WO2025049746A1 - Compositions comprising werner syndrome helicase inhibitors and methods of using the same - Google Patents
Compositions comprising werner syndrome helicase inhibitors and methods of using the same Download PDFInfo
- Publication number
- WO2025049746A1 WO2025049746A1 PCT/US2024/044435 US2024044435W WO2025049746A1 WO 2025049746 A1 WO2025049746 A1 WO 2025049746A1 US 2024044435 W US2024044435 W US 2024044435W WO 2025049746 A1 WO2025049746 A1 WO 2025049746A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- ring
- membered heterocyclyl
- substituted
- alkyl
- heterocyclyl ring
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/04—Ortho-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/12—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
- C07D487/14—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D519/00—Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
Definitions
- compositions comprising Werner Syndrome Helicase (WRN, also known as Werner Syndrome RecQ Helicase or Werner Syndrome ATP-Dependent Helicase) inhibitors and methods of using the same.
- WRN Werner Syndrome Helicase
- the WRN gene encodes a helicase that falls within the RecQ DNA helicase subfamily and is directly involved in DNA damage repair. While WRN has previously been shown to exhibit tumor suppressive activity, an activity that correlates well with its activity in DNA damage repair, certain cancer cells have been shown to be dependent on the presence of WRN. For example, certain cancers associated with deficient DNA mismatch repair (dMMR), which often exhibit high levels of microsatellite instability (microsatellite instability-high or MSI-H cancers), depend on WRN activity for continued survival. In view of WRN’ s essential role in such cancers, there remains a need in the art to identify WRN inhibitors for use in the treatment of such dMMR and/or MSI-H cancers.
- dMMR deficient DNA mismatch repair
- compositions and methods described herein relate to compositions comprising WRN helicase inhibitors and methods of their use in treating disease, e.g., cancer.
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof
- K and J are independently selected from C, N, O, or S
- R 6 and R 7 are independently selected from H or halogen; wherein R 6 or R 7 can optionally join with R 5 to form a C 3 -C 8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl,
- K and J are independently selected from C, N, O, or S;
- R 6 and R 7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C 6 -C 10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) n is 0, 1, 2, or 3; refers to a single or a
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is C 1 -C 6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl or C3-C6 cycloalkyl.
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R5 is C2 alkyl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are independently selected from H or halogen.
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are H.
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are F.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R 1 is aryl, C 3 -C 7 cycloalkyl, 5-10 membered heteroaryl, or 3- 8 membered heterocyclyl, the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is optionally substituted with one or more R 36 , or the said aryl,
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O- (C 1 -C 6 )alkyl-COOH, three C 1 -C 6 alkoxy, -C(O)-R 33 , -C(O)-NH-R 33 , -S(O)R33, -
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is NR 30 R 31 , R 30 and R 31 are independently selected from optionally substituted C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C1-C6 alkynyl, or H.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C3 alkyl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are -CH3.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is substituted by one or more -O-(C 1 -C 6 )alkyl-COOH, three C 1 -C 6 alkoxy, -C(O)-R 33 , - C(O)-NH-R 33 , -S(O)R 33 , -S(O 2 )R 33 , -NH-S(O 2 )R 33 , -NH-C(O)-NH-R 33 , or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is aryl, the aryl is fused with a 5- or 6- membered heterocyclyl ring, wherein the said 5- or 6- membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, and wherein the said 5- or 6- membered heterocyclyl ring is optionally substituted with C 1 -C 3 alkyl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is phenyl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 5-membered heterocyclyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 6-membered heterocyclyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6- membered heterocyclyl ring comprises carbon atoms and one or two oxygen atoms.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6- membered heterocyclyl ring comprises carbon atoms and one or two nitrogen atoms.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6- membered heterocyclyl ring comprises carbon atoms, one or two oxygen and one or two nitrogen atoms.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ,
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: , ,
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a C3-C8 cycloalkyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3-8 membered cycloalkenyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3-8 membered heterocyclyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is a fully saturated C 3 -C 7 cycloalkyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the C 3 -C 7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C 3 -C 7 cycloalkyl ring is optionally linked with a second C 3 -C 8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH,- C(O)-C 1 -C 6 alkyl, or -O-(C 1 -C 6 )alkyl-COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3- C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring.
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein,
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is substituted with C1-C4 alkoxy.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said 4- or 7- membered heterocyclyl ring is optionally joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6- membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C 2 -C 6 halogenated alkenyl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6- membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsub
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein R 1 is selected from
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein R1 is selected from:
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein the said the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, X is N, Y is C, R 2 is NR 8 R 9, and R 3 is H, wherein when R 2 is NR 8 R 9 , R 8 and R 9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6- membered heterocyclyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, X is N, Y is In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H, wherein R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R 11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R 2 is NR 8 R 9 , R 8 and R 9 are independently selected from H, C 1 -C 4 alkyl, and -C(O)R 11 , wherein R 11 is
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused ring .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5- membered heterocyclyl ring, wherein when the 5-membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R 11 is a C 1 -C 3 alkyl or substituted 5- or 6-membered heterocyclyl ring.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused 4- or 5-membered heterocyclyl ring is selected from:
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C1-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C 3 -C 5 cycloalkyl ring, halogen, -
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R 2 is H, R 3 is -C(O)R 10 , and R 10 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, a C 3 -C 5 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C3- C 5 cycloalkyl ring, halogen, -S(O 2 )CH 3 , and/or -C(O)N(CH 3 ) 2 .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from:
- the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein R 10 is C2-C6 alkenyl.
- the compositions and methods described herein relate to a compound of fo nts disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R 2 is H, R 3 is -S(O 2 )R 10 , and R 10 is C 1 -C 4 alkyl, C 1 -C 4 alkenyl, a C 3 -C 5 cycloalkyl ring, a 5- or 6-membered heterocyclyl ring, wherein R10 is optionally substituted with -OH, C1- C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or - C(O)N(CH 3 ) 2 .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from: .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, wherein , wherein W, P, S, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; w wherein when T is N, R10 is , and M and Z are independently selected from N or C; and wherein when T is C, R 10 i wherein when N M and M Z are both single bonds, M is C(O) and
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from: ,
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is , wherein A, W, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond;
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from: .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from:
- the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C 3 -C 7 cycloalkyl ring, a C 1 -C 6 aryl, or a 5-
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from: .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R 2 is H, R 3 is wherein when U is N + , R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -OH or -NR19R20; wherein when R15 is -OH, R16 is a carbonyl, R17 and R18 join together to form a fused 5- or 6- membered heterocyclyl ring, and U V is a single bond; wherein when R15 is NR19R20,
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from: .
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein R 23 and R 24 are independently selected from -OH, -COOH, -NH 2 , - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalky
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R 4 is selected from independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R 23 is C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, C 1 -C 6 hydroxyalkyl, halogenated C 1 -C 6 alkoxy, C 3 -C 8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membere
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein is C or O, n is 0 or 1, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl.
- compositions and methods described herein relate to a compound of fo substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein is C or O, n is 0, and R 23 is C 1 -C 4 alkyl, wherein the C 1 -C 4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl.
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from: .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein is C or O, n is 1, and R 23 is C 1 -C 4 alkyl, wherein the C 1 -C 4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl.
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from: .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein is C or O, n is 0 or 1, and R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C 3 -C 8 cycloalkyl, 3-8 membered heterocyclyl ring, C 6 -C 10 aryl, or 5-10 membered heteroaryl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
- the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, O, n is 1 or 2, x is 1 or 2, and R 24 is C 1 -C 4 alkyl, wherein the C 1 -C 4 alkyl is optionally substituted with halogens from F or Cl.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein , L is C or O, n is 0, 1 or 2, x is 0, 1 or 2, the atoms in the bridge can be selected from C, N, O, or S, and R 24 is C 1 -C 4 alkyl, wherein the C 1 -C 4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from .
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein , n is 0, 1 or 2, R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C 6 -C 10 aryl, or 5-10 membered heteroaryl or the said R 24 is a spiro ring
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein , L G is a single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H C1 C6 alkyl C1 C6 haloalkyl, - OR 28 , or -S(O) m or either R 25 or R27 join with R26 to form a C4-C7 cycloalkyl ring or a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen.
- compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from:
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: .
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions and methods described herein relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: ,
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: , , , , ,
- compositions and methods described herein relate to a combination comprising a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, and one or more additional active agents.
- the compositions and methods described herein relate to a combination wherein an additional active agent is an anti-cancer agent.
- the compositions and methods described herein relate to a combination wherein the additional active anti- cancer agent is a chemotherapy.
- the compositions and methods described herein relate to a pharmaceutical composition comprising a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers.
- compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, for use as a medicament.
- the compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease.
- the compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
- compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR).
- MSI-H microsatellite instability-high
- dMMR mismatch repair deficient
- compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR) that is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer.
- MSI-H microsatellite instability-high
- dMMR mismatch repair deficient
- compositions and methods described herein relate to a method of modulating WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein.
- the compositions and methods described herein relate to a method of inhibiting WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein.
- compositions and methods described herein relate to a method of treating a disorder or disease with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein.
- compositions and methods described herein relate to a method of treating cancer with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein.
- compositions and methods described herein relate to a method of treating cancer with a WRN inhibitor in a subject, comprising administering a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, wherein the cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR).
- MSI-H microsatellite instability-high
- dMMR mismatch repair deficient
- compositions and methods described herein relate to a method of treating a cancer wherein the cancer is characterized as microsatellite instability- high (MSI-H) and/or mismatch repair deficient (dMMR) and is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer.
- the compositions and methods described herein relate to a use of a compound, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
- compositions and methods described herein relate to a use of a compound or pharmaceutically acceptable salt thereof, wherein the use is for the treatment of a cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR).
- MSI-H microsatellite instability-high
- dMMR mismatch repair deficient
- the compositions and methods described herein relate to a process to manufacture a compound described herein, or a pharmaceutically acceptable salt thereof.
- BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 depicts the results of an in vivo efficacy assay of certain compounds of the present disclosure.
- MSI-h HCT116 or MSS HT-29 tumors were established in 4–5- week-old female Crl:NU(NCr)-Foxn1nu athymic nude mice (Charles River Laboratories). Mice were housed in 5 animals per cage and food and water was provided ad libitum. All procedures were performed at the Mispro Vivarium facility in New York, NY and were conducted according to the guidelines of the Mispro Institutional Animal Care and Use Committee and Eikon Therapeutics protocol. HCT116 and HT-29 cells were sourced from ATCC and cultured in McCoy’s 5a Medium (ATCC) with 10% FBS.
- TGI Tumor Growth Inhibition
- Compounds were dissolved to required concentrations in an aqueous solution of 20% 2-hydroxypropyl-beta-cyclodextrin (HP- ⁇ -CD) w/v in water and pH adjusted to 7.4.
- FIG. 13 depicts the results of an assay to validate that efficacy of WRN inhibitors is limited to MSI-h tumors. Briefly, the activity of Compound 164 was assessed in the MSS HT-29 tumor model. Very limited activity of Compound 164 was observed in the HT-29 tumor model.
- Figure 3 depicts the results of a pharmacodynamic assessment of WRN levels in tumor samples demonstrating the specific degradation of WRN protein upon treatment with Compound 164. Briefly, at the initial 2 h timepoint, moderate reduction in WRN relative vehicle control is observed, with a dose responsive decrease of approximately 20-35 %.
- the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the range 6.0-7.0, the number 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, and 7.0 are explicitly contemplated.
- the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 3 or more than 3 standard deviations, per the practice in the art.
- “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value.
- the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value.
- modulate or “modulating” refers to increasing or decreasing, e.g., modulation of the activity of an enzyme includes increasing the activity of the enzyme as well as decreasing the activity of the enzyme.
- alkyl includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and may be unsubstituted or substituted.
- C1-Cn as in “C1-Cn alkyl” is defined to include groups having 1, 2, ...., n-1 or n carbons in a linear or branched arrangement.
- C1-C6, as in “C1-C6 alkyl” is defined to include groups having 1, 2, 3, 4, 5, or 6 carbons in a linear or branched arrangement, and specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, pentyl, hexyl, and octyl.
- alkenyl refers to a non-aromatic hydrocarbon radical, straight or branched, containing at least 1 carbon to carbon double bond, and up to the maximum possible number of non aromatic carbon-carbon double bonds may be present, and may be unsubstituted or substituted.
- C2-C6 alkenyl means an alkenyl radical having 2, 3, 4, 5, or 6 carbon atoms, and up to 1, 2, 3, 4, or 5 carbon-carbon double bonds respectively.
- Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl.
- alkynyl refers to a hydrocarbon radical straight or branched, containing at least 1 carbon to carbon triple bond, and up to the maximum possible number of non- aromatic carbon-carbon triple bonds may be present, and may be unsubstituted or substituted.
- C2-C6 alkynyl means an alkynyl radical having 2 or 3 carbon atoms and 1 carbon-carbon triple bond, or having 4 or 5 carbon atoms and up to 2 carbon-carbon triple bonds, or having 6 carbon atoms and up to 3 carbon-carbon triple bonds.
- Alkynyl groups include ethynyl, propynyl and butynyl.
- heteroalkyl includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and at least 1 heteroatom within the chain or branch.
- cycloalkyl refers to a saturated or partially saturated, monocyclic or fused or spiro polycyclic, carbocycle, including, but not limited to, those containing from 3 to 9 carbons per ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like, unless otherwise specified. It includes monocyclic systems such as cyclopropyl and cyclohexyl, bicyclic systems such as decalin, and polycyclic systems such as adamantane.
- the group may be a terminal group or a bridging group.
- cycloalkenyl refers to a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond, including, but not limited to, those having from 5-10 carbon atoms per ring.
- exemplary monocyclic cycloalkenyl rings include cyclopentenyl, cyclohexenyl or cycloheptenyl.
- the cycloalkenyl group may be substituted by one or more substituent groups.
- the group may be a terminal group or a bridging group.
- heterocyclyl or “heterocyclic” refers to a mono- or poly- cyclic ring system which can be saturated or contains one or more degrees of unsaturation and contains one or more heteroatoms. Heteroatoms include N, O, and/or S, including N- oxides, sulfur oxides, and dioxides. In certain embodiments, the ring is three to ten- membered and is either saturated or has one or more degrees of unsaturation. The heterocycle may be unsubstituted or substituted, with multiple degrees of substitution being allowed.
- Such rings may be optionally fused to one or more of another “heterocyclic” ring(s), heteroaryl ring(s), aryl ring(s), or cycloalkyl ring(s).
- heterocycles include, but are not limited to, tetrahydrofuran, pyran, 1,4-dioxane, 1,3-dioxane, piperidine, piperazine, pyrrolidine, morpholine, thiomorpholine, tetrahydrothiopyran, tetrahydrothiophene, 1,3- oxathiolane, and the like.
- alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl substituents may be substituted or unsubstituted, unless specifically defined otherwise.
- aryl is intended to mean any stable monocyclic, bicyclic or polycyclic carbon ring of up to 10 atoms in each ring, wherein at least one ring is aromatic, and may be unsubstituted or substituted.
- aryl elements include phenyl, p- toluenyl (4-methylphenyl), naphthyl, tetrahydro-naphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
- the aryl substituent is bicyclic and one ring is non- aromatic, it is understood that attachment is via the aromatic ring.
- heteroaryl refers to any stable monocyclic, bicyclic or polycyclic ring of up to 10 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S.
- Bicyclic aromatic heteroaryl groups include phenyl, pyridine, pyrimidine or pyridazine rings that are (a) fused to a 6-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom; (b) fused to a 5- or 6-membered aromatic (unsaturated) heterocyclic ring having two nitrogen atoms; (c) fused to a 5-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom together with either one oxygen or one sulfur atom; or (d) fused to a 5- membered aromatic (unsaturated) heterocyclic ring having one heteroatom selected from O, N or S.
- Heteroaryl groups within the scope of this definition include but are not limited to: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridyl, pyrimidyl, pyrroly
- haloalkyl examples include fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl, chloromethyl, chlorofluoromethyl and trichloromethyl groups.
- substitution refers to a functional group as described above in which one or more bonds to a hydrogen atom contained therein are replaced by a bond to non-hydrogen or non-carbon atoms, provided that normal valencies are maintained and that the substitution results in a stable compound.
- Substituted groups also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom are replaced by one or more bonds, including double or triple bonds, to a heteroatom.
- substituent groups include the functional groups described herein, and halogens (i.e., F, Cl, Br, and I); alkyl groups, such as methyl, ethyl, n-propyl, and trifluorom ethyl; hydroxyl; alkoxy groups, such as methoxy, ethoxy, n-propoxy, and isopropoxy; aryloxy groups, such as phenoxy; arylalkyloxy.
- the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties, singly or plurally.
- independently substituted it is meant that the (two or more) substituents can be the same or different.
- substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure result.
- the compounds of the subject invention may have spontaneous tautomeric forms.
- each tautomeric form is contemplated as being included within this invention whether existing in equilibrium or predominantly in one form.
- This invention also provides isotopic variants of the compounds disclosed herein, including wherein the isotopic atom is 2 H and/or wherein the isotopic atom 13 C. Accordingly, in the compounds provided herein hydrogen can be enriched in the deuterium isotope. It is to be understood that the invention encompasses all such isotopic forms. In the compound structures depicted herein, hydrogen atoms are not shown for carbon atoms having less than four bonds to non-hydrogen atoms.
- the compounds of the present invention include all hydrates, solvates, and complexes of the compounds used by this invention. If a chiral center or another form of an isomeric center is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereomers, are intended to be covered herein.
- Compounds containing a chiral center may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well- known techniques and an individual enantiomer may be used alone.
- the compounds described in the present invention are in racemic form or as individual enantiomers.
- the compounds used in the method of the present invention may be in a salt form.
- a “salt” is a salt of the instant compounds which has been modified by making acid or base salts of the compounds.
- the salt is pharmaceutically acceptable.
- pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as phenols.
- the salts can be made using an organic or inorganic acid.
- acid salts are chlorides, bromides, sulfates, nitrates, phosphates, sulfonates, formates, tartrates, maleates, malates, citrates, benzoates, salicylates, ascorbates, and the like.
- Phenolate salts are the alkali earth metal salts, sodium, potassium or lithium.
- pharmaceutically acceptable salt in this respect, refers to the relatively non-toxic, inorganic and organic acid or base addition salts of compounds of the present invention.
- salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base or free acid form with a suitable organic or inorganic acid or base, and isolating the salt thus formed.
- Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like.
- the compounds used in the method of the present invention can be administered in admixture with suitable pharmaceutical diluents, extenders, excipients, or in carriers such as the novel programmable sustained-release multi-compartmental nanospheres (collectively referred to herein as a pharmaceutically acceptable carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
- a pharmaceutically acceptable carrier suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
- the unit will be in a form suitable for oral, nasal, rectal, topical, intravenous or direct injection or parenteral administration.
- the compounds can be administered alone or mixed with a pharmaceutically acceptable carrier.
- This carrier can be a solid or liquid, and the type of carrier is generally chosen based on the type of administration being used.
- the active agent can be co-administered in the form of a tablet or capsule, liposome, as an agglomerated powder or in a liquid form.
- suitable solid carriers include lactose, sucrose, gelatin and agar.
- Capsule or tablets can be easily formulated and can be made easy to swallow or chew; other solid forms include granules, and bulk powders. Tablets may contain suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow- inducing agents, and melting agents.
- suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules.
- Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents.
- Oral dosage forms optionally contain flavorants and coloring agents.
- Parenteral and intravenous forms may also include minerals and other materials to make them compatible with the type of injection or delivery system chosen. 2.
- compositions of Matter comprising Werner Syndrome Helicase (WRN) inhibitors and methods of using the same.
- WRN Werner Syndrome Helicase
- K and J are independently selected from C, N, O, or S;
- R 6 and R 7 are independently selected from H or halogen; wherein R 6 or R 7 can optionally join with R 5 to form a C 3 -C 8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) n is 0, 1, 2, or 3; refers to a single or a double bond
- K and J are independently selected from C, N, O, or S;
- R 6 and R 7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C 6 -C 10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R4 is selected from , wherein L and G are independently selected from C,
- compositions described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, where formula (II) is .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R5 is C1-C6 alkyl, C1-C6 alkenyl, C1- C 6 alkynyl or C 3 -C 6 cycloalkyl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 5 is C 2 alkyl.
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are independently selected from H or halogen.
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are H.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are F.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R 1 is NR 30 R 31 , R 30 and R 31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3- 8 membered heterocyclyl,
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R 1 is NR 30 R 31 , aryl, C 3 -C 7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O- (C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R 33 , -S(O 2 )R 33 ,
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is NR 30 R 31 , R 30 and R 31 are independently selected from optionally substituted C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl, or H.
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is NR 30 R 31 , R 30 and R 31 are independently selected from optionally substituted C1-C3 alkyl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is NR 30 R 31 , R 30 and R 31 are -CH 3 .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, - S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C 3 -C 8 cycloalkyl ring, a C 3 -C 8 cycloalkenyl ring, a 5- or 6- membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 5- or 6- membered heterocyclyl ring, wherein the said 5- or 6- membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, and wherein the said 5- or 6- membered heterocyclyl ring is optionally substituted with C1-C3 alkyl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is phenyl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 5- membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 6- membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6-membered heterocyclyl ring comprises carbon atoms and one or two oxygen atoms.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6-membered heterocyclyl ring comprises carbon atoms and one or two nitrogen atoms.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6-membered heterocyclyl ring comprises carbon atoms, one or two oxygen and one or two nitrogen atoms.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from: , , , , , , , ,
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-NH-R33, -S(O)R33, - S(O 2 )R 33 , -NH-S(O 2 )R 33 , -NH-C(O)-NH-R 33 , wherein R 33 is selected from C 1 -C 6 alkyl, C 2 - C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from: ,
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: , , , , , , , ,
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a C3- C8 cycloalkyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: , In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3- 8 membered cycloalkenyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is aryl, the aryl is fused with a 3- 8 membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5- 10 membered heteroaryl to form a spiro ring, or the said C 3 -C 7 cycloalkyl ring is optionally linked with a second C 3 -C 8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R 4 is a substituted phenyl group.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH,-C(O)-C 1 -C 6 alkyl, or -O-(C1-C6)alkyl-COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from , , , , , , .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is a 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is substituted with C 1 -C 4 alkoxy.
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said 4- or 7-membered heterocyclyl ring is optionally joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6-membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6- membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is a 5- or 6-membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6- membered heterocyclyl ring is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R 1 is selected from
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R1 is selected from: , , In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein the said the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 1 is .
- compositions described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, X is N, Y is C, R 2 is NR 8 R 9, and R3 is H, wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1- C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein and R3 is H.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R 11 , and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein when R2 is NR8R
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R 3 is H, wherein R 8 and R 9 are independently selected from H, C 1 -C 4 alkyl, and - C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R 2 and R 3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R 2 is NR 8 R 9 , R 8 and R 9 are independently selected from H, C 1 -C 4 alkyl, and -C(O)R 11 , wherein R 11 is a substituted 5- or 6-membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused ring .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5-membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused 4- or 5-membered heterocyclyl ring is selected from:
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R 10 , or -S(O 2 )R 10 , and R 10 is C 1 -C 6 alkyl, C 1 -C 6 alkenyl, a C 3 -C 7 cycloalkyl ring, wherein R 10 is optionally substituted with -OH, C 1 -C 3 alkoxy, an optionally substituted C 3 - C5 cycloalkyl ring, hal
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C3-C5 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C 1 -C 3 alkoxy, an optionally substituted C 3 -C 5 cycloalkyl ring, halogen, -S(O 2 )CH 3 , and/or -C(O)N(CH 3 ) 2 .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from:
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is C2-C6 alkenyl.
