C13 with tetrahydro-2H-pyran-4-yl methanesulfonate in the presence of césium carbonate and potassium lodide, at elevated température in Ν,Ν-dimethylformamide, provided the requisite tert-butyl 4-[1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-3-yl]piperidine-1-carboxyiate.
7. The carbonate reagent employed in this case was 1-({[(3-{[tert-butyl(dimethyl)silyl]oxy}-1,1,1- trifluoropropan-2-yl)oxy]carbonyl}oxy)pyrrolidine-2,5-dione, prepared from 3-{[fertbutyl(dimethyl)silyl]oxy}-1,1,1-trifluoropropan-2-ol (see footnote 1) using the general method described for synthesis of Cl7 In Example 3.
8. tert-Butyl 2,9-diazaspiro[5.5]undecane-2-carboxylate was converted to 2-tert-butyl 9-{(2R)-
I, 1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl} 2,9-diazaspiro[5.5]undecane-2,9- dicarboxylate using the method described tor synthesis of C29 in Example 6. Treatment with trifluoroacetic acid removed both pratecting groups, affordïng (2R)-1,1,1-trifluoro-3hydroxypropan-2-yl 2,9-diazaspiro[5.5]undecane-9-carboxylate, which was then subjected to reductive amination with 4-fluorobenzaldehyde and sodium triacetoxyborohydride to afford Example 41.
9. Reaction of C23 with 4-fluorobenzaldehyde in the presence of 1 H-benzotriazole and acetic acid provided the corresponding imine; In situ treatment with méthylmagnésium bromide then afforded the requisite tert-butyl 4-[1-(4-fluorophenyl)ethyl]-1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate. Example 43 Is a diastereomeric mixture, with both stereochemistries présent at the methyl group.
10. Séparation of Example 43 Into its component diastereomers was effected via supercritical fluid chromatography [Column: Chiral Technologies Chiralpak AD, 5 pm; Mobile phase: 4:1 carbon dioxide : (0.1% ammonium hydroxide in éthanol)]. The first-eluting diastereomer was Example 44, and the second-eluting diastereomer was Example 45.
II. In this case, tert-butyl 1-oxa-6-azaspiro[2.5]octane-6-carboxylate was reacted with tetrahydro-2H-pyran-4-amine, and the resulting tert-butyl 4-hydroxy-4-[(tetrahydro-2H-pyran-4ylamino)methyl]pîperidine-1-carboxylate was N-alkylated with 1-(bromomethyl)-4-fluorobenzene in the presence of potassium carbonate; this afforded the requisite tert-butyl 4-{[(4fluorobenzyl)(tetrahydro-2H-pyran-4-yl)amino]methyl}-4-hydroxypiperidine-1-carboxylate.
12. tert-Butyl 4-[(tetrahydro-2H-pyran-4-yiamino)methyi]piperidine-1-carboxylate was synthesized via reductive amination of tetrahydro-4H-pyran-4-one with tert-butyl 4(amlnomethyi)plperidine-l-carboxylate, using magnésium sulfate and silica gel followed by sodium triacetoxyborohydride.
153
13. Conditions for analyticat HPLC. Column: Waters Atlantis dC18,4,6 x 50 mm, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid In acetonitrile (v/v); Gradient: 5.0% to 95% B, linear over 4.0 minutes; Fiow rate: 2 mL/minute.
14. Intermediate tert-butyl 4-({[(4-fluorophenyl)sulfonyl]amino}methyl)piperidine-1-carboxyiate was reacted with lodomethane and potassium carbonate to provide tert-butyl 4-((1(4fluorophenyl)sulfonyl](methyl)amino}methyl)piperidine-l-carboxylate.
15. The requisite 2-chloro-4-(1-methyl-1H-pyrazol-5-yl)pyridine was prepared via a Suzuki reaction between 2-chloro-4-iodopyridine and 1-methyl-5-(4,4,5,5-tetramethyl-1l3,2dioxaborolan-2-yl)-1H-pyrazole, mediated via [1,T- bis(diphenylphosphino)ferrocene]dichloropalladium(ll).
16. Suzuki reaction of 2-bromo-6-chloropyridine and 1-methy 1-5-(4,4,5,5-tetramethyl-1,3,2dioxaborolan-2-yl)-1H-pyrazole, mediated via [1,1’bis(diphenylphosphino)fenOcene]dichloropalladium(ll), afforded 2-chloro-6-(1-methyl-1Hpyrazo l-5-yl)pyrid ine.
17. The requisite sulfonic acid was prepared from the corresponding bromo compound via reaction with a mixture of potassium disulfite, palladium(ll) acetate, triphenylphosphine, tetraethylammonium bromide, sodium formate and 1,10-phenanthroline in N,Ndimethylformamide at elevated température. The sulfonic acid was cooled to 0 ’C, treated with C31 and W-chlorosuccinimide, and stirred at 30 ’C for 1 hour. The resulting sulfonamide was deprotected via treatment with trifluoroacetic acid ln dichloromethane to provide the product of the Example.
18. Conditions for analytical HPLC. Column: Waters XBridge C18, 2.1 x 50 mm, 5 pm; Mobile phase A: 0.0375% trifluoroacetic acid in water; Mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; Gradient: 10% to 100% B over 4.0 minutes; Flow rate: 0.8 mL/minute.
19. W-Benzyl-2-oxo-1,3-oxazolidine-3-sulfonamide was used as the sulfonylating reagent in this case, in a mixture of triethylamine and acetonitrile.
20. Reaction of C31 and 4-bromobenzenesulfonyl chloride provided (2R)-1,1,1-trifluoro-3-[(4methoxybenzyl)oxy]propan-2-yl 4-[(4-bromophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undecane9-carboxylate, which was then converted to (2R)-1,1,1-trifluoro-3-[(4- methoxybenzyl)oxy]propan-2-yl 4-([4-[(trimethylsilyl)ethynyl]phenyl}sulfonyl)-1-oxa-419diazaspiro[5.5]undecane-9-carboxylate by reaction with ethynyl(trimethyl)silane, copper(l) iodide, and [1,T-bis(diphenylphosphino)ferrocene]dichloropalladium(ll). Desilylation was effected via treatment with potassium carbonate and methanol to affoid (2F?)-1,1,1 -lrifiuoro-3[(4-methoxybenzyl)oxy]propan-2-yl 4-[(4-ethynylphenyl)sulfonylJ-1-oxa-4,935 diazaspiro[5.5]undecane-9-carboxylate; final deprotection using trifluoroacetic acid provided Example 68.
21. Reaction of tert-butyl 4-oxopîperidine-1-carboxylate with nitroethane and triethylamine provided tert-butyl 4-hydroxy-4-(1-nîtroethyl)piperidine-1-carboxylate, which was hydrogenated
154
to afford fert-butyl 4-(1-aminoethyl)-4-hydroxypiperidine-1-carboxylate. This material was treated with 4-fluorobenzenesulfonyl chloride and potassium carbonate, followed by 1,2-dibromoethane, to provide the requisite intermediate fert-butyl 4-[(4-fluorophenyl)sulfonyl]-5-methyl-1-oxa-4,9· diazaspiro[5.5]undecane-9-carboxylate.
22. Séparation to provide the diastereomers of Examples 70 and 71 was effected via supercritical fluid chromatography [Column: Chiral Technologies Chiralpak AD, 5 pm; Mobile phase A: carbon dioxide; Mobile phase B: 2-propanol; Gradient: 25% to 100% B). The first* eluting diastereomer was Example 70, and the second-eluting diastereomer was Example 71.
23. tert-Butyl 2-oxo*1-oxa-3,8-diazaspiro[4.5]decane-8-carboxylate was treated with sodium hydride and 1 *(chloromethyl)-4-fluorobenzene to afford fert-butyl 3-(4-fluorobenzyl)-2-oxo-1 -oxa3,8-diazaspiro[4.5]decane-8-carboxylate; deprotection with hydrogen chloride in 1,4-dioxane provided the requisite 3-(4-fluorobenzyl)-1-oxa-3,8-diazaspiro[4.5]decan-2-one.
24. In this case, the final deprotection was effected with hydrogen chloride in 1,4-dioxane, rather than trifluoroacetic acid.
25. Compound C31 was allowed to react with the appropriate sulfonyl chloride and triethylamine; the resulting sulfonamide was deprotected with trifluoroacetic acid in dichloromethane to afford the product of the Example.
26. Conditions for analytical HPLC. Column: Waters XBridge C18, 2.1 x 50 mm, 5 pm; Mobile phase A: 0.0375% trifluoroacetic acid in water; Mobile phase B: 0.01875% trifluoroacetic acid in acetonitrile; Gradient: 1% to 5% B over 0.6 minutes; 5% to 100% B over 3.4 minutes; Flow rate: 0.8 mL/mlnute.
27. 2-Bromoethanol was slowly added to a solution of chlorosulfonyl isocyanate In dichloromethane at 0 *C. After an hour, a mixture of 1-(pyridin-2-yl)methanamine and triethylamine was slowly added to the 0 ’C reaction mixture. Removal of solvent provided 2-oxo-
N-(pyrîdin-2-ylmethyl)*1,3-oxazolidine-3-sulfonamide, which was used as the sulfonylating reagent in this case, in a mixture of triethylamine and acetonitrile.
28. The requisite fert-butyl 4-[1-(4-ethynylphenyl)*1H-pyrazol-3-yl]piperidine-1-carboxylate was prepared as follows: fert-butyl 4-acetylpiperidine*1-carboxylate was converted to fert-butyl 4-[1 (4-bromophenyl)-1H-pyrazol-3-yl]piperidine-1-carboxylate using the methods described for synthesis of C18 from C4 that are found in Examples 1 and 3. Reaction with ethynyl(trimethyl)sîlane, copper(l) lodide and tetrakis(triphenylphosphine)palladium(0), followed by removal of the silyl group via treatment with potassium carbonate in methanol, provided tertbutyl 4-[1*(4-ethynylphenyl)-1H-pyrazol-3-yl]piperidine-1-carboxylate.
29. In this case, the enantiomer of C1 was employed; this may be prepared similarly, via the use 35 of (2S)-2-(trifluoromethyl)oxirane rather than (2R>2-(trifluoromethyl)oxirane.
30. A 0 *C solution of 6 In acetonitrile was treated with 5 équivalents of diphosphoryl tetrachloride; after one to two hours at 0 *C, the appropriate alcohol (40 équivalents) was added, generating a mixture of the mono- and di-alkyl phosphate esters.
155
31. Reversed phase HPLC was used to separate the monomethyt and dimethyl phosphate esters. Column: Phenomenex Gemini NX C18, 5 pm; Mobile phase A: 0.1% ammonium hydroxide in water; Mobile phase B: 0.1% ammonium hydroxide in acetonitrile; Gradient: 50% to 100% B. The first-eluting product was Example 85, and the second-eluting product was Example 86.
32. Reversed phase HPLC was used to separate the monoethyl and diethyl phosphate esters. Column: Phenomenex Gemini NX C18, 5 pm; Mobile phase A: 0.1% ammonium hydroxide in water; Mobile phase B: 0.1% ammonium hydroxide in acetonitrile; Gradient: 50% to 100% B. The first-eluting product was Example 87, and the second-eluting product was Example 88.
33. In this case, 2-(dimethylamino)ethanol was used In place of methanol. The product was purified using reversed phase HPLC (Column: Phenomenex Luna C18, 5 pm; Mobile phase A: 0.1% formic acid in water; Mobile phase B: 0.1% formic acid in acetonitrile; Gradient: 50% to 100% B).
34. In this case, 2-hydroxy-N,N,N-trimethylethanaminium chloride was used In place of methanol.
Example 92 (2R)-1,1,1-Trifluom-3-hydroxypropan-2-yl (3R)-3-[ethyl(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate (92)
Step 1. Synthesis oftert-butyl (3R)-3-[ethyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]decane8-carboxylate (C88).
Sodium hydride (60% dispersion in minerai oil; 15 mg, 0.38 mmol) was added to a 0 ’C solution of C48 (50.0 mg, 0.126 mmol) in N,N-dimethylformamide (1 mL). After the reaction
156
mixture had been stirred at 0 “C for 30 minutes, a solution of bromoethane (27.5 mg, 0.252 mmol) in Ν,Ν-dimethylformamide (0.1 mL) was added, and the reaction mixture was allowed to stir at 25 *C for 16 hours. It was subsequently cooled to 0 ‘C, and additional sodium hydride (60% dispersion in minerai oil; 15 mg, 0.38 mmol) was added; stirring was continued at 0 ’C for 5 30 minutes, whereupon a solution of bromoethane (20 mg, 0.18 mmol) in N,Ndlmethylformamide (0.1 mL) was added. The reaction mixture was then stirred at 25 ’C for 16 hours. Water (30 mL) was added, and the resulting mixture was extracted with dichloromethane (3 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated in vacuo, and subjected to préparative thin layer chromatography on silica gel (Eiuent: 2:1 10 petroleum ether / ethyl acetate), providing the product as a light yellow gum. Yield: 40 mg, 94 pmol, 75%. LCMS m/z 447.2 [M+Na*]. 1H NMR (400 MHz, CDCI3) δ 7.81 (br d, 3=7 Hz, 2H), 7.58 (br dd, 3=7.4, 7.3 Hz, 1H), 7.51 (br dd, 3=7.9, 7.2 Hz, 2H), 4.64-4.54 (m, 1H), 3.80 (dd, 3=9.8, 7.6 Hz, 1H), 3.66-3.5 (m. 2H), 3.50 (dd, 3=9.9, 6.2 Hz, 1H), 3.28-3.10 (m, 4H), 1.94 (dd, 3=13.2, 8.8 Hz, 1H), 1.63-1.53 (m, 3H), 1.47 (dd, 3=13.3, 8.0 Hz, 1H), 1.43 (s, 9H), 1.42-1.33 15 (m, 1H), 1.30 (t, 3=7.0 Hz, 3H).
Step 2. Synthesis of(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl (3RJ-3[ethyl(phenylsulfonyl)aminoJ-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C89).
Trifluoroacetic acid (1 mL) was added in a drop-wise manner to a 0 ’C solution of C88 (39.0 mg, 91.9 pmol) in dichloromethane (1 mL), and the reaction mixture was stirred at 15 ’C 20 for 1 hour. Removal of volatiles under reduced pressure provided A/-ethyl-A/-[(3R)-1-oxa-8azaspiro[4.5]dec-3-yl]benzenesulfonamide, trifluoroacetate sait, as a yellow gum, LCMS m/z
325.1 [M+H]\ This material was dissolved In acetonitrile (1 mL), cooled to 0 C, and treated with C2 (reaction solution in acetonitrile containing 0.11 mmol) and triethylamine (73.3 mg, 0.724 mmol). After the reaction mixture had been stirred at 20 eC for 16 hours, it was concentrated in 25 vacuo and purified via chromatography on silica gel (Gradient: 0% to 50% ethyl acetate in petroleum ether) to afford the product as a coloriess gum. Yield: 35 mg, 58 pmol, 63%. LCMS m/z 623.1 [M+Na*]. 1H NMR (400 MHz, CDCh) δ 7.84-7.80 (m, 2H), 7.62-7.57 (m, 1H), 7.567.49 (m, 2H), 7.23 (br d, 3=8.7 Hz, 2H), 6.87 (br d, 3=8.7 Hz, 2H), 5.52-5.40 (m, 1H), 4.66-4.53 (m, 1H), 4.49 (AB quartet, upfield doublet is broadened, 3ab=11.7 Hz, Avab=28.4 Hz, 2H), 3.8830 3.62 (m, 5H). 3.81 (s, 3H), 3.58-3.47 (m, 1H), 3.36-3.10 (m, 4H), 1.93 (dd, 3=13.2, 8.8 Hz, 1H),
1.7-1.55 (m, 3H, assumed; partially obscured by water peak), 1.50 (dd, 3=13.2, 8.1 Hz, 1H),
1.39 (ddd, 3=13.5,11.2, 4.3 Hz, 1H), 1.31 (t, 3=7.1 Hz, 3H).
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3R)-3[ethyt(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (92).
To a 0 ’C suspension of C89 (35 mg, 58 pmol) in dichloromethane (1 mL) was added trifluoroacetic acid (1 mL). The reaction mixture was stirred at 18 ’C for 1 hour, whereupon It was cooled to 0 ’C and slowly treated with aqueous sodium bicarbonate solution (30 mL), while the purple mixture became coloriess. It was then extracted with dichloromethane (3 x 30 mL),
157 and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified using reversed phase HPLC (Column: Agela Durashell, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 44% to 84% B), to afford the product as a colorless oil. Yield: 7.5 mg, 16 pmol, 28%. LCMS m/z
481.0 [M+H]*. 1H NMR (400 MHz, CDCb) δ 7.82 (br d, J=8 Hz, 2H), 7.60 (br dd, J=7.5,7.5 Hz,
1H), 7.52 (br dd, J=7.5, 7.5 Hz, 2H), 5.30-5.17 (m, 1H), 4.66-4.55 (m, 1 H), 4.04-3.93 (m, 1 H), 3.91-3.69 (m, 4H), 3.58-3.48 (m, 1H), 3.39-3.09 (m, 4H), 2.50-2.36 (m, 1H), 2.01-1.88 (m, 1 H).
1.7-1.6 (m, 2H, assumed; largely obscured by water peak), 1.57-1.35 (m, 2H), 1.31 (t, J=7.0 Hz, 3H).
Examples 93 and 94 (2R)-1,1, l-Trifluoro-3-hydroxypropan-2-yl (3R)-3-[(cyclopmpylsulfonyl)(methyl)aminol-1-oxa-8azaspiro[4.5]decane-8-carboxylate (93) and (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl (3S)-3[(cyclopropylsulfonyl)(methyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (94)
OPMB
J DIASTN. ,O
Ο V C91
-v Ο V C92
CF3COOH cf3cooh
o'
o'
Step 1. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3[(cyclopmpylsulfonyl)aminoj- 1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C90).
158
Cyclopropanesulfonyl chloride (650 mg, 4.62 mmol) and triethylamine (1.17 g, 11.6 mmol) were added to an 18 C suspension of C73 (1.00 g, 2.31 mmol) In dichloromethane (8 mL), and the réaction mixture was stirred at 10 *C for 12 hours. After the reaction mixture had been concentrated in vacuo, the residue was diluted with water (30 mL) and extracted with ethyl 5 acetate (3 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered, concentrated under reduced pressure, and purified using silica gel chromatography (Gradient: 0% to 15% methanol in dichloromethane), affording the product as a coloriess gum. Yield: 641 mg, 1.19 mmol, 52%. LCMS m/z 559.1 [M+Na*].
Step 2. Synthesis of (2R)~1,1,1-tnfluoro-3-[(4-methoxybenzyl)oxy]propan-2-yf (3R)-310 [(cyclopropylsutfonyl)(methyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (DIAST-1) (C91) and (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyi)oxyJpropan-2-yi (3S)-3[(cyclopmpylsulfonyl)(methyl)amino]-1-oxa-8-azaspirO[4.5]decane-8-carboxylate (DIAST-2) (C92).
To a 0 eC solution of C90 (641 mg, 1.19 mmol) in W,N-dimethylformamide (8 mL) was added sodium hydride (60% dispersion In minerai oil; 95.6 mg, 2.39 mmol), and the reaction mixture was stirred at 0 °C for 30 minutes, lodomethane (254 mg, 1.79 mmol) was added at 0 °C, and the reaction mixture was allowed to stirat 15 C for 3 hours, whereupon it was diluted with water (50 mL) and extracted with ethyl acetate (3 x 30 mL). The combined organic layers were concentrated In vacuo, and the residue was purified by silica gel chromatography (Gradient: 0% to 50% ethyl acetate in petroleum ether) to afford a mixture of C91 and C92 as a coloriess gum. Yield: 310 mg, 0.563 mmol, 47%. The component diastereomers were separated via supercritical fluid chromatography [Column: Chiral Technologies Chiralpak IC, 10 pm; Mobile phase: 40% (0.1% ammonium hydroxide in 2-propanol) in carbon dioxïde], affording C91 as the first-eluting dlastereomer, and C92 as the second-eluting diastereomer, both as coloriess gums. The indicated stereochemistries at the sulfonamide positions were assigned on the basis of a chiral synthesis of 93 (see Altemate Synthesis of Example 93 below).
C91 - Yield: 147 mg, 0.267 mmol, 22%. LCMS m/z 573.0 [M+Na*]. Ή NMR (400 MHz, CDCb) δ
7.25 (br d, >8.5 Hz. 2H), 6.88 (br d, >8.5 Hz, 2H), 5.55-5.42 (m, 1H), 4.73-4.62 (m, 1H), 4.51 (AB quartet, Λβ=11.7 Hz, ΔνΑ0=29.3 Hz. 2H), 3.96 (dd, half of ABX pattern, >10.0,7.5 Hz, 1H).
3.85 (dd, half of ABX pattern, >10.1, 5.2 Hz, 1H), 3.82 (s, 3H), 3.8-3.64 (m, 4H), 3.41-3.22 (m.
2H), 2.88 (s, 3H), 2.26 <tt. >8.0,4.9 Hz. 1H), 2.11-1.97 (m, 1H), 1.85-1.64 (m, 4H), 1,45 (ddd, >13.7,11.2, 4.4 Hz, 1H), 1.21-1.15 (m, 2H), 1.03-0.97 (m, 2H).
C92 - Yield: 155 mg. 0.282 mmol, 24%. LCMS m/z 573.0 [M+Na*]. ’H NMR (400 MHz, CDCh), characteristic peaks: δ 7.25 (br d, >8.7 Hz, 2H), 6.88 (br d, >8.5 Hz, 2H), 5.54-5.43 (m, 1 H),
4.73-4.63 (m, 1H), 4.51 (AB quartet, upfield d is broadened, Jab=11.9 Hz, ΔνΑβ=29,0 Hz, 2H),
4.01-3.91 (m, 1H), 3.89-3.78 (m, 2H), 3.82 (s, 3H), 3.79-3.64 (m, 3H), 3.40-3.20 (m, 2H), 2.88 (s, 3H), 2.26 (tt, >8, 5 Hz, 1H), 2.14-1.95 (m, 1H), 1.84-1.7 (m, 4H, assumed; partially obscured by water peak), 1.21-1.15 (m, 2H), 1.03-0.97 (m, 2H).
159
Step 3. Synthesis of (2RJ-Ï, 1,1-trifluoro-3-hydroxypropan-2-yl (3RJ-3[(cyclopmpylsulfonyl)(methy!)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (93).
Trifluoroacetic acid (2 mL) was added to a 0 ’C solution of C91 (147 mg, 0.267 mmol) In dichloromethane (8 mL). The réaction mixture was stirred at 16 ’C for 1 hour, whereupon it was 5 cooled to 0 ’C, and slowly treated with aqueous sodium bicarbonate solution (20 mL), while the purple mixture became colorless. The resulting mixture was extracted sequentially with dichloromethane (20 mL) and ethyl acetate (2 x 20 mL); the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Chromatography on silica gel (Gradient: 0% to 70% ethyl acetate in petroleum ether) afforded the product as a yellow oil.
Yield: 49.7 mg, 0.115 mmol, 43%. LCMS m/z 431.2 [M+H]*. ’H NMR (400 MHz, CDCb) δ 5.325.20 (m, 1H), 4.73-4.64 (m, 1H), 4.05-3.93 (m, 2H), 3.94-3.75 (m, 4H), 3.46-3.24 (m, 2H), 2.89 (s, 3H), 2.39-2.21 (m, 1H), 2.26 (tt, >8.0, 4.9 Hz, 1H), 2.13-2.04 (m, 1H), 1.86-1.69 (m, 4H), 1.56-1.41 (m, 1H), 1.22-1.15 (m, 2H), 1.04-0.96 (m, 2H).
Step 4. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3S)-315 [(cyclopropylsulfonyl)(methyljamino}- 1-oxa-8-azaspiro[4.5]decane-8-carboxylate (94).
Conversion of C92 to the product was carried out using the method described for synthesis of 93 from C91. The product was Isolated as a yellow oil. Yield: 63 mg, 0.15 mmol, 53%. LCMS m/z 431.2 (M+H]*. Ή NMR (400 MHz, CDCb) δ 5.31-5.20 (m, 1H), 4.74-4.64 (m, 1H), 4.05-3.93 (m, 2H), 3.92-3.71 (m, 4H), 3.42-3.20 (m, 2H), 2.89 (s, 3H), 2.37-2.22 (m, 2H), 20 2.08 (dd, >13.3, 9.0 Hz, 1 H), 1.86-1.67 (m, 3H), 1.81 (dd, >13.6, 7.0 Hz, 1H), 1.55-1.45 (m,
1H, assumed; partially obscured by water peak), 1.22-1.15 (m, 2H), 1.04-0.96 (m, 2H).
Altemate Synthesis of Example 93 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl (3R)-3-[(cyc!opmpylsu!fonyl)(methyl)amlno]-1-oxa-825 azaspiro[4.5]decane-8-carboxy!ate (93)
160
O CF,
ΑοΑ^οη
Step 1. Synthesis oftert-butyi (3R)-3-amino-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C93).
A pH 8.0 buffer solution was prepared, containing 0.1 M aqueous potassium phosphate and 2 mM magnésium chloride. A stock solution of substrate was prepared as follows: tert-butyl
3-oxo-1-oxa-8-azaspira[4.5]decane-8-carboxylate (18.0 g, 70.5 mmol) was dissolved in water containing 4% dimethyl sulfoxlde (14.4 mL). Warming and stîrring were requlred for dissolution, 10 and the resulting solution was maintained at 40 ’C. Propan-2-amine, hydrochloride sait (16.8 g, 176 mmol) was added to a mixture of pyridoxal 5'-phosphate monohydrate (1.87 g, 7.05 mmol) and the pH 8.0 buffer (300 mL). The resulting pH was approximately 6.5; the pH was adjusted to 8 via addition of aqueous potassium hydroxide solution (6 M; approximately 4 mL). The stock solution of substrate was added via syringe, in 5 mL portions, resulting In a suspension, still at pH 8. Codex® ATA-200 transaminase (batch #11099; 1.4 g) was almost completely dissolved in pH 8 buffer (20 mL), and poured Into the réaction mixture. Additional pH 8 buffer (25.6 mL) was
161
used to ensure complété transfer of the enzyme. The reaction mixture was stirred at 35 ’C with a nitrogen sweep (32 mL/minute) through a needle placed approximately 0.5 cm above the reaction surface. Due to difficulties in stirring, vacuum (220 Ton, 300 mbar) was applied after 3 hours, to remove the acetone generated by the transamination reaction. The suspended solids were broken up manually, which improved the stirring of the reaction mixture. After 26 hours, the reaction mixture was allowed to cool to room température, and aqueous hydrochloric acid (6 M, 5 mL) was added, to bring the pH from 8 to 6.5. After addition of ethyl acetate (200 mL), the mixture was vigorously stirred for 5 minutes and then filtered through diatomaceous earth (43 g; this filter ald had been slurried in water prior to being introduced into the filter funnel. The water was then removed, providing a tightly packed bed). The filter pad was washed sequentially with water (120 mL) and ethyl acetate (100 mL), and the aqueous layer of the combined filtrâtes was adjusted to pH 9 - 9.5 with aqueous potassium hydroxide solution (6 M; approximately 10 mL). The aqueous layer was then treated with dichloromethane (200 mL), and the resulting mixture was vigorously stirred for 5 minutes before being filtered through a pad of diatomaceous earth.
The filter pad was washed with dichloromethane (100 mL), and the aqueous layer of the combined filtrâtes was extracted twice with dichloromethane, in the same manner as that described above, with adjustment of the pH to 9 -10 (this required approximately 2 mL of the 6 M aqueous potassium hydroxide solution in both cases). Ail of the dichloromethane extracts were combined and dried over sodium sulfate with vigorous stirring. Filtration and concentration in vacuo afforded the product as an oily yellow solid (14.76 g). A fourth extraction was carried out In the same manner, but in this case the aqueous layer was adjusted to a pH of >10. The product obtained from this extraction was a white solid (1.9 g). Combined yield: 16.61 g, 64.79 mmol, 92%. 1H NMR (500 MHz, CDCh) δ 3.95 (dd, >9.0, 5.6 Hz, 1H), 3.69-3.63 (m, 1H), 3.623.52 (m, 3H), 3.38-3.27 (m, 2H), 2.6-2.2 (v br s, 2H), 2.07 (dd, >13.0,7.6 Hz, 1H), 1.78-1.71 (m, 1 H), 1.69-1.56 (m, 2H), 1.55-1.47 (m, 2H), 1.45 (s, 9H).
Step 2. Synthesis of tert-butyl (3R)-3-amino-1-oxa-8-azaspiro[4.5]decane-8-carboxylate, (2R)-5oxopyirolidine-2-carboxylate sait (C94).
A solution of C93 (16.61 g. 64.79 mmol) In éthanol (400 mL) was heated to 63 ’C and treated portion-wise with (2R)-5-oxopyrrolidine-2-carboxylic acid (7.78 g, 60.3 mmol). The 30 reaction mixture was then removed from the heating bath, and allowed to cool overnight. The mixture was cooled to 12 ’C In an ice bath, and filtered. The collected solids were washed with cold éthanol (2 x 50 mL) and then with diethyl ether (100 mL), affording the product as a pale yellow solid (19.2 g). The combined filtrâtes were concentrated in vacuo, with removai of approximately 400 mL of solvents. A thin line of solid formed around the Inner surface of the 35 flask. This was swirled back into the remaining solvents; diethyl ether (100 mL) was added, and the mixture was cooled in an ice bath with stirring. After approximately 15 minutes, the mixture was filtered and the collected solids were washed with diethyl ether (100 mL), affording additional product as a yellow solid (1.5 g). Combined yield: 20.7 g, 53.7 mmol, 89%. 1H NMR
162
(500 MHz, DjO) δ 4.16 (dd, J=8.9, 5.9 Hz, 1H), 4.11 (dd, half of ABX pattern, J=10.4, 5.8 Hz, 1H), 4.09-4.03 (m, 1H), 3.93 (dd, J=10.3, 3.1 Hz, 1H), 3.61-3.46 (m, 2H), 3.46-3.30 (m, 2H),
2.53-2.36 (m, 4H), 2.06-1.97 (m, 1H), 1.85 (dd, J=14.1, 4.6 Hz, 1H), 1.82-1.72 (m, 2H), 1.72-
1.65 (m, 1H), 1.59 (ddd, half of ABXY pattern, J=18, 9,4.5 Hz, 1H), 1.43 (s, 9H).
Conversion ofC94 to C48, for assessment of absolute stereochemistry
A small sample of C94 was derivatized via reaction with benzenesulfonyl chloride and saturated aqueous sodium bicarbonate solution for 1 hour at 40 ’C. The reaction mixture was extracted with ethyl acetate, and the solvent was removed from the extract under a stream of 10 nitrogen. Supercritical fluid chromatographie analysis (Column: Chiral Technologies Chiralcel OJ-H, 5 pm; Mobile phase A: carbon dîoxide; Mobile phase B: methanol; Gradient: 5% to 60% B) revealed the product to hâve an enantiomeric excess of >99%. Injection under the same conditions of samples of C48 and C49 established the derivatization product as Identical to C48, the absolute configuration of which was determined via X-ray crystallographic analysis (see 15 above).
Step 3. Synthesis of tert-butyl (3R)-3-[(cyclopropylsulfonyl)amino]-1-oxa~8-azaspiro[4.5Jdecane8-carboxylate (C95).
Cyclopropanesulfonyl chloride (56.8 mg, 0.404 mmol) and triethylamine (136 mg, 1.34 20 mmol) were added to a suspension of C94 (100 mg, 0.26 mmol) In dichloromethane (1 mL) at 16 ’C. The reaction mixture was stirred at 10 ’C for 14 hours, whereupon it was concentrated in vacuo and combined with material from a similar reaction carried out using C94 (30 mg, 78 pmol). The resulting mixture was purified via silica gel chromatography (Gradient: 0% to 15% methanol in dichloromethane) to provide the product as a yellow gum. Yield: 90 mg, 0.25 mmol, 25 74%. LCMS m/z 383.3 [M+Na*].
Step 4. Synthesis of tert-butyl (3R)-3-[(cyclopropylsu!fonyl)(methyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate (C96).
To a 0 ’C suspension of C95 (90 mg, 0.25 mmol) in W,A/-dlmethylformamlde (1 mL) was added sodium hydride (60% dispersion in minerai oil; 20 mg, 0.50 mmol), and the reaction mixture was stirred at 0 ’C for 30 minutes, lodomethane (53.2 mg, 0.375 mmol) was added at 0 ’C, and the reaction mixture was stirred at 15 ’C for 2 hours. It was then treated with saturated aqueous sodium chloride solution (40 mL) and extracted with ethyl acetate (3 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 50% ethyl acetate in petroleum ether) provided the product as a colorless gum. Yield: 78 mg, 0.21 mmol, 84%. LCMS m/z 397.3 [M+Na*]. Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl (3R)-3[(cyclopropylsulfonyl)(methyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C97).
163
Conversion of C96 to the product was carried out using the method described for synthesis of C89 from C88 in Example 92. The product was obtained as a colorless gum. Yield: 67 mg, 0.12 mmol, 57%. LCMS m/z 573.0 [M+Na*].
Step 6. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3R)· 35 [(cyclopropylsulfonyl)(methyl)amino]-1-oxa-8-azaspiro[4.5Jdecane-8-carboxylate (93).
Conversion of C97 (67 mg, 0.12 mmol) to the product was carried out using the method described for synthesis of 93 from C91 in Example 93. In this case, purification was effected using reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 35% to 55% B), affording 10 the product as a brown gum. Yield: 10.0 mg, 23.2 pmol, 19%. LCMS m/z 431.2 [M+H]*. 1H NMR (400 MHz, CDCh) δ 5.32-5.20 (m, 1H), 4.73-4.63 (m, 1 H), 4.04-3.93 (m, 2H), 3.93-3.74 (m, 4H), 3.46-3.24 (m, 2H), 2.88 (s, 3H), 2.55-2.25 (v brs, 1H), 2.30-2.21 (m, 1H), 2.14-2.02 (m, 1H), 1.86-1.68 (m.4H), 1.56-1.41 (m, 1H), 1.22-1.14 (m, 2H), 1.04-0.96 (m, 2H).
Example 95 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 3-phenyl-1-oxa-8-azaspiro[4.5]decane-8-carboxylate [From C101, ENT-2] (95)
164
Step 1. Synthesis oftert-butyl 4-(2,3-dibromopropyl)-4-hydroxypiperidine-1-carboxylate (C98). This réaction was carried out in two identical batches. A solution of fert-butyl 4-hydroxy-
4-(prop-2-en-1-yl)piperidine-1-carboxylate (209 g, 0.866 mol) in dichloromethane (1.2 L) was cooled in a cold water bath. A solution of bromine (152 g, 0.951 mol) in dichloromethane (250 mL) was added at such a rate that the color of the réaction mixture did not become intense. At the conclusion of the addition, an aqueous solution containing sodium thiosulfate and sodium bicarbonate was added to the reaction mixture, and stirring was continued until the mixture had completely decolorized. At this point, the two batches were combined. The aqueous layer was 10 extracted with dichloromethane (3 x 400 mL), and the combined organic layers were washed with saturated aqueous sodium chloride solution (2 x 200 mL), dried over sodium sulfate, and concentrated in vacuo to afford the product as a red gum. Yield: 600 g, 1.5 mol, 87%. 1H NMR (400 MHz, CDCh) δ 4.43-4.33 (m, 1H), 3.96-3.74 (m, 2H), 3.91 (dd, 7=10.3, 4.0 Hz, 1H), 3.66 (dd, 7=10.0, 9.8 Hz, 1H), 3.27-3.13 (m, 2H), 2.47 (dd, half of ABX pattern, 7=15.8, 2.8 Hz, 1H), 15 2.13 (dd, half of ABX pattern, 7=15.7, 8.9 Hz, 1H), 1.78-1.68 (m, 2H), 1.65-1.53 (m, 2H, assumed; partially obscured by water peak), 1.47 (s, 9H).
Step 2. Synthesis oftert-butyl 3-bromo-1-Oxa-8-azaspiro[4.5]decane-8-carboxylate (C99). Potassium carbonate (119 g, 861 mmol) was added to a cooled solution of C98 (230 g, 573 mmol) in methanol (1.5 L), and the reaction mixture was stirred at 10 ’C to 15 ’C for 16 20 hours. The crude reaction mixture was combined with the crude réaction mixtures from two similar reactions using C98 (350 g, 873 mmol, and 20 g, 50 mmol) and filtered. The filtrate was concentrated in vacuo, and the resulting red oil was recrystallized from petroleum ether (150 mL) at 0 ’C to provide a light yellow solid (360 g). This was subjected to silica gel chromatography (Eluent: dichloromethane), and the purified material was recrystallized from 25 petroleum ether (120 mL) and washed with petroleum ether (3 x 40 mL) to afford the product as a white solid (180 g). The mother liquors from recrystallization were concentrated under reduced
165
pressure and purified by silica gel chromatography (Gradient: 0% to 20% ethyl acetate in petroleum ether). The resulting material was recrystallized from petroleum ether (100 mL) and washed with petroleum ether (3 x 40 mL), affording additional product as a white solid (95 g). Combined yield: 275 g, 0.859 mol, 57%. 1H NMR (400 MHz, DMSO-Λ) δ 4.71-4.63 (m, 1H), 5 4.12 (dd, J=10.4,4.9 Hz, 1H), 3.90 (dd, J=10.5, 3.8 Hz, 1H), 3.52-3.40 (m, 2H), 3.3-3.15 (m,
2H), 2.41 (dd, J=14.3, 7.3 Hz, 1H), 2.10 (dd, J=14.0, 4.0 Hz, 1H), 1.79-1.71 (m, 1H), 1.65 (br ddd, half of ABXY pattern, J=13,10,4 Hz, 1H), 1.55-1.41 (m, 2H), 1.39 (s, 9H).
Step 3. Synthesis ofted-butyl 3-phenyl-1-oxa-8-azaspiro[4.5]decane-8-carboxy!ate, ENT-1 (C100) andted-butyl 3-phenyl-1-oxa-8-azaspiro[4.5]decane-8-carboxy!ate, ENT-2 (C101).
A mixture of C99 (150 mg, 0.468 mmol), phenylboronic acid (114 mg, 0.935 mmol), frans-2-aminocyclohexanol (10.8 mg, 93.7 pmol) and nîckel(ll) iodide (29.3 mg, 93.7 pmol) In 2propanol (3 mL, previously dried over molecular sieves) was treated with sodium bis(trimethylsilyl)amide (1 M solution In tetrahydrofuran; 0.937 mL, 0.937 mmol). The reaction vessel was then capped, warmed to 60 ’C, and stirred for 14 hours. The resulting suspension was combined with a similar reaction mixture carried out using C99 (50 mg, 0.16 mmol), filtered through a pad of diatomaceous earth, and concentrated In vacuo. The residue was purified via chromatography on silica gel (Gradient: 0% to 40% ethyl acetate In petroieum ether) to afford the racemic product as a white soiid. Yield: 170 mg, 0.536 mmol, 85%. 1H NMR (400 MHz, CDCh) δ 7.36-7.30 (m, 2H), 7.3-7.21 (m, 3H, assumed; partially obscured by solvent peak),
4.23 (dd, 7=8, 8 Hz, 1 H), 3.80 (dd, 7=9, 9 Hz, 1 H), 3.70-3.47 (m, 3H), 3.44-3.33 (m, 2H), 2.27 (dd, <7=12.5, 8 Hz, 1H), 1.84 (dd,7=12,11 Hz, 1H), 1.79-1.67 (m, 3H), 1.64-1.55 (m, 1H, assumed; partially obscured by water peak), 1.47 (s, 9H).
The component enantiomers were separated using supercritical fluid chromatography [Column: Chiral Technologies Chiralpak AD, 10 pm; Mobile phase: 35% (0.1% ammonium hydroxide in 25 methanol) In carbon dioxîdej. The first-eluting enantiomer was assigned as C100. Yield: 65 mg,
38% for the séparation. LCMS m/z 262.1 [(M - 2-methylprop-1-ene)+H]*. The second-eluting enantiomer was assigned as C101. Yield: 70 mg, 41% for the séparation. LCMS m/z 262.1 [(M - 2-methylprop-1-ene)+HJ*.
Step 4. Synthesis of(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxyJpropan-2-yl3-phenyl-1-oxa-830 azasplro[4.5]decane-8-cart)Oxylate [From C101, ENT-2J (C102).
Trifluoroacetîc acid (0.6 mL) was added drop-wise to a solution of C101 (70.0 mg, 0.220 mmol) In dichloromethane (2 mL), and the reaction mixture was stirred at 25 eC for 2 hours. Volatiles were removed under reduced pressure to provide 3-phenyl-1-oxa-8azaspiro[4.5]decane, trifluoroacetate sait, as a yellow gum. This material was dissolved In acetonitrile (2 mL), cooled to 0 ’C, and slowly treated with triethylamine (89.5 mg, 0.884 mmol). After this solution had stirred for 30 minutes, C2 (reaction solution In acetonitrile containing 0.221 mmol) was added at 0 °C. The reaction mixture was stirred at 25 ’C for 18 hours, whereupon It was concentrated In vacuo and purified by préparative thin layer chromatography
166
on silica gel (Eluent: 3:1 petroleum ether / ethyl acetate), affording the product as a yellow gum (120 mg). This material was taken directly to the following step. LCMS m/z 516.1 [M+Na*]. Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-phenyl-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C101, ENT-2] (95).
Trifluoroacetic acid (0.5 mL) was added to a 0 ’C solution of C102 (from the previous step: 120 mg, £0.220 mmol) In dichloromethane (1.5 mL). The reaction mixture was stirred at 25 ’C for 2 hours, whereupon It was concentrated in vacuo and subjected to préparative thin layer chromatographyon silicagel (Eluent: 3:1 petroleumether/ethyl acetate). Thematerialobtained (40 mg) was then purified using reversed phase HPLC (Column: Dalso C18, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 42% to 72% B) to afford the product as a coloriess gum. Yield: 10.1 mg, 27.0 pmol, 12% over 2 steps. LCMS m/z 373.9 [M+H]*. ’H NMR (400 MHz, CDCh) δ 7.36-7.30 (m, 2H), 7.27-7.22 (m, 3H), 5.32-5.21 (m, 1H), 4.24 (dd, >8.0, 8.0 Hz, 1H), 4.01 (dd, half of ABX pattern, >12.4, 2.9 Hz, 1H), 3.92-3.74 (m, 4H), 3.60-3.35 (m, 3H), 2.32-2.22 (m, 1H), 1.92-1.55 (m, 5H, assumed;
partially obscured by water peak).
Example 96 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl S-phenyi-l-oxa-e-azaspiro^.SJdecane-e-carboxylate [From C100, ENT-1J(96)
Step 1. Synthesis of (2R)-1,1,1-tnfluoro-3 [(4-mcthoxybonzyl)oxy]propan-2-yl 3-phenyl-1-oxa-8azaspiro[4.5Jdecane-8-carboxylate [From C100, ENT-1] (C103).
Trifluoroacetic acid (2 mL) was added to a 0 ’C suspension of C100 (65 mg, 0.20 mmol) in dichloromethane (3 mL). The reaction mixture was stirred at 16 ’C for 2 hours, whereupon it 25 was concentrated In vacuo to provïde the deprotected material as a yellow gum. The gum was dissolved in acetonitrile (1 mL), cooled to 0 ’C, and treated with C2 (réaction solution In
167 acetonitriie containing 0.24 mmol) and triethylamine (166 mg, 1.64 mmol). This reaction mixture was stirred at 18 ’C for 16 hours, and then treated with additional C2 (reaction solution in acetonitriie containing 0.24 mmol). Stirring was continued at 18 ’Cfor an additional 16 hours. Volatiles were removed under reduced pressure, and the residue was subjected to chromatography on silica gel (Gradient: 0% to 100% ethyl acetate in petroleum ether) to afford the product as a yellow gum (101 mg). This material was used in the following step without additional purification. LCMS m/z 516.1 [M+Na*]
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-phenyl-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C100, ENT-1](96).
Trifluoroacetic acid (2 mL) was added to a 0 eC suspension of C103 (from the previous step: £0.20 mmol) in dichloromethane (2 mL), and the reaction mixture was stirred at 20 *C for 1 hour. After the reaction mixture had been cooled to 0 eC, aqueous sodium bicarbonate solution (40 mL) was slowly added, and the purple mixture became colorless. It was extracted with dichloromethane (3 x 20 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 50% ethyl acetate in petroleum ether) was followed by reversed phase HPLC (Column: Agela Durashell, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitriie; Gradient: 5% to 95% B), affording the product as a yeliow oil. Yield: 15.2 mg, 40.7 pmol, 20% over 2 steps. LCMS m/z 373.9 [M+H]*. ’H NMR (400 MHz, CDCb) δ 7.36-7.30 (m, 2H), 7.2820
7.21 (m, 3H), 5.32-5.21 (m, 1H), 4.24 (dd, J=8.3,7.8 Hz, 1H), 4.06-3.97 (m, 1H), 3.93-3.74 (m,
4H), 3.60-3.32 (m, 3H), 2.49-2.38 (m, 1H). 2.27 (dd, J=12.6, 8.3 Hz, 1H), 1.89-1.6 (m. 4H), 1.87 (dd, J=12.0,10.8 Hz, 1H).
Example 97 (2R)-1,1,1-Trifluoro-3-hydroxypmpan-2-yl 3-(5d]uompyridin-2-yl)-1-oxa-8-azaspirO[4.5]decane·
8-carboxy!ate, trifluoroacetate sait (97)
O CF3 nAcA^0PMB
Br · CFaCOOH NEt3
C1Û4
Br
C105
168
Step 1. Synthesis of 3-bmmo-1-oxa-8-azaspiro[4.5}decane, trifluoroacetate sait (C104). Trifluoroacetic acid (100 mL) was added drop-wise to a 0 ’C solution of C99 (25.0 g,
78.1 mmol) in dichloromethane (400 mL). After the reaction mixture had been stirred at 13 ’C for 15 hours, it was concentrated In vacuo to afford the product as a brown oil (30 g). This material was used in the next step without additional purification. 1H NMR (400 MHz, CDjOD) δ 4.63-4.55 (m, 1H), 4.20 (dd, half of ABX pattern, J=10.5,4.5 Hz, 1H), 4.04 (dd, half of ABX pattern, J=10.5, 3.5 Hz. 1H), 3.3-3.21 (m, 4H), 2.50 (dd, half of ABX pattern, J=14.6, 7.0 Hz, 1H), 2.30-2.18 (m, 2H), 1.97 (ddd, J=14,10, 6.5 Hz, 1H), 1.91-1.77 (m, 2H).
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3-bromo-1-oxa-8azaspiro[4.5]decane-8-carboxylate (C105).
Triethylamine (39.5 g, 390 mmol) was added to a 15 ’C solution of C104 (from the previous step; 30 g, £78.1 mmol) in acetonitrile (400 mL). The resulting solution was stirred at 15 ’C for 1 hour, whereupon it was cooled to 0 ’C and treated with C2 [reaction solution In acetonitrile (400 mL) containing 85.9 mmol). After the reaction mixture had been stirred at 13 ’C for 15 hours, it was concentrated in vacuo and purified twice via chromatography on silica gel (Gradient: 5% to 9% ethyl acetate in petroleum ether). A final silica gel chromatographie purification (Gradient: 0% to 9% ethyl acetate in petroleum ether) afforded the product as a colorless gum. Yield: 20.3 g, 40.9 mmol, 52% over 2 steps. LCMS m/z 519.8 (bromine isotope pattern observed) [M Wa*]. 1H NMR (400 MHz. CDCh) δ 7.25 (d. J=8.5 Hz, 2H). 6.89 (d, J=8.7 Hz, 2H), 5.54-5.43 (m, 1H), 4.51 (AB quartet, upfield doublet is broadened, Jab=11.7 Hz, Avab=29.1 Hz, 2H), 4.44-4.36 (m, 1H), 4.19 (dd, J=10.4, 5.3 Hz, 1H), 4.07-3.99 (m, 1H), 3.91-
3.63 (m, 4H), 3.82 (s, 3H), 3.44-3.27 (m, 2H), 2.42-2.25 (m, 1H), 2.24-2.08 (m, 1H), 2.04-1.89 (m, 1H), 1.81-1.47 (m, 3H).
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzy!)oxy]propan-2-yl 3-(4,4,5,5tetramethyl-1,3,2-dioxaborolan-2-yl)· 1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C106).
169
A mixture of C105 (6.50 g, 13.1 mmol), 4,4,4',4',5,5,5,,5,-octamethyl-2,2'-bi-1,3,2dioxaborolane (4.99 g, 19.6 mmol), polymer-bound triphenylphosphine (687 mg, 2.62 mmol), lithium methoxîde (995 mg, 26.2 mmol), and copper(l) lodide (249 mg, 1.31 mmol) in Λ/,Λ/dimethytformamide (50 mL) was stirred at 1 ’C to 10 ’C for 16 hours. The reaction mixture was 5 then diluted with dichloromethane (150 mL) and filtered; the filter cake was washed with dichloromethane (150 mL), and the combined filtrâtes were concentrated in vacuo. The resulting oil was mixed with saturated aqueous ammonium chloride solution (150 mL) and extracted with diethyl ether (3 x 150 mL). The combined organic layers were washed sequentially with water (150 mL) and saturated aqueous sodium chloride solution (150 mL), dried over sodium sulfate, 10 filtered, and concentrated under reduced pressure to provide the product as a pale yellow gum.
Yield: 7.00 g, 12.9 mmol, 98%. 1H NMR (400 MHz, CDCh) δ 7.24 (br d, >8.8 Hz, 2H), 6.88 (br d, >8.8 Hz, 2H), 5.53-5.42 (m, 1H), 4.51 (AB quartet, Jab-1 1.7 Hz, ΔνΑΒ=27.2 Hz, 2H), 4.03 (dd, >8.3, 8.2 Hz. 1 H), 3.81 (s, 3H), 3.80-3.63 (m, 5H), 3.45-3.30 (m, 2H), 1.98-1.74 (m, 2H), 1.72-1.40 (m, 5H), 1.25 (s, 12H).