- the compositions described herein relate to a compound of formula (I) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R 2 is H, R 3 is - S(O 2 )R 10 , and R 10 is C 1 -C 4 alkyl, C 1 -C 4 alkenyl, a C 3 -C 5 cycloalkyl ring, a 5- or 6-membered heterocyclyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C 3 -C 5 cycloalkyl ring, halogen, -S(O 2 )CH 3 , and/or -C(O)N(CH 3 ) 2 .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is selected from: .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 3 is .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R 2 is H, R 3 is - C(O)R10, or -S(O2)R10, wherein , wherein W, P, S, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; wherein T is N or C; wherein when T is N, R10 is , and M and Z are independently selected from N or C; and wherein when T is C, R 10 wherein when N M and M Z are both single bonds, M is C(O) and
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from: ,
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 , wherein A, W, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond;
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R 10 , or -S(O 2 )R 10 , and R 10 are independently selected from H, -OH, or C1-C3 alkyl, or R12 and R13 join together to form a fused substituted or unsubstituted 5- or 6- membered heterocyclyl ring, wherein when the said 5- or 6- membered heterocyclyl ring is substituted, the substituents are selected from C 1 -C 3 alkyl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from:
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R 2 is H, R 3 is - C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, a C1- C6 aryl,
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from: .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - wherein when U is N + , R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -OH or -NR19R20; wherein when R15 is -OH, R16 is a carbonyl, R17 and R18 join together to form a fused 5- or 6- membered heterocyclyl ring, and U V is a single bond; wherein when R15 is NR19R20, U V
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 10 is selected from:
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from , wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein R23 and R24 are independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkoxy, C
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof
- R4 is selected from are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when alkyl, wherein the C 1 -C 4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C 1 -C 6 hydroxyalkyl, halogenated C 1 -C 6 alkoxy, C 3 -C 8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein or 1, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein , and R 23 is C 1 -C 4 alkyl, wherein the C 1 -C 4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl.
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from: .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein , and R 23 is C 1 -C 4 alkyl, wherein the C 1 -C 4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl.
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from: .
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein or 1, and R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C 6 -C 10 aryl, or 5-10 membered heteroaryl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is selected from
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, 1 or 2, and R24 is C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens from F or Cl.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R 4 is .
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein , 1 or 2, x is 0, 1 or 2, the atoms in the bridge can be selected from C, N, O, or S, and R 24 is C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C 1 -C 6 hydroxyalkyl, halogenated C 1 -C 6 al
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C 6 -C 10 aryl, or 5-10 membered heteroaryl or the said R 24 is a spiro ring or a fused ring;
- the compositions described herein relate to a compound of formula (I
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, -OR28, or -S(O)mR28 wherein m is 0-2, R 28 is a methyl or trihalomethyl group, or either R 25 or R 27 join with R 26 to form a C4-C7 cycloalkyl ring or a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen.
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from: ,
- the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein single or double bond;
- G and L are independently selected from C, N, O;
- R 26 and R 27 are independently selected from H, C 1 -
- compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from: , , , , , , , , , In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions described herein relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: ,
- compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
- compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: , , , , ,
- the present disclosure is directed to a compound of formula (I) or formula (II), with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof: and one or more additional active agents.
- the present disclosure is directed to compounds of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or pharmaceutically acceptable salts thereof, wherein the additional active agent is an anti-cancer agent.
- the active anti-cancer agent is a chemotherapy.
- the additional active agent is a cytotoxic agent, a cytostatic agent, a hormone treatment, or a checkpoint inhibitor.
- the additional active agent is an immunotherapy.
- the present disclosure is directed to a compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, where the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof is formulated in a composition comprising one or more pharmaceutically acceptable carriers.
- the present disclosure is directed to a compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof is for use as a medicament.
- the disease is cancer.
- the cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR).
- the cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR) is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer.
- the present disclosure is directed to a method of modulating WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof.
- the present disclosure is directed to a method of inhibiting WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof.
- the present disclosure is directed to a method of treating a disorder or disease with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof.
- the present disclosure is directed to a method of treating cancer with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof.
- the cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR).
- the cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR) is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer.
- the present disclosure is directed to a process to manufacture a compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof. 4. Examples The following Examples are presented by way of illustration, not limitation. One skilled in the art can modify the procedures set forth in the illustrative examples to arrive at the desired products. Abbreviations Used
- the mixture was heated to 100 °C and stirred for 16 hours at 100 °C under N 2 .
- the resulting mixture was cooled to 25 °C, filtered and the filter cake was washed with EtOAc (30 mL). The filtrate was concentrated in vacuo.
- Step 5 Preparation of 2-(2-(methylthio)ethoxy)acetimidamide.
- Intermediate L5 To a mixture of methyl 2-(2-(methylthio)ethoxy)acetate (1.4 g, 8.5 mmol) was added dropwise the solution of amino(methyl)aluminum chloride in toluene at 25 °C. The mixture was heated to 80 °C and stirred at 80 °C for 18 hours.
- Step 6 Preparation of ethyl 5-(benzyloxy)-2- ⁇ [2-(methylsulfanyl)ethoxy]methyl ⁇ - 6-oxo-1H-pyrimidine-4-carboxylate.
- Intermediate L6 To a mixture of EtONa (138 mg, 2.0 mmol) in EtOH (50 mL), 2-(2- (methylthio)ethoxy)acetimidamide (1.2 g crude, HCl salt) and 1,4-diethyl 2-(benzyloxy)-3- oxobutanedioate (1.2 g, 4.0 mmol) was added to a 100 mL round-bottom flask at 25 °C. The mixture was stirred at 60 °C for 3 hours.
- Step 7 Preparation of (2-((5-(benzyloxy)-4-(ethoxycarbonyl)-6-oxo-1,6- dihydropyrimidin-2-yl)methoxy)ethyl)dimethylsulfonium Intermediate L7 A mixture ethyl 5-(benzyloxy)-2- ⁇ [2-(methylsulfanyl)ethoxy]methyl ⁇ -6-oxo-1H- pyrimidine-4-carboxylate (100 mg, 0.3 mmol) and MeI (4.1 mL, 66.8 mmol) in CH 2 Cl 2 (5 mL) was stirred at 25 °C under N 2 for 8 days.
- Step 2 Preparation of tert-butyl 4-(2-(dimethylamino)-4-(2-ethoxy-2-oxoethyl)-5- ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate.
- reaction mixture was cooled to 25 °C, quenched with H2O (40 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated in vacuo.
- Step 3 Preparation of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid.
- Step 1 Preparation of tert-butyl 4-[4-( ⁇ [2-chloro-4- (trifluoromethyl)phenyl]carbamoyl ⁇ methyl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate.
- Step 2 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide.
- Step 2 Preparation of ethyl 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetate.
- Step 3 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetic acid.
- Step 2 Preparation of ⁇ 2-bromo-6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl ⁇ acetic acid.
- Step 3 Preparation of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate.
- Step 1 Preparation of 2-[2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide.
- Step 2 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
- Step 2 Preparation of ethyl 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetate.
- Intermediate S2
- Step 3 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetic acid.
- Step 2 Preparation of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide.
- Step 2 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl]acetamide.
- Step 1 Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-2-((3aR,6aS)-tetrahydro-1H- furo[3,4-c]pyrrol-5(3H)-yl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate.
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide.
- the mixture was heated to 50 °C and stirred at 50 °C for 2 hours under N 2 .
- the mixture was cooled to 25 °C.
- the mixture was quenched with H 2 O (5 mL) and extracted with EtOAc (3 x 10 mL).
- the combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo.
- Step 2 Preparation of 2-[2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide.
- the reaction vessel was fitted with an addition funnel, and the reaction mixture containing above reaction mixture was transferred to the addition funnel, rinsing with CH2Cl2 (3 x 2 mL). The reaction mixture was allowed to stir at -10 C. After 1.5 hours, the reaction mixture was diluted with 1:1 H 2 O : saturated aqueous NH 4 Cl (100 mL) and the phases were partitioned. The organic layer was washed with water (1 x 100 mL), and the combined aqueous layers were further extracted with 3:1 CHCl3 : i-PrOH (3 x 200 mL).
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3- dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Compound 7 The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(chroman-8-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane.
- Compound 8 The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-5,8-dihydro-6H-pyrano[3,4-b]pyridine.
- Compound 9 The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 8-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3,4-dihydro-1H-pyrano[4,3-c]pyridine.
- Compound 16 The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2H-1,3-benzodioxol-4-ylboronic acid.
- Compound 17 The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2,3-dihydrobenzofuran-4-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- Compound 18 The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2,3-dihydrobenzofuran-7-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- the title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2,3-dihydrofuro[2,3-b]pyridine.
- Step 2 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(1-methyl-1H-pyrazolo[3,4-c]pyridin-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
- the mixture was heated to 130 °C and stirred at 130 °C for 16 hours under N2.
- the mixture was cooled to 25 °C, diluted with H2O (50 mL) and extracted with EtOAc (3 x 30 mL).
- the combined organic layers were washed with H 2 O (30 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated in vacuo.
- Step 3 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-1H-pyrazolo[3,4- c]pyridin-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,4-oxazepan-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-oxa-6- azaspiro[3.3]heptan-6-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Step 1 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -5-ethyl-7-oxo-2- ⁇ 5H,6H,7H-pyrazolo[1,5-a]pyrimidin-4-yl ⁇ - [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
- Step 2 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2- ⁇ 5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2- ⁇ 5H,6H,7H- pyrazolo[1,5-a]pyrimidin-4-yl ⁇ -[1,2,4]triazolo[1,5-a]pyrimidin-4-yl ⁇ acetamide.
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(4-(3-hydroxy-4-oxo-4,6,7,9-tetrahydropyrimido[2,1- c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide.
- Compound 64 The title compound can be prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with 5-(benzyloxy)thiazole-4-carboxylic acid.
- EXAMPLE 65 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(4-hydroxyisothiazole-3-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Compound 65 The title compound can be prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with 4-(benzyloxy)isothiazole-3-carboxylic acid.
- Step 2 Preparation of 4-(piperazin-1-yl)pyrimidine.
- Compound 66.2 A solution of tert-butyl 4-(pyrimidin-4-yl)piperazine-1-carboxylate (820 mg, 3.1 mmol) in dioxane (3 mL, 0.023 mmol) was treated with HCl (4 M in dioxane, 20 mL) solution at 25 °C for 3 hours. The mixture was concentrated in vacuo to afford (500 mg, crude) of 4-(piperazin-1-yl)pyrimidine as a colorless oil. The crude product was used directly in the next step without further purification.
- Step 3 Preparation of methyl 3-oxo-2-[4-(pyrimidin-4-yl)piperazin-1- yl]pentanoate.
- Compound 66.3 A solution of 4-(piperazin-1-yl)pyrimidine hydrochloride (500 mg, 2.5 mmol) and methyl 2-chloro-3-oxopentanoate (451 mg, 2.7 mmol) in MeCN (10 mL) was treated with K2CO3 (1.03 g, 7.5 mmol) in one portion at 25 °C under N2. The mixture was heated to 60 °C and stirred at 60 °C for 16 hours under N 2 .
- Step 4 Preparation of 2-bromo-5-ethyl-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyrimidin-7-one.
- Compound 66.4 A solution of methyl 3-oxo-2-[4-(pyrimidin-4-yl)piperazin-1-yl]pentanoate (340 mg, 1.2 mmol) and 5-bromo-2H-1,2,4-triazol-3-amine (190 mg, 1.2 mmol) in EtOH (3 mL) was treated with H 3 PO 4 (114 mg, 1.2 mmol) in one portion at 25 °C under N 2 .
- Step 5 Preparation of 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(pyrimidin-4- yl)piperazin-1-yl]-4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one.
- Step 6 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide.
- Step 3 Preparation of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
- the title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4-(trifluoromethyl)-2- oxabicyclo[2.1.1]hexan-1-amine.
- Step 1 Preparation of 2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one.
- Step 3 Preparation of 2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide.
- Step 4 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-methyl-4-(trifluoromethyl)phenyl)acetamide.
- Step 5 Preparation of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-methyl-4-(trifluoromethyl)phenyl)acetamide.
- Step 2 Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(3-hydroxy-4- oxo-4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
- Step 1 Preparation of 2-[2-(2H-1,3-benzodioxol-5-yl)-6- ⁇ 4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl ⁇ -5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
- the mixture was heated to 100 °C and stirred at 100 °C for 16 hours under N 2 .
- the mixture was cooled to 25 °C, quenched with H 2 O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over Na 2 SO 4 .
- Step 2 Preparation of 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
- Compound 100 The title compound was prepared using similar procedure as Example 96, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2-(2,3-dihydro- 1-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- Step 1 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
- Compound 104.1
- Step 1 Preparation of tert-butyl 4-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl- 7-oxo-4-( ⁇ [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl ⁇ methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate.
- Step 2 Preparation of 2-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
- Step 3 Preparation of 2- ⁇ 6-[4-(2-cyclopropanesulfonamidobenzoyl)piperazin-1- yl]-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl ⁇ -N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-6-(4-(5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- the title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 1-(tert-butoxycarbonyl)-1,2,3,4-tetrahydro-1,7-naphthyridine-8-carboxylic acid.
- the title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 1-(tert-butoxycarbonyl)-1,2,3,4-tetrahydroimidazo[1,5-a]pyrimidine-8- carboxylic acid.
- the title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 4-(tert-butoxycarbonyl)-1-methyl-4,5,6,7-tetrahydro-1H-pyrazolo[4,3- b]pyridine-3-carboxylic acid.
- the title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 4-(tert-butoxycarbonyl)-4,5,6,7-tetrahydroisothiazolo[4,5-b]pyridine-3- carboxylic acid.
- the title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 4-(tert-butoxycarbonyl)-4,5,6,7-tetrahydroisoxazolo[4,5-b]pyridine-3-carboxylic acid.
- the title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 1-methyl-7- (trifluoromethyl)-1H-benzo[d]imidazol-4-amine.
- Step 2 Preparation of tert-butyl (1-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate.
- Compound 124.2 The title compound can be prepared using similar procedure as Intermediate E, replacing tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)piperazine-1-carboxylate (Intermediate C) with methyl 2-(3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)-3- oxopentanoate.
- Step 3 Preparation of tert-butyl (1-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate.
- Step 4 Preparation of tert-butyl (1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate.
- Step 5 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(3-(methylamino)azetidin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide.
- Step 6 Preparation of 5-(benzyloxy)-N-(1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)-N,6- dimethylpyrimidine-4-carboxamide.
- Step 7 Preparation of N-(1-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)azetidin-3-yl)-5-hydroxy-N,6-dimethylpyrimidine-4-carboxamide.
- the title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl (1R,6R)-3,7- diazabicyclo[4.2.0]octane-7-carboxylate.
- the title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl (1S,5R)-3,6- diazabicyclo[3.2.0]heptane-6-carboxylate.
- Compound 128 The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl 5,6-dihydroimidazo[1,5- a]pyrazine-7(8H)-carboxylate.
- the title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with 1-(5,6,7,8-tetrahydroimidazo[1,5- a]pyrazin-3-yl)ethan-1-one.
- the title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with 3-(5-methoxypyrimidin-4-yl)- 5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine.
- Step 1 Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-1,1-difluoro-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate.
- Compound 131.1 The title compound can be prepared using similar procedure as Intermediate J, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-iodoacetamide (Intermediate D) with N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoro-2-iodoacetamide.
- Step 2 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-2,2-difluoroacetamide.
- Step 3 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoroacetamide.
- Step 4 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoroacetamide.
- Compound 132 The title compound can be prepared using similar procedure as Compound 131, replacing 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (isochroman-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- the title compound can be prepared using similar procedure as Compound 131, replacing 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- the title compound can be prepared using similar procedure as Compound 25, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (Intermediate H) with tert-butyl 4-(2-bromo-4-(2-((2- chloro-4-(trifluoromethyl)phenyl)amino)-1,1-difluoro-2-oxoethyl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate X).
- the title compound can be prepared using similar procedure as Compound 131, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)-1-methyl-1H-pyrazole-4-carboxylic acid.
- Step 1 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
- Step 2 Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
- Step 3 Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
- Compound 137 The title compound can be prepared using similar procedure as Compound 136, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (2,3-dihydrobenzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- the title compound can be prepared using similar procedure as Compound 136, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (isochroman-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.
- the title compound can be prepared using similar procedure as Compound 134, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)-2,2-difluoroacetamide with 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1- yl)acet
- EXAMPLE 140 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(3-hydroxypicolinoyl)piperazin-1- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
- the title compound can be prepared using similar procedure as Compound 139, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)picolinic acid.
- the title compound can be prepared using similar procedure as Compound 131, replacing tert-butyl 4-(2-bromo-4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-1,1- difluoro-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (Intermediate X) with tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- ((trifluoromethyl)thio)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Z).
- Compound 148 The title compound can be prepared using similar procedure as Compound 139, replacing tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Y) with tert-butyl 4-(2-bromo-4- (2-((2-chloro-4-((trifluoromethyl)thio)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Z).
- EXAMPLE 150 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(4- ((trifluoromethyl)thio)phenyl)acetamide.
- Compound 150 The title compound can be prepared using similar procedure as Compound 139, replacing tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Y) with tert-butyl 4-(2-bromo-5- ethyl-7-oxo-4-(2-oxo-2-((4-((trifluoromethyl)thio)phenyl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate AA).
- the title compound can be prepared using similar procedure as Compound 145, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)-1-methyl-1H-pyrazole-4-carboxylic acid.
- Step 4 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
- Step 5 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro- 8H-imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Step 5 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
- the resulting mixture was stirred at 90 °C for 2 hours under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to 25 °C. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 5 mL). The filtrate was concentrated under reduced pressure.
- Step 6 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(6,7-dihydro- 4H-pyrazolo[5,1-c][1,4]oxazin-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Step 2 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -5-ethyl-2-(2-methyl-1,3-benzoxazol-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
- Step 3 Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylbenzo[d]oxazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
- Compound 155 Compound 155
- the crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C 18 5 ⁇ m, 30*150mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 31% B to 50%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.33) to afford (13.9 mg, 75% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2- ⁇ 5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-1,3-benzoxazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl ⁇ acetamide as a white solid.
- Step 1 Preparation of 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3-benzothiazole.
- Compound 156.1 To a stirred solution of 4-bromo-2-methyl-1,3-benzothiazole (600 mg, 2.6 mmol) and bis(pinacolato)diboron (1.3 g, 5.3 mmol) in DMF (10 mL) were added KOAc (0.8 g, 7.9 mmol) and Pd(dppf)Cl 2 .CH 2 Cl 2 (214 mg, 0.3 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred overnight at 80 °C under nitrogen atmosphere.
- Step 2 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2- ⁇ 5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-1,3-benzothiazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl ⁇ acetamide.
- Compound 156
- Step 1 Preparation of 5-bromo-2-(but-3-yn-1-yloxy)pyrimidine.
- Compound 157.1 A solution of NaH (1.2 g, 29.0 mmol, 60%) and 3-butyn-1-ol (2.0 g, 29.0 mmol) in THF (10 mL) was stirred for 30 min at 0 °C. To the above mixture was added 5-bromo-2- chloropyrimidine (4.0 g, 20.7 mmol) dropwise over 20 min at 0 °C. The resulting mixture was stirred overnight at room temperature. The reaction was quenched with water (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 20 mL).
- Step 3 Preparation of 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H,3H- furo[2,3-b]pyridine.
- Compound 157.3 To a stirred mixture of 5-bromo-2H,3H-furo[2,3-b]pyridine (165 mg, 0.8 mmol) and bis(pinacolato)diboron (251 mg, 0.9 mmol) in dioxane (2 mL) were added KOAc (243 mg, 2.5 mmol) and Pd(dppf)Cl2CH2Cl2 (65 mg, 0.08 mmol) in portions at 25 °C.
- Step 4 Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrofuro[2,3-b]pyridin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
- Step 5 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2- ⁇ 2H,3H-furo[2,3-b]pyridin-5-yl ⁇ -6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide.
- Step 2 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-7-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
- Compound 158 A mixture of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl ⁇ -2-(2,3-dihydro-1-benzofuran-7-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (125 mg, 0.2 mmol,
- Desired product could be detected by LCMS.
- the resulting mixture was concentrated under reduced pressure.
- the crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18 5 ⁇ m, 30*150mm; Mobile Phase A: Water(10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 34% B to 90% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 6.80/9.98) to afford (9.7 mg, 8.6% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-7-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]
- Step 2 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -2-(3,4-dihydro-2H-1-benzopyran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide
- Compound 159.2 To a stirred solution of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2-
- Step 3 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 2H-1-benzopyran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 159
- Step 1 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -2-(3,4-dihydro-2H-1-benzopyran-8-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
- the reaction mixture was heated to 80 °C and stirred at 80 °C for 24 hours under nitrogen atmosphere.
- the mixture was allowed to cool down to room temperature, diluted with water (10 mL) and extracted with EtOAc (3 x 10 mL).
- the combined organic layers were washed with brine (10 mL) and dried over anhydrous Na 2 SO 4 . After filtration, the filtrate was concentrated under reduced pressure.
- Step 4 Preparation of 8-bromo-1H,3H,4H-pyrano[4,3-c]pyridine.
- Compound 161.4 To a solution of 2-[5-bromo-4-(hydroxymethyl)pyridin-3-yl]ethanol (30 mg, 0.1 mmol) in ACN (5 mL) was added TsOH (44 mg, 0.2 mmol) at 25 °C and the mixture was heated to 110 °C and stirred at 110 °C for 36 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under vacuum.
- Step 6 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -5-ethyl-7-oxo-2- ⁇ 1H,3H,4H-pyrano[4,3-c]pyridin-8-yl ⁇ - [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoro methyl) phenyl]acetamide.
- the resulting mixture was heated to 80 °C and stirred at 80 °C for additional 1h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25°C. The resulting mixture was filtered, the filter cake was washed with EA (3 x 5 mL). The filtrate was concentrated under reduced pressure.
- Step 7 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2- ⁇ 5-ethyl-6-[4-(5- hydroxy-6-methylpyri midine-4-carbonyl)piperazin-1-yl]-7-oxo-2- ⁇ 1H,3H,4H-pyrano[4,3- c]pyridin-8-yl ⁇ -[1,2,4]triazolo[1,5-a]pyri midin-4-yl ⁇ acetamide.
- Step 1 Preparation of methyl 5-bromo-2-(bromomethyl)pyridine-3-carboxylate.
- Compound 162.1 A solution of methyl 5-bromo-2-methylpyridine-3-carboxylate (4.0 g, 17.3 mmol), AIBN (0.3 g, 1.7 mmol) and NBS (3.0 g, 17.3 mmol) in CCl4 (40 mL) was stirred overnight at 80 °C. The resulting mixture was concentrated under reduced pressure. To the above mixture was added ethylbis(propan-2-yl)amine (1.8 mL, 13.9 mmol) and diethyl phosphonate (1.4 mL, 10.1 mmol) in THF (50 mL) at room temperature.
- Step 2 Preparation of methyl 3-bromo-5-hydroxy-8H-pyrano[3,4-b]pyridine-6- carboxylate.
- Compound 162.2 NaH (807 mg, 33.6 mmol) was added to a mixture of methyl 5-bromo-2- (bromomethyl)pyridine-3-carboxylate (5.2 g, 16.8 mmol) and methyl 2-hydroxyacetate (1.5 g, 16.8 mmol) in DMF (50 mL) at 0 °C. The mixture was stirred at 25 °C for 1 h. The reaction was quenched by the addition of sat. NH4Cl (aq.) (100 mL) at room temperature.
- Step 7 Preparation of 5H,6H,8H-pyrano[3,4-b]pyridin-3-ylboronic acid.
- Compound 162.7 To a stirred solution of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridine (330 mg, 1.5 mmol) and Pd(dppf)Cl 2 CH 2 Cl 2 (135 mg, 0.2 mmol) in 2-methyloxolane (3 mL) were added 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (782 mg, 3.0 mmol), MeOH (1 mL) and potassium 2,2-dimethylpropanoate (324 mg, 2.3 mmol) at room temperature under nitrogen atmosphere.
- Step 8 Preparation of 2-(6- ⁇ 4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl ⁇ -5-ethyl-7-oxo-2- ⁇ 5H,6H,8H-pyrano[3,4-b]pyridin-3-yl ⁇ - [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
- Step 9 Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2- ⁇ 5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2- ⁇ 5H,6H,8H-pyrano[3,4- b]pyridin-3-yl ⁇ -[1,2,4]triazolo[1,5-a]pyrimidin-4-yl ⁇ acetamide.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The presently disclosed subject matter relates to compositions comprising Werner Syndrome Helicase (WRN) inhibitors and methods of using the same.
Description
COMPOSITIONS COMPRISING WERNER SYNDROME HELICASE INHIBITORS AND METHODS OF USING THE SAME
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of priority to U.S. Provisional Patent Application Serial No. 63/535,307, filed on August 29, 2023, and U.S. Provisional Patent Application Serial No. 63/665,217, filed on June 27, 2024, the contents of which are incorporated herein by reference in their entireties.
TECHNICAL FIELD
The subject matter described herein relates to compositions comprising Werner Syndrome Helicase (WRN, also known as Werner Syndrome RecQ Helicase or Werner Syndrome ATP-Dependent Helicase) inhibitors and methods of using the same.
BACKGROUND
The WRN gene encodes a helicase that falls within the RecQ DNA helicase subfamily and is directly involved in DNA damage repair. While WRN has previously been shown to exhibit tumor suppressive activity, an activity that correlates well with its activity in DNA damage repair, certain cancer cells have been shown to be dependent on the presence of WRN. For example, certain cancers associated with deficient DNA mismatch repair (dMMR), which often exhibit high levels of microsatellite instability (microsatellite instability-high or MSI-H cancers), depend on WRN activity for continued survival. In view of WRN’ s essential role in such cancers, there remains a need in the art to identify WRN inhibitors for use in the treatment of such dMMR and/or MSI-H cancers.
SUMMARY OF THE INVENTION
In certain aspects, the compositions and methods described herein relate to compositions comprising WRN helicase inhibitors and methods of their use in treating disease, e.g., cancer.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof
K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein: (A) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl
ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4--membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-
membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring,
wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; (B) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra and Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, - NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the
substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C2-C6 alkenyl, a C3-C7 cycloalkyl ring, C1-C6 hydroxyalkyl, , wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or
X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
wherein W, U, V, and Z are independently selected from H, C, N, S, or O and refers to a single or a double bond; wherein when
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond;
wherein when U is C, R15 is -NR19R20,U V is a double-bond and R16, R17, R18, are independently selected from H, -CH3, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring, or
L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl;
wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H, -OCF2Cl, or -S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen, wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -
OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen;
wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl, the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is optionally substituted with one or more R36, or the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or said the aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is fused with a second 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring or a C3-C8 cycloalkenyl ring, wherein R36 is independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, 3-8 membered heterocyclyl, -O-(C1-C6)alkyl-COOH, -C(O)-R33, -C(O)- NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; and either (B) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused
C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6
alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
, wherein W, U, V, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; ; wherein when
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20 U V is a double-bond and R16, R17, and R18 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and - S(O)R21R22, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted
or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring;
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring;
wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen, and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8
cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 3-8 membered heterocyclyl ring, wherein when the 3-8 membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein R8 and R9 are independently selected from H, C1-C6 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein R10 is selected from C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3- C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C3-C8 cycloalkyl, halogen, a 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or R10 is fused with a second C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C6-C10 aryl, a 3-8 membered heterocyclyl, or a 5-10 membered heteroaryl; and either (B) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and
wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom,
wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; or
ĨC) R4 is selected from
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring;
wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen, and wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring. 1. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S;
R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (B) R4 is selected from
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein R23 and R24 are independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, a bridged or unbridged C3-C8 cycloalkyl, a 3-8 membered heterocyclyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
single or double bond; R25, R29, and R35 are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 haloalkenyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, -OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring;
and either (B) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered
heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or
the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring1; or (C) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10
membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
, wherein W, U, V, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20 U V is a double-bond and R16, R17, and R18 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and -S(O)R21R22, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring, and wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl, and wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, where formula (II) is
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R5 is C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl or C3-C6 cycloalkyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R5 is C2 alkyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are independently selected from H or halogen. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are H. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are F. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein:
R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3- 8 membered heterocyclyl, the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is optionally substituted with one or more R36, or the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or said the aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is fused with a second 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring or a C3-C8 cycloalkenyl ring, wherein R36 is independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, 3-8 membered heterocyclyl, -O-(C1-C6)alkyl- COOH, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, - NH-C(O)-NH-R33, and wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1- C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring;
wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O- (C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3- C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3- C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group;
wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl-COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6- membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6- membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom,
wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6- membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl, or H. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C3 alkyl.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are -CH3. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-R33, - C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 5- or 6- membered heterocyclyl ring, wherein the said 5- or 6- membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, and wherein the said 5- or 6- membered heterocyclyl ring is optionally substituted with C1-C3 alkyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 5-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6- membered heterocyclyl ring comprises carbon atoms and one or two oxygen atoms. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6- membered heterocyclyl ring comprises carbon atoms and one or two nitrogen atoms. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6- membered heterocyclyl ring comprises carbon atoms, one or two oxygen and one or two nitrogen atoms. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from:
,
, , , , , , , , , ,
, , , ,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-NH-R33, - S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, wherein R33 is selected from C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 5-10 membered heteroaryl ring, wherein the said aryl and the said 5-10 membered heteroaryl ring are optionally substituted by one or more halogen, -OH, =O, - C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from:
,
,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a C3-C8 cycloalkyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3-8 membered cycloalkenyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is . In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents
disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3-8 membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein R1 is aryl, the aryl is fused with a 3-8 membered heterocyclyl ring, wherein the said aryl and the said 3-8 membered heterocyclyl ring are optionally substituted by one or more halogen, -OH, =O, - C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, 6-10 membered aryl, 5-10 membered heteroaryl, or a second 3-8 membered heterocyclyl, or the said C3-C8 cycloalkyl, C3-C8 cycloalkenyl, 6-10 membered aryl, 5-10 membered heteroaryl, or the second 3-8 membered heterocyclyl is linked with the said aryl and the said first 3-8 membered heterocyclyl ring to form a spiro ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from:
,
,
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a fully saturated C3-C7 cycloalkyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH,- C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl-COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3- C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from
,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is substituted with C1-C4 alkoxy. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said 4- or 7- membered heterocyclyl ring is optionally joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6- membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6- membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-
membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom; wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein R1 is selected from
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a pyridinyl
ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the pyridinyl ring, the second 4-, 5-, or 6-membered heterocyclyl ring, are optionally substituted by one or more H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein R1 is selected from:
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein the said the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 3-8 membered heterocyclyl ring, wherein when the 3-8 membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1- C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring;
wherein R8 and R9 are independently selected from H, C1-C6 alkyl, and - C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein R10 is selected from C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C3-C8 cycloalkyl, halogen, a 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or R10 is fused with a second C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C6-C10 aryl, a 3-8 membered heterocyclyl, or a 5-10 membered heteroaryl In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra and Rb are independently selected from H, - OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6- membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8
cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with - C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5-membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C2-C6 alkenyl, a C3-C7 cycloalkyl ring, C1-C6 hydroxyalkyl, , wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
wherein W, U, V, and Z are independently selected from H, C, N, S, or O and refers to a single or a double bond; wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20, U V is a double-bond and R16, R17, R18 are independently selected from H, -CH3, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently
selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring In certain embodiments, the compositions and methods described herein relate to a compound of formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb, R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or - C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
selected from
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H, wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6- membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, X is N, Y is
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein
when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H, wherein R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused ring
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5- membered heterocyclyl ring, wherein when the 5-membered heterocyclyl ring is substituted,
the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused 4- or 5-membered heterocyclyl ring is selected from:
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C1-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or -C(O)N(CH3)2. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C3-C5 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C3- C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or -C(O)N(CH3)2. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from:
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein R10 is C2-C6 alkenyl. In certain embodiments the compositions and methods described herein relate to a compound of fo nts disclosed
herein, or a pharmaceutically acceptable salt thereof, wherein R10
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -S(O2)R10, and R10 is C1-C4 alkyl, C1-C4 alkenyl, a C3-C5 cycloalkyl ring, a 5- or 6-membered heterocyclyl ring, wherein R10 is optionally substituted with -OH, C1- C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or - C(O)N(CH3)2. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n=2, X is C, Y is N, R2 is H, R3 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is . In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, wherein
, wherein W, P, S, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; w
wherein when T is N, R10 is
, and M and Z are independently selected from N or C; and wherein when T is C, R10 i
wherein when N M and M Z are both single bonds, M is C(O) and Z is N or C substituted with H or C1-C3 alkyl; and when one of N M and M Z is a single-bond and one is a double-bond, M and Z are independently selected from C or N, optionally substituted with H or C1-C3 alkyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from: ,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is
, wherein A, W, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; R12, R13, and R14 are independently selected from H, -OH, =O, C1-C3 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, a C3-C8 cycloalkyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring,, or R12 and R13 join together to form a fused substituted or unsubstituted 5- or 6- membered heterocyclyl ring, wherein when the said 5- or 6- membered heterocyclyl ring is substituted, the substituents
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10
are independently selected from H, -OH, or C1-C3 alkyl, or R12 and R13 join together to form a fused substituted or unsubstituted 5- or 6- membered heterocyclyl ring, wherein when the said 5- or 6- membered heterocyclyl ring is substituted, the substituents are selected from C1-C3 alkyl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, a C1-C6 aryl, or a 5- or 6-membered heterocyclyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, and/or -C(O)N(CH3)2, wherein when R10 is a C3-C7 cycloalkyl ring, a C1-C6 aryl, a 5-10 membered heteroaryl ring or a 5- or 6-membered heterocyclyl ring R10 is optionally
fused with a second C3-C7 cycloalkyl ring, C1-C6 aryl, 5-10 membered heteroaryl, or 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -OH or -NR19R20; wherein when R15 is -OH, R16 is a carbonyl, R17 and R18 join together to form a fused 5- or 6- membered heterocyclyl ring, and U V is a single bond; wherein when R15 is NR19R20, U V is a double-bond and R16, R17, R18, R19, and R20 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from: ,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein R23 and R24 are independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, a
bridged or unbridged C3-C8 cycloalkyl, a 3-8 membered heterocyclyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
single or double bond; R25, R29, and R35 are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 haloalkenyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, -OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R4 is selected from
independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl;
wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
is C or O, n is 0 or 1, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl. In certain embodiments the compositions and methods described herein relate to a compound of fo substituents
disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
is C or O, n is 0, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
is C or O, n is 1, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
is C or O, n is 0 or 1, and R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof,
O, n is 1 or 2, x is 1 or 2, and R24 is C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens from F or Cl. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, L is C or O, n is 0, 1 or 2, x is 0, 1 or 2, the atoms in the bridge can be selected from C, N, O, or S, and R24 is C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, n is 0, 1 or 2, R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, L G is a single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H C1 C6 alkyl C1 C6 haloalkyl, - OR28, or -S(O)m or either R25
or R27 join with R26 to form a C4-C7 cycloalkyl ring or a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, L G is a single or double bond; G and L are independently selected from C, N, O; R25, R29, and R35, are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, - OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
,
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
, ,
. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: ,
, , ,
. , In certain embodiments, the compositions and methods described herein relate to a combination comprising a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, and one or more additional active agents. In certain embodiments, the compositions and methods described herein relate to a combination wherein an additional active agent is an anti-cancer agent. In certain embodiments, the compositions and methods described herein relate to a combination wherein the additional active anti- cancer agent is a chemotherapy. In certain embodiments, the compositions and methods described herein relate to a pharmaceutical composition comprising a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, for use as a medicament.
In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer. In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR). In certain embodiments, the compositions and methods described herein relate to a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, for use in the treatment of a cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR) that is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer. In certain embodiments, the compositions and methods described herein relate to a method of modulating WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein. In certain embodiments, the compositions and methods described herein relate to a method of inhibiting WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein. In certain embodiments, the compositions and methods described herein relate to a method of treating a disorder or disease with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein. In certain embodiments, the compositions and methods described herein relate to a method of treating cancer with a WRN inhibitor in a subject, comprising administering to
the subject a therapeutically effective amount of the compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, as described herein. In certain embodiments, the compositions and methods described herein relate to a method of treating cancer with a WRN inhibitor in a subject, comprising administering a compound of formula (I) or (II) as described herein, or a pharmaceutically acceptable salt thereof, wherein the cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR). In certain embodiments, the compositions and methods described herein relate to a method of treating a cancer wherein the cancer is characterized as microsatellite instability- high (MSI-H) and/or mismatch repair deficient (dMMR) and is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer. In certain embodiments, the compositions and methods described herein relate to a use of a compound, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer. In certain embodiments, the compositions and methods described herein relate to a use of a compound or pharmaceutically acceptable salt thereof, wherein the use is for the treatment of a cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR). In certain embodiments, the compositions and methods described herein relate to a process to manufacture a compound described herein, or a pharmaceutically acceptable salt thereof. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 depicts the results of an in vivo efficacy assay of certain compounds of the present disclosure. Briefly, MSI-h HCT116 or MSS HT-29 tumors were established in 4–5- week-old female Crl:NU(NCr)-Foxn1nu athymic nude mice (Charles River Laboratories). Mice were housed in 5 animals per cage and food and water was provided ad libitum. All procedures were performed at the Mispro Vivarium facility in New York, NY and were conducted according to the guidelines of the Mispro Institutional Animal Care and Use Committee and Eikon Therapeutics protocol. HCT116 and HT-29 cells were sourced from ATCC and cultured in McCoy’s 5a Medium (ATCC) with 10% FBS. To establish the tumors, mice were injected with 2 x 106 HCT116 or HT-29 cells in 50% Geltrex/50% HBSS
in the right flank. Animal body weights and tumor volumes were measured twice weekly throughout the study and recorded in StudyLog. Tumor volume was calculated using the formula TV = 0.5*length*width2. Tumor Growth Inhibition (TGI) was calculated using the formula TGI = (1-((TD25- Tinitial)/(CD25-Cinitial)))*100, where T is the Test group TV and C is the Control group TV at Day 25 vs the initial TV at study commencement. Percent tumor regression was calculated using the formula % regression = 100- ((Tfinal/Tinitial)x100), where Tfinal is the latest TV measurement and Tinitial is the initial TV at study commencement. After HCT116 tumors grew to an average volume of 150-200 mm3 (7 days post-implantation), mice were randomized into treatment groups (n=8 mice/group) and received either test compounds or vehicle control daily via oral gavage at 10mL/kg body weight. Compounds were dissolved to required concentrations in an aqueous solution of 20% 2-hydroxypropyl-beta-cyclodextrin (HP-β-CD) w/v in water and pH adjusted to 7.4. Compound 13 was administered at 15, 30 or 90 mg/kg while compounds 164 and 169 were administered at 5, 15 or 30 mg/kg. Figure 2 depicts the results of an assay to validate that efficacy of WRN inhibitors is limited to MSI-h tumors. Briefly, the activity of Compound 164 was assessed in the MSS HT-29 tumor model. Very limited activity of Compound 164 was observed in the HT-29 tumor model. Figure 3 depicts the results of a pharmacodynamic assessment of WRN levels in tumor samples demonstrating the specific degradation of WRN protein upon treatment with Compound 164. Briefly, at the initial 2 h timepoint, moderate reduction in WRN relative vehicle control is observed, with a dose responsive decrease of approximately 20-35 %. At 8 h post final dose, a further reduction in WRN protein in observed, with a 40% reduction seen following 5 mg/kg Compound 164 and 50% reduction seen for both 15 and 30 mg/kg of Compound 164. A rebound in overall WRN levels is seen at the final 48 h timepoint, with WRN levels in tumors treated with 5 mg/kg Compound 164 being equivalent to vehicle control. In the higher doses, an around 20 % reduction in WRN level relative to vehicle was observed at the 48 h timepoint. Plasma concentrations of Compound 164 demonstrated clear dose dependent increase from 5 to 30 mg/kg and showed expected decrease over the time- course of the study.
DETAILED DESCRIPTION The presently disclosed subject matter relates to compositions comprising WRN helicase inhibitors and methods of their use in treating cancer. For purposes of clarity of disclosure and not by way of limitation, the detailed description is divided into the following subsections: 1. Definitions 2. Compositions of Matter 3. Methods of Use 4. Examples 1. Definitions Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In case of conflict, the present document, including definitions, will control. Preferred methods and materials are described below, although methods and materials similar or equivalent to those described herein can be used in practice or testing of the presently disclosed subject matter. All publications, patent applications, patents and other references mentioned herein are incorporated by reference in their entirety. The materials, methods, and examples disclosed herein are illustrative only and not intended to be limiting. The terms “comprise(s),” “include(s),” “having,” “has,” “can,” “contain(s),” and variants thereof, as used herein, are intended to be open-ended transitional phrases, terms, or words that do not preclude the possibility of additional acts or structures. The singular forms “a,” “an” and “the” include plural references unless the context clearly dictates otherwise. The present disclosure also contemplates other instances “comprising,” “consisting of”, and “consisting essentially of,” the instances or elements presented herein, whether explicitly set forth or not. For the recitation of numeric ranges herein, each intervening number within the range is explicitly contemplated with the same degree of precision. For example, for the range of 6-9, the numbers 7 and 8 are contemplated in addition to 6 and 9, and for the range 6.0-7.0, the number 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, and 7.0 are explicitly contemplated. As used herein, the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which
will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 3 or more than 3 standard deviations, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. As used herein, “modulate” or “modulating” refers to increasing or decreasing, e.g., modulation of the activity of an enzyme includes increasing the activity of the enzyme as well as decreasing the activity of the enzyme. As used herein, “treat” or “treating” refers to an effort to alter the natural course of a disease, including prophylaxis of the disease, alleviation of symptoms and/or ameliorating pathology associated with the disease. As used herein, “alkyl” includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and may be unsubstituted or substituted. Thus, C1-Cn as in “C1-Cn alkyl" is defined to include groups having 1, 2, ...., n-1 or n carbons in a linear or branched arrangement. For example, C1-C6, as in “C1-C6 alkyl” is defined to include groups having 1, 2, 3, 4, 5, or 6 carbons in a linear or branched arrangement, and specifically includes methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, pentyl, hexyl, and octyl. As used herein, “alkenyl” refers to a non-aromatic hydrocarbon radical, straight or branched, containing at least 1 carbon to carbon double bond, and up to the maximum possible number of non aromatic carbon-carbon double bonds may be present, and may be unsubstituted or substituted. For example, “C2-C6 alkenyl” means an alkenyl radical having 2, 3, 4, 5, or 6 carbon atoms, and up to 1, 2, 3, 4, or 5 carbon-carbon double bonds respectively. Alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. The term “alkynyl” refers to a hydrocarbon radical straight or branched, containing at least 1 carbon to carbon triple bond, and up to the maximum possible number of non- aromatic carbon-carbon triple bonds may be present, and may be unsubstituted or substituted. Thus, “C2-C6 alkynyl” means an alkynyl radical having 2 or 3 carbon atoms and 1 carbon-carbon triple bond, or having 4 or 5 carbon atoms and up to 2 carbon-carbon triple bonds, or having 6 carbon atoms and up to 3 carbon-carbon triple bonds. Alkynyl groups include ethynyl, propynyl and butynyl.