Sfep 4. Synthesis of potassium trifluoro{8-[({(2R)-1,1,1-trtfiuoro-3-[(4~ methoxybenzyl)oxy]propan-2-yi)oxy)carbonyl]-1-oxa‘8‘azaspiro[4.5ldec-3-yl}borate(1-) (C107).
Aqueous potassium hydrogenfluoride solution (4.5 M, 11.5 mL, 51.8 mmol) was added to a 0 ’C solution of C106 (7.00 g, 12.9 mmol) in tetrahydrofuran (50 mL) and the reaction mixture was stined at 0 ’C to 5 ’C for 16 hours. Removal of volatiles In vacuo provided a thick 20 oil, which was extracted with acetone (4 x 75 mL). The combined acetone layers were filtered, and the filtrate was concentrated to a volume of approximately 20 mL, cooled to 0 ’C, and diluted with diethyl ether (150 mL). A white tacky material appeared; the solvent was removed via décantation, and the remaining gum was triturated with diethyl ether (150 mL) to afford the product as a white solid. Yield: 3.8 g, 7.26 mmol, 56%. LCMS m/z 483.9 [M‘]. Ή NMR (400 25 MHz, acetone-de) δ 7.27 (br d, >8.7 Hz, 2H), 6.91 (br d, >8.7 Hz, 2H), 5.55-5.43 (m, 1 H), 4.52 (AB quartet, JAB=11.6 Hz, ΔνΑΒ=19.0 Hz, 2H), 3.84-3.70 (m, 3H), 3.79 (s, 3H), 3.70-3.53 (m, 3H), 3.44-3.23 (m, 2H), 1.70-1.58 (m, 1H), 1.58-1.45 (m,4H), 1.45-1.34 (m, 1H), 1.30-1.14 (m, 1H).
Step 5. Synthesis of (2R)-1,1,1-triïJuofO-3-hydroxypropan-2-y! 3-(5-fluoropyridin-2-yl)-1-oxa-830 azaspiro[4.5]decane-8-carboxylate, trifluoroacetate sait (97).
A mixture of 2-bromo-5-fluoropyridine (17.6 mg, 0.100 mmol), C107 (78.3 mg, 0.150 mmol), [!r{dFCF3ppy}2(bpy)f PF6* (2.5 mg, 2.4 pmol), césium carbonate (48.9 mg, 0 150 mmol), nickel(ll) chioride, 1,2-dimethoxyethane adduct (1.1 mg, 5.0 pmol), and 4,4'-di-tert-butyl-2,2'blpyridine (1.4 mg, 5.2 pmol) was degassed under vacuum and then purged with nitrogen; this 35 evacuation-purge cycle was carried out a total of three times. 1,4-Dioxane (7 mL) was added, and the reaction mixture was sonicated and shaken to provide a suspension. The reaction mixture was then Irradiated with blue visible light (wavelength: 460 nm) from a 60 watt blue LED strip for 18 hours. After removal of volatiles in vacuo, a mixture of dichloromethane (0.5 mL) and
170 trifluoroacetic acid (0.5 mL) was added, and the resulting reaction mixture was stirred at room température for 30 minutes. The reaction mixture was concentrated in vacuo, and the residue was purified via reversed phase HPLC (Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid In water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 10% to 100% B). The product was assumed to be a mixture of two diastereomers. Yield: 1.4 mg, 2.7 pmol, 3%. LCMS m/z 393.3 [M+H]*. Rétention time: 2.96 minutes [Analytical HPLC conditions - Column: Waters Atlantis dC18,4.6 x 50 mm, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 5.0% to 95% B, linear over 4.0 minutes Flow rate: 2 mL/mlnute].
Example 98 (2R)-1,1,1-TnfJuoro-3-hydroxypropan-2-yl 2-(2-fluorobenzoyl)-2,8-diazaspîro[4.5]decane-8carboxylate (98)
HN
Step 1. Synthesis of tert-butyl 2-(2-fluorobenzoyl)-2,8-diazaspiro[4.5]decane-8-carboxy!ate (C108).
To a suspension of tert-butyl 2,8-diazaspiro[4.5]decane-8-carboxylate (200 mg, 0.832 mmol) in acetonitrile (2 mL) were added 2-fluorobenzoic acid (175 mg, 1.25 mmol), O-(7azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU; 506 mg, 1.33 mmol), and Λ/,Ν-diisopropylethylamine (323 mg, 2.50 mmol). The réaction mixture was stirred at 25 ’C for 16 hours, whereupon it was concentrated in vacuo. The residue was dissolved in methanol (8 mL), treated with Ion exchange resin Amberlyst® A26, hydroxide form [3.6 g, prewashed with methanol (7 mL)], stirred at 25 ’C for 1 hour, and filtered. The filtrate was
171
concentrated under reduced pressure and subjected to silica gel chromatography (Gradient: 0% to 50% ethyl acetate in petroleum ether), affording the product as a colorless gum. By ’H NMR analysis, this was judged to be a mixture of rotamers. Yield: 231 mg, 0.637 mmol, 77%. ’H NMR (400 MHz, CDaOD) δ 7.55-7.46 (m, 1H), 7.45-7.38 (m, 1H), 7.32-7.26 (m, 1H), 7.26-7.18 (m,
1 H), 3.72-3.66 (m, 1 H), 3.56-3.47 (m, 1 H), 3.51 (s, 1 H), 3.46-3.3 (m, 4H), 3.21 (br s, 1 H), [1.94 (dd, >7.5, 7.3 Hz) and 1.87 (dd, >7.3, 7.0 Hz), total 2H], 1.66-1.48 (m, 4H), [1.47 (s) and 1.43 (s), total 9H].
Step 2. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy}propan-2-yl 2-(2fluorobenzoyl)-2,8-diazaspiro[4.5]decane~8-carboxylate (C109).
Conversion of C108 to the product was carried out using the method described for synthesis of C89 from C88 in Example 92. The product was obtained as a colorless gum. Yield: 500 mg, 0.93 mmol, quantitative. LCMS m/z 539.1 [M+H]+.
Step 3. Synthesis of (2R)-1,1,1-tnfluoro-3-hydroxypropan-2-yl 2-(2-fluorobenzoyl)-2,8diazaspiro[4.5]decane~8-carboxylate (98).
Conversion of C109 to the product was carried out using the method described for synthesis of from C89 in Example 92. In this case, the gradient used for HPLC purification was 36% to 56% B, and the product was Isolated as a colorless gum. By ’H NMR analysis, this was judged to be a mixture of rotamers. Yield: 89 mg, 0.21 mmol, 23%. LCMS m/z 419.0 [M+Hj*. ’H NMR (400 MHz, CDCh) δ 7.46-7.37 (m, 2H), 7.25-7.19 (m, 1H), 7.15-7.08 (m, 1H), 5.32-5.19 (m, 1H),
4.04-3.94 (m, 1 H), 3.92-3.79 (m, 1 H), 3.79-3.62 (m, 2H), 3.58 (s, 1 H), 3.56-3.30 (m, 4H), 3.20 (s, 1H), 2.6-2.3 (br m, 1H), [1.90 (dd, >7.5, 7.3 Hz) and 1.82 (dd, >7.0, 7.0 Hz), total 2H],
1.74-1.47 (m, 4H, assumed; partially obscured by water peak).
Examples 99 and 100 (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 3-[benzoyl(methyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C112, DIAST-2] (99) and (2R)-1,1,1-Trifluom-3hydroxypropan-2-yi 3-[benzoyl(methyl)amino]-1-oxa-8-azasplro[4.5]decane-8-carboxylate [From C111, DIAST-1] (100)
172
o cf3
ΑθΑ^ΟΡΜΒ
C73
cf3cooh
Ο
Ο CF3
AqA^opmb
C110 cf3
AqA^opmb
DIAST-2
C112 cf3cooh
Sfep 1, Synthesis of (2R)-1,1,1-trifluoro~3-[(4-methoxybenzyl)oxy]propan-2-yl 3-(benzoylamino)1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C11O).
A solution of benzoyl chloride (58.5 mg, 0.416 mmol) in dichloromethane (0.5 mL) was added to a 0 ’C solution of C73 (150 mg, 0.347 mmol) and triethylamine (105 mg, 1.04 mmol) In dichloromethane (2 mL). The reaction mixture was stirred at 25 ’C for 3 hours, whereupon saturated aqueous ammonium chloride solution (2 mL) was added, and the resulting mixture was extracted with dichloromethane (2x3 mL). The combined organic layers were washed with 10 saturated aqueous sodium chloride solution (2x3 mL), filtered. and concentrated in vacuo.
Silica gel chromatography (Gradient: 5% to 20% ethyl acetate in petroleum ether) provided the product as a colorless gum. Yield: 135 mg, 0.252 mmol, 73%. LCMS m/z 559.1 [M+Na*].
Sfep 2. Synthesis of (2R)-1,1,1-trifluoro~3-[(4-methoxybenzyl)oxy]propan-2-yl 3[benzoy!(methyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate, DIAST-1 (C111) and (2R)15 1,1,1-tnfluoro-3-[(4-mcthoxybûnzyl)oxy]piOpan-2-yl 3-[benzoyl(nicthyl)amino]-1-oxa-8azaspirc[4.5]decane-8-cartoxylate. DIAST-2 (C112).
Sodium hydride (60% dispersion in minerai oil; 17.1 mg, 0.428 mmol) was added to a 0 ’C solution of C110 (115 mg, 0.214 mmol) in dry tetrahydrofuran (2 mL), and the reaction mixture was stirred at 0 ’C for 30 minutes, lodomethane (45.6 mg, 0.321 mmol) was added, and 20 stirring was continued at 25 ’C for 2 hours. The réaction mixture was then combined with a
173
similar reaction mixture using C110 (20 mg, 37 pmol) and cooled to 0 °C. Saturated aqueous ammonium chloride solution (5 mL) was added, and the resulting mixture was extracted with ethyl acetate (2x5 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (2x5 mL), filtered, and concentrated in vacuo to afford the product, a 5 diastereomeric mixture of C111 and C112, as a colorless gum. Yield of diastereomeric product mixture: 130 mg, 0.236 mmol, 94%. LCMS m/z 573.2 [M+Na*]. 1H NMR (400 MHz, CDCh), characteristic peaks: δ 7.46-7.40 (m, 3H), 7.40-7.34 (m, 2H), 7.24 (br d, 4=8.5 Hz, 2H), 6.88 (br d, 4=8.7 Hz, 2H), 5.53-5.42 (m, 1H), 4.51 (AB quartet, 4ab=11.7 Hz, Avab=28.4 Hz, 2H), 3.91-
3.85 (m, 1H), 3.85-3.63 (m, 3H), 3.82 (s, 3H), 3.42-3.19 (m, 2H), 3.07-2.89 (m, 3H), 1.85-1.67 10 (m, 3H).
The component diastereomers were separated via supercritical fluid chromatography (Column: Chiral Technologies Chiralpak IC, 10 pm; Mobile phase: 40% (0.1% ammonium hydroxide in 2-propanol) in carbon dioxide]. The first-eluting diastereomer was C111 (50 mg) and the second-eluting diastereomer was C112 (55 mg).
Sfep 3. Synthesis of (2RJ-1,1,1-trifluoro-3~hydmxypmpan-2-yl 3-[benzoyl(methyl)amino]- 1-oxa8-azaspim[4.5]decane-8-carboxylate [Fmm C112, DIAST-2] (99).
Trifluoroacetic acid (0.5 mL) was added to a solution of C112 (55 mg, 0.10 mmol) in dichloromethane (1 mL), and the reaction mixture was stirred at 18 *C for 2 hours. Saturated aqueous sodium bicarbonate solution was added until the pH reached 8-9, and the resulting 20 mixture was extracted with dichloromethane (2x2 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution, filtered, dried over sodium sulfate, and concentrated in vacuo. Reversed phase HPLC (Column: Agela Durashell, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 8% to 58% B) afforded the product as a white solid. Yield: 15.6 mg, 36.2 pmol, 36%. LCMS m/z 431.0 25 [M+H]*. 1H NMR (400 MHz, CDCh), characteristic peaks: δ 7.46-7.34 (m, 5H), 5.30-5.20 (m,
1H), 4.04-3.96 (m, 1H), 3.92-3.68 (m, 4H), 3.44-3.15 (m, 2H), 3.07-2.89 (m, 3H), 2.46-1.96 (m, 2H), 1.87-1.72 (m, 3H).
Sfep 4. Synthesis of (2R)-1,1,1-trifluom-3-hydmxypmpan-2-yl 3-[benzoyl(methyl)amlno]-1-oxa8-azaspim[4.5]decane-8-carboxy!ate [Fmm C111, DIAST-1] (100).
Conversion of Cl 11 to the product was effected using the method employed for synthesis of 99 from C112. Yield: 17.4 mg, 40.4 pmol, 44%. LCMS m/z 431.0 [M+H]*. 1H NMR (400 MHz, CDCh), characteristic peaks: δ 7.46-7.40 (m, 3H), 7.40-7.34 (m, 2H). 5.30-5.20 (m, 1H), 4.06-3.95 (m, 1H), 3.94-3.70 (m, 4H), 3.48-3.21 (m, 2H), 3.08-2.88 (m, 3H), 2.43-2.27 (m, 1H), 1.88-1.72 (m, 3H).
Example 101 (2R)~1,1,1-Trifluom-3-hydmxypmpan-2-yl 3-(1,1-dioxfdo~1,2-benzothiazo/-2(3H)-y/)-1-oxa-8azaspim[4.5]decane-8-carboxylate (101)
174
O CF;
Ο CF3
Step 1. Synthesis of2,3-dihydro-1,2-benzothiazole 1,1-dioxide (C113).
A solution of 1,2-benzothiazol-3(2H)-one 1,1-dioxide (200 mg, 1.09 mmol) in tetrahydrofuran (2 mL) was added drop-wise to a 0 ’C suspension of lithium aluminum hydride (45.6 mg, 1.20 mmol) in tetrahydrofuran (3 mL). After the reaction mixture had stirred for 30 minutes at 0 ’C, it was gradually warmed to 15 ’C and stirred at 15 ’C for 16 hours. The white suspension was treated with saturated aqueous ammonium chloride solution, and then extracted with ethyl acetate (20 mL). The organic layer was dried over sodium sulfate, filtered, and concentrated in vacuo to provide the product as a grey solid. Yield: 160 mg, 0.946 mmol,
87%. 1H NMR (400 MHz, CDCb) δ 7.81 (d, >7.8 Hz, 1H), 7.63 (dd, half of ABX pattern, >7.5,
7.3 Hz, 1H), 7.54 (dd, half of ABX pattern, >7.5,7.5 Hz, 1H), 7.41 (d, >7.8 Hz, 1H), 4.95-4.80 (brs, 1H>, 4.55 (s, 2H).
Step 2. Synthesis of (2R)-1,1,1-trifiuoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3-(1,1-dioxldo-1,2benzothiazol-2(3H)-yl)-1 -oxa-8-azaspiro[4.5]decane-8-carboxyla te (C114).
A mixture of C105 (80 mg, 0.16 mmol), C113 (39.3 mg, 0.232 mmol), césium carbonate (114 mg, 0.350 mmol), and potassium iodide (28.9 mg, 0.174 mmol) in W,N-dimethylformamide (2 mL) was stirred at 80 ’C for 16 hours. The reaction mixture was then diluted with ethyl acetate (30 mL), washed with saturated aqueous sodium chloride solution (3 x 30 mL), dried over sodium sulfate, filtered, and concentrated in vacuo. Préparative thin layer chromatography on silica gel (Eluent: 1:3 ethyl acetate / petroleum ether) provided the product as a light yellow oil. Yield: 55 mg, 94 pmol, 59%. LCMS m/z 607.0 [M+Na*].
Step 3. Synthesis of(2R)-1,1,1-trifiuom-3-hydroxypropan-2-yl 3-(1,1-dioxldo-1,2-benzothiazol· 2(3H)-yl)-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (101).
Trifluoroacetic acid (2 mL) was added in a drop-wise mannerto a 0 ’C solution of C114 (55 mg, 94 pmol) in dichloromethane (6 mL). The reaction mixture was stirred at 0 ’C for 1 hour, whereupon it was diluted with saturated aqueous sodium bicarbonate (30 mL) and extracted
175 ►
with ethyl acetate (30 mL). The organic layer was dried over sodium sulfate, filtered, concentrated in vacuo, and purified via reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: water containing 0.225% formlc acid; Mobile phase B: acetonitrile;
Gradient: 30% to 50% B). The product was obtained as a white solid, presumed to be a mixture 5 of diastereomers. Yield: 6.0 mg, 13 pmol, 14%. LCMS m/z 487.0 [M+Na*]. 1H NMR (400 MHz,
CDCh) δ 7.81 (br d, 7=7.5 Hz, 1H), 7.64 (ddd. 7=7.5, 7.5,1.2 Hz, 1H), 7.56 (br dd, 7=7, 7 Hz, 1H), 7.42 (br d, 7=7.8 Hz, 1H), 5.31-5.21 (m, 1H), 4.41 (br AB quartet, 7ab=14 Hz, ΔνΑβ=12 Hz, 2H), 4.4-4.30 (m, 1H), 4.16 (dd, half of ABX pattern, 7=9.7,6.4 Hz, 1H), 4.05 (dd, half of ABX pattern, 7=9.8, 5.5 Hz, 1H), 4.05-3.96 (m, 1H), 3.93-3.73 (m, 3H), 3.50-3.28 (m, 2H), 2.42-2.25 10 (m, 2H), 2.21-2.08 (m, 1H), 1.89-1.70 (m, 3H).
Example 102 (2R)-1,1,1-Trifluom-3-hydroxypropan-2-yl 3-[(5-methylpyridin-2-yi)methyl]-1-oxa-8azaspiro[4.5Jdecane-8-carboxylate ( 102)
(Ph3PMe)+ Br
CF3COOH
C117
F F
Pd(dppf)CI2 K2CO3 • CF3COOH
Step 1. Synthesis oftert-butyl 3-methylidene-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C115).
176
Methyltriphenylphosphonium bromlde (8.4 g, 24 mmol) was added portion-wise to a mixture of sodium hydride (60% dispersion in minerai oil; 940 mg, 23.5 mmol) In dimethyl sulfoxide (40 mL), and the reaction mixture was stirred for 30 minutes at room température. A solution of tert-butyl 3-oxo-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (2.0 g, 7.8 mmol) in 5 dimethyl sulfoxide (18 mL) was then added drop-wise, and the reaction mixture was allowed to continue stirring at room température for 72 hours, The reaction was then carefully quenched with water (250 mL), and extracted with diethyl ether (5 x 50 mL). The combined organic layers were washed with water (2 x 25 mL), dried over magnésium sulfate, filtered, and concentrated in vacuo. The residue was triturated three times with heptane to afford an off-white solid, which 10 proved to be largely triphenylphosphine oxide on analysis. The combined heptane portions from the triturations were concentrated in vacuo and subjected to silica gel chromatography (Eluents.· 0%, followed by 10% and 20% ethyl acetate ln heptane), which afforded the product as a coloriess oil. Yield; 1.77 g, 6.99 mmol, 90%. GCMS m/z 253.1 [M*]. 1H NMR (400 MHz, CDCh) δ 5.02-4.98 (m, 1H), 4.95-4.91 (m, 1H), 4.37-4.33 (m. 2H), 3.60 (ddd, J=13, 5, 5 Hz, 2H), 3.34 15 (ddd. J=13.3, 9.9, 3.3 Hz, 2H), 2.42-2.38 (m, 2H), 1.70-1.63 (m, 2H), 1.55 (ddd, J=13.3,10.0, 4.5 Hz, 2H), 1.46 (s, 9H).
Step 2. Synthesis of tert-butyl 3-[(5-methyipyridin-2-yl)methyl]-1-oxa-8-azaspiro[4.5]decane-8carboxylate (C116).
Compound C115 (200 mg, 0.789 mmol) was dissolved in a 9-borabicyclo[3.3.1]nonane 20 solution (0.5 M in tetrahydrofuran; 1.58 mL, 0.79 mmol). After the reaction vessel had been capped, the reaction mixture was stirred at 70 ’C for 1 hour, whereupon it was cooled to room température and added to a mixture of 2-bromo-5-methylpyridine (123 mg, 0.715 mmol), [1,Tbis(diphenylphosphino)ferrocene]dichloropalladium(ll), dichloromethane complex (32 mg, 39 pmol), and potassium carbonate (109 mg, 0.789 mmol) ln a mixture of /V.N-dimethylfbrmaniide 25 (1.7 mL) and water (170 pL). The reaction vessel was capped and stirred at 60 ’C ovemîght.
After the réaction mixture had cooled to room température, it was poured Into water and extracted three times with ethyl acetate. The combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo. The residue was purified via silica gel chromatography (Eluents: 10%, 30 followed by 25%, 50%, and 75% ethyl acetate in heptane) to afford the product as a coloriess oil. Yield: 91 mg, 0.26 mmol, 36%. LCMS m/z 347.3 [M+H]*. 1H NMR (400 MHz, CDCI3) δ 8.398.35 (m, 1H), 7.44 (br d, J=7 Hz, 1H), 7.04 (d, J=7.7 Hz, 1H), 3.95 (dd, J=8.6, 6.6 Hz, 1H), 3.61-
3.47 (m, 2H), 3.55 (dd, J=8.5, 7.8 Hz, 1H), 3.40-3.26 (m, 2H). 2.92-2.75 (m, 3H), 2.32 (s, 3H), 1.92 (dd, J=12.5, 7.3 Hz, 1H), 1.7-1.5 (m,4H), 1.51-1.41 (m, 1H), 1.45 (s, 9H).
Step 3. Synthesis of3-[(5-methylpyridin-2-yl)methyl]-1-oxa-8-azaspiro[4.5]decane, trifluoroacetate sait (C117).
A solution of C116 (91 mg, 0.26 mmol) in dichloromethane (3 mL) was cooled to 0 ’C. Trifluoroacetic acid (1.5 mL) was added, and the reaction mixture was stirred at room
177
température for 1 hour. Solvents were removed under reduced pressure to provide the product as a pale yellow oil (185 mg), which was used directly In the following step. GCMS m/z 246.1 [M*]. ’H NMR (400 MHz, CDCh) δ 8.65-8.62 (br s, 1H), 8.17 (br d, J=8 Hz, 1H), 7.60 (d, J=8.1 Hz, 1H), 3.99 (dd, J=8.8,7.0 Hz, 1H), 3.58 (dd, J=8.6, 8.2 Hz, 1H), 3.40-3.26 (m, 4H), 3.25 (dd, half of ABX pattern, J=14.4,7.0 Hz, 1H), 3.13 (dd, half of ABX pattern, J=14.3, 8.3 Hz, 1H), 2.90-2.77 (m, 1H), 2.58 (s, 3H), 2.11-1.80 (m, 5H), 1.63-1.54 (m, 1H, assumed; partially obscured by water peak).
Step 4. Synthesis of(2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3-[(5methylpyridin-2-yl)methyl]-1-oxa-8-azaspim[4.5]decane-8-carboxyiate (C118).
Bis(pentafluorophenyl) carbonate (132 mg, 0.335 mmol) was added to a 0 *C solution of
C1 (84 mg, 0.34 mmol) in acetonitrile (5 mL). Triethylamine (180 pL, 1.29 mmol) was then added, and the reaction mixture was stirred at room température for 1 hour. In a separate flask, a solution of C117 (from the previous step; 185 mg, £0.26 mmol) In acetonitrile (3 mL) was cooled to 0 °C and treated with triethylamine (360 pL, 2.6 mmol); after this mixture had stirred in the Ice bath for a few minutes, the carbonate solution prepared from C1 was added drop-wise to the solution containing C117. The reaction mixture was stirred at 0 ’C for a few minutes, and then allowed to stir at room température ovemight. It was then concentrated in vacuo, and the resulting oil was taken up in ethyl acetate and washed sequentially with aqueous 1 M hydrochloric acid, saturated aqueous sodium bicarbonate solution, and saturated aqueous sodium chloride solution. The organic layer was dried over sodium sulfate, filtered, and concentrated under reduced pressure. Silica gel chromatography (Eluents: 10%, followed by 25%, 50%, and 75% ethyl acetate in heptane) afforded the product as a coloriess oil. Yield: 93 mg, 0.18 mmol, 69% over two steps. LCMS m/z 523.4 [M+H]*. ’H NMR (400 MHz, CDCh) δ 8.38-8.35 (m, 1H), 7.42 (br dd, J=7.8,1.8 Hz, 1H), 7.24 (br d, J=8.6 Hz, 2H), 7.02 (d, J=7.9 Hz,
1 H), 6.88 (br d, J=8.5 Hz, 2H), 5.53-5.41 (m, 1 H), 4.50 (AB quartet, upfield doublet is broadened, Jab’11.7 Hz, ΔνΑ0=26.8 Hz, 2H), 4.00-3.92 (m, 1H), 3.81 (s, 3H), 3.79-3.62 (m. 4H), 3.59-3.51 (m, 1H), 3.44-3.27 (m, 2H). 2.90-2.75 (m, 3H), 2.32 (s, 3H), 1.96-1.83 (m, 1H), 1.741.38 (m, 5H).
Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[(5-methylpyridin-2-yl)methylJ~1~ 30 oxa-8-azaspiro[4.5]decane-8-carboxylate (102).