As used herein, “heteroalkyl” includes both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms and at least 1 heteroatom within the chain or branch. As used herein, “cycloalkyl” refers to a saturated or partially saturated, monocyclic or fused or spiro polycyclic, carbocycle, including, but not limited to, those containing from 3 to 9 carbons per ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like, unless otherwise specified. It includes monocyclic systems such as cyclopropyl and cyclohexyl, bicyclic systems such as decalin, and polycyclic systems such as adamantane. The group may be a terminal group or a bridging group. As used herein, the term “cycloalkenyl” refers to a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond, including, but not limited to, those having from 5-10 carbon atoms per ring. Exemplary monocyclic cycloalkenyl rings include cyclopentenyl, cyclohexenyl or cycloheptenyl. The cycloalkenyl group may be substituted by one or more substituent groups. The group may be a terminal group or a bridging group. As used herein, the term “heterocyclyl” or “heterocyclic” refers to a mono- or poly- cyclic ring system which can be saturated or contains one or more degrees of unsaturation and contains one or more heteroatoms. Heteroatoms include N, O, and/or S, including N- oxides, sulfur oxides, and dioxides. In certain embodiments, the ring is three to ten- membered and is either saturated or has one or more degrees of unsaturation. The heterocycle may be unsubstituted or substituted, with multiple degrees of substitution being allowed. Such rings may be optionally fused to one or more of another “heterocyclic” ring(s), heteroaryl ring(s), aryl ring(s), or cycloalkyl ring(s). Examples of heterocycles include, but are not limited to, tetrahydrofuran, pyran, 1,4-dioxane, 1,3-dioxane, piperidine, piperazine, pyrrolidine, morpholine, thiomorpholine, tetrahydrothiopyran, tetrahydrothiophene, 1,3- oxathiolane, and the like. The alkyl, alkenyl, alkynyl, aryl, heteroaryl and heterocyclyl substituents may be substituted or unsubstituted, unless specifically defined otherwise. As used herein, “aryl” is intended to mean any stable monocyclic, bicyclic or polycyclic carbon ring of up to 10 atoms in each ring, wherein at least one ring is aromatic, and may be unsubstituted or substituted. Examples of such aryl elements include phenyl, p- toluenyl (4-methylphenyl), naphthyl, tetrahydro-naphthyl, indanyl, biphenyl, phenanthryl,
anthryl or acenaphthyl. In cases where the aryl substituent is bicyclic and one ring is non- aromatic, it is understood that attachment is via the aromatic ring. As used herein, the term “heteroaryl” refers to any stable monocyclic, bicyclic or polycyclic ring of up to 10 atoms in each ring, wherein at least one ring is aromatic and contains from 1 to 4 heteroatoms selected from the group consisting of O, N and S. Bicyclic aromatic heteroaryl groups include phenyl, pyridine, pyrimidine or pyridazine rings that are (a) fused to a 6-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom; (b) fused to a 5- or 6-membered aromatic (unsaturated) heterocyclic ring having two nitrogen atoms; (c) fused to a 5-membered aromatic (unsaturated) heterocyclic ring having one nitrogen atom together with either one oxygen or one sulfur atom; or (d) fused to a 5- membered aromatic (unsaturated) heterocyclic ring having one heteroatom selected from O, N or S. Heteroaryl groups within the scope of this definition include but are not limited to: benzoimidazolyl, benzofuranyl, benzofurazanyl, benzopyrazolyl, benzotriazolyl, benzothiophenyl, benzoxazolyl, carbazolyl, carbolinyl, cinnolinyl, furanyl, indolinyl, indolyl, indolazinyl, indazolyl, isobenzofuranyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, naphthpyridinyl, oxadiazolyl, oxazolyl, oxazoline, isoxazoline, oxetanyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridopyridinyl, pyridyl, pyrimidyl, pyrrolyl, quinazolinyl, quinolyl, quinoxalinyl, tetrazolyl, tetrazolopyridyl, thiadiazolyl, thiazolyl, thienyl, triazolyl, azetidinyl, aziridinyl, 1,4- dioxanyl, hexahydroazepinyl, dihydrobenzoimidazolyl, dihydrobenzofuranyl, dihydrobenzothiophenyl, dihydrobenzoxazolyl, dihydrofuranyl, dihydroimidazolyl, dihydroindolyl, dihydroisooxazolyl, dihydroisothiazolyl, dihydrooxadiazolyl, dihydrooxazolyl, dihydropyrazinyl, dihydropyrazolyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dihydroquinolinyl, dihydrotetrazolyl, dihydrothiadiazolyl, dihydrothiazolyl, dihydrothienyl, dihydrotriazolyl, dihydroazetidinyl, methylenedioxybenzoyl, tetrahydrofuranyl, tetrahydrothienyl, acridinyl, benzothiazolyl, thienyl, benzothienyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, tetra- hydroquinoline. In cases where the heteroaryl substituent is bicyclic and one ring is nonaromatic or contains no heteroatoms, it is understood that attachment is via the aromatic ring or via the heteroatom containing ring, respectively. If the heteroaryl contains nitrogen atoms, it is understood that the corresponding N-oxides thereof are also encompassed by this definition. As used herein, the term “halogen” refers to F, Cl, Br, and I.
As used herein, the term “haloalkyl” means an alkyl group that is substituted with one or more fluorine, chlorine, bromine or iodine atoms. Examples of such haloalkyl include fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl, chloromethyl, chlorofluoromethyl and trichloromethyl groups. The term “substitution,” “substituted” and “substituent” refers to a functional group as described above in which one or more bonds to a hydrogen atom contained therein are replaced by a bond to non-hydrogen or non-carbon atoms, provided that normal valencies are maintained and that the substitution results in a stable compound. Substituted groups also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom are replaced by one or more bonds, including double or triple bonds, to a heteroatom. Examples of substituent groups include the functional groups described herein, and halogens (i.e., F, Cl, Br, and I); alkyl groups, such as methyl, ethyl, n-propyl, and trifluorom ethyl; hydroxyl; alkoxy groups, such as methoxy, ethoxy, n-propoxy, and isopropoxy; aryloxy groups, such as phenoxy; arylalkyloxy. Where multiple substituent moieties are disclosed or claimed, the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties, singly or plurally. By independently substituted, it is meant that the (two or more) substituents can be the same or different. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure result. The compounds of the subject invention may have spontaneous tautomeric forms. In cases wherein compounds may exist in tautomeric forms, such as keto-enol tautomers, each tautomeric form is contemplated as being included within this invention whether existing in equilibrium or predominantly in one form. This invention also provides isotopic variants of the compounds disclosed herein, including wherein the isotopic atom is 2H and/or wherein the isotopic atom 13C. Accordingly, in the compounds provided herein hydrogen can be enriched in the deuterium isotope. It is to be understood that the invention encompasses all such isotopic forms.
In the compound structures depicted herein, hydrogen atoms are not shown for carbon atoms having less than four bonds to non-hydrogen atoms. However, it is understood that enough hydrogen atoms exist on said carbon atoms to satisfy the octet rule. Except where otherwise specified, if the structure of a compound of this invention includes an asymmetric carbon atom, it is understood that the compound occurs as a racemate, racemic mixture, and isolated single enantiomer. All such isomeric forms of these compounds are expressly included in this invention. Except where otherwise specified, each stereogenic carbon may be of the R or S configuration. It is to be understood accordingly that the isomers arising from such asymmetry (e.g., all enantiomers and diastereomers) are included within the scope of this invention, unless indicated otherwise. Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemically controlled synthesis. The compounds of the present invention include all hydrates, solvates, and complexes of the compounds used by this invention. If a chiral center or another form of an isomeric center is present in a compound of the present invention, all forms of such isomer or isomers, including enantiomers and diastereomers, are intended to be covered herein. Compounds containing a chiral center may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well- known techniques and an individual enantiomer may be used alone. The compounds described in the present invention are in racemic form or as individual enantiomers. In choosing the compounds of the present invention, one of ordinary skill in the art will recognize that the various substituents, i.e., R1, R2, etc. are to be chosen in conformity with well-known principles of chemical structure connectivity. The compounds used in the method of the present invention may be in a salt form. As used herein, a “salt” is a salt of the instant compounds which has been modified by making acid or base salts of the compounds. In the case of compounds used to treat an infection or disease caused by a pathogen, the salt is pharmaceutically acceptable. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as phenols. The salts can be made using an organic or inorganic acid. Such acid salts are chlorides, bromides, sulfates, nitrates, phosphates, sulfonates, formates, tartrates, maleates, malates, citrates, benzoates, salicylates, ascorbates, and the like. Phenolate salts are the alkali earth metal salts, sodium, potassium or lithium. The term "pharmaceutically
acceptable salt" in this respect, refers to the relatively non-toxic, inorganic and organic acid or base addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base or free acid form with a suitable organic or inorganic acid or base, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts and the like. The compounds used in the method of the present invention can be administered in admixture with suitable pharmaceutical diluents, extenders, excipients, or in carriers such as the novel programmable sustained-release multi-compartmental nanospheres (collectively referred to herein as a pharmaceutically acceptable carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices. The unit will be in a form suitable for oral, nasal, rectal, topical, intravenous or direct injection or parenteral administration. The compounds can be administered alone or mixed with a pharmaceutically acceptable carrier. This carrier can be a solid or liquid, and the type of carrier is generally chosen based on the type of administration being used. The active agent can be co-administered in the form of a tablet or capsule, liposome, as an agglomerated powder or in a liquid form. Examples of suitable solid carriers include lactose, sucrose, gelatin and agar. Capsule or tablets can be easily formulated and can be made easy to swallow or chew; other solid forms include granules, and bulk powders. Tablets may contain suitable binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, flow- inducing agents, and melting agents. Examples of suitable liquid dosage forms include solutions or suspensions in water, pharmaceutically acceptable fats and oils, alcohols or other organic solvents, including esters, emulsions, syrups or elixirs, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Such liquid dosage forms may contain, for example, suitable solvents, preservatives, emulsifying agents, suspending agents, diluents, sweeteners, thickeners, and melting agents. Oral dosage forms optionally contain flavorants and coloring agents. Parenteral and intravenous forms may also include minerals and other materials to make them compatible with the type of injection or delivery system chosen.
2. Compositions of Matter The presently disclosed subject matter relates to compositions comprising Werner Syndrome Helicase (WRN) inhibitors and methods of using the same. For example, but not by way of limitation, the present disclosure is directed to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring;
wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring,
the said 4--membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or
the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; (B) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra and Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, - NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring,
a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C2-C6 alkenyl, a C3-C7 cycloalkyl ring, C1-C6 hydroxyalkyl, , wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
wherein W, U, V, and Z are independently selected from H, C, N, S, or O and refers to a single or a double bond;
wherein when
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20,U V is a double-bond and R16, R17, R18, are independently selected from H, -CH3, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring;
L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2;
wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H, -OCF2Cl, or -S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6-
membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen; wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein
K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl, the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is optionally substituted with one or more R36, or the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or said the aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is fused with a second 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring or a C3-C8 cycloalkenyl ring, wherein R36 is independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, 3-8 membered heterocyclyl, -O-(C1-C6)alkyl-COOH, -C(O)-R33, -C(O)- NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8
cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; and either (B) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2,
X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
, wherein W, U, V, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; ; wherein when
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20 U V is a double-bond and R16, R17, and R18 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and - S(O)R21R22, R19, and R20 are independently selected
from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; or
L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl;
wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring. 2. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or
Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 3-8 membered heterocyclyl ring, wherein when the 3-8 membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein R8 and R9 are independently selected from H, C1-C6 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein R10 is selected from C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3- C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C3-C8 cycloalkyl, halogen, a 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or R10 is fused with a second C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C6-C10 aryl, a 3-8 membered heterocyclyl, or a 5-10 membered heteroaryl; and either (B) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H;
wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or
the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-
, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring; and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring or
L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl;
wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen; and wherein R1 is H,f -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R4 is selected from
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein R23 and R24 are independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, a bridged or unbridged C3-C8 cycloalkyl, a 3-8 membered heterocyclyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl;
wherein when
single or double bond; R25, R29, and R35 are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 haloalkenyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, -OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; and either (B) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl;
C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl
ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring; or
(C) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from
a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
, wherein W, U, V, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20 U V is a double-bond and R16, R17, and R18 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and -S(O)R21R22, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; and wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring. In certain embodiments, the compositions described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, K and J are independently selected from C, N, O,
or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl, wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring. 3. In certain embodiments, the compositions described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, where formula (II) is ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R5 is C1-C6 alkyl, C1-C6 alkenyl, C1- C6 alkynyl or C3-C6 cycloalkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R5 is C2 alkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are independently selected from H or halogen.
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are H. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 are F. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3- 8 membered heterocyclyl, the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is optionally substituted with one or more R36, or the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or said the aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is fused with a second 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring or a C3-C8 cycloalkenyl ring, wherein R36 is independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, 3-8 membered heterocyclyl, -O-(C1-C6)alkyl- COOH, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, - NH-C(O)-NH-R33, and wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1- C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy,
C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein: R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O- (C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3- C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl;
wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3- C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl-COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6- membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered
heterocyclyl ring to form a spiro ring, or the said 5- or 6- membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6- membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are
independently selected from optionally substituted C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl, or H. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C3 alkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is NR30R31, R30 and R31 are -CH3. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, - S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6- membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 5- or 6- membered heterocyclyl ring, wherein the said 5- or 6- membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, and
wherein the said 5- or 6- membered heterocyclyl ring is optionally substituted with C1-C3 alkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is phenyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 5- membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the phenyl is fused with a 6- membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6-membered heterocyclyl ring comprises carbon atoms and one or two oxygen atoms. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6-membered heterocyclyl ring comprises carbon atoms and one or two nitrogen atoms. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the 5- or 6-membered heterocyclyl ring comprises carbon atoms, one or two oxygen and one or two nitrogen atoms. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
, , , , , , ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl-COOH, three C1-C6 alkoxy, -C(O)-NH-R33, -S(O)R33, - S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, wherein R33 is selected from C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 5- 10 membered heteroaryl ring, wherein the said aryl and the said 5-10 membered heteroaryl ring are optionally substituted by one or more halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: , , ,
, , , , ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a C3- C8 cycloalkyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein,
or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3- 8 membered cycloalkenyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is aryl, the aryl is fused with a 3- 8 membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof wherein R1 is aryl, the aryl is fused with a 3-8 membered heterocyclyl ring, wherein the said aryl and the said 3-8 membered heterocyclyl ring are optionally substituted by one or more halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, 6-10 membered aryl, 5-10 membered heteroaryl, or a second 3-8 membered heterocyclyl, or the said C3-C8 cycloalkyl, C3-C8 cycloalkenyl, 6-10 membered aryl, 5-10 membered heteroaryl, or the second 3-8 membered heterocyclyl is linked with the said aryl and the said first 3-8 membered heterocyclyl ring to form a spiro ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from: , ,
, , , , ,
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a fully saturated C3-C7 cycloalkyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5- 10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH,-C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl-COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein,
or a pharmaceutically acceptable salt thereof, wherein R1 is selected from
, , , , , ,
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is substituted with C1-C4 alkoxy.
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is . In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said 4- or 7-membered heterocyclyl ring is optionally joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6-membered heterocyclyl ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6- membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is . In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a 5- or 6-membered heterocyclyl
ring, wherein the 5- or 6-membered heterocyclyl ring is a fully saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6- membered heterocyclyl ring is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom; wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R1 is selected from
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the pyridinyl ring, the second 4-, 5-, or 6-membered heterocyclyl ring, are optionally substituted by one or more H, halogen, - OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R1 is selected from:
,
,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein the said the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R1 is
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or
R2 and R3 join together to form an unsubstituted or substituted fused 3-8 membered heterocyclyl ring, wherein when the 3-8 membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1- C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein R8 and R9 are independently selected from H, C1-C6 alkyl, and - C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein R10 is selected from C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C3-C8 cycloalkyl, halogen, a 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or R10 is fused with a second C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C6-C10 aryl, a 3-8 membered heterocyclyl, or a 5-10 membered heteroaryl In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra and Rb are independently selected from H, - OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6- membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl
ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with - C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5-membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C2-C6 alkenyl, a C3-C7 cycloalkyl ring, C1-C6 hydroxyalkyl, , wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
wherein W, U, V, and Z are independently selected from H, C, N, S, or O and refers to a single or a double bond;
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20, U V is a double-bond and R16, R17, R18 are independently selected from H, -CH3, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring 4. In certain embodiments, the compositions described herein relate to a compound of formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb, R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or - C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring.
5. In certain embodiments, the compositions described herein relate to a compound of formula (II) with the formula further defined by the substituents disclosed herein, or a
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H, wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1- C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
and R3 is H.
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is NR8R9 and R3 is H, wherein R8 and R9 are independently selected from H, C1-C4 alkyl, and - C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 is
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 1, X and Y are C, R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused ring
.
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5-membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the said fused 4- or 5-membered heterocyclyl ring is selected from:
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C1-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C3- C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or -C(O)N(CH3)2. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C3-C5 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or -C(O)N(CH3)2. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from:
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is C2-C6 alkenyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) with the formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - S(O2)R10, and R10 is C1-C4 alkyl, C1-C4 alkenyl, a C3-C5 cycloalkyl ring, a 5- or 6-membered heterocyclyl ring, wherein R10 is optionally substituted with -OH, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, -S(O2)CH3, and/or -C(O)N(CH3)2. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n=2, X is C, Y is N, R2 is H, R3 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R3 is . In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R10, or -S(O2)R10, wherein
,
wherein W, P, S, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; wherein T is N or C; wherein when T is N, R10 is
, and M and Z are independently selected from N or C; and wherein when T is C, R10
wherein when N M and M Z are both single bonds, M is C(O) and Z is N or C substituted with H or C1-C3 alkyl; and when one of N M and M Z is a single-bond and one is a double-bond, M and Z are independently selected from C or N, optionally substituted with H or C1-C3 alkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from: ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10
, wherein A, W, T, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; R12, R13, and R14 are independently selected from H, -OH, =O, C1-C3 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, a C3-C8 cycloalkyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl
ring,, or R12 and R13 join together to form a fused substituted or unsubstituted 5- or 6- membered heterocyclyl ring, wherein when the said 5- or 6- membered heterocyclyl ring is substituted, the substituents are selected from C1-C3 alkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R10, or -S(O2)R10, and R10
are independently selected from H, -OH, or C1-C3 alkyl, or R12 and R13 join together to form a fused substituted or unsubstituted 5- or 6- membered heterocyclyl ring, wherein when the said 5- or 6- membered heterocyclyl ring is substituted, the substituents are selected from C1-C3 alkyl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is - C(O)R10, or -S(O2)R10, and R10 is C1-C6 alkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, a C1- C6 aryl, or a 5- or 6-membered heterocyclyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, and/or -C(O)N(CH3)2,
wherein when R10 is a C3-C7 cycloalkyl ring, a C1-C6 aryl, a 5-10 membered heteroaryl ring, or a 5- or 6-membered heterocyclyl ring, R10 is optionally fused with a second C3-C7 cycloalkyl ring, C1-C6 aryl, 5-10 membered heteroaryl, or 5- or 6-membered heterocyclyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein,
or a pharmaceutically acceptable salt thereof, wherein n is 2, X is C, Y is N, R2 is H, R3 is -
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -OH or -NR19R20; wherein when R15 is -OH, R16 is a carbonyl, R17 and R18 join together to form a fused 5- or 6- membered heterocyclyl ring, and U V is a single bond; wherein when R15 is NR19R20, U V is a double-bond and R16, R17, R18, R19, and R20 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R10 is selected from:
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein R23 and R24 are independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, a bridged or unbridged C3-C8 cycloalkyl, a 3-8 membered heterocyclyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
single or double bond; R25, R29, and R35 are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 haloalkenyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, -OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereofwherein R4 is selected from
are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from
F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
or 1, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, and R23 is C1-C4 alkyl, wherein the C1-C4 alkyl is alkyl is optionally substituted with halogens selected from F or Cl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
or 1, and R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl,
C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof,
1 or 2, and R24 is C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens from F or Cl. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
, 1 or 2, x is 0, 1 or 2, the atoms in the bridge can be selected from C, N, O, or S, and R24 is C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl.
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, -OR28, or -S(O)mR28 wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a C4-C7 cycloalkyl ring or a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen.
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from: ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein
single or double bond; G and L are independently selected from C, N, O; R25, R29, and R35, are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, -OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from: ,
, , , , , ,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
,
In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
, ,
. In certain embodiments, the compositions described herein relate to a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof, wherein the compound is selected from: ,
, , ,
. , 3. Methods of Use In certain embodiments, the present disclosure is directed to a compound of formula (I) or formula (II), with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof: and one or more additional active agents. For example, but not by way of limitation, the present disclosure is directed to compounds of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or pharmaceutically acceptable salts thereof, wherein the additional active agent is an anti-cancer agent. In certain embodiments, the active anti-cancer agent is a chemotherapy. In certain embodiments, the additional active agent is a cytotoxic agent, a cytostatic agent, a hormone treatment, or a checkpoint inhibitor. In certain embodiments, the additional active agent is an immunotherapy. In certain embodiments, the present disclosure is directed to a compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, where the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a
pharmaceutically acceptable salt thereof is formulated in a composition comprising one or more pharmaceutically acceptable carriers. In certain embodiments, the present disclosure is directed to a compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof is for use as a medicament. For example, but not by way of limitation, wherein the use is for the treatment of a disease. In certain embodiments, the disease is cancer. In certain embodiments, the cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR). In certain embodiments, the cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR) is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer. In certain embodiments, the present disclosure is directed to a method of modulating WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof. In certain embodiments, the present disclosure is directed to a method of inhibiting WRN activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof. In certain embodiments, the present disclosure is directed to a method of treating a disorder or disease with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof. In certain embodiments, the present disclosure is directed to a method of treating cancer with a WRN inhibitor in a subject, comprising administering to the subject a therapeutically effective amount of the compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically
acceptable salt thereof. In certain embodiments, the cancer is characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR). In certain embodiments, the cancer characterized as microsatellite instability-high (MSI-H) and/or mismatch repair deficient (dMMR) is selected from colorectal, gastric, bladder, endometrial, adrenocortical, uterine, cervical, esophageal, central nervous system, head and neck, breast, kidney, liver, lung, skin, prostate and ovarian cancer. In certain embodiments, the present disclosure is directed to a process to manufacture a compound of formula (I) or formula (II) with either formula further defined by the substituents disclosed herein, or a pharmaceutically acceptable salt thereof. 4. Examples The following Examples are presented by way of illustration, not limitation. One skilled in the art can modify the procedures set forth in the illustrative examples to arrive at the desired products. Abbreviations Used
Preparation of Intermediates Intermediate A Synthesis of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid. Intermediate A
Step 1: Preparation of 5-(benzyloxy)-4,6-dichloropyrimidine. Intermediate A1
A mixture of 4,6-dichloropyrimidin-5-ol (5.0 g, 30.3 mmol), K2CO3 (7.1 g, 51.5 mmol) and benzyl bromide (5.7 g, 33.0 mmol) in DMF (25 mL) was stirred at 25 °C for 16 hours. The mixture was quenched with H2O (25 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 12% EtOAc in petroleum ether) to afford (6.0 g, 77.6% yield) of 5-(benzyloxy)-4,6-dichloropyrimidine as a colorless solid. LCMS observed m/z = 255.0 [M+H]+. Step 2: Preparation of 5-(benzyloxy)-4-chloro-6-methylpyrimidine. Intermediate A2
A mixture of 5-(benzyloxy)-4,6-dichloropyrimidine (30.2 g, 118.3 mmol), Pd(dppf)Cl2.CH2Cl2 (4.8 g, 6.6 mmol), K3PO4 (75.3 g, 355.1 mmol) in toluene (300 mL) and H2O (90 mL) was stirred under N2 at 105 °C. To the mixture was added dropwise a solution of methylboronic acid (8.5 g, 142.0 mmol) in dioxane (400 mL). The resulting mixture was stirred at 105 °C for 18 hours. To the mixture was added a second batch solution of methylboronic acid (8.5 g, 142.0 mmol) in dioxane (300 mL) and stirred at 105 °C for an additional 8 hours under N2. To the mixture was added a third batch solution of methylboronic acid (8.5 g, 142.0 mmol) in dioxane (300 mL) and stirred at 105 °C for an additional 8 hours under N2. The resulting mixture was cooled to 25 °C, quenched with H2O (500 mL), filtered and the filter cake was washed with EtOAc (3 x 100 mL). The filtrate was extracted with EtOAc (3 x 250 mL), washed with brine and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (18.7 g, 67.3% yield) of 5-(benzyloxy)-4-chloro-6-methylpyrimidine as a light yellow oil. LCMS observed m/z = 235.1 [M+H]+.