Trifluoroacetic acid (2.5 mL) was added portion-wise to a 0 ’C solution of C118 (93 mg, 0.18 mmol) in dichloromethane (5 mL). The reaction mixture was allowed to stir at room température for 75 minutes, whereupon it was concentrated in vacuo and partitioned between dichloromethane and saturated aqueous sodium bicarbonate solution. The organic layer was extracted twice with dichloromethane, and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated under reduced pressure. Chromatography on silica gel (Eluents: 50%, followed by 100% ethyl acetate ïn heptane) provided the product as a coloriess oil, presumed to be a
178 mixture of diastereomers. Yield: 54 mg, 0.13 mmol, 72%. LCMS m/z 403.2 ÎM+H1*. Ή NMR (400 MHz, CDCh) δ 8.38-8.34 (m, 1 H), 7.43 (br dd, 3=7.8, 2.0 Hz, 1 H), 7.03 (d, 3=7.8 Hz, 1 H),
5.30-5.18 (m, 1H), 4.03-3.91 (m, 2H), 3.85 (dd, half of ABX pattern, 3=12.3,6.8 Hz, 1H), 3.823.62 (m, 2H), 3.59-3.51 (m, 1H), 3.48-3.25 (m, 2H), 2.90-2.72 (m, 3H), 2.31 (s, 3H), 1.95-1.86 (m, 1H), 1.75-1.59 (m, 3H), 1.56-1.41 (m, 2H).
Examples 103 and 104 (2R)-1,1,1-TnfJuoro-3-hydroxypmpan-2-yl 3-(1H-pyrazol-1-yl)-1-oxa-8-azaspiro[4.5]decane-8carboxylate [From C120, DIAST-2] (103) and (2R)-1,1,1-Trifluoro-3-hydroxypropan-2-yl 3-(1Hpyrazol-1-yl)-1-oxa-8-azaspiro[4.5]decane-8-carboxylate [From C119, DIAST-1] (104)
o CF3
AoA^.opmb
DIAST-1
C119
O CF3
AqA^opmb
DIAST-2
C120 o CF3
AqA^opmb
CF3C00H
O CF3 <z^N'^Ox1s^'OH Ap ( [fromC119, Λ °IAST-1
104
CF3COOH
Step 1. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3-(1H-pyrazol-1yl)-1-oxa-8-azaspiro[4.5]decane-8-carboxylate, DIAST-1 (C119) and (2R)-1,1,1-trif!uoro-3-[(4methoxybenzyl) oxy]propan-2-yl 3-(1 H-pyrazol- 1-yl)-1-oxa-8-azaspiro[4.5Jdecane-8-carboxylate, DIAST-2 (C120).
A mixture of C105 (200 mg, 0.403 mmol), 1H-pyrazole (54.9 mg, 0.806 mmol), and césium carbonate (394 mg, 1.21 mmol) in Λ/,Ν-dimethylformamide (6 mL) was stirred at 20 ’C for 16 hours. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (3 x 20 rnL), tlie combined organic layers were washed with water (3x10 mL) and with saturated aqueous sodium chlorlde solution (3x10 mL), dried over sodium sulfate, filtered, and concentrated In vacuo. Silica gel chromatography (Eluents: 0%, then 10%, then 25% ethyl acetate In petroleum ether) provided a mixture of diastereomeric products C119 and C120 as a coloriess oil, Yield: 60 mg, 0.124 mmol, 31%. This material was combined with the diastereomeric product mixture (30 mg) from a similar reaction carried out on C105, and
179
subjected to séparation via supercritical fluid chromatography (Column: Chiral Technologies Chiralpak AD, 10 ym; Mobile phase: 2:3 (0.1% ammonium hydroxide in methanol) / carbon dioxide). The flrst-eluting diastereomer was assigned as C119, and the second-eluting diastereomer as C120. Both were obtained as colorless oils. C119: Yield: 43 mg, 48% for the 5 séparation. LCMS m/z 506.1 [M+Na*]. C120: Yield: 38 mg, 42% for the séparation. LCMS m/z
506.1 [M+Na*].
Step 2. Synthesis of (2R)-1,1,1-trif1uoro-3-hydroxypropan-2-yi 3-(1H-pyrazol-1-yi)-1-oxa-8azaspiro[4.5Jdecane-8-carboxylate [From C120, DIAST-2] (103).
To a 0 ’C solution of C120 (38 mg, 78 ymol) in dichloromethane (4 mL) was added 10 trifluoroacetic acid (1 mL), and the reaction mixture was stirred for 1 hour. After solvents had been removed in vacuo, the residue was partitioned between dichloromethane (10 mL) and saturated aqueous sodium bicarbonate solution (20 mL). The aqueous layer was extracted with ethyl acetate (2x10 mL), and the combined organic layers were dried over sodium sulfate, filtered, and concentrated under reduced pressure. Reversed phase HPLC (Column:
Phenomenex Synergi C18,4 ym; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 19% to 49% B) provided the product as a brown gum. Yield: 17.0 mg, 46.7 ymol, 60%. LCMS m/z 363.8 [M+H]*. 1H NMR (400 MHz, CDCh) δ 7.54 (d, >1.5 Hz, 1 H), 7.50 (d, >2.1 Hz, 1H), 6.28 (dd, >2,2 Hz, 1H), 5.30-5.21 (m, 1 H), 5.05-4.97 (m, 1H), 4.26-4.17 (m, 2H), 4.01 (br dd, >12.5, 3 Hz, 1H), 3.92-3.73 (m, 3H), 3.50-3.31 (m, 2H), 2.3820 2.25 (m, 2H), 1.94-1.56 (m, 4H, assumed; partially obscured by water peak).
Step 3. Synthesis of (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-(1iï-pyrazoi-1-yl)-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C119, DIAST-1] (104).
Conversion of C119 to the product was effected using the method described for synthesis of 103 from C120. In this case, the reversed phase HPLC was carried out using a 25 gradient of 37% to 57% B, to provide the product as a brown gum. Yield: 18.2 mg, 50.0 ymol, 56%. LCMS m/z 363.8 [M+H]*. 1H NMR (400 MHz, CDCh), characteristic peaks: δ 7.55 (d, >1.5 Hz. 1H), 7.50 (br s, 1H), 6.28 (brs, 1 H), 5.31-5.20 (m, 1H), 5.05-4.96 (m, 1H), 4.26-4.16 (m, 2H), 4.05-3.97 (m, 1H), 3.93-3.74 (m, 3H), 3.49-3.30 (m, 2H), 2.39-2.25 (m, 2H).
Example 105 (2R)~1,1,1-Trifluoro-3-hydroxypropan-2-yl 2-(phenylsulfonyl)-2,8-diazaspiro[4.5]decane-8carboxyiate (105)
180
A solution of tert-butyl 2,8-diazaspiro[4.5]decane-8-carboxylate (36 mg, 0.15 mmol) In pyridine (0.4 mL) was added to a solution of benzenesulfonyl chloride (39.7 mg, 0.225 mmol) and A/,/V-dimethylpyridin-4-arTiine (0.25 mg, 2.0 pmol) In pyridine (0.4 mL), and the reaction mixture was shaken at room température ter 2 days. The pyridine was removed in vacuo, and the residue was partitioned between half-saturated aqueous sodium bicarbonate solution (1.5 mL) and ethyl acetate (2.4 mL). After the mixture had been vortexed, the organic layer was eluted through a solid phase extraction cartridge (6 mL) charged with sodium sulfate (-1 g); this extraction procedure was repeated twice, and the combined eluents were concentrated in vacuo. The residue was treated with a mixture of 1,2-dichloroethane and trifluoroacetic acid (1:1; 1 mL), shaken at room température for 2.5 hours, and concentrated under reduced pressure. The remaining material was dissolved in 1,2-dichloroethane (2.4 mL), vortexed, and loaded onto an SCX (strong cation exchanger) solid phase extraction cartridge (Silicycle, 6 mL, 1 g); the vial was rinsed with a mixture of methanol and 1,2-dichloroethane (1:1 ; 2 x 2.4 mL).
The cartridge was eluted with methanol (5 mL), followed by a solution of triethylamine in methanol (1 M, 7.5 mL) to elute the deprotected intermediate. Fractions containing the desired material were concentrated in vacuo, and the residue was azeotroped with toluene (2x1 mL) to remove trace methanol. The resulting material was dissolved in dichloromethane (0.5 mL).
A crude solution of C2 was prepared separately, as follows: Bls(pentafluorophenyl) carbonate (5.8 g, 15 mmol) and triethylamine (41 mL, 290 mmol) were added to a stirring solution of C1 (3.75 g. 15.0 mmol) in tetrahydrofuran (30 mL). Sufficlent tetrahydrofuran was added to bring the total volume to 98 mL, and the reaction mixture was stirred at room température for 1 hour. A portion of this crude C2 solution (1.0 mL, 0.15 mmol of C2 and 3 mmol of triethylamine) was added to the deprotected amine solution prepared above, and the reaction mixture was shaken at room température for 5 days. It was then partitioned between half-saturated aqueous sodium bicarbonate solution (1.5 mL) and ethyl acetate (2.4 mL) and
181
subjected to vortexing. The organic layer was eluted through a solid phase extraction cartridge (6 mL) charged with sodium sulfate (*1 g); this extraction procedure was repeated twice, and the combined eluents were concentrated in vacuo. This material was treated with a mixture of trifluoroacetic acid and 1,2-dichloroethane (1:1,1 mL) and shaken at room température for 1 5 hour, whereupon it was concentrated in vacuo and purified using reversed phase HPLC (Column: Waters Sunfire C18, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 25% to 95% B). Yield:
4.8 mg, 11 pmol, 7%. Analytical rétention time: 2.64 minutes (Analytical HPLC conditions Column: Waters Atlantis dC18,4.6 x 50 mm, 5 pm; Mobile phase A: 0.05% trifluoroacetic acid in 10 water (v/v); Mobile phase B: 0.05% trifluoroacetic acid in acetonitrile (v/v); Gradient: 5.0% to
95% B, linear over 4.0 minutes; Flow rate: 2 mL/minute). LCMS m/z 437.1 fM+H]*.
Example 106 (2R)-1,1,1-Trif1uoro-3-hydroxypropan-2-yl (3R)-3-{[(cyclopmpylmethyl)sulfonyl](methyl)amino}-1· oxa-8-azaspiro[4.5]decane-8-carboxylate (106)
C123
182
Pd(PPh3)4
o cf3
AqA^opmb
C125
Cl o=s=o
NEt3
o cf3
AoA^opmb
-N o C126 \pF3C00H tfS^ X Â0£oh
-NO (A_<
Step 1. Synthesis oftert-butyl(3R)-3-{[(prop-2-en-1-yloxy)carbonyl]amino}-1-oxa-8azaspiro[4.5]decane-8-carboxylate (C121).
Prop-2-en-1 -yI carbonochloridate (7.13 g, 59.2 mmol) was added drop-wise to a 0 ’C solution of C94 (15.2 g, 39.4 mmol) in saturated aqueous sodium bicarbonate solution (160 mL) and tetrahydrofuran (40 mL). The reaction mixture was stirred at 10 ’C for 14 hours, whereupon it was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo to afford the product as a pale yellow gum (13.6 g). This material was used directly In the following step. Ή NMR (400 MHz, CDCh) δ 5.98-
5.85 (m, 1H), 5.31 (apparent brdd, 7=17.2,1.4 Hz, 1H), 5.23 (brd, 7=10.3 Hz, 1H), 4.95-4.84 (m, 1H), 4.62-4.51 (m, 2H), 4.39-4.27 (m, 1H), 4.00 (dd, 7=9.4,5.6 Hz, 1H). 3.73-3.52 (m, 3H), 3.38-3.24 (m. 2H), 2.13 (dd, 7=13.3, 7.8 Hz, 1H), 1.74-1.57 (m, 4H, assumed; partially obscured by water peak), 1.56-1.46 (m, 1 H), 1.46 (s, 9H).
Step 2. Synthesis ofted-butyl (3R)-3-{methyl[(prop-2-en-1-yloxy)carbonyl]amino}-1-oxa-8azaspiro[4.5]decane-8-carboxylate (C122).
Sodium hydride (60% dispersion In minerai oil; 2.36 g, 59.0 mmol) was added to a 0 ’C solution of C121 (from the prevlous step; 13.4 g, £38.8 mmol) In tetrahydrofuran (200 mL), and the reaction mixture was stirred at 0 ’C for 30 minutes, lodomethane (16.8 g, 118 mmol) was 20 added drop-wise, and stirring was continued for 16 hours at 0 ’C to 5 ’C. Sodium hydride (60% dispersion in minerai oil; 2.36 g, 59.0 mmol) was again added, and the reaction mixture was stirred at 25 ’C for 16 hours, whereupon it was poured into saturated aqueous ammonium chloride solution (200 mL) and extracted with ethyl acetate (3 x 300 mL). The combined organic layers were washed with saturated aqueous sodium chloride solution (600 mL), dried over 25 sodium sulfate, filtered, and concentrated under reduced pressure to afford the product as a brown gum (16 g). This was used in the following step without additional purification. ’H NMR (400 MHz, CDCh) δ 5.99-5.89 (m, 1H), 5.34-5.27 (m, 1H), 5.24-5.19 (m, 1H), 5.09-4.85 (brm, 1 H), 4.59 (ddd, 7=5.5,1.5,1.4 Hz, 2H), 3.94 (dd, half of ABX pattern, 7=9.7,7.6 Hz, 1 H), 3.76
183
(dd, half of ABX pattern, 7=9.9, 5.4 Hz, 1 H>, 3.69-3.52 (m, 2H), 3.38-3.23 (m, 2H), 2.87 (s, 3H), 2.09 (dd, 7=13.1,9.0 Hz, 1 H), 1.75-1.60 (m, 4H, assumed; partially obscured by water peak), 1.51-1.41 (m, 1H), 1.46 (s, 9H).
Step 3. Synthesis of prop-2-en-1-yl methy![(3R)-1-oxa-8-azaspiro[4.5]dec-3-yl]carbamate, trifluoroacetate sait (C123).
Trifluoroacetic acid (20 mL) was added to a solution of C122 (from the previous step; 16 g, £38.8 mmol) in dichloromethane (100 mL), and the reaction mixture was stirred at 15 ’C for 2 hours. Removal of volatiles In vacuo afforded the product as a brown gum (20 g). This material was used directly In the following step. LCMS m/z 255.2 [M+H]*.
Step 4. Synthesis of (2R)-1,11-tntJuom-3-[(4-methoxybenzyl)oxyJpropan-2-yf (3R)-3{methy/[(prop-2-en- 1-yloxy)carbonyl]amino}- 1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C124).
Triethylamine (19.9 g, 197 mmol) was slowly added to a 0 ’C solution of C123 (from the previous step; 20 g, £38.8 mmol) in acetonitrile (250 mL). The reaction mixture was stirred at 0 ’C for 30 minutes, whereupon C2 [reaction solution In acetonitrile (80 mL) containing 40 mmol], 15 was added, and stirring was continued at 13 ’C for 18 hours. The réaction mixture was concentrated in vacuo, and the residue was purified via silica gel chromatography (Gradient: 9% to 50% ethyl acetate in petroleum ether) to provide the product as a pale yellow gum. Yield: 16.67 g, 31.4 mmol, 81% over 4 steps. LCMS m/z 553.1 [M+Na*]. Ή NMR (400 MHz, CDCh) δ 7.24 (br d, 7=8.8 Hz. 2H), 6.88 (br d, 7=8.8 Hz, 2H), 6.01-5.89 (m, 1H), 5.53-5.43 (m, 1H), 5.3520 5.27 (m, 1 H), 5.26-5.20 (m, 1 H), 5.08-4.86 (br m, 1 H), 4.60 (ddd, 7=5.5,1.5.1.2 Hz, 2H), 4.51 (AB quartet, 7ab=11.5 Hz, Avab=28.3 Hz, 2H), 3.94 (dd, 7=9.8, 7.5 Hz, 1H), 3.81 (s, 3H), 3.80-
3.64 (m, 5H), 3.43-3.25 (m, 2H), 2.88 (s, 3H), 2.13-2.00 (m, 1H), 1.80-1.60 (m, 4H), 1,47 (ddd, 7=13.6,10.8,4.3 Hz, 1H).
Step 5. Synthesis of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl (3R)-325 (methylamino)-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (C125).
Tetrakis(triphenylphosphine)palladium(0) (2.12 g, 1.83 mmol) was added to a 10 ’C solution of C124 (6.50 g, 12.2 mmol) and 1,3-dimethylpyrimidine-2,4,6(1H,3H,5H)-trione (2.87 g,
18.4 mmol) in tetrahydrofuran (100 mL). After the reaction mixture had been stined at 25 ’C for 2 hours, solid sodium carbonate (65 mg, 0.61 mmol) was added, and stirring was continued at 30 10 ’C for 20 minutes. The reaction mixture was filtered, and the filtrate was concentrated In vacuo. The residue was purified twice by silica gel chromatography (Gradient: 0% to 10% methanol in dichloromethane) to afford the product as a yellow gum. Yield: 3.8 g, 8.5 mmol, 70%. LCMS m/z 447.3 [M+H]*. Ή NMR (400 MHz, CDCh) δ 7.24 (br d, 7=8.7 Hz, 2H), 6.88 (br d, 7=8.7 Hz, 2H), 5.53-5.42 (m, 1H), 4.51 (AB quartet, 7ab=11.6 Hz, ûvAb=28.0 Hz, 2H), 3.96 35 (dd, 7=9.2, 6.0 Hz, 1H), 3.81 (s, 3H), 3.8-3.64 (m, 5H), 3.43-3.28 (m, 3H), 2.43 (s, 3H), 2.081,97 (m, 1H), 1.85-1.46 (m, 5H, assumed; partially obscured by water peak).
184
Sfep 6. Synthesis of (2R)-1,1,1-tn8uoro-3-[(4-methoxybenzyl)oxy]pmpan-2-yl (3R)-3{[(cyclopropylmethyl)sulfonyl](methyl)amino)-1-oxa-B-azaspiro[4.5]decane-B-carboxylate (C126).
To a 15 *C solution of C125 (400 mg, 0.896 mmol) In dichloromethane (5 mL) were added cyclopropylmethanesulfonyl chloride (208 mg, 1.35 mmol) and triethylamine (453 mg,
4.48 mmol). The reaction mixture was stirred at 15 ’C for 16 hours, whereupon it was concentrated In vacuo and purified via chromatography on silica gel (Gradient: 0% to 50% ethyl acetate In petroleum ether). The product was obtained as a colorless gum. Yield: 430 mg, 0.762 mmol, 85%. LCMS m/z 587.1 [M+Na*].
Sfep 7. Synthesis of (2R)-1,1,1-trifIuoro-3-hydroxypropan-2-yl (3R)-3{[(cyclopropylmethyl)sulfonyl](methyl)amino}-1-oxa-8-azaspiro[4.5]decane-8-carboxylate (106).
Trifluoroacetic acid (3 mL) was added to a 0 ’C solution of C126 (430 mg, 0.762 mmol) in dichloromethane (12 mL). The reaction mixture was stirred at 15 ’C for 2 hours, whereupon the pH was adjusted to 6 - 7 via addition of sodium bicarbonate. The resulting mixture was 15 extracted with dichloromethane (15 mL) and with ethyl acetate (2x15 mL). The combined organic layers were dried over sodium sulfate, filtered, and concentrated in vacuo. Silica gel chromatography (Gradient: 0% to 50% ethyl acetate In petroleum ether) was followed by reversed phase HPLC (Column: Agela Durashell C18, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 30% to 50% B), affording the product as a colorless gum. Yield: 211 mg, 0.475 mmol, 62%. LCMS m/z 445,2 [M+H]*. 1H NMR (400 MHz, CDCh) δ 5.30-5.19 (m, 1H), 4.72-4.62 (m, 1H), 4.01-3.90 (m, 2H), 3.89-3.69 (m, 4H), 3.44-3.23 (m, 2H), 2.88-2.83 (m, 2H), 2.86 (s, 3H), 2.82-2.64 (br m, 1 H), 2.13-2.01 (m, 1 H), 1.81-1.65 (m, 4H), 1.55-1.39 (m, 1H), 1.13-1.01 (m, 1H), 0.76-0.62 (m, 2H), 0.42-0.29 (m, 2H).
Table 6A. Method of synthesis, structure, and physicochemical properties for Examples 107-150.