Step 3: Preparation of methyl 5-(benzyloxy)-6-methylpyrimidine-4-carboxylate. Intermediate A3
A mixture of 5-(benzyloxy)-4-chloro-6-methylpyrimidine (5.0 g, 21.3 mmol) in MeOH (20 mL) was added Pd(dppf)Cl2.CH2Cl2 (0.7 g, 1.0 mmol) in a pressure tank. The mixture was purged with N2 for 5 minutes and then was charged with carbon monoxide (5 atm.) three times at 25 °C. The resulting mixture was heated to 50 °C and stirred at 50 °C for 48 hours. The mixture was cooled to 25 °C, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography, (eluent: with 50% EtOAc in petroleum ether) to afford (4.0 g, 72.6% yield) of methyl 5-(benzyloxy)-6- methylpyrimidine-4-carboxylate as a colorless oil. LCMS observed m/z = 259.1 [M+H]+. Step 4: Preparation of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid. Intermediate A
A mixture of methyl 5-(benzyloxy)-6-methylpyrimidine-4-carboxylate (10.0 g, 38.7 mmol) in THF (20 mL) and MeOH (20 mL) was added dropwise a solution of NaOH (3.8 g, 97.1 mmol) in H2O (20 mL). The mixture was stirred at 25 °C for 10 minutes. Then the resulting mixture was concentrated in vacuo to remove THF and MeOH. The mixture was acidified to pH 3 with HCl (6 M in H2O) aqueous solution. The precipitated solids were collected by filtration and washed with H2O (3 x 5 mL) to afford (2.0 g, 21% yield) of 5- (benzyloxy)-6-methylpyrimidine-4-carboxylic acid as a white solid. 1H NMR (400MHz, DMSO-d6) δ 14.13 (s, 1H), 8.86 (s, 1H), 7.49 – 7.36 (m, 5H), 5.05 (s, 2H), 2.49 (s, 3H). LCMS observed m/z = 245.10 [M+H]+. Intermediate B Synthesis of 3-bromo-1H-1,2,4-triazol-5-amine. Intermediate B
A mixture of 5-amino-1H-1,2,4-triazole (50.0 g, 594.6 mmol), NaBr (152.9 g, 1486.6 mmol) and H2SO4 (145.8 g, 1486.6 mmol) in H2O (450 mL) was stirred at 25 °C for 10 minutes. To the mixture was added dropwise a solution of sodium bromate (44.8 g, 297.3 mmol) in H2O (150 mL) over 1 hour at 55-60 °C. The mixture was stirred at 55-60 °C for an additional 20 hours. The mixture was cooled to 0 °C and quenched with saturated sodium hyposulfite aqueous solution at 0 °C. The mixture was adjusted to pH 6 with 20% NaOH aqueous solution. The resulting mixture was concentrated in vacuo. The residue was washed by n-BuOH (750 mL) and EtOAc (1.5 L) in order. Then the solid was purified by trituration with dioxane (300 mL) at 80 °C. After filtration, the filter cake was vacuum-dried at 50-60 °C for 12 hours to afford (8.5 g, 8% yield) of 5-bromo-2H-1,2,4-triazol-3-amine as a white solid.1H NMR (400 MHz, DMSO-d6) δ 12.26 (s, 1H), 6.29 (s, 2H). LCMS observed m/z = 163.05 [M+H]+. Intermediate C Synthesis of tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)piperazine-1- carboxylate. Intermediate C
A mixture of tert-butyl piperazine-1-carboxylate (120.0 g, 644.2 mmol) and methyl 2-chloro-3-oxopentanoate (106.0 g, 644.2 mmol) in MeCN (500 mL) was added NEt3 (268.6 mL, 1932.8 mmol) in one portion at 25 °C. The mixture was heated to 60 °C and stirred at 60 °C for 16 hours. The mixture was cooled to 25 °C and concentrated in vacuo. The residue was diluted with H2O (300 mL) and extracted with CH2Cl2 (3 x 300 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 20% EtOAc in petroleum ether) to afford (30.5 g, 14% yield) of tert-butyl 4- (1-methoxy-1,3-dioxopentan-2-yl)piperazine-1-carboxylate as a light-yellow semi-solid.1H NMR (400 MHz, CDCl3) δ 4.10 (s, 1H), 3.79 (d, J = 8.2 Hz, 3H), 3.51 (s, 3H), 3.09 – 3.05 (m, 1H), 2.89 (s, 1H), 2.75 (s, 3H), 2.70 – 2.55 (m, 2H), 1.48 (d, J = 7.2 Hz, 9H), 1.15 – 1.11 (m, 3H). LCMS observed m/z = 313.2 [M+H]+.
Intermediate D Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-iodoacetamide. Intermediate D
Step 1: Preparation of 2-chloro-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Intermediate D1
To a mixture of 2-chloro-4-(trifluoromethyl)aniline (100.0 g, 511.3 mmol) in CH2Cl2 (500 mL) was added NEt3 (155.2 g, 1534.0 mmol) at 25 °C. To the mixture was added dropwise 2-chloroacetyl chloride (121.3 g, 1073.8 mmol) over 5 minutes at 0 °C. The mixture was stirred at 25 °C for 12 hours. Then the mixture was quenched by the addition of H2O at 25 °C and extracted with EtOAc (3 x 300 mL). The combined organic layers were washed with brine, dried over Na2SO4 and concentrated in vacuo. The residue was purified by silica gel column (eluent: with 8% EtOAc in petroleum ether) to afford (56.0 g, 40.2% yield) of 2-chloro-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 270.0 [M+H]+. Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide. Intermediate D
A mixture of 2-chloro-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (56.0 g, 205.8 mmol) in acetone (100 mL) was treated with KI (102.5 g, 617.5 mmol) at 60 °C for 5 hours. The mixture was filtered to remove solid and the filtrate was concentrated in vacuo to afford (71.0 g, 95% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide as a white solid. The crude product was used in other reactions without further purification. LCMS observed m/z = 362.0 [M+H]+. Intermediate E Synthesis of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate E
A mixture of tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)piperazine-1- carboxylate (14.5 g, 46.2 mmol, Intermediate C) and 5-bromo-2H-1,2,4-triazol-3-amine (7.6 g, 46.3 mmol, Intermediate B) in EtOH (45 mL) was treated with H3PO4 (4.5 g, 46.3 mmol) at 80 °C under N2 for 16 hours. Then the mixture was cooled to 25 °C and adjusted to pH 8 with saturated Na2CO3 aqueous solution. The resulting mixture was concentrated in vacuo. The residue was diluted with EtOH (100 mL) and stirred at 25 °C for 30 minutes. The resulting mixture was filtered, and the filter cake was washed with EtOH. The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 8% MeOH in CH2Cl2) to afford (4.0 g, 20% yield) of tert-butyl 4-{2-bromo- 5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate as a light- yellow solid. LCMS observed m/z = 362.0 [M+H]+. Intermediate F Synthesis of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate F
A mixture of tert-butyl 4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl}piperazine-1-carboxylate (30.0 g, 70.2 mmol, Intermediate E) and DIPEA (27.2 g, 210.6 mmol) in DMF (30 mL) was treated with N-[2-chloro-4- (trifluoromethyl)phenyl]-2-iodoacetamide (28.0 g, 77.2 mmol, Intermediate D) at 90 °C under N2 for 3.5 hours. The mixture was quenched with H2O (100 mL) at 25 °C and was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (2 x 250 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated
in vacuo. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (10.0 g, 21% yield) of tert-butyl 4-[2-bromo-4-({[2- chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]piperazine-1-carboxylate as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.73 (dd, J = 8.7, 2.1 Hz, 1H), 5.32 (s, 2H), 3.96 – 3.93 (m, 2H), 3.39 (dd, J = 4.7, 2.2 Hz, 2H), 2.98 – 2.95 (m, 4H), 2.66 (d, J = 11.2 Hz, 2H), 1.44 (s, 9H), 1.17 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 662.0 [M+H]+. Intermediate G Synthesis of 2-(2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Intermediate G
A mixture of tert-butyl 4-[2-bromo-4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]piperazine-1-carboxylate (4.8 g, 7.2 mmol, Intermediate F) in TFA (24 mL) and CH2Cl2 (24 mL) was stirred at 25 °C under N2 for 30 minutes. The mixture was concentrated in vacuo to afford (4.5 g crude) of 2-(2-bromo-5-ethyl-7-oxo-6-(piperazin-1- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as the crude mixture and was used directly in the next step without further purification. LCMS observed m/z = 562.0 [M+H]+. Intermediate H Synthesis of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Intermediate H
A mixture of 2-[2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (1.5 g, 2.6 mmol, Intermediate G), HATU (1.5 g, 3.9 mmol) and DIPEA (1.7 g, 13.3 mmol) in DMF (15 mL) was stirred at 25 °C under N2 for 2.5 hours. The mixture was quenched with H2O (200 mL) at 25 °C and extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (2 x 600 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 10% petroleum ether in EtOAc) to afford (749 mg, 33% yield) of 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid.1H NMR (400 MHz, CDCl3) δ 8.88 (s, 1H), 8.84 (s, 1H), 8.49 (d, J = 8.7 Hz, 1H), 7.70 (d, J = 2.1 Hz, 1H), 7.59 – 7.55 (m, 1H), 7.50 – 7.39 (m, 5H), 5.22 – 5.19 (m, 1H), 5.16 – 5.06 (m, 3H), 4.81 – 4.79 (m, 1H), 3.80 – 3.76 (m, 2H), 3.44 – 3.29 (m, 2H), 3.15 (q, J = 7.5 Hz, 2H), 3.07 – 3.05 (m, 1H), 2.88 – 2.83 (m, 1H), 2.65 (m, 1H), 2.48 (s, 3H), 1.32 (t, J = 7.7 Hz, 3H). LCMS observed m/z = 788.40 [M+H]+. Intermediate I Synthesis of tert-butyl 4-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate I
To a mixture of tert-butyl 4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl}piperazine-1-carboxylate (1.2 g, 3.0 mmol, Intermediate E) and 2-(3,6- dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (954 mg, 4.5 mmol) in dioxane (15 mL) and H2O (5 mL) were added Pd(dppf)Cl2.CH2Cl2 (247 mg, 0.3 mmol) and
Na2CO3 (641 mg, 6.0 mmol) in one portion at 25 °C under N2. The mixture was heated to 100 °C and stirred for 16 hours at 100 °C under N2. The resulting mixture was cooled to 25 °C, filtered and the filter cake was washed with EtOAc (30 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in CH2Cl2) to afford (1.1 g, 84% yield) of tert-butyl 4-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine- 1-carboxylate as a light-yellow solid. LCMS observed m/z = 429.2 [M+H]+. Intermediate J Synthesis of tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate J
To a mixture of tert-butyl 4-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (775 mg, 1.8 mmol, Intermediate I) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (719 mg, 1.9 mmol, Intermediate D) in DMF (9 mL) was treated with DIPEA (698 mg, 5.4 mmol) at 45 °C under N2 for 3.5 hours. The mixture was concentrated in vacuo and the residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 10-100% MeCN in H2O) to afford (525 mg, 44% yield) of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a brown solid. LCMS observed m/z = 664.2 [M-H]-. Intermediate K Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Intermediate K
of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (50 mg, 0.07 mmol, Intermediate J) in TFA (10 mL) was stirred at 25 ℃ for 2 hours. The mixture was quenched with H2O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparative TLC (eluent: 17% EtOAc in petroleum ether) to afford (10.7 mg, 25% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 8.17 (d, J = 8.4 Hz, 1H), 7.84 (d, J = 2.1 Hz, 1H), 7.63 (dd, J = 8.9, 2.1 Hz, 1H), 7.00 – 6.95 (m, 1H), 5.41 (s, 2H), 4.34 (q, J = 2.8 Hz, 2H), 4.03 – 3.81 (m, 4H), 3.38 – 3.34 (m, 2H), 3.24 – 3.14 (m, 2H), 3.09 (q, J = 7.5 Hz, 2H), 2.94 (d, J = 9.9 Hz, 2H), 2.67 – 2.63 (m, 2H), 1.36 – 1.22 (t, J = 7.3 Hz, 3H). LCMS observed m/z = 566.30 [M+H]+. Intermediate L Synthesis of 3-(benzyloxy)-4-oxo-4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine- 2-carboxylic acid. Intermediate L
Step 1: Preparation of amino(methyl)aluminum chloride. Intermediate L1
A mixture of NH4Cl (3.4 g, 64.4 mmol) in toluene (50 mL) was added dropwise a solution of trimethylaluminum (65 mL, 0.9 mmol, 1 M in toluene) at 0 °C under N2. The resulting mixture was used directly in the next step without work up or further purification. Intermediate L2
Step-2: A mixture of ethyl 2-(benzyloxy) acetate (2.8 g, 14.4 mmol), NaH (0.4 g, 15.9 mmol, 60% purity) and ethyl oxalate (2.1 g, 14.4 mmol) in THF (50 mL) and EtOH (8.37 uL, 0.1 mmol) was stirred at 25 °C under N2 for 18 hours. Then the mixture was quenched with H2O and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (2 x 15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford (3.5 g crude) of diethyl 2-(benzyloxy)-3- oxosuccinate as the crude product and was used directly in the next step without further purification. LCMS observed m/z = 295.3 [M+H]+. [M+H]+. Step 3: Preparation of 2-(2-(methylthio)ethoxy)acetic acid. Intermediate L3
A mixture of NaH (0.9 g, 39.8 mmol, 60% purity) in THF (60 mL) were added 2- (methylthio)-ethanol (1.0 g, 10.9 mmol) at 25 °C. After 20 minutes, a solution of 2- bromoacetic acid (1.5 g, 10.8 mmol) in THF (25 mL) was added dropwise to the mixture at 25 °C under N2 and stirred at 70 °C under N2 for 6 hours. Then the mixture was cooled to 25 °C, adjusted to pH 3 with HCl (1 M in H2O) aqueous solution and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford (1.4 g crude) of 2-(2-(methylthio)ethoxy)acetic acid as a yellow oil and was used directly in the next step without further purification.1HNMR (400 MHz, CDCl3) δ 4.20 (s, 2H), 3.79 (t, J = 6.3 Hz, 2H), 2.77 (t, J = 6.4 Hz, 2H), 2.19 (s, 3H). Step 4: Preparation of methyl 2-(2-(methylthio)ethoxy)acetate. Intermediate L4
A mixture of 2-(2-(methylthio)ethoxy)acetic acid (1.4 g, 9.3 mmol) and oxalyl chloride (2.3 g, 18.6 mmol) in CH2Cl2 (50 mL) was stirred at 25 °C under N2 for 18 hours. The mixture was concentrated in vacuo and the residue was dissolved in CH2Cl2 (50 mL) and to the mixture was added MeOH (6.0 mL, 148.2 mmol) and pyridine (10 mL, 126.4 mmol) at 0 °C. The mixture was stirred at 25 °C for 1 hour. Then the mixture was quenched with H2O (30 mL) and extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were washed with HCl (15 mL, 1 M in H2O) aqueous solution, saturated NaHCO3 aqueous solution (25 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford (1.4 g, 91% yield) of methyl 2-(2- (methylthio)ethoxy)acetate as a yellow oil.1HNMR (400 MHz, DMSO-d6) δ 4.15 (s, 2H), 3.65 (d, J = 8.6 Hz, 5H), 2.65 (t, J = 6.8 Hz, 2H), 2.09 (s, 3H). Step 5: Preparation of 2-(2-(methylthio)ethoxy)acetimidamide. Intermediate L5
To a mixture of methyl 2-(2-(methylthio)ethoxy)acetate (1.4 g, 8.5 mmol) was added dropwise the solution of amino(methyl)aluminum chloride in toluene at 25 °C. The mixture was heated to 80 °C and stirred at 80 °C for 18 hours. The mixture was cooled to 0 °C and quenched with MeOH (100 mL) at 0 °C. The mixture was filtered and the filter cake was washed with MeOH (3 x 100 mL). The filtrate was concentrated in vacuo and the residue was purified by trituration with IPA (160 mL) and acetone (40 mL). The precipitated solids were collected by filtration and washed with IPA (2 x 10 mL) to afford (1.2 g crude) of 2- (2-(methylthio)ethoxy)acetimidamide as the HCl salt and was used directly in the next step without further purification. LCMS observed m/z = 149.1 [M+H]+. Step 6: Preparation of ethyl 5-(benzyloxy)-2-{[2-(methylsulfanyl)ethoxy]methyl}- 6-oxo-1H-pyrimidine-4-carboxylate. Intermediate L6
To a mixture of EtONa (138 mg, 2.0 mmol) in EtOH (50 mL), 2-(2- (methylthio)ethoxy)acetimidamide (1.2 g crude, HCl salt) and 1,4-diethyl 2-(benzyloxy)-3- oxobutanedioate (1.2 g, 4.0 mmol) was added to a 100 mL round-bottom flask at 25 °C. The mixture was stirred at 60 °C for 3 hours. The mixture was cooled to 25 °C, quenched with acetic acid (2 mL) and concentrated in vacuo to remove EtOH. The mixture was diluted with EtOAc (150 mL) and washed with saturated NaHCO3 aqueous solution, brine and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (100 mg, 5% yield) of ethyl 5-(benzyloxy)-2-{[2- (methylsulfanyl)ethoxy]methyl}-6-oxo-1H-pyrimidine-4-carboxylate as a yellow oil. LCMS observed m/z = 379.1 [M+H]+. Step 7: Preparation of (2-((5-(benzyloxy)-4-(ethoxycarbonyl)-6-oxo-1,6- dihydropyrimidin-2-yl)methoxy)ethyl)dimethylsulfonium Intermediate L7
A mixture ethyl 5-(benzyloxy)-2-{[2-(methylsulfanyl)ethoxy]methyl}-6-oxo-1H- pyrimidine-4-carboxylate (100 mg, 0.3 mmol) and MeI (4.1 mL, 66.8 mmol) in CH2Cl2 (5 mL) was stirred at 25 °C under N2 for 8 days. Then the mixture was concentrated in vacuo to afford (130 mg, 95% yield) of (2-{[5-(benzyloxy)-4-(ethoxycarbonyl)-6-oxo-1H- pyrimidin-2-yl]methoxy}ethyl)dimethylsulfanium iodide as the crude product and was used directly in the next step without further purification. LCMS observed m/z = 394.1 [M+H]+. Step 8: Preparation of ethyl 3-(benzyloxy)-4-oxo-4,6,7,9-tetrahydropyrimido[2,1- c][1,4]oxazine-2-carboxylate. Intermediate L8
A mixture of (2-((5-(benzyloxy)-4-(ethoxycarbonyl )-6–oxo-1,6-dihydropyrimidin– 2-yl)methoxy)ethyl) dimethylsulfonium (130 mg, 0.3 mmol) and K2CO3 (913.2 mg, 6.6 mmol) in DMF (5 mL) was stirred at 25 °C under N2 for 2 days. Then the solid was filtered and the filtrate was concentrated in vacuo. The residue was diluted with EtOAc (30 mL), washed with HCl (0.1 M in H2O) aqueous solution, saturated Na2CO3 aqueous solution (25 mL) and dried over MgSO4. The resulting mixture was concentrated in vacuo and the residue was purified by chromatography on silica column (eluent: with 40% EtOAc in petroleum ether) to afford (80 mg, 73% yield) of ethyl 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1- c][1,4]oxazine-2-carboxylate as a white solid. LCMS observed m/z = 331.3 [M+H]+. Step 9: Preparation of 3-(benzyloxy)-4-oxo-4,6,7,9-tetrahydropyrimido[2,1- c][1,4]oxazine-2-carboxylic acid. Intermediate L
A mixture of ethyl 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2- carboxylate (80 mg, 0.2 mmol) and NaOH (32 mg, 0.8 mmol) in EtOH (20 mL) was stirred at 25 °C under N2 for 30 minutes. The mixture was quenched with HCl (4 M in H2O, 10 mL) aqueous solution and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (25 mL), dried over anhydrous Na2SO4 and concentrated in vacuo to afford (60 mg, 82% yield) of 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1- c][1,4]oxazine-2-carboxylic acid as a white solid and the crude product was used directly in the next step without further purification. LCMS observed m/z = 303.09 [M+H]+. Intermediate M Synthesis of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(dimethylamino)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid. Intermediate M
Step 1: Preparation of tert-butyl 4-(2-bromo-4-(2-ethoxy-2-oxoethyl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate M1
A mixture of tert-butyl 4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl}piperazine-1-carboxylate (10.0 g, 23.4 mmol, Intermediate E) and ethyl bromoacetate (5.9 g, 35.1 mmol) in DMF (40 mL) was added DIPEA (9.1 g, 70.2 mmol) at 25 °C under N2. The mixture was heated to 90 ℃ and stirred at 90 ℃ under N2 for 5 hours. The mixture was cooled to 25 ℃, quenched with H2O (40 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 33% EtOAc in petroleum ether) to afford (6.0 g, 50% yield) of tert-butyl 4-[2-bromo-4-(2-ethoxy-2-oxoethyl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a yellow solid. LCMS observed m/z = 513.1 [M+H]+. Step 2: Preparation of tert-butyl 4-(2-(dimethylamino)-4-(2-ethoxy-2-oxoethyl)-5- ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate M2
A mixture of tert-butyl 4-[2-bromo-4-(2-ethoxy-2-oxoethyl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (3.0 g, 5.8 mmol) and KOAc (1.1 g, 11.7 mmol) in a mixed solvent of DMF (5 mL) and DMSO (5 mL) was added dimethylamine (2 M in THF, 11.7 mL, 23.4 mmol) solution at 25 °C under N2. The reaction mixture was heated to 120 ℃ and stirred at 120 ℃ under N2 for 3 hours. The reaction mixture was cooled to 25 ℃, quenched with H2O (40 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (1.6 g, 57% yield) of tert-butyl 4-[2-(dimethylamino)-4-(2-ethoxy-2-oxoethyl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a light yellow solid. LCMS observed m/z = 478.3 [M+H]+. Step 3: Preparation of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid. Intermediate M
A mixture of tert-butyl 4-[2-(dimethylamino)-4-(2-ethoxy-2-oxoethyl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (1.5 g, 3.1 mmol) in a mixed solvent of THF (10 mL) and H2O (10 mL) was added LiOH.H2O (150 mg, 6.3 mmol) in portions at 25 °C. The mixture stirred at 25 °C for 2 hours. The mixture was quenched with saturated NH4Cl aqueous solution and concentrated in vacuo. The residue was dissolved in H2O (20 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine, dried over Na2SO4 and concentrated in vacuo to afford (1.3 g, 92% yield) of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(dimethylamino)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid as a yellow oil and the crude product was used directly in the next step without further purification. LCMS observed m/z = 450.25 [M+H]+. Intermediate N
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl- 7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Intermediate N
Step 1: Preparation of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate. Intermediate N1
A mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(dimethylamino)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (1.3 g, 2.9 mmol, Intermediate M), 2-chloro-4-(trifluoromethyl)aniline (848 mg, 4.3 mmol) and NEt3 (2.2 g, 21.4 mmol) in EtOAc (3 mL) was added T3P (25 mL) at 25 °C under N2 for 3 hours. Then the mixture was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 10-50% MeCN in H2O) to afford (580 mg, 32% yield) of tert-butyl 4-[4-({[2-chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate as a yellow solid. LCMS observed m/z = 626.2 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide. Intermediate N
A mixture of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (580 mg, 0.9 mmol) in CH2Cl2 (5 mL) was added TFA (5 mL) and stirred at 25 °C under N2 for 3 hours. The mixture was concentrated in vacuo. The residue was purified by prep-HPLC (10-34% MeCN in H2O) to afford (320 mg, 66% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide as a white solid. LCMS observed m/z = 526.2 [M+H]+. Intermediate O Synthesis of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid. Intermediate O
Step 1: Preparation of ethyl 2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetate. Intermediate O1
A mixture of tert-butyl 4-[2-(dimethylamino)-4-(2-ethoxy-2-oxoethyl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (600 mg, 1.2 mmol) in CH2Cl2 (6 mL) was added TFA (2 mL) and stirred at 25 °C under N2 for 3 hours. The mixture was concentrated in vacuo to afford (426 mg, crude) of ethyl 2-[2-(dimethylamino)- 5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetate as a yellow solid and the crude product was used directly in the next step without further purification. LCMS observed m/z = 378.2 [M+H]+. Step 2: Preparation of ethyl 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetate. Intermediate O2
A mixture of ethyl 2-[2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetate (275 mg, 0.7 mmol), 5-(benzyloxy)-6- methylpyrimidine-4-carboxylic acid (195 mg, 0.8 mmol) and DIPEA (188 mg, 1.4 mmol) in DMF (10 mL) was added HATU (332 mg, 0.8 mmol) and stirred at 25 °C under N2 for 3 hours. The mixture was quenched by the addition of H2O (10 mL) and extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (197 mg, 44% yield) of ethyl 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetate as a light yellow solid. LCMS observed m/z = 604.3 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetic acid. Intermediate O
A mixture of ethyl 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetate (192 mg, 0.3 mmol) in THF (6 mL) and H2O (3 mL) was added LiOH.H2O (15 mg, 0.6 mmol) and was stirred at 25 °C under N2 for 3 hours. The mixture was adjusted to pH 6 with HCl (1 M in H2O) aqueous solution and concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 0-50% MeCN in H2O) to afford (97 mg, 52% yield) of (6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetic acid as a white solid. LCMS observed m/z = 576.26 [M+H]+. Intermediate P Synthesis of 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine. Intermediate P
Step 1: Preparation of 2-bromo-5-iodobicyclo[4.2.0]octa-1,3,5-triene. Intermediate P1
A mixture of 2,5-dibromobicyclo[4.2.0]octa-1(6),2,4-triene (5.2 g, 19.6 mmol) in THF (60 mL) was added n-BuLi (2.5 M in hexane, 9.4 mL, 23.6 mmol, 1.2 equiv.) solution dropwise at -78 ℃ under N2. The mixture was stirred for 30 minutes at -78 ℃. To the mixture was added a solution of I2 (6.0 g, 23.6 mmol) in THF (60 mL) dropwise at -78 ℃. The mixture was stirred for additional 25 minutes at -78 ℃. The mixture was warmed to 25 ℃ and stirred at 25 ℃ for 50 minutes. The mixture was quenched with saturated
Na2S2O3 aqueous solution (20 mL) at 0 ℃, extracted with EtOAc (3 x 50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 100% petroleum ether) to afford (5.4 g, 90% yield) of 2-bromo-5-iodobicyclo[4.2.0]octa-1(6),2,4-triene as a white solid. LCMS observed m/z = 308.9 [M+H]+. Step 2: Preparation of 2-bromo-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- triene. Intermediate P2
A mixture of 2-bromo-5-iodobicyclo[4.2.0]octa-1(6),2,4-triene (5.4 g, 17.5 mmol) and methyl 2,2-difluoro-2-sulfoacetate (16.8 g, 87.4 mmol) in NMP (30 mL) was treated with CuI (5.0 g, 26.2 mmol) in one portion at 25 ℃. The mixture was heated to 80 ℃ and stirred at 80 ℃ for 16 hours under N2. The mixture was cooled to 25 ℃, quenched with H2O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 100% petroleum ether) to afford (4.1 g, 93% yield) of 2- bromo-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-triene as a colorless oil. LCMS observed m/z = 250.9 [M+H]+. Step 3: Preparation of 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine. Intermediate P
A mixture of 2-bromo-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-triene (3.9 g, 15.7 mmol) and Cu(acac)2 (4.1 g, 15.7 mmol) in DMF (40 mL) was treated with NH3.H2O (40 mL) in one portion at 25 ℃. The mixture was heated to 90 ℃ and stirred at 90 ℃ for 16 hours. The mixture was cooled to 25 ℃, quenched by the addition of H2O (65 mL) and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (2 x 100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography
(eluent: with 2% EtOAc in petroleum ether) to afford (1.9 g, 65% yield) of 5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine as a brown yellow oil. LCMS observed m/z = 188.1 [M+H]+. Intermediate Q Synthesis of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate Q
Step 1: Preparation of tert-butyl 4-{2-bromo-4-[2-(tert-butoxy)-2-oxoethyl]-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate. Intermediate Q1
A mixture of tert-butyl 4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl}piperazine-1-carboxylate (5.0 g, 11.7 mmol, Intermediate E) and tert- butyl 2-bromoacetate (2.4 g, 12.3 mmol) in DMF (8 mL) was treated with DIPEA (4.5 g, 35.1 mmol) in one portion at 25 ℃. The mixture was heated to 45 ℃ and stirred at 45 ℃ for 16 hours under N2. The mixture was cooled to 25 ℃. The precipitated solids were collected by filtration and washed with DMF (2 x 2 mL) to afford (2.5 g, 39% yield) of tert- butyl 4-{2-bromo-4-[2-(tert-butoxy)-2-oxoethyl]-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl}piperazine-1-carboxylate as a white solid. LCMS observed m/z = 539.1 [M+H]+. Step 2: Preparation of {2-bromo-6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid.