Example Number Method of Synthe sis; Noncomme rcial sterling materia Is Structure 1H NMR (400 MHz, CDCh) δ; Mass spectrum, observed ion m/z [M+H]* or HPLC rétention time; Mass spectrum m/z [M+Hj* (unless otherwise indicated)
185
107 Exampl e 92M; C48, C2 0 cf3 rNAoA,OH °'b 7.83 (d, J=7.5 Hz, 2H), 7.57 (br dd, half of ABX pattern, J=7.5,7.0 Hz, 1H), 7.51 (br dd, half of ABX pattern, J=7.5, 7.0 Hz, 2H), 5.31-5.19 (m, 1H), 4.16-3.95 (m, 3H), 3.923.68 (m, 5H), 3.50-3.30 (m, 2H), 2.52-2.35 (m, 1H), 2.29-2.15 (m, 1H), 1.99-1.82 (m, 2H), 1.79-1.47 (m, 3H, assumed; partially obscured by water peak), 1.26-1.18 (m, 6H); 495.1
108 Exampl e 11 ».37. C73 0 cf3 <-νΛ0Λ-οη eP Γ DIAST-1 -NO °”b-F 1H NMR (400 MHz, DMSO-de) δ 7.72-7.63 (m, 3H), 7.61-7.55 (m, 1H), 5.24-5.14 (m, 1H), 4.68-4.59 (m, 1H), 3.77-3.59 (m. 4H), 3.57-3.44 (m, 3H), 3.3-3.12 (m, 2H), 2.70 (s, 3H), 1.96-1.85 (m, 1H), 1.65-1.49 (m, 3H), 1.49-1.35 (m, 2H); 485.3
109 Exampl e 11 ».37. C73 0 cf3 0£nWh DIAST-2 -%O °b-F 1H NMR (400 MHz, DMSO-de) δ 7.73-7.62 (m, 3H), 7.62-7.54 (m, 1H), 5.23-5.14 (m, 1H), 4.69-4.58 (m, 1H), 3.78-3.58 (m, 4H), 3.58-3.44 (m, 3H), 3.3-3.11 (m, 2H), 2.70 (s, 3H), 1.91 (dd, J=13.4, 9.1 Hz, 1H), 1.67-1.49 (m, 3H). 1.48-1.31 (m, 2H); 485.3
110 Exampl e109M; C73 o cf3 οΡ'νΛο'<-οη -N> DIAST-1 A 1H NMR (400 MHz, DMSO-de) δ 7.91-7.84 (m, 2H), 7.46 (br dd, J=8.8, 8.8 Hz, 2H), 5.24-5.14 (m, 1H), 4.64-4.55 (m, 1H), 3.783.68 (m, 3H), 3.68-3.58 (m, 1H), 3.57-3.43 (m, 3H), 3.3-3.12 (m, 2H), 2.67 (s, 3H), 1.89 (br dd, J=13, 9.5 Hz, 1 H), 1.65-1.49 (m, 3H), 1.49-1.34 (m, 2H); 485.3
111 Exampl e 109M; C73 0 cf3 θ AnPA™ -No DIAST·2 A 1H NMR (400 MHz, DMSO-de) δ 7.91-7.85 (m, 2H), 7.46 (br dd, J=8.8, 8.8 Hz, 2H), 5.24-5.14 (m, 1H), 4.64-4.55 (m, 1H), 3.783.68 (m, 3H), 3.68-3.59 (m, 1H), 3.57-3.44 (m, 3H), 3.3-3.13 (m, 2H), 2.67 (s, 3H), 1.89 (brdd, J=13, 9.5 Hz, 1H), 1.65-1.48 (m, 3H), 1.48-1.34 (m, 2H); 485.3
188
112 Exampl es 93 and 9439. C73 0 cf3 -n_o 0 Mixture of 2 diastereomers; characteristic peaks: 5.30-5.20 (m. 1H), 4.76-4.66 (m, 1H), 4.05-3.92 (m, 2H), 3.92-3.70 (m, 4H), 3.48-3.23 (m, 3H), 2.87 (s, 3H), 2.35-2.22 (m, 1H), 2.13-2.02 (m, 1H), 2.02-1.91 (m, 3H), 1.88-1.59 (m, 7H); 459.3
113 Exampl es 93 and 9439; C73 0 cf3 <·νλολ-οη -N ..° Mixture of 2 diastereomers; 5.32-5.19 (m, 1H), 4.72-4.61 (m, 1H), 4.05-3.96 (m, 1H), 3.96-3.68 (m, 6H), 3.44-3.19 (m, 2H). 2.83 (s, 3H), 2.60-2.46 (m, 2H), 2.46-2.19 (m, 3H), 2.10-1.96 (m, 3H), 1.81-1.67 (m, 4H), 1.55-1.43 (m, 1 H); 445.2
114 Exampl e 1140·39; C73 0 cf3 -N ° Mixture of 2 diastereomers: 9.02 (d, */=2.0 Hz, 1H), 8.85 (dd, J=4.8,1.5 Hz, 1H), 8.09 (ddd, */=8.0, 2.1,1.8 Hz, 1H), 7.51 (dd, */=8.0, 5.0 Hz, 1H), 5.29-5.18 (m, 1H), 4.80-4.69 (m, 1H), 4.04-3.94 (m, 1H), 3.913.69 (m, 4H), 3.65 (dd, */=10.4.5 Hz, 1H), 3.38-3.13 (m, 2H), 2.81 (s, 3H), 2.37-2.22 (m, 1H), 1.98-1.86 (m, 1H), 1.74-1.6 (m, 3H), 1.54-1.39 (m, 2H); 468.0
115 Exampl e 114*1; C73 0 cf3 ^N^OxP'zOH -N.° w Mixture of 2 diastereomers; 9.42 (s, 1H), 9.10 (s, 2H), 5.29-5.19 (m, 1H), 4.81-4.71 (m, 1H), 4.04-3.95 (m, 1H), 3.95-3.74 (m, 4H), 3.70 (dd, */=10.4, 4.6 Hz, 1H). 3.383.14 (m, 2H), 2.85 (s, 3H), 2.45-2.22 (br m, 1H), 2.05-1.91 (m, 1H), 1.76-1.6 (m, 3H, assumed; partially obscured by water peak), 1.52 (dd, */=13.6, 6.5 Hz, 1H), 1.5- 1.40 (m, 1H); 469.0
116 Exampl e 92«.«; C99, C2 0 cf3 ζ-νΑΑλη Λ [from DIAST-2; ~~N 0 see footnote 43] 0 5.31-5.20 (m, 1H), 4.70-4.61 (m, 1H), 4.144.05 (m, 2H), 4.05-3.92 (m, 2H), 3.92-3.70 (m, 4H), 3.43-3.20 (m, 4H), 3.16-3.05 (m, 1H), 2.89 (s, 3H), 2.44-2.23 (m, 1H), 2.10 (dd, */=13.3, 9.0 Hz, 1H), 1.98-1.67 (m, 8H), 1.6-1.46 (m, 1H); 475.1
187
117 Exampl e 9242·43; C99, C2 O cf3 f-AcA-0 Ap Γ [from DIAST-1; N o see footnote 43] A 5.32-5.20 (m, 1H), 4.70-4.60 (m, 1H), 4.144.06 (m, 2H), 4.05-3.92 (m, 2H), 3.92-3.73 (m, 4H), 3.46-3.26 (m, 4H), 3.16-3.06 (m, 1H), 2.89 (s, 3H), 2.41-2.24 (m, 1 H), 2.152.04 (m, 1H), 1.98-1.68 (m, 8H), 1.6-1.42 (m, 1H); 475.1
118 Exampl e 10244; C99, C1 0 cf3 rNA0+-0H r ocf3 Mixture of 2 diastereomers; 3.35 minutes13; 458.2
119 Exampl e 10249; C1 0 cf3 Cl Mixture of 2 diastereomers; 3.25 minutes13; 408.2
120 Exampl e97; C107 o cf3 ο<'ΝΛΟΛ-ΟΗ 0 F Mixture of 2 diastereomers; 2.45 minutes13; 393.3
121 Exampl e97; C107 o cf3 S'* N 'py'P'ON /A j ÇN -0 Mixture of 2 diastereomers; 2.18 minutes13; 405 3
188
122 Exampl β97; C107 o cf3 opWH · CFjCOOH Q Mixture of 2 diastereomers; 1.40 minutes13; 375.1
123 Exampl e97; C107 o çf3 OJ Ί · CF3COOH P Mixture of 2 diastereomers; 2.36 minutes13; 389.3
124 Exampl e98; C2 0 cf3 Α'νλολ-οη N-* 5.33-5.20 (m, 1H), 4.02-3.91 (m, 1H), 3.893.77 (m, 1H), 3.76-3.07 (m, 9H), 2.81-2.66 (m, 1 H), 1.90-1.66 (m, 8H), 1.64-1.46 (m, 6H); 392.9
125 Exampl e 105*®; C2 0 cf3 <'νΛοΛ-οη N-* °\s 2.35 minutes13; 408.2
126 Exampl e 105**; C2 0 cf3 N** °b 2.38 minutes13; 401.3
127 Exampl e98; C2 0 cf3 ~nAoA_oh N' Y Charactcristic peaks: 5.33-5.20 (m, 1H), 4.05-3.96 (m, 1H), 3.92-3.82 (m, 1H), 3.783.24 (m, 8H), 2.47-2.33 (m, 1 H), 2.29-2.15 (m, 2H). 1.99-1.65 (m, 9H); 443.0
189
128 Exampl elOS49; C2 0 cf3 rvWH N-' 1.97 minutes13; 402.3
129 Exampl e 9247·49; C2 0 CF3 <·νλοΆοη Ap J DIAST-1 O Ή NMR (400 MHz, CDjOD) δ 8.11 (ddd, >5.0, 2.0, 0.8 Hz, 1H), 7.67 (ddd, >8.4, 7.1, 2.0 Hz, 1H), 6.94 (ddd, >7.2, 5.1, 0.9 Hz, 1H), 6.79 (ddd, >8.3, 0.9, 0.8 Hz, 1H), 5.55-5.50 (m, 1 H), 5.33-5.24 (m, 1H), 4.18 (dd, >10.5,4.6 Hz, 1H), 4.01-3.96 (m, 1H), 3.90-3.84 (m, 1H), 3.84-3.69 (m, 3H), 3.50-3.3 (m, 2H), 2.20 (dd, half of ABX pattern, >13.9, 6.8 Hz, 1H), 2.07 (ddd, half of ABXY pattern, >14.0, 2.0,1.1 Hz, 1H), 1.98-1.88 (m, 1H), 1.78-1.62 (m, 3H); 390.9
130 Exampl e 9247·48; C2 o cf3 0 pVv“ J DIAST-2 0 1H NMR (400 MHz, CD3OD) δ 8.11 (ddd, >5.0, 2.0, 0.8 Hz, 1H), 7.67 (ddd, >8.5, 7.0, 2.0 Hz, 1H), 6.94 (ddd, >7.0, 5.0,1.0 Hz, 1H), 6.78 (br d, >8.5 Hz, 1H), 5.55- 5.50 (m, 1H), 5.33-5.24 (m, 1H), 4.18 (dd, >10.4, 4.6 Hz, 1H), 3.99 (ddd, >10.4, 2.0,1.1 Hz, 1 H), 3.87 (br dd, half of ABX pattern, >12.3,4.0 Hz, 1H), 3.84-3.70 (m, 3H), 3.50-3.33 (m, 2H), 2.20 (dd, half of ABX pattern, >13.9, 6.7 Hz, 1H), 2.07 (br d, >14.0 Hz, 1H), 1.98-1.85 (m, 1H), 1.811.58 (m, 3H); 390.9
131 Exampl e 101; C105 o cf3 Mixture of 2 diastereomers; 7.85 (br d, >7.8 Hz, 1H), 7.52-7.46 (m, 1H), 7.44- 7.38 (ιη, 1H), 7.29-7.23 (m, 1H, assumed, partially obscured by solvent peak), 5.305.19 (m, 1H), 4.59-4.45 (m, 1H), 4.11-3.95 (m, 2H), 3.92-3.68 (m, 6H), 3.46-3.20 (m. 2H), 3.12-3.03 (m, 2H), 2.41-2.24 (m, 1H), 2.21-2.11 (m, 1H), 1.88-1.79 (m, 1H), 1.79-
190
1.64 (m, 3H), 1.6-1.43 (m, 1 H, assumed; partially obscured by water peak); 479.2
132 Exampl e102«; C1 0 cf3 o^naoa^>h w Mixture of 2 diastereomers; 2.92 minutes13; 404.2
133 çjSO.SI 0 CF3 N-J O=< 0 5.31-5.21 (m, 1H), 3.96 (dd, half of ABX pattern, >12.4,3.4 Hz, 1H), 3.83 (dd, half of ABX pattern, >12.3, 6.8 Hz, 1H), 3.663.29 (m, 10H), 3.29-3.22 (br s, 2H), 1.871.77 (m, 4H), 1.74 (dd, >7.3, 7.0 Hz, 2H), 1.62-1.47 (m. 4H); 394.1
134 Exampl e6M 0 cf3 o U'C ySO N=< M 8.00 (d, >3.0 Hz, 1 H), 7.66 (d, >3.0 Hz, 1 H), 5.29-5.19 (m, 1 H), 4.03-3.94 (m, 1 H), 3.90-3.80 (m, 1H), 3.67-3.47 (m, 4H), 3.45- 3.18(m, 4H), 2.58-2.43 (brs, 1H), 1.79 (dd, >7.0, 7.0 Hz, 2H), 1.51-1.37 (m, 4H); 443.8
135 Exampl e6M o cf3 r-'N-V-™ °< (5 \=N 9.09-9.05 (br s, 1H), 8.85 (d, >4.5 Hz, 1H), 8.13 (br d, >8.0 Hz, 1H). 7.52 (dd, >7.9, 4.9 Hz, 1H), 5.29-5.20 (m, 1 H), 3.99 (br dd, half of ABX pattern, >12.5, 3 Hz, 1H), 3.85 (brdd, half of ABX pattern, >12, 7 Hz, 1H), 3.59-3.25 (m, 6H), 3.21-3.11 (m, 2H), 2.47-2.30 (br s, 1H), 1.74 (dd, >7.0, 7.0 Hz, 2H), 1.47-1.39 (m, 4H); 437.9
136 Exampl e 106; C125 o cf3 <·νΑ<Α-fP -NQo °5-s 7.98 (d, >3.0 Hz, 1 H), 7.65 (d, >3.0 Hz, 1H), 5.30-5.19 (m, 1H), 4.94-4.84 (m, 1H), 4.04-3.95 (m, 1H), 3.92 (dd, half of ABX pattern, >10.4, 7.4 Hz, 1 H), 3.91-3.71 (m, 4H), 3.42-3.21 (m, 2H), 2.95 (s, 3H), 2.ΙΟ- Ι.97 (m, 1H), 1.7-1.37 (m, 5H, assumed; partially obscured by water peak); 474.0
191
137 Exampl e 106; C125 o cf3 ^2jNAo^oh -%O o cf3 5.32-5.20 (m, 1H), 4.75-4.65 (m, 1H), 4.05- 3.93 (m, 2H), 3.93-3.7 (m, 4H), 3.74 <q, Jhf=9.3 Hz, 2H), 3.45-3.23 (m, 2H), 2.93 (s, 3H), 2.19-2.07 (m, 1 H), 1.84-1.67 (m, 4H), 1.57-1.41 (m, 1H); 473.2
138 Exampl e 106; C125 O CF3 -n'o I 7.76 (s, 1H), 7.70 (s, 1H), 5.30-5.18 (m, 1H), 4.66-4.54 (m, 1H), 4.03-3.93 (m, 1H), 3.96 (s, 3H), 3.88-3.71 (m, 3H), 3.87 (dd, half of ABX pattern, >10.2,7.4 Hz, 1H), 3.67 (br dd, half of ABX pattern, >10.2, 5.1 Hz, 1H), 3.40-3.19 (m, 2H), 2.74 (s, 3H), 2.69-2.52 (m, 1H), 2.01-1.88 (m, 1H), 1.82-1.63 (m, 3H, assumed; partially obscured by water peak), 1.58 (dd, >13.3, 7.0 Hz, 1H), 1.51-1.36 (m, 1 H); 471.2
139 Exampl e 106; C125 0 CF3 —N ,o 0 P 5.31-5.19 (m, 1H), 4.70-4.59 (m, 1H), 4.033.90 (m, 2H), 3.90-3.70 (m. 4H), 3.45-3.23 (m, 2H), 2.82 (s, 3H), 2.75 (br d, >6.5 Hz, 2H), 2.73-2.64 (m, 1H), 2.30-2.16 (m, 1H), 2.12-2.00 (m, 1H), 1.80-1.66 (m, 4H), 1.55- 1.40 (m, 1 H), 1.09 (br d, >6.8 Hz, 6H); 447.3
140 Exampl e 106; C125 0 cf3 £p 5.31-5.19 (m, 1H), 4.67-4,56 (m, 1H), 4.033.90 (m, 2H), 3.90-3.70 (m, 4H), 3.45-3.24 (m, 2H), 2,99 (br d, >7.5 Hz, 2H), 2.842.70 (m, 1H), 2.81 (s, 3H), 2.69-2.58 (m, 1H), 2.25-2.14 (m, 2H), 2.12-2,01 (m, 1H), 2.01-1.91 (m, 1H), 1.91-1.79 (m,3H), 1.791.66 (m, 4H), 1.55-1.40 (m, 1H); 459.2
141 C125“ 0 cf3 2.82 minutes”; 449
192
142 C12552 0 cf3 rNAoA.OH Æ 2.47 minutes”; 409
143 C125” o cf3 ['''νΑιΆ-ΟΚ -N 2.86 minutes2®; 423
144 C12552 o CF3 -N -P 2.80 minutes2®; 411
145 C12552 o cf3 ^-^ΑθΑ,ΟΗ -N <n> 2.44 minutes53; 409
146 C12552 0 cf3 ’ ·HCOOH -N N==\ OHJ 2.26 minutes53; 432
147 Exampl e 97; C107 o ÇFj r-N AoA-OH f CN Mixture of 2 diastereomers; 2.77 minutes13; 399.3
193
148 Exampl e 33«·”; C79 eP ο -° 0 -£o ---NH·· ah,· ’H NMR (400 MHz, D2O), characteristic peaks: δ 7.84 (br d, >7.6 Hz, 2H), 7,76- 7.69 (m. 1H), 7.63 (br dd, half of ABX pattern, >7.8,7.6 Hz, 2H), 5.44-5.34 (m, 1H), 4.10-4.01 (m, 1H), 4.00-3.90 (m, 1H), 3.90-3.81 (m, 1H), 3.70 (t, >6.0 Hz, 2H), 3.65-3.22 (m, 5H), 2.98 (dd, >7.6, 7.4 Hz, 4H), 2.76 (s, 3H), 1.95 (dd, >13.7, 9.2 Hz, 1H), 1.91-1.81 (m, 4H), 1.74-1.62 (m, 7H), 1.54-1.33 (m, 6H); 547.3
149 Exampl e 3356.57. Examp le 91 0 Çp3 A 0 έ O *2 0 *' Ή NMR (400 MHz, D2O) δ 7.83-7.78 (m, 2H), 7.77-7.72 (m, 1 H), 7.65 (br dd, >7.8, 7.4 Hz, 2H), 5.47-5.37 (m, 1H), 4.13-4.05 (m, 1H). 4.02-3.93 (m, 1H), 3.93-3.79 (m, 1H), 3.83 (brdd, >5.1, 4.9 Hz, 2H), 3.783.63 (m, 1H), 3.71 (t, >6.1 Hz, 2H), 3.363.13 (m, 2H), 3.11-3.0 (m, 2H), 2.99 (dd, >7.6, 7.4 Hz, 4H), 2.95-2.86 (m, 2H), 2.00-1.82 (m, 6H), 1.74-1.64 (m, 6H), 1.531.34 (m, 4H); 533.1
150 Exampl e 3358,59. C125 eP o -No - 2 o-s^ âh3* H Ή NMR (400 MHz, D2O), characteristic peaks: δ 7.92-7.85 (m, 2H). 7.35 (br dd, >8.9, 8.8 Hz, 2H), 5.44-5.35 (m, 1H), 4.78-4.66 (m, 1H, assumed; partially obscured by solvent peak), 4.10-4.02 (m, 1H), 3.99-3.91 (m, 1H), 3.87 (dd, >10.0, 7.9 Hz, 1H), 3.68 (t, >6.1 Hz, 2H), 3.673.60 (m, 2H), 2.98 (dd, >7.7, 7.5 Hz, 4H), 2.77 (s, 3H), 1.97 (dd, >13.5, 9.3 Hz, 1 H), 1.91-1.79 (m, 5H), 1.73-1.63 (m, 5H), 1.551.33 (m, 7H); 565.3
35. Intermediate tert-butyl (3R)-3-[(phenylsulfonyl)(propan-2-yl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate was synthesized via a Mitsunobu reaction between C48 and 2-propanol.
36. Prior to the final deprotection, intermediate (2R)-1,1,1-trifluoro-3-[(4- methoxybenzyl)oxy]propan-2-yl 3-{[(3-fluorophenyl)sulfonyl]amîno}-1-oxa-8azaspiro[4.5]decane-8-carboxylate was deprotonated with potassium tert-butoxide and
194
methylated with dimethyl sulfate to afford (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2y 13-{[ (3-fl uoropheny I ) suifo ny I] (methyl )a min o}-1 -oxa-8-azaspiro[4,5]decane-8-ca rboxylate.
37. Examples 108 and 109 were synthesized as a mixture and separated into the component diastereomers using supercritical fluid chromatography (Column: Phenomenex Lux Amylose-2,
5 pm; Mobile phase: 87.5:12.5 carbon dioxide / 2-propanol). Example 108 was the first-eluting diastereomer, and Example 109 was the second-eluting diastereomer.
38. Examples 110 and 111 were synthesized as a mixture and separated into the component diastereomers using supercritical fluid chromatography (Column: Phenomenex Lux Amylose-2, 5 pm; Mobile phase: 85:15 carbon dioxide / 2-propanol). Example 110 was the first-eluting diastereomer, and Example 111 was the second-eluting diastereomer.
39. In this case, the 2 diastereomers of the product were not separated.
40. Prior to the final deprotection, intermediate (2R)-1,1,1-trifluoro-3-[(4methoxybenzyl)oxy]propan-2-yl 3-[(pyridin-3-ylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]decane-8carboxylate was deprotonated with sodium bis(trimethylsilyl)amîde and methylated with lodomethane, affording (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 3[methyl(pyridin-3-ylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-ca rboxylate.
41. In this case, the sulfonylation of C73 was effected using pyridine in tetrahydrofuran, rather than aqueous sodium bicarbonate in dichloromethane.
42. The requisite tert-butyl 3-[methyl(tetrahydro-2H-pyran-4-ylsulfonyl)amino]-1-oxa-&- azaspiro[4.5]decane-8-carboxylate was synthesized via potassium carbonate-mediated reaction of C99 with tetrahydro-2H-pyran-4-sulfonamide. The resulting tert-butyl 3-[(tetrahydro-2H-pyran4-ylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]decane-8-carboxylate was then methylated using sodium hydride and iodomethane.
43. Prior to the final deprotection, intermediate (2R)-1,1,1-trifluoro-3-[(4- methoxybenzyl)oxy]propan-2-yl 3-[methyl(tetrahydro-2H-pyran-4-ylsulfonyl)amino]-1 -oxa-8azaspiro[4.5]decane-8-carboxylate was separated into Its component diastereomers via supercritical fluid chromatography (Column: Chiral Technologies Chiralcel OD, 5 pm; Mobile phase A: carbon dioxide; Mobile phase B: 0.1% ammonium hydroxlde in 2-propanol; Gradient: 30% to 35% B). The first-eluting diastereomer was assigned as DIAST-1, and the second30 eluting diastereomer as DIAST-2.
44. Reaction of C99 with [3-(trifluoromethoxy)phenyl]boronlc acid was carried out using the method described for synthesis of the mixture of C100 and C101 from C99 in Example 95. The product was deprotected with hydrogen chloride in 1,4-dioxane and dichloromethane to afford the requisite 3-[3-(trifluoromethoxy)phenyl]-1-oxa-8-azaspiro[4.5]decane, hydrochloride sait.
45. Reaction of tert-butyl 3-oxo-1-oxa-8-azaspîro[4.5]decane-8-carboxylate with 3chlorophenylmagnesium bromide provided tert-butyl 3-(3-chlorophenyl)-3-hydroxy-1-oxa-8azaspiro[4.5]decane-8-carboxylate. This was subjected to triethylsilane, boron trifluoride diethyl
195
etherate and trifluoroacetic acid, providing partial deoxygenation, followed by hydrogénation In methanol and acetic acid, to afford 3-(3-chlorophenyl)-1-oxa-8-azaspiro[4.5]decane.
46. In this case, the first step was an amide formation, rather than a sulfonamide formation, tertButyl 2,8-diazaspiro[4.5]decane-8-carboxylate was reacted with the appropriate carboxylic acid using 2-[2-oxo-1 (2H)-pyridy!]-1,1,3,3-tetramethyluronium tetrafluoroborate (TPTU) and N,Ndiisopropylethylamine in /V,N-dimethylformamide.
47. tert-Butyl 3-hydroxy-1-oxa-8-azaspiro[4.5]decane-8-carboxylate was deprotonated with potassium tert-butoxide and reacted with 2-chloropyridine to afford the requisite tert-butyl 3(pyrldin-2-yloxy)-1-oxa-8-azaspiro[4.5]decane-8-carboxylate.
48. The mixture of Examples 129 and 130 was separated into its component diastereomers using reversed phase HPLC (Column: Phenomenex Luna C18, 5 pm; Mobile phase A: water containing 0.225% formic acid; Mobile phase B: acetonitrile; Gradient: 25% to 45% B). The firsteluting diastereomer was Example 129, and the second-eluting diastereomer was Example 130.
49. Reaction of tert-butyl 3-hydroxy-1-oxa-8-azaspiro[4.5]decane-8-carboxylate with sodium hydride and benzyl bromide afforded tert-butyl 3-(benzyloxy)-1-oxa-8-azaspiro[4.5]decane-8carboxylate, which was deprotected with hydrochloric acid to provide the requisite 3(benzyloxy)-1-oxa-8-azaspiro[4.5]decane.
50. tert-Butyl 2,8-dîazaspiro[4.5]decane-8-carboxylate was converted to (2R)-1,1,1-trifluoro-3[(4-methoxybenzyl)oxy]propan-2-yl 2,8-diazaspiro[4.5]decane-8-carboxylate using the method described for synthesis of C73 from tert-butyl 3-amino-1-oxa-8-azaspiro[4.5]decane-8carboxylate in Example 27. In this case, the palladium catalyst employed for the final step was tetrakis(triphenylphosphine)pailadium(0).
51. Reaction of (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl 2,8diazaspiro[4.5]decane-8-carboxylate (see footnote 50) with 4-nitrophenyl pyrrolidine-1- carboxylate (see E. Bridgeman and N. C. O. Tomkinson, Synlett 2006, 243-246) in the presence of N./V-diisopropylethylamine provided (2R)-1,1,1-trifluoro-3-[(4-methoxybenzyl)oxy]propan-2-yl
2-(pyrrolidin-1-ylcarbonyl)-2,8-diazaspiro[4.5]decane-8-carboxylate, which was deprotected with trifluoroacetic acid to afford Example 133.
52. Compound C125 was reacted with the appropriate carboxylic acid using 2,4,6-tripropyl-
1,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide and Ν,Ν-diisopropylethylamine in 1,4-dioxane. The resulting product was deprotected with trifluoroacetic acid to afford the Example.
53. Conditions for analytical HPLC. Column: Waters XBridge C18, 2.1 x 50 mm, 5 pm; Mobile phase A: 0.05% ammonium hydroxide in water; Mobile phase B: acetonitrile; Gradient: 5% B for 0.5 minutes; 5% to 100% B over 2.9 minutes; 100% B for 0.8 minutes; Flow rate: 0.8 mL/minute.
54. In this case, C79 was syntheslzed from C50, via deprotection with p-toluenesulfonic acid and conversion of the resulting amine to C79 using the method described for synthesis of C84 from C85 in Altemate Synthesis of Example 32.
196
55. In this case, the final product did not precipitate out of the reaction mixture. The reaction mixture was therefore concentrated in vacuo; the residue was dissolved In hot methanol, filtered, concentrated under reduced pressure, and crystallized from methanol / tert-butyl methyl ether to afford Example 148.
56. An aqueous solution of Example 91 was acidified with concentrated hydrochloric acid at 0 *C. After 30 minutes at room température, the reaction mixture was extracted three times with ethyl acetate, and the combined organic layers were washed with saturated aqueous sodium chloride solution, dried over sodium sulfate, filtered, and concentrated in vacuo to afford the requisite (neutre I) phosphate of Example 91.
57. In this case, the final product did not precipitate out of the reaction mixture. The reaction mixture was therefore concentrated in vacuo; the residue was dissolved In hot methanol, cooled to 0 *C and treated with tert-butyl methyl ether. Filtration afforded Example 149.
58. Using the method described in Example 11, C125 was converted to (2/7)-1,1,1-trifluoro-3hydroxypropan-2-yl (3/?)-3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8- azaspiro[4.5]decane-8-carboxylate. Phosphate formation, using the chemistry employed for conversion of 15 to C79 In Example 30, then provided the requisite (2/7)-1,1,1-trifluoro-3(phosphonooxy)propan-2-yl (3/7)-3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8azaspiro[4.5]decane-8-carboxylate.
59. In this case, after the reaction mixture had been heated at 40 *0, it was slowly treated with 20 tert-butyl methyl ether (3 mL). After the suspension had cooled to room température and then to ’C, itwas filtered to afford Example 150.