Intermediate Q2
A mixture of tert-butyl 4-{2-bromo-4-[2-(tert-butoxy)-2-oxoethyl]-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate (2.3 g, 4.2 mmol) in THF (150 mL) and MeOH (15 mL) was treated with NaOH (340 mg, 8.5 mmol) in H2O (20 mL) in one portion at 25 ℃. The mixture was stirred at 25 ℃ for 2 hours. The mixture was acidified to pH 6~7 with HCl (0.1 M in H2O) aqueous solution and concentrated in vacuo. The aqueous layer was extracted with CH2Cl2 (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford (1.8 g) of {2-bromo-6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}acetic acid as a brown yellow solid. The crude product was used directly in the next step without further purification. LCMS observed m/z = 487.1 [M+H]+. Step 3: Preparation of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate Q
A mixture of {2-bromo-6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (1.3 g, 2.7 mmol) and 5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine (467 mg, 2.5 mmol, Intermediate P) in DMF (20 mL) was treated with TCFH (898 mg, 3.2 mmol) and NMI (1.1 g, 13.4 mmol) in one portion at 25 ℃. The mixture was heated to 40 ℃ and stirred at 40 ℃ for 36 hours. The mixture was cooled to 25 ℃, quenched with H2O (120 mL) and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed
with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography to afford (1.0 g, 55% yield) of tert-butyl 4-[2-bromo-5-ethyl-7-oxo-4-({[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a white solid. 1H NMR (400 MHz, Methanol-d4) δ 7.42 (q, J = 8.8 Hz, 2H), 5.23 (s, 2H), 4.11 – 4.05 (m, 2H), 3.57 – 3.53 (m, 2H), 3.37 (s, 2H), 3.24 (t, J = 4.3 Hz, 2H), 3.09 – 3.03 (m, 4H), 2.79 – 2.75 (m, 2H), 1.51 (s, 9H), 1.29 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 654.25 [M+H]+. Intermediate R Synthesis of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate R
6. Step 1: Preparation of 2-[2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. Intermediate R1
A mixture of tert-butyl 4-[2-bromo-5-ethyl-7-oxo-4-({[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)-
[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (1.5 g, 2.3 mmol, Intermediate Q) in HCl (4 M in 1,4-dioxane, 20 mL) solution was stirred for 30 minutes at 25 ℃. The mixture was concentrated in vacuo to afford (1.3 g crude) of 2-[2-bromo-5- ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. The crude product was used directly in the next step without further purification. LCMS observed m/z = 554.0 [M+H]+. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Intermediate R
A mixture of 2-[2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (1.3 g, 2.3 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (560 mg, 2.3 mmol, Intermediate A) in DMF (10 mL) was treated with HATU (1.1 g, 2.8 mmol) and DIPEA (1.5 g, 11.5 mmol) in one portion at 25 ℃ and stirred at 25 ℃ for 1 hour. The mixture was diluted with H2O (100 mL) and extracted with EtOAc (3 x 40 mL). The combined organic layers were washed with brine (2 x 120 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 50-99% MeOH in H2O) to afford (1.0 g, 53% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine- 4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.71 (s, 1H), 8.87 (s, 1H), 7.55 – 7.48 (m, 3H), 7.48 – 7.38 (m, 4H), 5.22 – 5.06 (m, 4H), 4.57 (m, 1H), 3.48 (m, 2H), 3.37 (s, 1H), 3.27 (dd, J = 5.5,
2.9 Hz, 2H), 3.19 (q, J = 4.7, 4.2 Hz, 2H), 3.02 – 2.98 (m, 3H), 2.87 – 2.85 (m, 1H), 2.66 – 2.64 (m, 1H), 2.49 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 780.30 [M+H]+. Intermediate S Synthesis of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetic acid. Intermediate S
Step 1: Preparation of ethyl 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetate. Intermediate S1
A mixture of tert-butyl 4-[2-(3,6-dihydro-2H-pyran-4-yl)-4-(2-ethoxy-2-oxoethyl)- 5-ethyl-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (2.4 g, 4.6 mmol) in mixed solvent of CH2Cl2 (25 mL) and TFA (14 mL) was stirred at 25 °C for 40 minutes. The mixture was concentrated in vacuo to afford (1.9 g, crude) of ethyl 2-(2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4] triazolo [1,5-a]pyrimidin- 4(7H)-yl)acetate. The crude product was used directly in the next step without further purification. LCMS observed m/z = 417.1 [M+H]+. Step 2: Preparation of ethyl 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetate. Intermediate S2
A mixture of ethyl 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetate (1.9 g, 4.5 mmol), 5-(benzyloxy)-6- methylpyrimidine-4-carboxylic acid (1.2 g, 5.0 mmol, Intermediate A), DIPEA (2.9 g, 22.8 mmol) and HATU (2.6 g, 6.8 mmol) in DMF (25 mL) was stirred at 25 °C for 4 hours. The mixture was quenched with H2O (30 mL) at 25°C and extracted with EtOAc (2 x 30 mL). The combined organic layers were washed with brine and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 5% MeOH in CH2Cl2) to afford (1.8 g, 61% yield) of ethyl 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetate as a brown yellow oil. LCMS observed m/z = 643.1 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetic acid. Intermediate S
A mixture of ethyl 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetate (1.8 g, 5.3 mmol) and LiOH.H2O (380 mg, 15.9 mmol) in THF (20 mL) and H2O (4 mL) was stirred at 40 °C for 6 hours. The mixture was concentrated in
vacuo. The residue was purified by preparatory HPLC (column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 12-35% MeCN in H2O) to afford (911 mg, 50% yield) of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)acetic acid as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.82 (s, 1H), 7.54 – 7.38 (m, 5H), 6.97 – 6.93 (m, 1H), 5.21 – 5.12 (m, 2H), 4.95 (d, J = 3.6 Hz, 2H), 4.70 – 4.67 (m, 1H), 4.34 (q, J = 2.8 Hz, 2H), 3.93 – 3.90 (m, 2H), 3.78 – 3.69 (m, 2H), 3.47 – 3.34 (m, 2H), 3.16 – 2.92 (m, 4H), 2.77 – 7.74 (m, 1H), 2.69 – 2.63 (m, 2H), 2.49 (s, 3H), 1.30 – 1.26 (m, 3H). LCMS observed m/z = 615.40 [M+H]+. Intermediate T Synthesis of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetic acid. Intermediate T
Step 1: Preparation of tert-butyl 4-(2-(benzo[d][1,3]dioxol-5-yl)-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate T1
A mixture of tert-butyl 4-[2-bromo-4-({[2-chloro-4-(trifluoromethyl) phenyl]carbamoyl} methyl)-5-ethyl-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-6-yl] piperazine-1-carboxylate (2.0 g, 3.0 mmol, Intermediate F) and 2-(2H-1,3-benzodioxol-5-
yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.1 g, 4.5 mmol) in dioxane (60 mL) and H2O (20 mL) were added Na2CO3 (960 mg, 9.1 mmol) and Pd(dppf)Cl2.CH2Cl2 (246 mg, 0.3 mmol) in portions at 25 °C. The mixture was heated to 100 °C and stirred at 100°C for 3 hours under N2. The mixture was cooled to 25 °C and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (1.2 g, 56% yield) of tert-butyl 4-[2-(2H-1,3-benzodioxol-5-yl)-4-({[2- chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]piperazine-1-carboxylate as a white solid. LCMS observed m/z = 704.21 [M+H]+. Step 2: Preparation of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Intermediate T
A mixture of tert-butyl 4-[2-(2H-1,3-benzodioxol-5-yl)-4- ({[2-chloro-4- (trifluoromethyl) phenyl]carbamoyl}methyl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]piperazine-1-carboxylate (1.4 g, 2.0 mmol) and TFA (1.1 g, 1.0 mmol) in CH2Cl2 (10 mL) was stirred at 25 °C for 3 hours. The mixture was concentrated in vacuo, diluted with H2O (10 mL), basified to pH 8 with saturated NaHCO3 aqueous solution and extracted with CH2Cl2 (5 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by trituration with EtOAc to afford (530.4 mg, 44% yield) of 2-[2-(2H-1,3-benzodioxol-5-yl)- 5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.16 (d, J = 8.6 Hz, 1H), 7.85 – 7.76 (m, 2H), 7.66 – 7.60 (m, 2H), 6.93 (d, J = 8.2 Hz, 1H), 6.04 (s, 2H), 5.47 (s, 2H), 3.96- 3.91 (m, 2H), 3.39 – 3.34 (m, 2H), 3.26 – 3.21 (m, 2H), 3.13 – 3.10 (m, 2H), 3.04 – 2.97 (m, 2H), 1.33 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 604.25 [M+H]+.
Intermediate U Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(isochroman-6- yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Intermediate U
Step 1: Preparation of tert-butyl 4-(2-(benzo[d][1,3]dioxol-5-yl)-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate U1
A mixture of tert-butyl 4-[2-bromo-4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]piperazine-1-carboxylate (700 mg, 1.0 mmol, Intermediate F) and 2-(3,4- dihydro-1H-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (329 mg, 1.2 mmol) in dioxane (10 mL) and H2O (3 mL) were added K2CO3 (364 mg, 2.6 mmol) and Pd(dppf)Cl2.CH2Cl2 (77 mg, 0.1 mmol) in portions at 25 °C under N2. The mixture was heated to 90 °C and stirred at 90 °C for 4 hours under N2. The mixture was cooled to 25 °C and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (500 mg, 66% yield) of tert-butyl 4- [4-({[2-chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,4-dihydro-1H-2- benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate as a white solid. LCMS observed m/z = 716.2 [M+H]+.
Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl]acetamide. Intermediate U
A mixture of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (100 mg, 0.1 mmol) in a mixed solvent of TFA (3 mL) and CH2Cl2 (3 mL) was stirred at 25 °C for 2 hours. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (50 mg, 58% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5- ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.48 (s, 1H), 9.06 (s, 1H), 8.61 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.90 (dt, J = 4.2, 2.0 Hz, 2H), 7.73 (dd, J = 8.7, 2.1 Hz, 1H), 7.20 (d, J = 8.5 Hz, 1H), 5.42 (s, 2H), 4.74 (s, 2H), 3.92 (t, J = 5.7 Hz, 2H), 3.83 – 3.73 (m, 2H), 3.18 (s, 3H), 3.05-2.95 (m, 2H), 2.92 – 2.83 (m, 4H), 1.25-1.15 (m, 3H). LCMS observed m/z = 616.20 [M+H]+. Intermediate V Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Intermediate V
Step 1: Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-2-((3aR,6aS)-tetrahydro-1H- furo[3,4-c]pyrrol-5(3H)-yl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate. Intermediate V1
To a solution of tert-butyl 4-[2-bromo-4-({[2-chloro-4-(trifluoromethyl) phenyl] carbamoyl} methyl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-6-yl] piperazine-1- carboxylate (400 mg, 0.6 mmol, Intermediate F) and (3aR,6aS)-hexahydro-1H-furo[3,4-c] pyrrole (137 mg, 1.2 mmol) in IPA (4 mL) was added NEt3 (244 mg, 2.4 mmol) in one portion at 25 °C. The mixture was stirred at 100 °C under N2 for 48 hours. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (331 mg, 79% yield) of tert-butyl 4- {2-[(3aR,6aS)-hexahydrofuro[3,4-c] pyrrol-5-yl]-4-({[2-chloro-4-(trifluoromethyl) phenyl] carbamoyl} methyl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-6-yl} piperazine-1- carboxylate as a white solid. LCMS observed m/z = 695.30 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Intermediate V
A mixture of tert-butyl 4-{2-[(3aR,6aS)-hexahydrofuro[3,4-c] pyrrol-5-yl]-4-({[2- chloro-4-(trifluoromethyl) phenyl] carbamoyl} methyl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5- a] pyrimidin-6-yl} piperazine-1-carboxylate (196 mg, 0.3 mmol) in CH2Cl2 (5 mL) was added TFA (1 mL) in portions at 0 °C under N2. The mixture was stirred at 0 °C under N2 for 30 minutes. The mixture was quenched with H2O (10 mL) and extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2- (6-(4-(2-(cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide as a crude product. The crude product was used directly in the next step without further purification. LCMS observed m/z = 595.20 [M+H]+. Intermediate W Synthesis of 2-[2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. Intermediate W
Step 1: Preparation of tert-butyl 4-[2-(dimethylamino)-5-ethyl-7-oxo-4-({[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate.
Intermediate W1
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (200 mg, 0.4 mmol, Intermediate M) and 5-(trifluoromethyl) bicyclo[4.2.0]octa-1(6),2,4-trien-2- amine (83 mg, 0.4 mmol, Intermediate P) in MeCN (2 mL) were added NMI (109 mg, 1.3 mmol) and TCFH (124 mg, 0.4 mmol) in portions at 25 °C under N2. The mixture was heated to 50 °C and stirred at 50 °C for 2 hours under N2. The mixture was cooled to 25 °C. The mixture was quenched with H2O (5 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: with 60-75% MeCN in H2O) to afford (234 mg, 85% yield) of tert-butyl 4-[2-(dimethylamino)-5-ethyl-7-oxo-4-({[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carb amoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a yellow solid. LCMS observed m/z = 619.6 [M+H]+. Step 2: Preparation of 2-[2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. Intermediate W
A solution of tert-butyl 4-[2-(dimethylamino)-5-ethyl-7-oxo-4-({[5- (trifluoromethyl) bicyclo [4.2.0] octa-1(6),2,4-trien-2-yl] carbamoyl} methyl)- [1,2,4]
triazolo[1,5-a] pyrimidin-6-yl] piperazine-1-carboxylate (234 mg, 0.4 mmol) and TFA (111 mg, 1.2 mmol) in CH2Cl2 (5 mL) was stirred at 25 ℃ for 1 hour. The mixture was concentrated in vacuo to afford (160 mg, crude) of 2-[2-(dimethylamino)-5-ethyl-7-oxo-6- (piperazin-1-yl)- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl]-N-[5-(trifluoromethyl) bicyclo [4.2.0] octa-1(6),2,4-trien-2-yl] acetamide as a yellow oil. The crude product was used directly in the next step without further purification. LCMS observed m/z = 743.2 [M+H]+. Intermediate X Synthesis of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-1,1-difluoro-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate X
The title compound can be prepared using similar procedure as Intermediate F, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-iodoacetamide (Intermediate D) with N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoro-2-iodoacetamide. Intermediate Y Synthesis of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate Y
The title compound can be prepared using similar procedure as Intermediate Q, replacing 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine (Intermediate P) with 3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-amine. Intermediate Z Synthesis of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- ((trifluoromethyl)thio)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Intermediate Z
The title compound can be prepared using similar procedure as Intermediate Q, replacing 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine (Intermediate P) with 2-chloro-4-((trifluoromethyl)thio)aniline. Intermediate AA Synthesis of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((4- ((trifluoromethyl)thio)phenyl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate. Intermediate AA
The title compound can be prepared using similar procedure as Intermediate Q, replacing 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine (Intermediate P) with 4-((trifluoromethyl)thio)aniline. Intermediate AB Synthesis of 5-hydroxy-6-methylpyrimidine-4-carboxylic acid
Intermediate AB
To a solution of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (30.0 g, 122 mmol, 1 eq) in EtOAc (150 mL) was added Pd/C (3.00 g, 2.82 mmol, 10% purity, 2.30e-2 eq) under N2 atmosphere. The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (15 Psi) at 28 °C for 4 hrs. TLC (Dichloromethane/Methanol = 5/1) indicated Intermediate A consumed completely and one new spot formed. The mixture was filtrated through a pad of Celite and washed with MeOH (3 times 200 mL), then was concentrated under reduced pressure to give a residue. The crude product (14.5 g, 94.0 mmol, 76.6% yield) was used into the next step without further purification and obtained as a reddish solid. LCMS observed m/z = 153.14 [M–H]–. Intermediate AC Synthesis of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide Intermediate AC
In a 250 mL round bottom flask equipped with a stir bar was added 5-hydroxy-6- methylpyrimidine-4-carboxylic acid, Intermediate AB (1.75 g, 11.4 mmol) and CH2Cl2 (75 mL). The mixture was allowed to stir at room temperature for 2 minutes to form a slurry, then Ghosez reagent (1.80 mL, 13.6 mmol) was added dropwise at room temperature via syringe over a period of 5 minutes. The reaction mixture was allowed to stir at room temperature. After 1.5 hours, the reaction mixture was a homogeneous brown solution. In a separate 500 mL round bottom flask equipped with a stir bar was added 2-(2-bromo-5-ethyl- 7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide, Intermediate G (4.52 g, 7.54 mmol), CH2Cl2 (60 mL), and DIPEA (7.9 mL, 45.3 mmol). The mixture was allowed to stir at room temperature for
5 minutes to form a suspension, then cooled to -10 C via brine/ice bath. The reaction vessel was fitted with an addition funnel, and the reaction mixture containing above reaction mixture was transferred to the addition funnel, rinsing with CH2Cl2 (3 x 2 mL). The reaction mixture was allowed to stir at -10 C. After 1.5 hours, the reaction mixture was diluted with 1:1 H2O : saturated aqueous NH4Cl (100 mL) and the phases were partitioned. The organic layer was washed with water (1 x 100 mL), and the combined aqueous layers were further extracted with 3:1 CHCl3 : i-PrOH (3 x 200 mL). The combined organic extracts were washed with saturated aqueous NaHCO3 (1 x 200 mL), brine (1 x 200 mL), dried over Na2SO4, filtered, and concentrated in vacuo. The combined crude residue was purified by column chromatography (SiO2, 1% to 10% MeOH in CH2Cl2) affording Intermediate AC (5.01 g, 96% yield). LCMS observed m/z = 698.10 [M+H]+. Intermediate AD Synthesis of tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate Intermediate AD
To a solution of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate, Intermediate F (77.6 g, 117 mmol, 1 eq) and the corresponding boronate ester (30.2 g, 122 mmol, 1.05 eq) in dioxane (776 mL) and H2O (115 mL) was added K3PO4 (74.5 g, 351 mmol, 3 eq) and Pd(dppf)Cl2 (4.28 g, 5.85 mmol, 0.05 eq) under N2. The mixture was stirred at 80 °C for 16 hrs under N2. The residue was diluted with water (1200 mL) and extracted with ethyl acetate (1200 mL * 4). The combined organic layers were washed with aq. NaCl (1200 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. TLC (Petroleum ether: Ethyl acetate = 1: 1, Rf/product = 0.40). The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 1/0 to 0/1, CH2Cl2) at
twice. The product was dissolved with CH2Cl2 (1.5 L). The mixture was added Pd scavenger (70.0 g) and stirred at 38 °C for 30 mins, filtered and concentrated under the reduced pressure to give tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (67.0 g, 95.4 mmol, 81.5% yield) as a white solid. LCMS: m/z = 646.4 [M-56+1]+. Intermediate AE Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide Intermediate AE
To a solution of tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate, Intermediate AD (74.6 g, 106 mmol, 1 eq) was added HCl/dioxane (2 M, 746 mL, 14.04 eq) in portions. The mixture was stirred at 25 °C for 16 hrs. The reaction mixture was concentrated under reduced pressure to remove solvent. N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (68.1 g, crude, HCl) was obtained as a white solid. LCMS: m/z = 602.2 [M+1]+. Synthesis of tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate Intermediate AF
To a solution of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate, Intermediate F (10.0 g, 15.1 mmol, 1 eq) and the corresponding boronic acid (2.9 g, 15.1 mmol, 1.0 eq) in dioxane (50 mL) and H2O (15 mL) was added K3PO4 (9.61 g, 45.3 mmol, 3 eq) and Pd(dppf)Cl2 (616 mg, 0.754 mmol, 0.05 eq) under N2. The mixture was stirred at 80 °C for 16 hrs under N2. The residue was diluted with water (120 mL) and extracted with ethyl acetate (120 mL * 4). The combined organic layers were washed with aq. NaCl (120 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. TLC (Petroleum ether: Ethyl acetate = 1: 1, Rf/product = 0.40). The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 1/0 to 0/1, CH2Cl2). The product was dissolved with CH2Cl2 (1.5 L). The mixture was added Pd scavenger (10.0 g) and stirred at 38 °C for 30 mins, filtered and concentrated under the reduced pressure to givetert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (8.33 g, 11.4 mmol, 75.6% yield) as a white solid. LCMS: m/z = 674.2 [M-56+1]+. Intermediate AG Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide Intermediate AG
To a solution of tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate, Intermediate AF (8.33 g, 11.4 mmol, 1 eq) was added HCl/dioxane (4 M, 21 mL, 50 eq) in portions. The mixture was stirred at 25 °C for 16 hrs. The reaction mixture was concentrated under reduced pressure to remove solvent. N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (7.7 g, crude, HCl) was obtained as a white solid. LCMS: m/z = 630.2 [M+1]+. Example 1 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3- dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 1
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
To a mixture of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (125 mg, 0.158 mmol, Intermediate F), (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid (42.8 mg, 0.238 mmol) and Cs2CO3 (33.6 mg, 0.317 mmol) in a mixture of dioxane (1.5 mL) and H2O (0.5 mL) was added Pd(dppf)Cl2.CH2Cl2 (12.9 mg, 0.0158 mmol). The mixture was stirred at 100 °C under N2 for 16 hours. The mixture was concentrated in vacuo. The residue was dissolved in CH2Cl2 and filtered through a pad of Celite and the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 10-75% EtOAc in Hexanes) to afford (70.4 mg, 75% yield) of 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a brown solid. LCMS observed m/z = 844.3 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3- dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 1
To a mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (70.4 mg, 0.0834 mmol) in CH2Cl2 (1.67 mL) was added dropwise a 1 molar solution of boron trichloride in CH2Cl2 (0.5 mL, 0.5 mmol). The mixture was stirred at -40 °C under N2 for 15 minutes. The mixture was quenched with MeOH (1.0 mL) and warmed to 25 °C. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0- 100% MeCN in H2O) to afford (8.2 mg, 12.% yield) of N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide as a white solid.1H NMR (400MHz, DMSO-d6) δ 10.40 (s, 1H), 8.56 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 7.34 (d, J = 7.6 Hz, 1H), 7.00 (d, J = 8.0 Hz, 1H), 6.91 (t, J = 7.8 Hz, 1H), 5.37 (s, 2H), 4.53 (d, J = 12.5 Hz, 1H), 4.29 (s, 4H), 3.52 (q, J = 11.9 Hz, 3H), 3.24 (d, J = 12.4 Hz, 2H), 3.01 (d, J = 10.0 Hz, 3H), 2.83 (d, J = 11.3 Hz, 1H), 2.66 (d, J = 10.9 Hz, 1H), 2.44 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 754.3 [M+H]+. EXAMPLE 2 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3- dihydrobenzo[b][1,4]dioxin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 2
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with (2,3-dihydrobenzo[b][1,4]dioxin- 5-yl)boronic acid.1H NMR (400MHz, DMSO-d6) δ 10.36 (s, 1H), 8.48 (s, 1H), 7.97 (d, J = 8.6 Hz, 1H), 7.91 (s, 1H), 7.65 (d, J = 8.7 Hz, 1H), 7.52 (d, J = 8.5 Hz, 1H), 7.47 (s, 1H), 6.92 (d, J = 8.4 Hz, 1H), 5.30 (s, 2H), 4.46 (d, J = 12.5 Hz, 1H), 4.23 (s, 4H), 3.45 (q, J = 11.6 Hz, 4H), 2.94 (q, J = 10.4 Hz, 3H), 2.76 (d, J = 11.2 Hz, 1H), 2.58 (d, J = 11.4 Hz, 1H), 2.37 (s, 3H), 1.13 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 754.3 [M+H]+. EXAMPLE 3 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(isochroman-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 3
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(isochroman-6-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, DMSO-d6) δ 10.44 (s, 1H), 8.48 (s, 1H), 8.36 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (s, 1H), 7.90 (s, 2H), 7.72 (d, J = 8.7 Hz, 1H), 7.19 (d, J = 8.1 Hz, 2H), 5.40 (s, 2H), 4.74 (s, 2H), 4.55 (d, J = 12.6 Hz, 1H), 3.91 (t, J = 5.8 Hz, 2H), 3.53 (d, J = 12.7 Hz, 3H), 3.03 (d, J = 10.3 Hz, 3H), 2.85 (d, J = 15.4 Hz, 3H), 2.68 (s, 1H), 2.42 (s, 3H), 1.22 (t, J = 7.8 Hz, 3H). LCMS observed m/z = 752.55 [M+H]+. EXAMPLE 4
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(isochroman-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 4
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(3,4-dihydro-1H-2-benzopyran- 5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 11.80 (s, 1H), 9.10 (s, 1H), 8.61 (s, 1H), 8.51 (d, J = 8.7 Hz, 1H), 8.03 (d, J = 7.7 Hz, 1H), 7.61 (d, J = 2.0 Hz, 1H), 7.54 (dd, J = 8.7, 2.0 Hz, 1H), 7.31 (s, 1H), 7.13 (d, J = 7.6 Hz, 1H), 5.69 (d, J = 13.1 Hz, 1H), 5.20 (s, 2H), 4.86 (s, 3H), 3.99 (t, J = 5.7 Hz, 2H), 3.86 (t, J = 11.7 Hz, 2H), 3.53 (s, 1H), 3.36 (t, J = 5.9 Hz, 2H), 3.21 - 3.27 (m, 2H), 3.11 (s, 1H), 2.87 – 2.81 (m, 2H), 2.59 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.30 [M+H]+. EXAMPLE 5 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(isochroman-8-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 5
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(isochroman-8-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 11.81 (s, 1H), 9.05 (s, 1H),
8.62 (s, 1H), 8.51 (d, J = 8.7 Hz, 1H), 8.11 – 8.05 (m, 1H), 7.61 (d, J = 2.0 Hz, 1H), 7.55 (d, J = 9.2 Hz, 1H), 7.32 (d, J = 7.6 Hz, 1H), 5.72 – 5.68 (m, 1H), 5.27 (s, 2H), 5.19 (s, 2H), 4.86 – 4.79 (m, 1H), 4.01 (t, J = 5.7 Hz, 2H), 3.91 – 4.87 (m, 2H), 3.54 (s, 1H), 3.24 (d, J = 7.8 Hz, 2H), 3.11 (s, 1H), 2.99 (t, J = 5.7 Hz, 2H), 2.88 – 2.82 (m, 2H), 2.60 (s, 3H), 1.40 (d, J = 7.5 Hz, 3H). LCMS observed m/z = 752.35 [M+H]+. EXAMPLE 6 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(chroman-5-yl)-5-ethyl- 6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 6
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(chroman-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. EXAMPLE 7 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(chroman-8-yl)-5-ethyl- 6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 7
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(chroman-8-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. EXAMPLE 8 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,8-dihydro-6H- pyrano[3,4-b]pyridin-3-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 8
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-5,8-dihydro-6H-pyrano[3,4-b]pyridine. EXAMPLE 9 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,4-dihydro-1H- pyrano[4,3-c]pyridin-8-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 9
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 8-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3,4-dihydro-1H-pyrano[4,3-c]pyridine. EXAMPLE 10
Synthesis of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 10
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2H-1,3-benzodioxol-5-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 11.94 (s, 1H), 9.14 (s, 1H), 8.64 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 7.86 (dd, J = 8.1, 1.7 Hz, 1H), 7.76 (d, J = 1.7 Hz, 1H), 7.62 (s, 1H), 7.56 (s, 1H), 6.91 (d, J = 8.1 Hz, 1H), 6.06 (s, 2H), 5.68 (s, 1H), 5.18 (s, 2H), 4.83 (d, J = 12.9 Hz, 1H), 3.86 (t, J = 11.6 Hz, 2H), 3.24 (d, J = 7.9 Hz, 1H), 3.11 (s, 3H), 2.88 – 2.82 (m, 2H), 2.63 (d, J = 7.6 Hz, 3H), 1.38 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 740.35 [M+H]+. EXAMPLE 11 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3-dihydrobenzofuran- 5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 11
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2,3-dihydro-1-benzofuran-5- yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 11.84 (s, 1H), 9.23 (s, 1H), 8.62 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.17 (d, J = 1.8 Hz, 1H), 8.08 (dd, J =
8.3, 1.8 Hz, 1H), 7.60 (d, J = 2.0 Hz, 1H), 7.55 (dd, J = 8.7, 2.0 Hz, 1H), 6.87 (d, J = 8.4 Hz, 1H), 5.72 – 5.68 (m, 1H), 5.18 (s, 2H), 4.85 – 4.81 (m, 1H), 4.68 (t, J = 8.7 Hz, 2H), 3.88 – 3.84 (m, 2H), 3.56 – 3.52 (m, 1H), 3.34 - 3.19 (m, 4H), 3.10 (s, 1H), 2.86 – 2.81 (m, 2H), 2.59 (s, 3H), 1.38 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 738.15 [M+H]+. EXAMPLE 12 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3-dihydrobenzofuran- 6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 12
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2,3-dihydro-1-benzofuran-6- yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 12.14 (s, 1H), 9.18 (s, 1H), 8.66 (s, 1H), 8.51 (d, J = 8.7 Hz, 1H), 7.83 (d, J = 7.7 Hz, 1H), 7.69 (s, 1H), 7.61 (s, 1H), 7.54 (d, J = 8.8 Hz, 1H), 7.31 (s, 1H), 5.65 (s, 1H), 5.19 (s, 2H), 4.86 – 3.78 (m, 1H), 4.69 – 4.57 (m, 2H), 3.91 – 3.84 (m, 2H), 3.55 (s, 1H), 3.34 – 3.19 (m, 4H), 3.12 (s, 1H), 2.87 – 2.83 (m, 2H), 2.68 (s, 3H), 1.38 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 738.15 [M+H]+. EXAMPLE 13 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 13
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(1,3-dihydroisobenzofuran-5- yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO-d6) δ 10.32 (s, 1H), 8.54 (s, 1H), 8.12 – 8.01 (m, 3H), 7.98 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.47 (d, J = 8.0 Hz, 1H), 5.40 (s, 2H), 5.07 (d, J = 5.3 Hz, 4H), 4.60 – 4.49 (m, 1H), 3.64 – 3.45 (m, 4H), 3.27 – 3.24 (m, 1H) 3.03 (d, J = 9.8 Hz, 3H), 2.88 – 2.82 (m, 1H), 2.71 – 2.65 (m, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 738.20 [M+H]+. EXAMPLE 14 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,7-dihydrofuro[3,4- b]pyridin-3-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 14
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(5,7-dihydrofuro[3,4-b]pyridin-3-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoro methyl)phenyl)acetamide. Compound 14.1
A mixture of 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,7-dihydrofuro[3,4- b]pyridine (60 mg, 0.2 mmol,CAS# 1648749-95-4), K2CO3 (100 mg, 0.7 mmol), Pd(dppf)Cl2.CH2Cl2 (35 mg, 0.05 mmol) and 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (191 mg, 0.2 mmol, Intermediate H) in dioxane (5 mL) and H2O (0.5 mL) was stirred at 100 °C for 2 hours under N2. The precipitated solids were collected by filtration and washed with EtOAc (3 x 10 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in CH2Cl2) to afford (60 mg, 29% yield) of 2-(6- (4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(5,7-dihydrofuro[3,4- b]pyridin-3-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a yellow solid. LCMS (ESI): [M+H] + = 829.25. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,7- dihydrofuro[3,4-b]pyridin-3-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 14
A solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(5,7-dihydrofuro[3,4-b]pyridin-3-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (50 mg, 0.06 mmol) in TFA (1
mL) was stirred at 80 °C for 30 minutes. The resulting mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: XBridge BEH C185 μm, 30*150 mm; mobile phase: 27-37% MeCN in H2O) to afford (22.9 mg, 50% yield) of N-(2-chloro- 4-(trifluoromethyl)phenyl)-2-(2-(5,7-dihydrofuro[3,4-b]pyridin-3-yl)-5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl) acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.31 (s, 2H), 9.15 (d, J = 1.8 Hz, 1H), 8.57 (s, 1H), 8.40 (d, J = 1.9 Hz, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 5.42 (s, 2H), 5.16 (s, 2H), 5.01 (d, J = 1.9 Hz, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.61 – 3.46 (m, 3H), 3.29 – 3.21 (m, 1H), 3.03 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.68 (d, J = 9.9 Hz, 1H), 2.45 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS (ESI): [M+H] + = 739.20. EXAMPLE 15 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(1,3-dihydro-2-benzofuran-4- yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO-d6) δ 10.44 (s, 1H), 8.56 (s, 1H), 8.06 (dd, J = 6.3, 3.5 Hz, 2H), 8.00 – 7.95 (m, 1H), 7.73 – 7.71 (m, 1H), 7.49 – 7.42 (m, 2H), 5.46 – 5.33 (m, 4H), 5.08 (s, 2H), 4.58 – 4.49 (m, 1H), 3.59 – 3.47 (m, 3H), 3.27 – 3.25 (m, 2H), 3.08 –3.03 (m, 3H), 2.88 – 2.82 (m, 1H), 2.68 – 2.67 (m, 1H) 2.45 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 738.15 [M+H]+. EXAMPLE 16 Synthesis of 2-(2-(benzo[d][1,3]dioxol-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide.
Compound 16
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2H-1,3-benzodioxol-4-ylboronic acid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 2H), 8.58 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.73 (dd, J = 8.7, 2.2 Hz, 1H), 7.52 (dd, J = 8.1, 1.3 Hz, 1H), 7.06 (dd, J = 7.7, 1.3 Hz, 1H), 6.97 (t, J = 7.9 Hz, 1H), 6.17 (s, 2H), 5.39 (s, 2H), 4.58 – 4.52 (m, 1H), 3.60 – 3.47 (m, 3H), 3.33 – 3.28 (m, 1H), 3.03 (d, J = 9.1 Hz, 3H), 2.87 – 2.81 (m, 1H), 2.71 – 2.62 (m, 1H), 2.45 (s, 3H), 1.25 – 1.18 (m, 3H). LCMS observed m/z = 740.30 [M+H]+. EXAMPLE 17 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3-dihydrobenzofuran- 4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 17
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2,3-dihydrobenzofuran-4-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane. EXAMPLE 18 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3-dihydrobenzofuran- 7-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Compound 18
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-(2,3-dihydrobenzofuran-7-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane. EXAMPLE 19 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3-dihydrofuro[2,3- b]pyridin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2,3-dihydrofuro[2,3-b]pyridine. EXAMPLE 20 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(6,7-dihydro-4H- pyrazolo[5,1-c][1,4]oxazin-3-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 20
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazine. 1H NMR (400 MHz, CDCl3) δ 11.82 (s, 1H), 9.02 (s, 1H), 8.62 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 8.15 (s, 1H), 7.66 – 7.61 (m, 1H), 7.56 (dd, J = 8.7, 2.0 Hz, 1H), 5.71 – 5.65 (m, 1H), 5.26 – 5.04 (m, 4H), 4.84 – 4.78 (m, 1H), 4.29 (s, 2H), 4.17 (s, 2H), 3.88 – 3.79 (m, 2H), 3.52 (s, 1H), 3.21 (d, J = 7.5 Hz, 2H), 3.10 (s, 1H), 2.88 – 2.81 (m, 2H), 2.59 (s, 3H), 1.42 – 1.32 (m, 3H). LCMS observed m/z = 742.20 [M+H]+. EXAMPLE 21 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylbenzo[d]oxazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazole. 1H NMR (400 MHz, Methanol- d4) δ 8.55 – 8.51 (m, 1H), 8.47 (d, J = 1.7 Hz, 1H), 8.29 (dd, J = 8.6, 1.7 Hz, 1H), 8.18 (d, J = 8.6 Hz, 1H), 7.84 (d, J = 2.1 Hz, 1H), 7.69 (d, J = 8.7 Hz, 1H), 7.63 (dd, J = 8.7, 2.1 Hz, 1H), 5.50 (s, 2H), 4.73 (s, 1H), 4.06 (s, 1H), 3.84 – 3.73 (m, 2H), 3.51 (s, 1H), 3.20 – 3.13 (m, 3H), 3.02 – 2.97 (m, 1H), 2.88 – 2.80 (m, 1H), 2.69 (s, 3H), 2.57 – 2.48 (m, 3H), 1.43 – 1.33 (m, 3H). LCMS observed m/z = 751.35 [M+H]+.