Example AA: MAGL and FAAH Enzymatic assays
Assessment of MAGL inhibition utilizes human recombinant Monoacylglycerol Lipase and the fluorogenic substrate 7-hydroxycoumarinyl arachidonate (7-HCA, Biomol ST-502). 400 nL of a test compound at decreasing concentration (ranging from 150 pM down to 1.5 nM) was spotted Into a 384-well back plate (PerkinElmer, 6007279) using a Labcyte Echo, followed by addition of 10 pL of MAGL enzyme in assay buffer (50mM HEPES, pH 7.4,100 mM NaCI, 5 mM MgCI2, 0.1% Triton X-100 and 25% glycerin). An equal volume of 7-HCA in assay buffer with 10% DMSO was added either immediately (T = 0 min) or after a 30 minute Incubation (T = 30 min) to initiate the reaction. The final concentration of MAGL enzyme was 88 pM and 7HCA substrate was 5 pM. After these dilutions, the final concentration of the test compound ranged from 3 pM to 0.03 nM. The reaction was allowed to progress for 60 minutes, after which the plate was read at an Ex/Em of 340/465. Percent Inhibitions were calculated based on control welis containing no compound (0% inhibition) and a control compound (e.g., a MAGL Inhibitor whose activity Is known or was previously reported in the literature, such as one with about 100% Inhibition). IC» values were generated based on a four parameterfit model using ABASE software from IDBS. See e.g., Wang, Y. et al., ‘A Fluorescence-Based Assay for
197
Monoacylglycerol Llpase Compatible with Inhibitor Screening, Assay and Drug Development Technologies, 2008, Vol. 6 (3) pp 387-393 (reporting an assay for measuring MAGL activity).
To measure MAGL Inactivation, the same protocol for the (T = 0 min) MAGL inhibition IC50 assay was performed with data collected every minute to acquire enzyme progress curves 5 at decreasing concentrations of compound. Kob* values were calculated from this data and kin«t/Ki ratios were determined from a plot of Kob« values vs. compound concentrations.
Assessment of FAAH inhibition utilizes human recombinant FAAH and the fluorescent substrate, Arachldonoyl-AMC. 400 nL of a test compound at decreasing concentrations was spotted into a 384-well back plate (PerkinElmer, 6007279) using a Labcyte Echo, followed by 10 addition of 10 pl of FAAH enzyme (Cayman 10010183) in assay buffer (50 mM Tris, pH 9.0,1 mM EDTA). After a 30 minute Incubation at room température, 10 pL of Arachidonyl-AMCA was added in assay buffer with 16% DMSO. Final concentration of FAAH enzyme was 0.0125 Units and AAMCA substrate was used at the Km of 5 pM. After these dilutions, the final concentration of the test compound ranged from 3 pM to 0.03 nM. The reaction was allowed to 15 progress for 60 minutes, after which the plate was read on a Molecular Devices FlexStation reader at an Ex/Em of 355/460. Percent inhibitions were calculated based on controls wells containing either no compound (0% Inhibition) or a control compound (e.g., an FAAH inhibitor whose activity is known or was previously reported in the lîterature, such as one with about 100% inhibition). ICso values were generated based on a four parameter fit mode! using
ABASE software from IDBS.
Table AA-1. Biological Data (MAGL ICso, FAAH ICw, and MAGL kinea/Ki) for Examples 1-150.
Exa mple Num ber Compound Name MAGL (T = 0 min) ICso (pM)· MAGL (T = 30 min) ICso (pM)· FAAH (T = 30 min) ICso (pM)· MAGL kin«ct/Kl (1/s per M)·
1 (2 F?)-1,1,1 -trifluoro-3-hydroxypropan-2yl (1a,5a,6a)-6-i1-(5-methoxypyridin-2yl)-1 H-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate 0.085 0.014 N.D.b 7806
2 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[1-(4-fluorophenyl)-1 H-pyrazol-3yl] piperid i n e-1 -carboxyla te 0.056 0.008 1.14 6109
3 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl (1 a,5a,6a)-6-i1-(4-fluorophenyl)-1 Hpyrazol-3-yl]-3azabicyclo[3.1,0]hexane-3-carboxylate 0.035 0.003 2.48 20005
198
4 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-(tetrahydro-2H-pyran-3-ylmethyl)-1oxa-4,9-diazaspiro[5.5]undecane-9carboxylate [from C25, DIAST-1] 0.166d 0.019d N.D. 5489
5 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-(tetrahydro-2H-pyran-3-ylmethyl)-1oxa-4,9-diazaspiro[5.5]undecane-9carboxylate [from C26, DIAST-2] 1.70d 0.161d N.D. N.D.
6 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[(4-fluorophenyl)sulfonyl]-1 -oxa4,9-diazaspiro[5.5]undecane-9carboxylate 0.083e 0.007e >30.0d 13406
7 (2/7)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(phenylsulfonyl)-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate 0.029e 0.003e >24.1 29124
8 (2/7)-1,1,1 -trifluoro-3-hydroxypropan-2yl (3S)-3-[(phenylsulfonyl)amino]-1-oxa8-azaspiro[4.5]decane-8-carboxylate 0.057 0.005 >30.0d 6754
9 (2/7)-1,1,1 -trifluoro-3-hydroxypropan-2yl (3/7)-3-[(phenylsulfonyl)amino]-1 -oxa8-azaspiro[4.5]decane-8-carboxylate 0.040 0.004 >30.0d 8588
10 (2/7)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-[(5-cyclopropylpyridin-2yl)oxy]piperidine-1-carboxylate 0.077e 0.007e >30.0d 5205
11 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[(3-fl uoropheny l)su tfonyl]-1 -oxa4,9-dlazaspiro[5.5]undecane-9carboxylate 0.014 0.001 >30.0d 147964
12 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 2-[(4-fluorophenyl)sulfonyl]-2,9diazaspiro[5.5]undecane-9-carboxylate 0.188 0.017 >30^ 1801
13 (2/7)-1,1,1-trifIuoro-3-hydroxypropan-2yl (38/7,635)-54(3,4difluorophenyl)sulfonyl]hexahydropyrrol o[3,4-c]pyrrole-2(1 H)-carboxylate 0.368 0.035 N.D. N.D.
199
14 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2y!4-(5-fluoropyridin-2-yl)-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate 0.485 0.045 N.D. N.D.
15 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl (3R)-3- [methyl(phenylsu Ifony 1) a mino]-1 -oxa-8azaspi ro[4.5]decane-8-ca rboxylate 0.017 0.002 N.D. 44421
16 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-hydroxy-4- {Kphenylsulfonyl)amino]methyl}piperidi ne-1-carboxylate 1.84 0.161 N.D. N.D.
17 (2/?)-1,1,1 -triflu oro-3-hyd roxypropan-2yl 4-(4-fluorobenzyl)-3-oxo-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate 0.951 0.110 N.D. N.D.
18 (2/?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 2-ethyl-4-[(4-fluorophenyl)sulfonyl]-1oxa-4,9-diazaspiro[5.5]undecane-9ca rboxylate 0.524 0.049 N.D. N.D.
19 1,1,1,3,3-pentafluoro-4-hydroxybutan- 2-yl 4-[(4-fluorophenyl)sulfonyl]-1-oxa- 4,9-diazaspiro[5.5]undecane-9- ca rboxylate 0.095 0.008 >30.0d 2397
20 1,1,1,3,3-pentafluoro-4-hydroxybutan- 2-yl 4-[(4-fluorophenyl)sulfonyl]-1-oxa- 4,9-dîazaspîro[5.5]undecane-9carboxylate, ENT-1 0.061 0.006 >30.0d 4611
21 1,1,1,3,3-pentafluoro-4-hydroxybutan- 2-yl 4-[(4-fluorophenyl)sulfonyl]-1 -oxa- 4,9-dÎazaspîro[5.5]undecane-9carboxylate, ENT-2 0.599 0.053 N.D. N.D.
22 (2R)-1,1,1 -trifluo ro-3-hyd roxypropa n-2y 14 - (m orph o!ïn-4-y I su Ifonyl) -1 -oxa-4,9dîazaspîro[5.5]undecane-9-ca rboxylate 2.638 0.192 ND ND.
23 (2/?)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-(4-fluorobenzyl)-3,8- d iaza bicyclof 3.2.1 ]octane-8-ca rboxylate >1.52 0.078 N.D. N.D.
200
24 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-hydroxy-4- {[methyl( pheny Is u Ifony l)a mino]methyl}pi pend ine-1 -carboxylate 1.90d 0.195d N.D. N.D.
25 (2/7)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(4-fluorobenzyl)piperazine-1carboxylate 0.794d 0.071d N.D. N.D.
26 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-(isoquinoiin-1-yloxy)piperidine-1carboxylate, trifluoroacetic acid sait 0.031 0.002 7.73d 16949
27 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-(pyridin-2-ylamino)-1-oxa-8azaspiro[4.5]decane-8-carboxylate 0.484 0.043 N.D. N.D.
28 (2/7)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(4-fluorobenzyl)-1-oxa-3-thia-4,9diazaspiro[5.5]undecane-9-carboxylate 3,3-dioxide 0.696 0.085 N.D. N.D.
29 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[( 4-fluoro p he ny I) sulfony IJ-3hydroxy-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate 0.109e 0.024e N.D. 85270
30 (2/7)-3,3,3-trifluoro-2-[({(3/?)-3[methyl(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]dec-8yl}carbonyl)oxy]propyl phosphate, disodium sait >3.00d* 0.549* N.D. N.D.
31 (2/7)-3,3,3-tnfluoro-2-[({(3/7)-3[(p heny Isu lfonyl)a mino]-1-oxa-8azaspiro[4.5]dec-8yl}carbonyl)oxy]propyl phosphate disodium sait >3.00d >3.00d N.D. N.D.
32 (2R)-3,3,3-trifluoro-2-[({4-[(4fluorophenyl) su Ifony I]-1 -oxa-4,9diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, disodium sait >3.00d* >3.00d· N.D. N.D.
201
33 (2R)-3t3,3-trifluoro-2-[({4-[(4fluo ro ph enyl)sulfonyl]-1 -oxa-4,9diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, (bis)L-lysine sait >3.00d· >3.00d· N.D. N.D.
34 (2R)-3,3,3-trifluoro-2-[((4-[(3fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, disodium sait N.D. N.D. N.D. N.D.
35 1,1,1 -tnfluoro-3-hydroxypropan-2-yl 4[2-(morpholin-4-yl)pyrimidin-4y IJpipe rldin e-1 -carboxylate 0.339 0.032d N.D, N.D.
36 rel-(2S,3R)-1,1,1,4,4,4-hexafluoro-3hydroxybutan-2-yl (1σ,5σ,6α)-6-[1-(4fluorophenyl)-1 £/-pyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate 0.132 0.012 13.2 3736
37 (2R)-1,1,1-tnfluoro-3-hydroxypropan-2yl (1 ff,5a,6a)-6-[1-(pyridin-2-ylmethyl)1H-pyrazol-3-yl]-3azablcyclo[3.1.0]hexane-3-carboxylate 0.357e 0.027 N.D. 1616
38 1,1,1 -trifluoro-3-hydroxypropan-2-yl 4[1 -(tetrahydro-2/7-pyran-4-yl)-1 Hpyrazol-3-y l]pipe ridin e-1 -ca rboxy late 1.47 0.077 N.D. N.D.
39 1,1,1,3,3-pentafluoro-4-hydroxybutan- 2-yl (1ff,5ff,6o)-6-[1-(4-fluorophenyl)1H-pyrazol-3-yl]-3azablcyclo[3.1.0]hexane-3-carboxylate 0.034 0.002 6.012 9893
40 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl (3aR,6aS}-5-[(4fl uoro ph enyl )su If on y I] hexa hydro pyrrolo[ 3,4*c]py rrole-2( 1 H)-ca rboxy late 0.494 0.050 N.D, N.D.
41 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 2-(4-fluorobenzyl)-2,9diazaspiro[5.5]undecane-9-carboxylate 1.58 0.166 N.D. N.D.
42 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(4-fluorobenzyl)-1-oxa-4,9diazaspîro[5.5]undecane-9-carboxylate 0.304e 0.032 N.D. 2591
202
43 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[1-(4-fluoropheny!)ethyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate, formate sait 0.145 0.013 >30.0® 3478
44 (2Æ?)-1,1, 1-tnfluoro-3-hydroxypropan-2yl 4-[1-(4-fluorophenyl)ethyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate, DIAST-1 0.179 0.017 >30.0® 3701
45 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[1-(4-fluorophenyl)ethyl]-1-oxa-4,9diazaspirotS.SJundecane-g-carboxylate. DIAST-2 0.191 0.018 >30.0® 3134
46 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-f[(4-fluorobenzyl)(tetrahydro-2Hpyran-4-yl)amlno]methyl}-4hydroxypiperidine-1-carboxylate 0.560 0.046 N.D. N.D.
47 (2R)-1,1,1 - trifl uoro-3-hyd roxy propan-2y!4-({[(4fluorophenyl)sulfonyl]amino}methyl)plp eridîne-1 -carboxylate 0.174 0.017 >30.0 5046
48 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl (3aR6aS)-5-(4-cyclopropylpyridin-2yl)hexahydropyrrolo[3,4-c]pyrrole2(1H)-carboxylate >3.00® 0.926 N.D. N.D.
49 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(tetrahydro-2H-pyran-4-ylsulfonyl)1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate >3.00® 0.654 N.D. N.D.
50 (2f?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-{[(4-fluorobenzyl)(tetrahydro-2Hpyran-4-yl)amino]methyl}plperidine-1carboxylate 0.857 0.071 N.D. N.D.
51 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-[(4-fluoro-3-methylphenyl)sulfonyl]1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate 0.043 0.004 >30.0® 10340
203
52 (2ί?)-1,1,1-trifluoro-3-hydroxypropan-2yl4-[(3,4-difluorophenyl)sulfonyl]-1-oxa4,9-diazaspiro[5.5]undecane-9carboxylate 0.056 0.005 >30.0“ 7262
53 (2FÎ)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-{[5-(trifluoromethyl)pyridin-2yl]oxy}piperidine-1-carboxylate 0.053 0.006 >30.0 6900
54 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-{[3-(pyrrolidin-1 -yl) pro py IJsulfony I}1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate, trifluoroacetic acid sait >3.00d >1.68 N.D. N.D.
55 ,1,1 -trifluoro-3-hydroxypropan-2yl 4-{[2-(pyridin-2-yl)ethyl]sulfonyl}-1 oxa-4,9-diazaspiro[5.5]undecane-9carboxylate, trifluoroacetic acid sait 0.447 0.043 N.D. N.D.
56 (2/7)-1,1,1 -trifluoro-3-hyd roxyp ro pa n-2yl 4-(13-(1 H-imidazol-1yl)propyl]sulfonyl}-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate, trifluoroacetic acid sait 2.34 0.167 N.D. N.D.
57 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-[(5-methylpyridin-2yl)oxy]piperidine-1 -carboxylate 0.126 0.010 N.D. 5998
58 (2 F?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-{[5-methyl-4-(1-methyl-1H-pyrazol5-yl)pyrimidin-2-yl]oxy}piperidine-1carboxylate 0.249 0.033 N.D. N.D.
59 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[(3-chloro-4-methylphenyl)sulfonyl]1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate 0.015 0.001 N.D. 59048
60 (2i?)-1,1,1 -trifluoro-3-hydroxypropan-2y 14-[ ( 3 -ch loro-4-fl u oroph eny l)su Ifony !]1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate 0.037 0.002 N.D. 19870
204
61 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-[(3,5-dimethylpyridin-2yl)oxy]piperidine-1-carboxylate 0.053 0.005 N.D. 7304
62 (2/?)-1,1,1 -trifluoro-3-hydroxypropan-2yl4-({[(4fluorophenyl)sulfonyl](methyl)amino}me thyl)piperidine-1-carboxylate 0.067d 0.007d >30.0d 216
63 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-fl3-chloro-5-(trifluoromethyl)pyridin2-yl]oxy}piperidine-1 -carboxylate 0.007 0.001 3.01 7347
64 ( 2R)-1,1,1 -trifluoro-3-hyd roxypropan-2yl 4-{[4-(1-methyl-1H-pyrazol-5yl)pyridin-2-yl]oxy}piperidine-1carboxylate 0.550 0.042 N.D. N.D.
65 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-{[6-(1-methyl-1H-pyrazol-5yl)pyridin-2-yl]oxy}piperidine-1carboxylate 0.359 0.030 N.D. N.D.
66 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-fl3-(propan-2-yl)phenyl]sulfonyl}-1oxa-4,9-diazaspiro[5.5]undecane-9carboxylate 0.003 0.0002 7.87d 430364
67 ( 2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(benzylsulfamoyl)-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate 0.029 0,004 N.D. 30591
68 ( 2R)-1,1,1 -trifluoro-3-hyd roxypropan-2yl 4-[(4-ethynylphenyl)sulfonyl]-1 -oxa4,9-diazaspiro[5.5]undecane-9carboxylate 0.056 0.006 >30.0* 16586
69 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl (1a,5o,6a)-6-[1-(6-methoxypyridin-3- yl)-1 H-pyrazol-3-yl]-3- azabîcyclo[3.1.0]hexane-3-carboxylate 0.104 0.011 N.D. 4621
70 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[(4-fluorop heny l)su lfonyl]-5-methy I1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate, DIAST-1 0.655 0.069 N.D. N.D.
71 (2R)-1,1,1-trifluoro-3-hydroxypropan-2- 0.099 0.017 N.D. 9358
205
yl 4-[(4-fluorophenyl)sulfonyl]-5-methyl· 1-oxa-4,9-diazaspiro[5.5]undecane-9carboxylate, DIAST-2
72 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-(4-fluorobenzyl)-2-oxo-1-oxa-3,8diazaspîro[4.5]decane-8-carboxylate 0.220 0.028 N.D. N.D.
73 (2R)-1,1,1 -trifluoro-3-hyd roxypropan-2yl 4-[(3-fluoro-4-methylphenyl)sulfonyl]1-oxa-4,9-diazasplro[5.5]undecane-9carboxylate 0.005 0.001 >30.0d 96515
74 (2R)-1,1,1-trifluoro-3-hydroxypropan-2y 14-[( pyrid in-2-y Imet hy I) sulfamoy I]-1 oxa-4,9-diazaspiro[5.5]undecane-9carboxylate 0.392 0.052 N.D. N.D.
75 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-fl5-(hydroxymethyl)pyridin-2yl]oxy}piperidine-1 -carboxylate 0.688 0.079 N.D. N.D.
76 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-(5-methylpyrimidin-2-yl)-1 -oxa-4,9diazaspiro[5.5]undecane-9-carboxylate 0.658 0.083 N.D. N.D.
77 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-[(3-ethy Ipheny I) sulfony I]-1 -oxa-4,9d iazas piro[5.5]u nd ecan e-9-carboxyla te 0.005 0.001 5.95d 403739
78 (2/?)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-{[4-(propan-2yloxy)phenyl]sulfonyl}-1-oxa-4,9diazasptro[5.5]undecane-9-carboxylate 0.006 0.001 2.58d 16865
79 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 4-[(3-ethynylphenyl)sulfonyl]-1 -oxa4,9-dlazasplro[5.5]undecane-9carboxylate 0.007 0.001 10.0 26513
80 (2R)-1,1,1-tnfluoro-3-hydroxypropan-2yl 4-[1-(4-ethynylphenyl)-1 H-pyrazol-3yl]piperidine-1 -carboxylate 0.007 0.001 7.92d 126124
81 (2R)-1,1,1 -tnfluoro-3-hydroxypropan-2yl (1a,5a,6o)-6-[1-(4-ethynylphenyl)-1Hpyrazol-3-yl]-3- 0.008 0.001 7.54d 45138
206
azabicyclo[3.1.0]hexane-3-carboxylate
82 (2S)-1,1,1 -trifluoro-3-hydroxypropan-2yl (1 σ,5σ,6σ)-6-[1 - (4-et hy ny lphenyl)-1 Hpyrazol-3-yl]-3azabicyclo[3.1.0]hexane-3-carboxylate >2.75 0.596 N.D. N.D.
83 (2R)-1,1,1 - trifl uoro-3-hyd roxy p ropan-2yl 4-[(3-chlorophenyl)sulfonyl]-1-oxa4,9-diazaspiro[5.5]undecane-9carboxylate 0.019e 0.002e >30.0* 21997*
84 (2/7)-1,1,1-trifluoro-3-hydroxypropan-2yl 4-[(2-fluorophenyl)sulfonyl]-1-oxa4,9-dlazasplro[5.5]undecane-9carboxylate 0.026 0.002 19.2* 49166
85 methyl (2R)-3,313-trifluoro-2-[({4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9dîazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, ammonium sait >3.00* 0.818 N.D. N.D.
86 (2/7)-3-[(dimethoxyphosphoryl)oxy]1,1,1-trifluoropropan-2-yl 4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9diazaspîro[5.5]undecane-9-carboxylate 0.097 0.008 N.D. 132
87 ethyl (2/7)-3,3,3-trifluoro-2-[({4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, ammonium sait 0.198 0.015 N.D. N.D.
88 (2R)-3-[(d ieth oxy pho sphory l)oxy]-1,1,1 trifluoropropan-2-yl 4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate >3.00* >3.00* N.D. N.D.
89 (9/7)-10,10,10-trifluoro-6-hydroxy-2methyl-6-oxido-5,7-dioxa-2-aza-6A9phosphadecan-9-yl 4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-carboxylate >3.00* >3.00* N.D. N.D.
207
90 (2R)-3,3,3-trifluoro-2-[({4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl 2(trimethylammonio)ethyl phosphate >3.00d >3.00d N.D. N.D.
91 (2/?)-3,3,3-trifluoro-2-({[4(phenylsulfonyl)-1-oxa-4,9diazaspiro[5.5]undec-9yl]carbonyl}oxy)propyl phosphate, disodium sait N.D. N.D. N.D. N.D.
92 (27?)-1,1,1-trifluoro-3-hydroxypropan-2yl (3R)-3-[ethyl(phenylsulfonyl)amino]- 1 -oxa-8-azaspiro[4.5]decane-8carboxylate 0.006 0.001 >30d 215500
93 (27?)-1,1,1-trifluoro-3-hydroxypropan-2yl (3R)-3- [(cyclopropylsulfonyl)(methyl)amino]-1oxa-8-azaspiro[4.5]decane-8carboxylate 0.318e 0.029e >30d 3282
94 (27?)-1,1,1-trifluoro-3-hydroxypropan-2yl (3S)-3- [(cyclopropylsulfonyl)(methyl)amino]-1oxa-8-azaspiro[4.5]decane-8carboxylate 0.316e 0.027e >30d 3764
95 (27?)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-phenyl-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C101, ENT-2] 0.016 0.001 8.72 131495
96 (27?)-1,1,1 -t rifl uoro-3-hyd roxy propan-2yl 3-phenyl-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C100, ENT-1] 0.062 0.005 9.52 18560
97 (27?>-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-(5-fluoropyridin-2-yl)-1-oxa-8azaspiro[4.5]decane-8-carboxylate 0.192 0.020 >30d 1897
96 (27?)-1,1,1-trifluoro-3-hydroxypropan-2yl 2-(2-fluorobenzoyl)-2,8diazaspiro[4.5Jdecane-8-carboxylate 0.071e 0.007e >30d 4933
208
99 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-[benzoyl(methyl)amino]-1-oxa-8azaspiro[4.5]deca ne-8-carboxy late [From C112, DIAST-2] 0.107 0.011 >30d 16883
100 (2R)-1 »1J-trifluoro-3-hydroxy propan-2yl 3-[benzoyl(methyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C111, DIAST-1] 0.086 0.010 >28.7 5664
101 (2Æ?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-(1,1-dioxido-1,2-benzothiazol2(3H)-yl)-1-oxa-8-azasplro[4.5]decane- 8-ca rboxylate 0.180 0.021 6.54 2454
102 (2 F?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-[( 5- me th y Ipy ridi n-2-y I) methyl]-1 oxa-8-azaspiro[4.5]decane-8ca rboxylate 0.215 0.026 18.3 1336
103 (2 F?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-(1H-pyrazol-1-yl)-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From C120, DlAST-2] 0.334 0.030 >30d N.D.
104 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-(1H-pyrazol-1-yl)-1-oxa-8azaspiro[4.5]decane-8-ca rboxylate [From C119, DIAST-1] 1.86 0.157 N.D. N.D.
105 (2R)-1,1,1 -trtfluoro-3-hydroxypropan-2yl 2-(phenylsulfonyl)-2,8diazaspiro[4.5]decane-8-carboxylate 0.022 0.002 9.76 59993
106 (2 F?)-1,1,1 -tnfluoro-3-hydroxypropan-2yl (3F?)-3- {[(cyclopropylmethyl)sulfonyl](methyl)a mino}-1-oxa-8-azaspiro[4.5]decane-8ca rboxylate 0.238 0.020 >30d 2856
107 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl (3R)-3-[(phenylsulfonyl)(propan-2yl)amino]-1-oxa-8-azaspiro[4.5]decane8-carboxylate 0.010 0.001 >30d 127771
209
108 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl3-fl(3fluorophenyl)sulfonyl](methyl)amino}-1oxa-8-azaspi ro[4.5]decan e-8carboxylate, DIAST-1 0.051 0.003 10.7 15960
109 (2jR)-1 ,1,1-trifluoro-3-hydroxypropan-2yl 3-(((3fluorophenyl)sulfonyl](methyl)amino}-1oxa-8-azaspiro[4.5]decane-8carboxylate, DIAST-2 0.021 0.002 >30d 34918
110 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yi 3-fl(4- fluorophenyl)sulfonylj(methyl)amino}-1oxa-8-azaspiro[4.5]decane-8carboxylate, DIAST-1 0.070 0.006 7.09 8796
111 (2 R)-1,1,1 -tnfluoro-3-hydroxypropan-2yl3-fl(4fluorophenyl)sulfonyl](methyl)amino}-1oxa-8-azasplro[4.5]decane-8carboxylate, DIAST-2 0.028 0.002 >30d 33483
112 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3- [(cyclopentylsulfonyl)(methyl)amino]-1oxa-8-azaspiro[4.5]decane-8carboxylate 0.064 0.007 >30d 14232
113 (2R)-1,1,1 -tnfluoro-3-hydroxypropan-2yl 3-[(cyclobutylsulfonyl)(methyl)amino]1-oxa-8-azasplro[4.5]decane-8carboxylate 0.139 0.010 >25.5 6254
114 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-[methyl(pyridin-3-ylsulfonyl)amlno]- 1 -oxa-8-azaspiro[4.5]decane-8carboxylate 0.163 0.015 >30d 6263
115 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-[methyl(pyrimidin-5ylsulfonyl)amino]-1 -oxa-8azasplro[4.5]decane-8-carboxylate 0.341 0.027 >30d 2917
210
116 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-[methyl(tetrahydro-2H-pyran-4ylsulfonyl)amlno]-1-oxa-8azaspiro[4.5]decane-8-carboxylate [From DIAST-2 In footnote 43, Table 6] 0.986 0.084 N.D. N.D.
117 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-[methyl(tetrahydro-2H-pyran-4ylsulfonyl)amino]-1 -oxa-8azaspiro[4.5]decane-8-carboxylate [From DIAST-1 in footnote 43, Table 6] 1.97 0.153 N.D. N.D.
118 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-[3-(trifluoromethoxy)phenyl]-1 -oxa8-azaspiro[4.5]decane-8-carboxylate 0.002 0.0004 >24.8 172844
119 (2R)-1,1,1-trtfluoro-3-hydroxypropan-2yl 3-(3-chlorophenyl)-1-oxa-8azaspiro[4.5]decane-8-carboxylate 0.032 0.003 3.57 23176
120 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yi 3-(6-fiuoropyridin-3-yl)-1-oxa-8azaspiro[4.5]decane-8-carboxylate 0.166 0.021 >30® 1516
121 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-(5-methoxypyridin-2-yl)-1 -oxa-8azaspiro[4,5]decane-8-carboxylate 0.149 0.020 14.7 1207
122 (2R)-1,1,1-tnfluoro-3-hydroxypropan-2yl 3-(pyridin-3-yl)-1-oxa-8azaspiro[4.5]decane-8-carboxylate, trifluoroacetate sait 0.385 0.032 N.D. N.D.
123 (2R)-1,1,1-trifluoro-3-hyd roxypropan-2yl 3-(6-methylpyridin-3-yl)-1 -oxa-8azasplro[4,5]decane-8-carboxylate, trifluoroacetate sait 0.349 0.030 24.6 N.D.
124 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 2-(cyclopentylcarbonyl)-2,8diazaspiro[4.5]decane-8-carboxylate 0.092 0.010 >30® 6720
125 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 2-(1,3-thiazol-2-ylcarbonyl)-2,8diazaspi ro[4.5]deca ne-8-carboxy late 0,065 0.007 >30® 5464
211
126 (2R)-1,1,1’trifluoro-3-hydroxypropan-2yl 2-benzoyl-2,8-diazaspiro[4.5]decane8-carboxylate 0.112 0.012 >30® 3723
127 (2R)-1,1,1 -tnfluoro-3-hydroxypropan-2yl 2-[(4,4-difluorocyclohexyl)carbonyl]2,8-diazaspiro[4.5]decane-8carboxylate 0.163 0.017 >30® 2067
128 (2R)-1,1,1 -tnfluo ro-3-hyd ro xyp ropan-2yl 2-(pyridin-2-ylcarbonyl)-2,8diazasprro[4.5]decane-8-carboxylate 0.423 0.050 N.D. N.D.
129 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 3-(pyridin-2-yloxy)-lOxa-8azaspiro[4.5]decane-8-carboxylate, DIAST-1 >3d 0.474 N.D. N.D.
130 (2R)-1,1,1 -trifluoro-3-hyd roxypropan-2yl 3-(pyridin-2-yloxy)-1-oxa-8azaspiro[4.5]decane-8-carboxylate, DIAST-2 0.076 0.006 25.7 12483
131 (2F?)-1,1,1-tnfluoro-3-hydroxypropan-2yl 3-(1,1-dioxIdo-3,4-dihydro-2H-1,2benzothiazin-2-yl)-1-oxa-8azaspîro[4.5]decane-8-carboxylate 0.086 0.011 17.4 4241
132 ( 2 F?)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-(benzyloxy)-1-oxa-8azaspiro[4.5]decane-8-carboxylate 0.130 0.012 N.D. 8426
133 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl 2-(pyrrolidin-1-ylcarbortyl)-2,8diazaspîro[4.5]decane-8-carboxylate 0.174 0.020 4.75 3614
134 ( 2R)-1,1,1 -trrfluoro-3-hyd raxypropan-2yl 2-( 1,3-thiazol-2-ylsulfonyl)-2,8diazaspîro[4.5]decane-8-carboxylate 0.096 0.007 >23.4 9930
135 (2 F?)-1,1,1 -trifl u o ro-3-hy d roxypropan-2yl 2-(pyridin-3-yisulfonyl)-2,8diazasplro[4.5]decane-8-carboxylate 0.168 0.013 >30d 4723
136 (2F?)-1,1, 1-trifluoro-3-hydroxypropan-2yl (3F?)-3-[methyl(1 .S-thiazol-Zylsulfonyl)amfno]-1 -oxa-8azaspfro[4.5]decane-8-carboxylate 0.052 0.005 >30d 15516
212
137 (2R)-1,1, 1-trifluoro-3-hydroxypropan-2yl (3F?)-3-{methyl[(2,2,2trifl uoroethy I ) s u Ifonylja mino}-1 -oxa-8azasplro[4.5]decane-8-carboxylate 0.126 0.011 >30* 7582
138 (2F?)-1,1,1-trifluoro-3-hydroxypropan-2yl (3R)-3-(methyl[(1-methyl-1 H-pyrazol4-yl)sulfonyl]amÎno}-1-oxa-8azaspiro[4.5]decane-8-carboxylate 0.185 0.015 16.3 2154
139 ( 2 R)-1,1,1 -trifl u o ro-3-hydroxy p ropa n-2yl (3R)-3-{methyl[(2methylpropyl)sulfonyl]amino}-1-oxa-8azaspiro [4.5]deca n e-8-ca rboxy late 0.153 0.015 >30* 3058
140 <2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl (3R)-3- {[(cyclobutylmethyl)sulfonyl](methyl)ami no}-1-oxa-8-azaspiro[4.5]decane-8ca rboxy late 0.094 0.009 >30* 4595
141 (2 F?)-1,1,1-trifl uoro-3-hyd roxy propan-2yl (3R)-3-[(3fluorobenzoyl)(methyl)amino]-1-oxa-8aza spiro[4.5]deca n e-8-ca rboxy late 0.077 0.007 >30* 3519
142 ( 2 R)-1,1,1 -trifl u oro-3-hyd roxy propan-2yl {3F?)-3- [(cyclobutylcarbonyl)(methyl)amino]-1oxa-8-azaspiro[4.5]decane-8ca rboxy late 0.084 0.009 >30* 3961
143 <2F?)-1,1,1-trifluoro-3-hydroxypropan-2yl (3R)-3Kcyclobutylacetyl)(methyl)amino]-1oxa-8-azaspi ro[4.5]decane-8ca rboxy la te 0.096 0.010 >30* 2585
144 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl (3/?}-3-[methyt(3methylbutanoyl)amino]-1-oxa-8azaspi ro[4.5]decan e-8-ca rboxy late 0.105 0.010 >30* 2802
145 ( 2R)· 1,1,1 -trifluoro-3-hyd roxy propa n-2yl (3R)-3[(cyclopropylacetyl)(methyl)amino]-1- 0.198 0.019 >30* 1476
213
oxa-8-azaspiro[4.5]decane-8carboxylate
146 (2R)-1,1,1-trifluoro-3-hydroxypropan-2yl (3R)-3-[methyl(pyridin-2ylcarbonyl)amlno]-1-oxa-8azaspiro[4.5]decane-8-carboxylate, formate sait 0.539 0.048 N.D. N.D.
147 (2R)-1,1,1 -trifluoro-3-hydroxypropan-2yl 3-(3-cyanophenyl)-1-oxa-8’ azaspiro[4.5]decane-8-carboxylate 0.060 0.006 >26.4 5973
148 (2R)-3,3,3-trifluoro-2-[({(3R)-3[methyl(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]dec-8yl}carbonyl)oxy]propyl phosphate, (bis)L-lysine sait >3d >1.23 N.D. N.D.
149 (2R)-3,3,3-trifluoro-2-({[4- (p henylsu tfonyl)-1 -oxa-4,9diaza spiro[5.5]u ndec-9yl]carbonyl}oxy)propyl phosphate, (bis)L-lysine sait N.D. N.D. N.D. N.D.
150 (2R)-3,3,3-trifluoro-2-({[(3R)-3-{[(4fluorophenyl)sulfonyl](methyl)amino}-1' oxa-8-azaspiro[4.5]dec-8yl]carbonyl}oxy)propyl phosphate, (bis)L-lysine sait N.D. N.D. N.D. N.D.
a. Reported ICso values or kinact/Ki values are the géométrie mean of 2 - 4 déterminations, unless otherwise Indicated.
b. N.D. = not determined
c. The reported IC» value or kl0«t/Ki value is the géométrie mean of a5 déterminations.
d. The IC» value or km«d/Ki value Is from a single détermination.
e. In this case, the corresponding phosphate itself was tested, rather than the sait.
Example BB: Prodrug In vivo data
Rats
Test compounds (Examples 31 and 32) were administered Intravenously to groups of two rats. The characteristics of the experimental rats are given in Table BB-1.
214
Table BB-1: Characteristics of experimental rats used in study
Species Rats
Type Wistar Hann
Number and sex 2 males
Approximate âge 7-11 weeks
Approx. Body weight 250-320 g at start of treatment
Source Charles River Labs
Blood samples were taken at various times after administration and submitted to analysis for the parent compound (Examples 9 or 6) and prodrug compound (Examples 31 or 32, respectively) using an LC-MS-MS assay. Pharmacokinetic parameters derived from the 5 plasma analytical data were determined using Watson LIMS 7.2.003 (Thermo Fisher Scientific, Waltham, MA). The results are glven in Tables BB-2 to BB-5.
Table BB-2: Pharmacokinetic Parameters of
Example 31 in Wistar Hann Rats Following IV Administration at 1.48 mg/kg
Parameter Units Subject Rat 01 Subject Rat 02 Mean S.D
Original Dose (Example 31) mg/kg 1.48 1.48
AUC Interval (0-0.5 Hours) (0-0.25 Hours)
AUC ng'Hours/mL 43.1 42.9 43.0
AUC Extrap ng*Hours/mL 43.6 43.6 43.6
% AUCExtrap % 1.19 1.62 1.41
Co ng/mL 399 553 476
CL mL/min/kg 566 566 566
T1/2 Hours 0.0805 0.0445 0.0625
Vdss L/kg 2.86 1.59 2.23
Rate Constant 1/Hours 8.61 15.6 12.1
Régression Points Hours 0.083, 0.25, 0.5 0.083, 0.25
Table BB-3: Pharmacokinetic Parameters of Example 9 in Wistar Hann Rats Following IV Administration of Example 31 at 1.48 mg/kg
Parameter Units Subject Rat 01 Subject Rat 02 Mean S.D.
Original Dose mg/kg 1.48 1.48
215
Parameter Units Subject Rat 01 Subject Rat 02 Mean S.D.
(Example 31)
Cmax ng/mL 253 378 316
Tmax Hours 0.083 0.083 0.083
AUC ng*Hours/mL 118 173 146
AUC Extrap ng*Hours/mL 121 178 150
% AUC Extrap % 2.08 2.83 2.46
Rate Constant 1/Hours 0.560 0.440 0.500
T1/2 Hours 1.24 1.57 1.41
Régression Points Hours 4.7 1,2, 4,7
Table BB-4: Pharmacokinetics of Example 32 in rats after IV administration of Example 32 (2 mg/kg active)
Parameter Units Subject Rat 03 Subject Rat 04 Mean S.D.
Original Dose (Example 32) mg/kg 2 2
AUC Interval (0-1 Hours) (0-0.5 Hours)
AUC ng*Hours/mL 185 133 159
AUC Extrap ng*Hours/mL 185 134 160
% AUC Extrap % 0.232 0.832 0.532
Co ng/mL 4480 3040 3760
CL mL/min/kg 180 249 215
T1/2 Hours 0.147 0.0971 0.122
Vdss L/kg 0.515 0.679 0.597
Rate Constant 1/Hours 4.73 7.14 5.94
Régression Points Hours 0.5,1 0.25, 0.5
Table BD-5. Pliarmacokinetic Parameters of Example 6
In Wistar Hann Rats Following IV Administration of Example 32 at 2 mg/kg
Parameter Units Subject Rat 03 Subject Rat 04 Mean S.D.
Original Dose (Example 32) mg/kg 2 2
216
Cmax ng/mL 234 384 309
Tmax Hours 0.083 0.033 0.058
AUC ng*Hours/mL 102 213 158
AUC Extrap ng*Hours/mL 109 215 162
% AUC Extrap % 6.04 0.880 3.46
Rate Constant 1/Hours 2.86 1.57 2.22
T1/2 Hours 0.242 0.442 0.342
Régression Points Hours 0.25, 0.5,1 0.5,1,3
Dogs
Test compounds (Examples 31 and 32) were administered intravenously to groups of two dogs. The characteristics of the experimental dogs are given ln Table BB-6.
Table BB-6: Characteristics of experimental dogs used in study
Species Dogs
Type Beagle
Number and sex 2 males
Approximate âge 2-5 years
Approx. Body weight 9 -13 kg at start of treatment
Source Marshall Farms
Biood samples were taken at various times after administration and submitted to analysis for the parent compound (Example 9 or 6) and its prodrug compound (Example 31 or 32, respectively) using an LC-MS-MS assay. Pharmacokinetic parameters derived from the plasma analytical data were determined using Watson LIMS 7.2.003 (Thermo Fisher Scientific, 10 Waltham, MA). The results are given ln Tables B B-7 to BB-10.
Table BB-7: Pharmacokinetic Parameters of
Example 31 ln Beagle Dogs Following IV Administration at 0,7 mg/kg
Parameter Umts Subject Dog 01 Subject Dog 02 Mean S.D.
Original Dose (Example 31) mg/kg 0.7 0.7
AUC Interval (0-0.5 Hours) (0-0.5 Hours)
AUC ng‘Hours/mL 108 53 8 80.9
AUC Extrap ng*Hours/mL 108 53.9 81.0
% AUC Extrap % 0.181 0.103 0.142
Co ng/mL 1630 821 1230
CL mUmin/kg 108 216 162
T1/2 Hours 0.0614 0.0620 0.0617
217
Vdss L/kg 0.235 0.465 0.350
Rate Constant 1/Hours 11.3 11.2 11.3
Régression Points Hours 0.083, 0.25, 0.5 0.083, 0.25, 0.5
Table BB-8: Pharmacokinetic Parameters of Example 9 in Beagle Dogs Following IV Administration of Example 31 at 0.7 mg/kg
Parameter Units Subject Dog 01 Subject Dog 02 Mean S.D.
Original Dose (Example 31) mg/kg 0.7 0.7
Cmax ng/mL 614 789 702
Tmax Hours 0.25 0.083 0.17
AUC ng*Hours/mL 1350 1460 1410
AUC Extrap ng*Hours/mL 1550 1560 1560
% AUC Extrap % 12.6 6.12 9.36
Rate Constant 1/Hours 0.0648 0.0863 0.0756
T1/2 Hours 10.7 8.03 9.37
Régression Points Hours 4. 7. 24 2, 4, 7, 24
Table BB-9: Pharmacokinetic Parameters of
Example 32 in Beagle Dogs Following IV Administration at 1 mg/kg
Parameter Umts Subject Dog 03 Subject Dog 04 Mean S.D.
Original Dose (Example 32) mg/kg 1 1
AUC Interval (0-1 Hours) (0-1 Hours)
AUC ng*Hours/mL 146 229 188
AUC Extrap ng*Hours/mL 146 229 188
% AUC Extrap % 0.0443 0.150 0.0972
Co ng/mL 1220 2370 1800
CL mL/min/kg 114 72.8 93.4
T1/2 Hours 0.136 0.137 0.137
Vdss L/kg 0.751 0.357 0.554
Rate Constant 1/Hours 5.08 5.06 5.07
Régression Points Hours 0.25, 0.5, 1 0.25, 0.5, 1
218
Table BB-10: Pharmacokinetic Parameters of
Example 6 In Beagle Dogs Following IV Administration of Example 32 at 1 mg/kg
Parameter Units Subject Dog 03 Subject Dog 04 Mean S.D.
Original Dose (Example 32) mg/kg 1 1
Cmax ng/mL 514 653 584
Tmax Hours 0.083 0.083 0.083
AUC ng*Hours/mL 591 705 648
AUC Extrap ng*Hours/mL 595 710 653
% AUC Extrap % 0.630 0.733 0.682
Rate Constant 1/Hours 0.169 0.129 0.149
T1/2 Hours 4.10 5.36 4.73
Régression Points Hours 4, 7, 24 7,24
Various modifications of the invention, In addition to those described herein, will be apparent to those skilled In the art from the foregoing description. Such modifications are also intended to fall within the scope of the appendant daims. Each reference (including ail patents, patent applications, journal articles, books, and any other publications) cited in the présent application is hereby incorporated by reference in its entirety.

Claims (20)

WHATIS CLAIMED IS:
1,1,1-trifluoro-3-hydroxypropan-2-yl4-[1-(tetrahydro-2H-pyran-4-yl)-1H-pyrazol-3- yl]piperidine-1-carboxylate;
(2R)- 1,1,1-trifluoro-3-hydroxypropan-2-yl 4-(4-fluorobenzyl)-1-oxa-4,9diaz as piro[5.5]un decane-9-ca rboxy late;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(3,4-difluorophenyl)sulfonyl]-1-oxa-419diazas pi ro[5.5]un deçà ne-9-ca rboxy late;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(4-ethynylphenyl)sulfonyl]-1-oxa-4l9diazas piro[5.5]un d eca ne-9-ca rboxy late;
1. A compound of Formula I:
or a pharmaceutically acceptable sait thereof, wherein:
each of R1 and R3 is independently Cm alkyl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and ¢^.7 cydoalkyl, wherein the C3.7 cydoalkyl is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, CM alkyl, Cm haloalkyl, Cm alkoxy, and Cm haloalkoxy;
or R’ and R2, together with the N atom to which they are attached, form 4- to 14membered heterocycloalkyl that Is optionally substituted with Rs and optionally substituted with one or more independently selected R® or R30;
each of R3 and R* is independently H, halogen, OH, Ci.e alkyl, or Cj_7 cydoalkyl, wherein the Cm alkyl of R3 and R4 is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and C» cydoalkyl, and wherein the C3.7 cydoalkyl of R3 and R4 is optionally substituted with one or more substituents each Independently selected from the group consisting of OH, halogen, C,. « alkyl, CM haloalkyl, Cm alkoxy, and CM haloalkoxy;
or R3 and R4, together with the C atom to which they are attached, form a C3.7 cydoalkyl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, CM alkyl, Cm haloalkyl, Cm alkoxy, and CM haloalkoxy;
each of R3 and R® is independently H, Cm alkyl, or C3.7 cydoalkyl, wherein lhe Cm alkyl of R5 and R® is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, CM alkoxy, haloalkoxy, and Ο1β cydoalkyl, and wherein the Cj.7 cydoalkyl of R9 and R® is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, CM alkyl, CM haloalkyl, CM alkoxy, and CM haloalkoxy;
or R9 and R®, together with the C atom to which they are attached, form Cî-7 cydoalkyl that is optionally substituted with one or more substituents each Independently selected from the group consisting of OH, halogen, Cm alkyl, Cm haloalkyl, CM alkoxy, and Cm haloalkoxy;
220
R7 is H, Cm alkyl. C3.7 cycloalkyl, or R , wherein the Cm alkyl of RTis optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and Cm cycloalkyl, and wherein the C3.7 cycloalkyl of RT is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, CM alkyl, CM haloalkyl, CM alkoxy, and C,. 4 haloalkoxy;
or R7 and R®, together with the intervening moiety of C(R®)-0' to which they are attached, form 4- to 7-membered heterocycloalkyl or 5- to 10-membered heteroaryl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, oxo, halogen, CM alkyl, CM haloalkyl, C,.4 alkoxy, and C14 haloalkoxy, and wherein each of the ring-forming atoms of the 4- to 7-membered heterocycloalkyl is independently C, N, O, S, or P and wherein each of the ring-forming atoms of the 5- to 10membered heteroaryl Is C, N, O, or S;
or R7 and R3, together with the intervening moiety of “C(R4)-C(R5R®}-0 to which they are attached, form 5- to 7-membered heterocycloalkyl or5-to 10-membered heteroaryl that is optionally substituted with one or more substituents each independently selected from the group consisting of OH, oxo, halogen, Cm alkyl, CM haloalkyl, CM alkoxy, and Cv4 haloalkoxy, and wherein each of the ring-forming atoms of the 5- to 7-membered heterocydoalkyl is independently C, N, O, S, or P, and wherein each of the ring-forming atoms of the 5- to 10membered heteroaryl is C, N, O, or S;
R® is -L’-R”, -L2-R12, -L3-R”, -L4-R'4, -0(^5)^1)^2). -C(R,s)(Cy’)[-NR23-S(=O)r Cy2], or-L5-N(-Le-Cy3)(-L7-Cy4);
each R’ is independently OH, oxo, halogen, optionally substituted CM alkyl, optionally substituted C,.4 alkoxy, or optionally substituted CM cycloalkyl;
R10 is -P(=O)(OR81)(OR82) or-S(=O)2OR“;
each of L1, L2, L3, and L4 is independently absent, -(CR21 R22)™-, -NR23-, -O-, -C(=O)-, S(=O)r, -S(=O)r(CR”R22)n-, -C(=O)-(CR2,R22)n-, -S(=O)2-NR23-, -C(=O}-NR23-, -(CR21R22)f1NR23-(CR2,R22)(2-, -(CR2,R22)n-O-(CR2,R22)s-, -C(=O}-NRI3-(CR2,R22)P-, or -S(=O)rNR23(CR^R22),,-;
L5 is absent or-(CR21 R22)-;
L® is absent or -(CR21 R22)-;
L7 is absent, -(CR71 R32)-, or-S(=O)2-;
R11 is 5- to 10-membered heteroaryl optionally substituted with one or more independently selected R3';
221
R12 Is 4- to 14-membered heterocycloalkyl optionally substituted with one or more independently selected R32;
R13 Is Co-ioaryl optionally substituted with one or more independently selected R33;
R14 Is Cj,14 cycloalkyl optionally substituted with one or more Independently selected R34;
R1S Is H, OH, halogen, Cm alkoxy, Cm alkyl, orcyclopropyl;
each of R21 and R22 is independently H, OH, halogen, Cm alkyl, or cyclopropyl, wherein the Cm alkyl Is optionally substituted with one or more substituents each independently selected from the group consisting of OH, halogen, Cm alkoxy, Cm haloalkoxy, and cyclopropyl;
R23 is H, CM alkyl, or cyclopropyl;
each of R30, R31, R32, R33, and R34 is independently selected from the group consisting of halogen, -N(R“)(Rb), -N(Rc)(C(=O)Rd), -N(Rc)(S(=O)2Rd), -C(=O)-N(R')(Rb), -C(=O)-Rd, -C(=O)ORd, -OC(=O)-RliI -N(Rc)(S(=O)2Rd), -S(=O)rN(R*)(Rb), -SR4, -S(=O)2Rd, -ORd, -OR”, -CN, Cie alkyl, CM alkenyl, CM alkynyl, Cj-w cycloalkyl, 4- to 10-membered heterocycloalkyl, Ce-io aryl,
(2/7)-3,3,3-trifluoro-2-({[4-(phenylsulfonyl)-1-oxa-4,9-diazaspifo[5.5]undec-9yl]cart>onyl}oxy)propyl phosphate, (bis)-L-lysine sait, and (2/7)-3,3,3-trifluoro-2-({[(3/7)-3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8azaspiro[4.5]dec-8-yl]carbonyl}oxy)propyl phosphate, (bls)-L-lysine sait.
(2/7)-3,3,3-trifluoro-2-[({(3/7)-3-[methyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]dec8-yl}carbonyl)oxy]propyl phosphate, (bls)-L-lysine sait;
(2/7)-3,3,3-trifluoro-2-({[4-(phenylsulfonyl)-1-oxa-4,9-diazaspiro[5.5]undec-9yl]carbonyl}oxy)propyl phosphate, disodium sait;
(2/7)-3,3,3-trifluoro-2-[({4-[(3-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}cart>onyl)oxy]propyl phosphate, disodium sait;
(2/7)-3,3,3-trifluoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-dÎazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, (bisj-L-lysine sait;
(2/7)-3,3,3-trifîuoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspîro[5.5]undec-9yl}carbonyl)oxy]propyl phosphate, disodium sait;
(2/7)-3,313-trifluoro-2-[(((3/7)-3-[methyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]deo8-yl}carbonyl)oxy]propyl phosphate, disodium sait;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3/7)-3-[(cydopropylacetyl)(methyl)amino]-1oxa-8-azaspiro[4.5]decane-8-carboxylate; and (2R)-3,3,3-trifluoro-2-(fl(3R)-3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8azaspiro[4.5]dec-8-yl]carbonyl}oxy)propyl dihydrogen phosphate, or a pharmaceutically acceptable sait thereof,
229 or a pharmaceutically acceptable sait of daim 1 selected from:
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3/7)-3-(methyl[(2methylpropyl)sulfonyl]amino}-1-oxa-8-azaspiro[4.5]decane-8-carboxylate;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3/7)-3-(1716^1((2,2,2trifluoroethyl)sulfonyl]amino}-1-oxa-8-azaspiro[4.5]decane-8-carboxylate;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 2-(cydope ntylcarbony 1)-2,8diazaspiro[4.5]decane-8-carboxylate;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[3-(trifluoromethoxy)phenyl]-1-oxa-8azas pi ro[4.5]d eca ne-8-ca rboxy late ;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3/7)-3{[(cyclopropy Imethy l)su Ifo rry l](methy I) a mino}-1 -oxa-8-azaspiro(4.5]decane-8-carboxy late ;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-[benzoyl(methyl)amino]-1-oxa-8azaspiro[4.5]d eca ne-8-ca rboxy la te ;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3/7)-3-[(cydopropylsulfonyl)(methyl)amino]-1oxa-8-azaspi ro[4.5]d eca ne-8-carboxylate;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1oxa-8-aza spi ro[4.5]d eca ne-8-carboxylate, DIAST-2 ;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1oxa-8-azaspiro[4.5]decane-8-carboxylate, DIAST-1 ;
(2/7)-3,3,3-trifluoro-2-({[4-(phenylsulfonyl)-1-oxa-4,9-diazaspiro[5.5]undec-9yl]carbonyl}oxy)propyl dihydrogen phosphate;
(2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-(4-fluorobenzyl)-2-oxo-1-oxa-3,8diazaspiro[4.5]decane-8-carboxylate;
2, The compound of Claim 1, or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I or pharmaceutically acceptable sait thereof is a compound of Formula I1, l-a, or l-a1 :
224 or pharmaceutically acceptable sait thereof.
3. The compound of Claim 2, or a pharmaceutically acceptable sait thereof, wherein the compound of Formula I Is a compound of l-a1.
3;
m is 1, 2, or 3;
n is 1,2, or 3;
p is 1, or 2; and r is 0 or 1, provided that when r is 1 and each of R3, R4, Rs and RB Is H, then the moiety of ·N(R’)(R2) Is other than optionally substituted 4-oxo-3H-5,6,7,8-tetrahydropyrido[3,4cflpyrimidin-7-yl.
4,9-diazaspiro[5.5]undec-9-yl}carbonyl)oxy]propyl dihydrogen phosphate, or a pharmaceutically acceptable sait thereof.
15
4,9-diazas pi ro[5.5]undecane-9-carboxy late ;
(2R)-3,3,3-trifluoro-2-[({(3R)'3-[methyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]dec8-yl}carbonyl)oxy]propyl dihydrogen phosphate;
(2R)-3,3,3-trifluoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl dihydrogen phosphate;
(2R)-3,3,3-trifluoro-2-[({4-[(3-fluorophenyl)sulfonyl]-1-oxa-4,9-diazaspiro[5.5]undec-9yl}carbonyl)oxy]propyl dihydrogen phosphate;
228
4. The compound of any one of Claims 1 to 3, or a pharmaceutically acceptable sait thereof, wherein R1 and R2, together with the N atom to which they are attached, form 4- to 14membered heterocycloalkyl that Is substituted with R8 and optionally substituted with one or more independently selected R*.
5 spondylitis, goût, labor, musculoskeletal disease, skin disease, toothache, pyresis, bum, sunburn, snake bite, venomous snake bite, spider bite, insect sting, neurogenic bladder, interstitial cystitis, urinary tract infection (ΙΠΊ), rhinitis, contact dermatitîs/hypersensitivity, itch, eczema, pharyngitis, mucositis, enteritis, Irritable bowel syndrome (IBS), cholecystitis, and pancreatitis; neuropathie pain (e.g., neuropathie low back pain, complex régional pain
5 meningitis; sleeping slckness; progressive multifocal feukoencephalopathy; De Vivo disease; cérébral edema; cérébral palsy; withdrawal syndrome [alcohol withdrawal syndrome, antidepressant discontinuation syndrome, antipsychotic withdrawal syndrome, benzodiazépine withdrawal syndrome, cannabis withdrawal, néonatal withdrawal, nicotine withdrawal, or opiold withdrawal]; traumatic brain Injury: non-traumatic brain injury; spinal cord injury; seizures;
5 azaspiro[4.5]decane-8-carboxyiate, DiAST-2; or a pharmaceutically acceptable sait thereof.
5. The compound of any one of claims 1 to 4, or a pharmaceutically acceptable sait thereof, wherein:
the moiety of -N(R1)(Ri)’ Is a moiety of Formula a-1, a-2, a-3, a-4, a-5, ora-6:
ring A1 is 4- to 7-membered cycloalkyl or heterocycloalkyl;
225 t1 is 0,1,2, or 3; t2 is 0,1,2, or 3; t3 is 0,1, 2, or 3; s1 is 1 or 2; and s2 is 1 or 2.
5- to 10-membered heteroaryl, (C3.10 cydoalkyl)-CM alkyl-, (4- to 10-membered heterocydoalkyl)-CM alkyl-, (Cmo aryl)-CM alkyl-, and (5-to 10-membered heteroaryl)-CM alkyl-, wherein each of the C14 alkyl, C2^ alkenyl, C2« alkynyl, C^o cydoalkyl, 4- to 10membered heterocydoalkyl, Ce-io aryl, 5- to 10-membered heteroaryl, (Cmo cycloalkyl)-CM alkyl-, (4-to 10-membered heterocydoalkyl)-CM alkyl-, (Ce.10aryl)-CMalkyl-, and (5-to 10membered heteroaryl)-CM alkyl- is optionally substituted with one or more Independently seleded R”; and wherein each of the C14 alkyl, Cyi0 cydoalkyl, 4- to 10-membered heterocydoalkyl, (C^o cydoalkyl)-CM alkyl-, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce.io aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl- is further optionally substituted one or more oxo;
each R” is independently seleded from the group consisting of H, C14 alkyl, Cj.10 cycloalkyl, 4- to 10-membered heterocydoalkyl, CMo aryl, 5- to 10-membered heteroaryl, (C3.10 cycloalkyl)-CM alkyl-, (4- to 10-membered heterocydoalkyl)-CM alkyl-, (Cmo aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, wherein each of the C14 alkyl, Cmo cydoalkyl, 4- to 10-membered heterocycloalkyl, Ce-io aryl, 5- to 10-membered heteroaryl, (Quo cydoalkyl)Cm alkyl·, (4- to 10-membered heterocydoalkyl)-CM alkyl-, (Cno aryl)-CM alkyl-, and (5- to 10membered heteroaryl)-CM alkyl- is optionally substituted with one or more substituents independently selected from the group consisting of halogen, -CN, -C(=0)Cm alkyl, -C(=O)OH, -C(=0)0-Cm alkyl, -C(=O)NHCm alkyl, -C(=0)N(Cm alkyl)2, oxo, -OH, -OC(=O)-Cm alkyl, OC(=O)O-Cm alkyl, -NH2, -NH(Cm alkyl), -N(Cm alkyl)2, -NHC(=O)Cm alkyl, -NHC(=O)OCm alkyl, -NHC(=O)NHCm alkyl, and Cm alkoxy;
222 each R38 is independently selected from the group consisting of halogen, -OH, -NO2, CN, -SFs, Cm alkyl. Cm haloalkyl, Cm haloalkoxy, Cm alkenyl, Cm alkynyl, C3.7 cycloalkyl, a 4to 10-membered heterocycloalkyl, -N(R*)(Rb), -N(Re)(C(=O)Rd), -C(=O)-N(R,)(Rb), -C(=O)-Rd, C(=O)-ORd, -OC(=O)-Rd, -N(RG)(S(=O)2Rd), -S^OJrNtR’JiR6), -SRd, -8(=Ο)2Κ*, and -ORd, wherein each ofthe Cm alkyl, Cs-7 cycloalkyl, and heterocydoalkyl is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, CN, -OH, Cm alkyl, CM alkoxy, CM haloalkyl, CM haloalkoxy, CM cycloalkyl, -N(R*)(Rb), NfR'XC^OJR11), -C(=O)-ORd, -C(=O)H, -C(=O)Rd, -C(=O)N(R')(Rb), -N(Rc)(S(=O)2Rd), -S(=O)2N(R*)(Rb), -SRd, -S(=O)2Rd, and -OR4;
each of R’1, R82, and R’° is independently selected from the group consisting of H, Cm alkyl, Cj.? cycloalkyl, and (Cy7 cycloalkyl)-Cm alkyl-, wherein each of the Cm alkyl, C3.7 cycloalkyl, and (C3.? cycloalkyl)-CM alkyl- is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CN, -OH, oxo, -NH2, NH(Cm alkyl), -N(CM alkyl)2, Cm alkyl, CM alkoxy, Cm haloalkyl, CM haloalkoxy, Cj.e cycloalkyl;
or OR81 and OR82, together with the P(=O) to which they are attached, form 4- to 10membered heterocycloalkyl that is further optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CN, -OH, oxo, -NH2, NH(Cm alkyl), -N(Cm alky1)a, Cm alkyl, Cm alkoxy, Cm haloalkyl, CM haloalkoxy, and Cj^ cycloalkyl;
each of Cy1, Cy2, Cy3, and Cy4 is independently selected from the group consisting of R”, R12, R13, and R'4;
each R‘ is independently H, Cm alkyl, Cm haloalkyl, Cy? cycloalkyl, or (Cy? cycloalkyl)Cm alkyl-;
each Rb Is Independently H or selected from the group consisting of Cm alkyl, CM haloalkyl, C3.T cydoalkyl, a 4- to 10-membered heterocycloalkyl, Cmo aryl, a 5- to 10-membered heteroaryl, (C3.7 cycloalkyl)-CM alkyl·, (4- to 10-membered heterocycloalkyl)-CM alkyl-, (Ce.10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-CM alkyl-, wherein each ofthe sélections from the group Is optionally substituted with one or more substituents each independently selected from the group consisting of-OH, -CN, CM alkyl, Cy7 cycloalkyl, Cm hydroxylalkyl, -SCm alkyl, -C(=O)H, -C(=O)-Cm alkyl, -C(=0)-0-Cm alkyl, -C(=O)-NH2, -C(=O)-N(Cm alkyl)2, Cm haloalkyl, Cm alkoxy, and CM haloalkoxy, or R1 and Rb, together with the N atom to which they are attached, form a 4- to 10membered heterocycloalkyl or a 5- to 10-membered heteroaryl, each optionally substituted with one or more substituents each Independently selected from the group consisting of halogen, 18588
223
OH, oxo, -C(=O)H, -C(=O)OH, -C(=O)-Cm alkyl, -C(=O)-NH21 -C(=O)-N(Cm alkyl)21 -CN, CM alkyl, Cm cycloalkyl, (C^ cycloalkyl)-Cv2 alkyl-, Cm alkoxy, Cm hydroxylalkyl, Cm haloalkyl, and Cm haloalkoxy;
each Rc is independently selected from the group consisting of H, Cm alkyl, C3.7 cycloalkyl, and (C3.7 cycloalkyl)-CM alkyl-;
each Rd is independently selected from the group consisting of C14 alkyl, C3.7 cycloalkyl, a 4- to 14-membered heterocycloalkyl, C^aryl, a 5- to 10-membered heteroaryl, (C3.7 cycioalkyl)-CM alkyl-, (4- to 10-membered heterocydoalkyl)-CM alkyl-, (Ce.10 aryl)-CM alkyl-, and (5- to 10-membered heteroaryl)-C14 alkyl-, wherein each of the sélections from the group is optionally substituted with one or more substituents each independently selected from the group consisting of halogen, -CF3, -CN, -OH, oxo, -S-Cm alkyl, Cm alkyl, Cm haloalkyl, 02.9 alkenyl, C2_e alkynyl, C3.7 cycloalkyl, Cm alkoxy, and CM haloalkoxy;
each of f1 and f2 Is independently 0,1, or 2, provided that the sum of f1 and f2 is 1,2, or
6. The compound of Claim 5, or a pharmaceutically acceptable sait thereof, wherein: the moiety of “-N(R’)(R2)“ is a moiety of Formula a-46-1, a-46-2, a-46-3, a-46-4, a-46-5, a-46-6, or a-46-7:
(R\o.
R8 a-46-2 a-46-3 t
a-46-1 t2 is 0,1,2, or 3;
t3 is 0, 1, or 2, t4 Is 0,1, or 2; and each R* Is Independently F, Cl, methyl, or Ci fluoroalkyi.
226
7. The compound of any one of daims 1-3, or a pharmaceutically acceptable sait thereof, wherein:
the moiety of -NtR’XR2)* Is a moiety of Formula b-46-1, b-46-2, b-46-3, b-46-4, b-46-5, b-46-6, or b-47:
I b-46-2 b-46-1
Oû (R30*
I t11 is 0,1, 2, or 3;
t3 Is 0,1, or 2; and each R* is Independently F, Cl, methyl, or Ci fluoroalkyl.
8. The compound of any one of daims 1 to 7, or a pharmaceutically acceptable sait thereof, wherein each of Rs and R® is H.
227
9. The compound of any one of daims 1 to 8, or a pharmaceutically acceptable sait thereof, wherein R7 is H or R10; and R10 is -P(=O)(OR”)(ORW).
10 syndrome, post trigeminal neuralgia, causalgla, toxic neuropathy, reflex sympathetic dystrophy, diabetic neuropathy, chronic neuropathy from chemotherapeutic agent, or sciatica pain)]; a demyelinating disease [e.g., multiple sclerosis (MS), Devic’s disease, CNS neuropathies, central pontine myelînolysis, syphilitic myelopathy, leukoencephalopathies, leukodystrophies, Guillain-Barre syndrome, chronic inflammatory demyelinating polyneuropathy, anti-myelin15 associated glycoprotein (MAG) peripheral neuropathy, Charcot-Marie-Tooth disease, peripheral neuropathy, myelopathy, optic neuropathy, progressive inflammatory neuropathy, optic neuritis, transverse myelitisj; and cognitive impairment [e.g., cognitive Impairment associated with Down's syndrome; cognitive impairment associated with Alzheimeris disease; cognitive impairment associated with PD; mild cognitive Impairment (MCI), dementia, post-chemotherapy
10 excitotoxin exposure; ischemia [stroke, hepatic ischemia or reperfusion, CNS Ischemia or reperfusion]; liver fibrosis, iron overload, cirrhosis of the liver; a lung disorder [asthma, allergies, COPD, chronic bronchitis, emphysema, cystic fibrosis, pneumonia, tuberculosis, pulmonary edema, lung cancers, acute respiratory distress syndrome, intersitital lung disease (ILD), sarcoidosis, idiopathic pulmonary fibrosis, pulmonary embolism, pleural effusion, or
10. The compound of any one of Claims 1 to 6, 8, and 9, or a pharmaceutically acceptable sait thereof, wherein the moiety of “-NiR’XR2) is a moiety of Formula a-46-1; R* is-L’-R11 orL’-R13; and each of L1 and L3 is -C(=O)- or-S(=O)r.
11. A compound of daim 1 selected from:
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (1 a,5a,6a)-6-[1-(5-methoxypyridin-2-yl)-1 H· py razol-3-yl]-3-aza bicydo[3.1.0]hexa ne-3-carboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (1a,5a,6a)-6-[1-(4-fluorophenyl)-1H-pyrazol-3yl]-3-azabicydo[3.1.0]hexane-3-carboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(4-fluorophenyl)su!fony!]-1-oxa-4,9diazaspiro[5.5]undecane-9-ca rboxylate;
(2R> 1,1,1 -trifl uoro-3-hyd roxy propan-2-y 14-(ph e nyl sulfony l)-1 -oxa-4,9d iaza s pi ro[5.5]un deçà ne-9-ca rboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3S)-3-[(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]decane-8-ca rboxylate;
(2R>1,1,1-trifluoro-3-hydroxypropan-2-yl (3R)-3-[(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(3-fluorophenyl)sulfonyl]-1-oxa-4,9diazaspiro[5.5]undecane-9-ca rboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl (3R)-3-[methyl(phenylsulfonyl)amino]-1-oxa-8azaspiro[4.5]decane-8-carboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-(4-fluorobenzyl)-3,8diazabicyclo[3.2,1]odane-B-carboxylate;
(2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(4-fluorophenyl)sulfonyl]-3-hydroxy-1-oxa-
12. A compound of Claim 1 that is (2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl (1α,5α,6α)-6-[1(S-methoxypyridin^-ylFIH-pyrazol-S-ylJ-S-azabicydop.l.Olhexane-S-carboxylate, ora pharmaceutically acceptable sait thereof.
13. A compound of Claim 1 that is (2/7)-1,1,1-trifluoro-3-hydroxypropan-2-yl 4-[(4fluorophenyl)sulfonyl]-1-oxa-4,9-diazasplro[5.5]undecane-9-cart>oxylate, or a pharmaceutically acceptable sait thereof.
14. A compound of Claim 1 that is (2/7)-1,1,1-tnfluoro-3-hydroxypropan-2-yl 4(phenylsulfonyl)-1-oxa-4,9-diazaspiro[5.5]undecane-9-carboxylate, or a pharmaceutically acceptable sait thereof.
15 mesothelioma]; a liver disorder [acute liver failure, Alagille syndrome, hepatitis, enlarged liver, Gilbert's syndrome, liver cysts, liver hemangioma, fatty liver disease, steatohepatitis, primary sclerosing cholangitis, fascioliasls, primary bilary cirrhosis, Budd-Chiari syndrome, hemochromatosis, Wilson's disease, or transthyretin-related hereditary amyloidosis], stroke [e.g., Ischémie stroke; hémorrhagie stroke]; subarachnoid hemorrhage; intracérébral
20 hemorrhage; vasospasm; AIDS wasting syndrome; rénal ischemia; a disorder associated with abnormal cell growth or prolifération [e.g., a benign tumor or cancer such as benign skin tumor, brain tumor, papilloma, prostate tumor, cérébral tumor (glioblastoma, medulloepithelioma, medulloblastoma, neuroblastome, astrocytoma, astroblastoma, ependymoma, oligodendroglioma, plexus tumor, neuroepithelioma, epiphyseal tumor, ependymoblastoma,
25 malignant meningioma, sarcomatosis, melanoma, schwannoma), melanoma, metastatic tumor, kidney cancer, bladder cancer, brain cancer, glioblastoma (GBM), gastrointestinal cancer, leukemia or blood cancer]; an autoimmune disease [e.g., psoriasis, lupus erythematosus, Sjogren’s syndrome, ankyfosing spondylitis, undifferentiated spondylitis, Behcet's disase, hemolytic anémia, graft rejection]; an inflammatory disorder [e.g., appendices, bursitis, colitis,
30 cystrtis, dermatitis, phlebitis, rhinitis, tendonitis, tonsillitis, vasculitis, acné vulgaris, chronic prostatitis, glomerulonephritis, hypersensitivities, IBS, pelvic inflammatory disease, sarcoidosis, HIV encephalitis, râbles, brain abscess, neuroinflammation, Inflammation in the central nervous System (CNS)j, a disorder oi the immune system (e.g., transplant rejection or cehac disease), post-traumatic stress disorder (PTSD); acute stress disorder; panic disorder; substance35 induced anxiety; obsessive-compulsive disorder (OCD); agoraphobie; spécifie phobia; social phobia; anxiety disorder; attention déficit disorder (ADD); attention déficit hyperactivity disorder (ADHD); Asperger's syndrome; pain [e.g., acute pain; chronic pain; inflammatory pain; viscéral
232 pain; post-operative pain; migraine; lower back pain; joint pain; abdominal pain; chest pain; postmastectomy pain syndrome; menstrual pain; endometriosls pain; pain due to physical trauma; headache; sinus headache; tension headache arachnoiditis, herpes virus pain, diabetic pain; pain due to a disorder selected from: osteoarthritis, rheumatoid arthritis, osteoarthritis,
15. A compound of Claim 1 that is (2/7)-1,1,1-trifîuoro-3-hydroxypropan-2-yl (3/7)-3[methyl(phenylsulibnyl)amino]-1-oxa-8-azaspiro[4.5Jdecane-8-carboxylale, ora pharmaceutically acceptable sait thereof.
230
16. A compound of Claim 1 that Is (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{[(4fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8-azaspiro[4.5]decane-8-carboxylate, DIAST-1; or (2R)-1,1,1-trifluoro-3-hydroxypropan-2-yl 3-{[(4-fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8-
17. A compound of Claim 1 that is (2R)-3,3,3-trifluoro-2-[({(3R)-3[methyl(phenylsulfonyl)amino]-1-oxa-8-azaspiro[4.5]dec-8-yl}carbonyl)oxy]propyl dihydrogen phosphate, or a pharmaceutically acceptable sait thereof.
18. A compound of Claim 1 that is (2R)-3,3,3-trifluoro-2-[({4-[(4-fluorophenyl)sulfonyl]-1-oxa-
19. A compound of Claim 1 that Is (2R)-3,3,3-trifluoro-2-({[(3R)-3-{[(4fluorophenyl)sulfonyl](methyl)amino}-1-oxa-8-azaspiro[4.5]dec-8-yl]carbonyl}oxy)propyl dihydrogen phosphate, or a pharmaceutically acceptable sait thereof.
20. A pharmaceutical composition comprising a compound or pharmaceutically acceptable
20 sait according to any one of Claims 1 to 19, and a pharmaceutically acceptable carrier.
21. A compound or pharmaceutically acceptable sait according to any one of Claims 1 to 19 for use in the treatment of a MAGL-mediated disease or disorder.
25 22. Use of a compound or pharmaceutically acceptable sait according to any one of Claims 1 17 in the manufacturing a médicament for treating a MAGL-mediated disease or disorder.
23. The compound or pharmaceutically acceptable sait thereof of claim 21, or the use of Claim
22. wherein the disorder is selected from the group consisting of a metabolic disorder (e.g.,
30 obesity); a kidney disease (e.g. acute inflammatory kîdney Injury and diabetic nephropathy); vomiting or emesls (e.g. chemotherapy Induced vomiting); nausea (e.g. refractory nausea or chemotherapy induced nausea); an eating disorder (e.g., anorexia or bulimia); neuropathy (e.g., diabetic neuropathy, peliagric neuropathy, alcoholie neuropathy, Béribéri neuropathy); burning feet syndrome; a neurodegenerative disorder [multiple sclerosis (MS), Parkinson's disease
35 (PD), Huntington’s disease, dementia, Alzheimeris disease, amyotrophie latéral sclerosis (ALS), epilepsy, fronto-temporal lobe dementia, a sleep disorder, Creutzfeldt-Jakob disease (CJD), or prion disease]; a cardiovascular disease (e.g., hypertension, dyslipidemia, atherosclerosis,
231 cardiac arrhythmias, or cardiac Ischemia); osteoporosis; osteoarthrîtis; schizophrénie;
dépréssion; bipolar disease; tremor; dyskinesia; dystonîa; spasticity; Tourette’s syndrome; sleep apnea; hearing loss; an eye disease (e.g., glaucoma, ocular hypertension, macular degeneration, or a disease arising from elevated intraocular pressure); cachexia; Insomnia;
20 cognitive Impairment (PCCI), postoperative cognitive dysfunction (POCD)].
OA1201800030 2015-07-31 2016-07-18 1,1,1-Trifluoro-3-Hydroxypropan-2-yl Carbamate Derivatives and 1,1,1-Trifluoro-4Hydroxybutan-2-yl Carbamate Derivatives as Magl Inhibitors. OA18588A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/199,330 2015-07-31
US62/335,290 2016-05-12

Publications (1)

Publication Number Publication Date
OA18588A true OA18588A (en) 2018-12-28

Family

ID=

Similar Documents

Publication Publication Date Title
US10723711B2 (en) 1,1,1-trifluoro-3-hydroxypropan-2-yl carbamate derivatives and 1,1,1-trifluoro-4-hydroxybutan-2-yl carbamate derivatives as MAGL inhibitors
US10858373B2 (en) Heterocyclic spiro compounds as MAGL inhibitors
CA3050625C (en) 1,1,1-trifluoro-3-hydroxypropan-2-yl carbamate derivatives as magl inhibitors
OA18588A (en) 1,1,1-Trifluoro-3-Hydroxypropan-2-yl Carbamate Derivatives and 1,1,1-Trifluoro-4Hydroxybutan-2-yl Carbamate Derivatives as Magl Inhibitors.