EXAMPLE 22 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-1H-indazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 22
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 1-methyl-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)indazole.1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 2H), 8.71 (s, 1H), 8.57 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 8.02 – 7.96 (m, 2H), 7.83 (d, J = 8.4 Hz, 1H), 7.74 – 7.67 (m, 1H), 7.59 – 7.51 (m, 1H), 5.49 (s, 2H), 4.59 – 4.53 (m, 1H), 4.12 (s, 3H), 3.61 – 3.51 (m, 3H), 3.29 (s, 1H), 3.11 – 2.98 (m, 3H), 2.92 – 2.85 (m, 1H), 2.73 – 2.67 (m, 1H), 2.46 (s, 3H), 1.24 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.20 [M+H]+. EXAMPLE 23 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylbenzo[d]oxazol-7-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 1, replacing (2,3-dihydrobenzo[b][1,4]dioxin-6-yl)boronic acid with 2-methyl-7-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)benzo[d]oxazole.1H NMR (400 MHz, CDCl3) δ 11.92 (s, 1H), 9.12 (s, 1H), 8.63 (s, 1H), 8.51 (d, J = 8.7 Hz, 1H), 8.18 (dd, J = 7.8, 1.2 Hz, 1H), 7.81 (dd, J = 7.9, 1.2 Hz, 1H), 7.60 (d, J = 2.0 Hz, 1H), 7.55 (d, J = 9.3 Hz, 1H), 7.43 (t, J = 7.9 Hz, 1H),
5.73 – 5.67 (m, 1H), 5.26 (s, 2H), 4.87 – 4.81 (m, 1H), 3.93 - 3.84 (m, 2H), 3.55 (s, 1H), 3.26 (d, J = 7.7 Hz, 2H), 3.13 (s, 1H), 2.89 – 2.83 (m, 2H), 2.73 (s, 3H), 2.63 (d, J = 3.2 Hz, 3H), 1.40 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 751.20 [M+H]+. EXAMPLE 24 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-1H-pyrazolo[3,4-c]pyridin-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 24
Step 1: Preparation of 1-methyl-4-(tributylstannyl)pyrazolo[3,4-c]pyridine. Compound 24.1
To a solution of 4-bromo-1-methylpyrazolo[3,4-c]pyridine (100 mg, 0.5 mmol) and 1,1,1,2,2,2-hexabutyldistannane (820 mg, 1.4 mmol) in dioxane (2 mL) were added Pd(dppf)Cl2.CH2Cl2 (55 mg, 0.05 mmol) in one portion at 25 °C under N2. The mixture was heated to 100 °C and stirred at 100 °C for 16 hours under N2. The mixture was cooled to 25 °C, quenched with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 30-90% MeCN in H2O) to afford (113 mg, 57% yield) of 1-methyl-4-(tributylstannyl)pyrazolo[3,4-c]pyridine as a yellow oil. LCMS observed m/z = 424.2 [M+H]+. Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(1-methyl-1H-pyrazolo[3,4-c]pyridin-4-yl)-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 24.2
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (100 mg, 0.1 mmol, Intermediate H) and 1-methyl-4-(tributylstannyl)pyrazolo[3,4-c]pyridine (107 mg, 0.3 mmol) in DMF (5 mL) was added Pd(dppf)Cl2.CH2Cl2 (9 mg, 0.01 mmol) in one portion at 25 °C under N2. The mixture was heated to 130 °C and stirred at 130 °C for 16 hours under N2. The mixture was cooled to 25 °C, diluted with H2O (50 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with H2O (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 10-60% MeCN in H2O) to afford (65 mg, 61% yield) of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(1-methyl-1H-pyrazolo[3,4-c]pyridin-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a brown oil. LCMS observed m/z = 841.3 [M+H]+. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-1H-pyrazolo[3,4- c]pyridin-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 24
To a solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-5-ethyl-2-{1-methylpyrazolo[3,4-c]pyridin-4-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (60 mg, 0.07 mmol) in CH2Cl2 (2 mL) was added boron trichloride (41 mg, 0.4 mmol) in one portion at 0 °C. The mixture was stirred for 2 hours at 25 °C under N2. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 10-50% MeCN in H2O) to afford (3.9 mg, 7% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{1-methylpyrazolo[3,4- c]pyridin-4-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.47 (s, 1H), 9.32 (s, 1H), 9.00 (s, 1H), 8.71 (s, 1H), 8.56 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.7, 2.1 Hz, 1H), 5.50 (s, 2H), 4.59 – 4.53 (m, 1H), 4.26 (s, 3H), 3.63 – 3.46 (m, 4H), 3.15 – 3.00 (m, 4H), 2.91 – 2.85 (m, 1H), 2.73 – 2.67 (m, 1H), 2.45 (s, 3H), 1.25 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 751.20 [M+H]+. EXAMPLE 25 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,4-oxazepan-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 25
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(1,4-oxazepan-4-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 25.1
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (200 mg, 0.253 mmol, Intermediate H) and 1,4- oxazepane hydrogen chloride (105 mg, 0.760 mmol) dissolved in a mixture of DMF (0.75 mL) and DMSO (0.75 mL) was added KOAc (207 mg, 1.52 mmol). The mixture was stirred at 100 °C for 16 hours. The mixture was concentrated in vacuo and the residue was purified by silica gel column chromatography (mobile phase: 0-50% of 20% MeOH in CH2Cl2) to afford (56 mg, 27%) of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a brown solid. LCMS observed m/z = 809.3 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,4-oxazepan-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 25
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-5-ethyl-2-(1,4-oxazepan-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide (56 mg, 0.0692 mmol) in CH2Cl2 (1.38 mL) was added dropwise a 1 molar solution of boron trichloride in dichloromethane (0.415 mL, 0.415 mmol). The mixture was stirred at -40 °C under N2 for 15 minutes. The mixture was quenched with MeOH (1.0 mL) and warmed to 25 °C. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (29 mg, 58% yield) of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6- (4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,4-oxazepan-4-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400MHz, DMSO-d6) δ 10.29 (s, 2H), 8.49 (d, J = 1.8 Hz, 1H), 8.10 – 7.81 (m, 2H), 7.66 (d, J = 8.7 Hz, 1H), 5.14 (s, 2H), 4.44 (d, J = 12.5 Hz, 1H), 3.58 (tt, J = 15.7, 5.2 Hz, 7H), 3.50 – 3.35 (m, 3H), 3.16 (t, J = 12.6 Hz, 1H), 2.96 – 2.80 (m, 3H), 2.71 (d, J = 11.3 Hz, 1H), 2.53 (d, J = 11.0 Hz, 1H), 1.77 (p, J = 5.7 Hz, 2H), 1.09 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 719.3 [M+H]+. EXAMPLE 26 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-oxa-6-azaspiro[3.3]heptan-6-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 26
Step 1: Preparation of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 26.1
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (250 mg, 0.317 mmol, Intermediate H) in CH2Cl2 (6.34 mL) was added dropwise a 1 molar solution of boron trichloride in dichloromethane (1.9 mL, 1.9 mmol). The mixture was stirred at -40 °C under N2 for 15 minutes. The mixture was quenched with MeOH (1.0 mL) and warmed to 25 °C. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (221 mg, 67% yield) of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide.1H NMR (400MHz, DMSO- d6) δ 10.35 (s, 1H), 10.24 (s, 1H), 8.58 (s, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.98 (s, 1H), 7.73 (d, J = 8.7 Hz, 1H), 5.32 (s, 2H), 4.53 (d, J = 12.5 Hz, 1H), 3.47 (q, J = 9.1 Hz, 4H), 3.25 (t, J = 13.2 Hz, 1H), 3.05 – 2.94 (m, 3H), 2.82 (d, J = 11.3 Hz, 1H), 2.64 (d, J = 11.1 Hz, 1H), 2.45 (s, 3H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 698.1 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-oxa-6- azaspiro[3.3]heptan-6-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
A mixture of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (70 mg, 0.10 mmol) and 2-oxa-6-azaspiro[3.3]heptane
(29.8 mg, 0.300 mmol) dissolved in a mixture of DMF (0.4 mL) and DMSO (0.4 mL) was added KOAc (81.9 mg, 0.601 mmol). The mixture was stirred at 100 °C for 16 hours. The mixture was concentrated in vacuo and the residue was purified by silica gel column chromatography (mobile phase: 0-50% of 20% MeOH in CH2Cl2) to afford (8.5 mg, 11% yield) of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-oxa-6-azaspiro[3.3]heptan-6-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400MHz, DMSO-d6) δ 10.29 (s, 1H), 8.40 (s, 1H), 8.00 (d, J = 8.5 Hz, 1H), 7.90 (s, 1H), 7.65 (d, J = 8.7 Hz, 1H), 5.15 (s, 2H), 4.62 (s, 3H), 4.43 (d, J = 12.4 Hz, 1H), 4.06 (s, 4H), 3.40 (d, J = 12.9 Hz, 4H), 2.86 (t, J = 9.2 Hz, 4H), 2.68 (d, J = 11.6 Hz, 1H), 2.51 (d, J = 11.6 Hz, 1H), 2.34 (s, 3H), 1.07 (t, J = 7.3 Hz, 3H). LCMS observed m/z = 717.3 [M+H]+. EXAMPLE 27 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3-methoxyazetidin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 27
The title compound was prepared using similar procedure as Example 26, replacing 2-oxa-6-azaspiro[3.3]heptane with 3-methoxyazetidine.1H NMR(400MHz, DMSO-d6) δ 10.35 (s, 1H), 8.50 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.6 Hz, 1H), 6.61 (s, 1H), 5.21 (s, 2H), 4.50 (d, J = 12.4 Hz, 1H), 4.30 (t, J = 5.6 Hz, 1H), 4.16 (t, J = 7.6 Hz, 2H), 3.78 (dd, J = 9.1, 4.1 Hz, 2H), 3.48 (d, J = 12.7 Hz, 4H), 3.21 (d, J = 1.7 Hz, 3H), 2.94 (d, J = 12.2 Hz, 3H), 2.76 (d, J = 11.5 Hz, 1H), 2.59 (d, J = 10.7 Hz, 1H), 2.42 (s, 3H), 1.15 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 705.3 [M+H]+. EXAMPLE 28 Synthesis of 2-(2-(azetidin-1-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
Compound 28
The title compound was prepared using similar procedure as Example 26, replacing 2-oxa-6-azaspiro[3.3]heptane with azetidine.1H NMR (400 MHz, Methanol-d4) δ 8.55 (d, J = 1.8 Hz, 1H), 8.19 (d, J = 8.6 Hz, 1H), 7.83 (d, J = 2.1 Hz, 1H), 7.64 (dd, J = 8.6, 2.1 Hz, 1H), 5.29 (s, 2H), 4.74 – 4.88 (m, 1H), 4.13 – 4.09 (m, 5H), 3.74 (s, 2H), 3.46 (s, 2H), 3.19 – 3.12 (m, 1H), 3.11 – 3.01 (m, 2H), 2.95 – 2.88 (m, 1H), 2.80 – 2.76 (m, 1H), 2.47 – 2.38 (m, 2H), 1.38 – 1.27 (m, 6H). LCMS observed m/z = 675.20 [M+H]+. EXAMPLE 29 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-oxa-6-azaspiro[3.4]octan-6-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 29
The title compound was prepared using similar procedure as Example 26, replacing 2-oxa-6-azaspiro[3.3]heptane with 2-oxa-6-azaspiro[3.4]octane. 1H NMR (400MHz, DMSO-d6) δ 10.31 (s, 1H), 8.41 (s, 1H), 7.98 (d, J = 8.6 Hz, 1H), 7.90 (s, 1H), 7.65 (d, J = 8.6 Hz, 1H), 6.57 (s, 1H), 5.15 (s, 2H), 4.49 (d, J = 6.1 Hz, 2H), 4.43 (t, J = 7.0 Hz, 3H), 3.54 (s, 2H), 3.42 (q, J = 12.1 Hz, 3H), 3.32 (d, J = 6.8 Hz, 1H), 3.20 – 3.09 (m, 1H), 2.91 – 2.82 (m, 4H), 2.69 (d, J = 11.3 Hz, 1H), 2.51 (d, J = 10.3 Hz, 1H), 2.34 (s, 3H), 2.14 (t, J = 6.9 Hz, 2H), 1.08 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 731.2 [M+H]+. EXAMPLE 30
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl- 6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 30
The title compound was prepared using similar procedure as Example 25, replacing 1,4-oxazepane hydrogen chloride with dimethylamine.1H NMR(400MHz, DMSO-d6) δ 10.36 (s, 2H), 8.56 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 5.21 (s, 2H), 4.51 (d, J = 12.5 Hz, 1H), 3.50 (q, J = 11.6 Hz, 3H), 3.23 (t, J = 12.6 Hz, 2H), 2.96 (s, 6H), 2.95-2.92 (m, 2H), 2.77 (d, J = 11.5 Hz, 1H), 2.59 (d, J = 10.9 Hz, 1H), 2.44 (s, 3H), 1.16 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 663.2 [M+H]+. EXAMPLE 31 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(6,7- dihydropyrazolo[1,5-a]pyrimidin-4(5H)-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{5H,6H,7H-pyrazolo[1,5-a]pyrimidin-4-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 31.1
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide (150 mg, 0.2 mmol, Intermediate H) and 4H,5H,6H,7H-pyrazolo[1,5-a]pyrimidine (47 mg, 0.4 mmol) in DME (2 mL) were added Pd2(dba)3 (17 mg, 0.02 mmol), XPhos (36 mg, 0.08 mmol) and K3PO4 (57 mg, 0.3 mmol) at 25 °C under N2. The mixture was stirred at 85 °C for 2 hours under N2. The resulting mixture was diluted with H2O (20 mL) and extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in CH2Cl2/MeOH) to afford (70 mg, 44% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl- 7-oxo-2-{5H,6H,7H-pyrazolo[1,5-a]pyrimidin-4-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)- N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 831.3 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{5H,6H,7H- pyrazolo[1,5-a]pyrimidin-4-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
To a solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-5-ethyl-7-oxo-2-{5H,6H,7H-pyrazolo[1,5-a]pyrimidin-4-yl}-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (60 mg, 0.07 mmol) in CH2Cl2 (1 mL) was added TFA (2 mL) at 25 ℃ under N2. The resulting mixture was stirred at 25 °C for 16 hours under N2. The mixture was concentrated in vacuo. The residue was purified by reversed phase flash chromatography (Column: YMC Triart C18 5 m, 30 mm * 150 mm; mobile phase: 23-46% MeCN in H2O) to afford (12.9 mg, 24% yield) of N-[2- chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-{5H,6H,7H-pyrazolo[1,5-a]pyrimidin-4-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a light yellow solid.1H NMR(400MHz, DMSO-d6) δ 10.43 (s, 1H), 8.58 (s, 1H), 8.04 (d, J = 8.4 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.26 (d, J = 2.0 Hz, 1H), 6.61 (d, J = 2.0 Hz, 1H), 5.32 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 4.21 – 4.07 (m, 2H), 4.01 – 3.90 (m, 2H), 3.57 – 3.48 (m, 3H), 3.27 – 3.17 (m, 1H), 3.11 – 2.92 (m, 3H), 2.82 (m, 1H), 2.68 – 2.59 (m, 1H), 2.45 (s, 3H), 2.19 (s, 2H), 1.28 – 1.13 (m, 3H). LCMS observed m/z = 741.20 [M+H]+. EXAMPLE 32 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(3-hydroxy-4-oxo-4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 32
Step 1: Preparation of 2-(6-(4-(3-(benzyloxy)-4-oxo-4,6,7,9- tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 32.1
A mixture of 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2- carboxylic acid (19 mg, 0.1 mmol, Intermediate L) and HATU (26 mg, 0.06 mmol) in DMF (10 mL) was added N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (30 mg, 0.05 mmol, Intermediate K) and the mixture was stirred at 25 °C under N2 for 2 hours. The mixture was quenched with H2O and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford (40 mg crude) of 2-(6-{4-[3- (benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carbonyl]piperazin-1-yl}-2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide as a yellow oil. The crude product was used directly in the next step without further purification. LCMS observed m/z =850.3 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(4-(3-hydroxy-4-oxo-4,6,7,9-tetrahydropyrimido[2,1- c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 32
A mixture of 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1- c][1,4]oxazine-2-carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (40 mg, 0.05 mmol) and boron trichloride (2 mL) in CH2Cl2 (20 mL) was stirred at 25 °C under N2 for 1 hour. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 19-42% MeCN in H2O) to afford (4 mg, 11% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-{3-hydroxy-4- oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carbonyl}piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5 -a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 6.84 (s, 1H), 5.32 (s, 2H), 4.59 (s, 2H), 4.49 – 4.35 (m, 1H), 4.26 (s, 2H), 4.04 (t, J = 5.4 Hz, 2H), 3.90 – 3.77 (m, 4H), 3.62 – 3,52 (m, 1H), 3.50 – 3.39 (m, 3H), 3.27 – 3.16 (m, 2H), 3.06 – 2.90 (m, 3H), 2.85 – 2.71 (m, 2H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 760.40 [M+H]+. Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(6,7- dihydropyrazolo[1,5-a]pyrimidin-4(5H)-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. EXAMPLE 33 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(3-hydroxy-1-methyl-1H-pyrazole-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 33
The title compound was prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with 3-(benzyloxy)-1-methyl-1H-pyrazole-4-carboxylic acid. 1H NMR(400MHz, DMSO-d6) δ 10.58 (s, 1H), 10.37 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.88 (s, 1H), 7.72 (d, J = 8.8 Hz, 1H), 6.83 (s, 1H), 5.32 (s, 2H), 4.25 (s, 3H), 3.80 (t,
J = 5.3 Hz, 2H), 3.65 (s, 2H), 3.52 – 3.39 (m, 2H), 3.00 (d, J = 7.8 Hz, 4H), 2.84 – 2.64 (m, 3H), 2.54 (s, 3H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 690.3 [M+H]+. EXAMPLE 34 Synthesis of 2-(6-(4-(3-aminopicolinoyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
A mixture of 3-aminopicolinic acid (8.79 mg, 0.0636 mmol) in DMF (0.5 mL) was added HATU (30.2 mg, 0.0795 mmol). The solution was stirred at 25 °C for 15 minutes. To the mixture was added N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (30 mg, 0.053 mmol, Intermediate K) and DIPEA (0.047 mL, 0.265 mmol). The mixture was stirred at 25 °C for 30 minutes. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (10.0 mg, 28% yield) of 2-(6-(4-(3-aminopicolinoyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide as a white solid. 1H NMR (400MHz, DMSO- d6) δ 10.38 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.80 (t, J = 2.9 Hz, 1H), 7.77 – 7.67 (m, 1H), 7.13 (d, J = 2.9 Hz, 2H), 6.83 (dd, J = 3.3, 1.7 Hz, 1H), 5.52 (s, 2H), 5.31 (s, 2H), 4.56 (d, J = 12.4 Hz, 1H), 4.25 (d, J = 2.9 Hz, 2H), 3.80 (t, J = 5.6 Hz, 3H), 3.59 – 3.41 (m, 2H), 3.24 (t, J = 12.3 Hz, 3H), 2.99 (d, J = 10.3 Hz, 3H), 2.80 (d, J = 11.4 Hz, 1H), 2.64 (d, J = 10.9 Hz, 1H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 686.3 [M+H]+. EXAMPLE 35 Synthesis of 2-(4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carbonyl)pyridine 1-oxide. Compound 35
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-carboxypyridine 1-oxide. 1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.29 (d, J = 6.2 Hz, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 7.63 (dd, J = 7.6, 2.3 Hz, 1H), 7.56 – 7.35 (m, 2H), 6.83 (s, 1H), 5.31 (s, 2H), 4.57 – 4.38 (m, 1H), 4.25 (d, J = 3.1 Hz, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.69 – 3.38 (m, 2H), 3.16 (dt, J = 22.0, 12.5 Hz, 2H), 2.99 (s, 3H), 2.81 (t, J = 13.7 Hz, 1H), 2.65 (d, J = 9.2 Hz, 1H), 2.54 (s, 2H), 1.18 (dt, J = 8.1, 5.3 Hz, 3H). LCMS observed m/z = 687.2 [M+H]+. EXAMPLE 36 Synthesis of 2-(6-(4-(3-amino-4-methylpicolinoyl)piperazin-1-yl)-2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 3-amino-4-methylpicolinic acid. 1H NMR (400MHz, DMSO- d6) δ 10.32 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.90 (d, J = 2.2 Hz, 1H), 7.70 – 7.61 (m, 2H), 6.99 (d, J = 4.6 Hz, 1H), 6.76 (dq, J = 3.0, 1.5 Hz, 1H), 5.25 (s, 2H), 5.18 (s, 2H), 4.50 (d, J = 12.4 Hz, 1H), 4.18 (q, J = 2.8 Hz, 2H), 3.73 (t, J = 5.4 Hz, 2H), 3.68 (d, J = 12.6 Hz, 1H), 3.43 (dd, J = 12.2, 9.0 Hz, 1H), 3.37 (d, J = 12.3 Hz, 1H), 3.16 (t, J = 12.1 Hz, 1H), 2.92 (d, J = 9.7 Hz, 2H), 2.74 (d, J = 11.2 Hz, 1H), 2.56 (d, J = 11.1 Hz, 1H), 2.08 (s, 3H), 1.12 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 700.3 [M+H]+.
EXAMPLE 37 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(3-((1-oxido-1l6-thietan-1-ylidene)amino)picolinoyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 37
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 3-((1-oxido-1l6-thietan-1-ylidene)amino)picolinic acid. 1H NMR (400MHz, DMSO-d6)) δ 10.38 (s, 1H), 8.14 (dd, J = 4.7, 1.4 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.96 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.9, 2.2 Hz, 1H), 7.39 (dd, J = 8.2, 1.4 Hz, 1H), 7.27 (dd, J = 8.2, 4.7 Hz, 1H), 6.87 – 6.78 (m, 1H), 5.31 (d, J = 2.6 Hz, 2H), 4.57 (d, J = 12.3 Hz, 1H), 4.41 – 4.35 (m, 2H), 4.35 – 4.18 (m, 4H), 3.80 (t, J = 5.4 Hz, 2H), 3.64 (dt, J = 11.2, 7.5 Hz, 1H), 3.52 (td, J = 11.6, 3.1 Hz, 1H), 3.30- 3.25 (m, 2H), 3.24 – 3.14 (m, 2H), 3.10 – 2.86 (m, 3H), 2.78 (d, J = 11.0 Hz, 1H), 2.61 (s, 1H), 2.39 – 2.31 (m, 1H), 2.22 (dt, J = 11.4, 9.2 Hz, 1H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 774.3 [M+H]+. EXAMPLE 38 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(4-(2-(methylsulfonamido)benzoyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 38
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-(methylsulfonamido)benzoic acid. 1H NMR (400MHz, DMSO-d6) δ 10.37 (s, 1H), 9.38 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (d, J = 2.0 Hz, 1H), 7.71 (dd, J = 8.7, 2.1 Hz, 1H), 7.43 (q, J = 4.7 Hz, 2H), 7.38 – 7.18 (m, 2H), 6.83 (t, J = 2.6 Hz, 1H), 5.32 (s, 2H), 4.50 (d, J = 12.5 Hz, 1H), 4.35 – 4.15 (m, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.43 (d, J = 29.4 Hz, 5H), 3.03 (s, 3H), 3.00 – 2.88 (m, 2H), 2.81 (d, J = 11.0 Hz, 1H), 2.63 (s, 1H), 2.52 (d, J = 10.9 Hz, 2H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 763.2 [M+H]+. EXAMPLE 39 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 39
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-(cyclopropanesulfonamido)benzoic acid.1H NMR (400MHz, DMSO-d6) δ 10.37 (s, 1H), 9.28 (s, 1H), 8.06 (d, J = 8.5 Hz, 1H), 7.96 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 7.56 – 7.38 (m, 2H), 7.30 (dd, J = 19.9, 7.3 Hz, 2H), 6.83 (t, J = 2.4 Hz, 1H), 5.32 (s, 2H), 4.52 (d, J = 12.3 Hz, 1H), 4.25 (q, J = 2.9 Hz, 2H), 3.80 (t, J = 5.5 Hz, 2H), 3.46 (s, 4H), 2.98 (q, J = 7.4 Hz, 3H), 2.81 (s, 1H), 2.75 – 2.56 (m, 2H), 2.52
(d, J = 11.7 Hz, 2H), 1.19 (t, J = 7.4 Hz, 3H), 1.09 – 0.76 (m, 4H). LCMS observed m/z =
EXAMPLE 40 Synthesis of N-(2-(4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carbonyl)phenyl)-1H-pyrazole-5-carboxamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-(1H-pyrazole-5-carboxamido)benzoic acid. 1H NMR (400MHz, DMSO-d6) δ 13.50 (s, 1H), 10.38 (s, 1H), 10.18 (s, 1H), 8.40 – 8.19 (m, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 12.2 Hz, 2H), 7.71 (d, J = 8.6 Hz, 1H), 7.49 (t, J = 7.8 Hz, 1H), 7.39 (d, J = 7.6 Hz, 1H), 7.21 (t, J = 7.5 Hz, 1H), 6.88 – 6.75 (m, 2H), 5.30 (s, 2H), 4.55 (s, 1H), 4.24 (d, J = 3.3 Hz, 2H), 3.79 (t, J = 5.5 Hz, 2H), 3.67 (s, 1H), 3.45 (s, 3H), 2.97 (q, J = 7.6 Hz, 3H), 2.74 (d, J = 52.8 Hz, 2H), 2.54 (s, 2H), 1.17 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 779.3 [M+H]+. EXAMPLE 41 Synthesis of 2-(6-(4-(1H-pyrazolo[3,4-c]pyridine-7-carbonyl)piperazin-1-yl)-2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (2-chloro-4-(trifluoromethyl)phenyl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1H-pyrazolo[3,4-c]pyridine-7-carboxylic acid. LCMS observed m/z = 711.3 [M+H]+. EXAMPLE 42 Synthesis of 2-(6-(4-(3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 42
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 3H-imidazo[4,5-c]pyridine-4-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 13.15 (s, 1H), 10.37 (s, 1H), 8.45 (s, 1H), 8.33 (d, J = 5.5 Hz, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.86 – 7.54 (m, 2H), 6.83 (s, 1H), 5.32 (s, 2H), 4.66 (d, J = 12.5 Hz, 1H), 4.25 (q, J = 2.8 Hz, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.51 (d, J = 39.7 Hz, 2H), 3.22 (s, 2H), 3.02 (t, J = 15.7 Hz, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.76 – 2.51 (m, 3H), 1.19 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 711.3 [M+H]+. EXAMPLE 43 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(thieno[3,2-d]pyrimidine-7-carbonyl)piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with thieno[3,2-d]pyrimidine-7-carboxylic acid.1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 9.62 (s, 1H), 9.24 (s, 1H), 8.72 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.96 (d, J = 2.0 Hz, 1H), 7.80 – 7.66 (m, 1H), 6.82 (s, 1H), 5.31 (s, 2H), 4.62 (d, J = 12.5 Hz, 1H), 4.39 – 4.13 (m, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.47 (dt, J = 56.9, 13.3 Hz, 5H), 3.15 – 2.93 (m, 3H), 2.84 (d, J = 11.3 Hz, 1H), 2.70 – 2.52 (m, 2H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 728.2 [M+H]+. EXAMPLE 44 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(1-methyl-1H-benzo[d]imidazole-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 44
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1-methyl-1H-benzo[d]imidazole-4-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 10.36 (s, 1H), 8.27 (s, 1H), 8.05 (d, J = 8.5 Hz, 1H), 7.96 (s, 1H), 7.70 (d, J = 8.7 Hz, 1H), 7.65 (d, J = 8.0 Hz, 1H), 7.35 (t, J = 7.7 Hz, 1H), 7.23 (d, J = 7.3 Hz, 1H), 6.82 (s, 1H), 5.30 (s, 2H), 4.65 (d, J = 12.5 Hz, 1H), 4.25 (d, J = 3.1 Hz, 2H), 3.87 (s, 3H), 3.80 (t, J = 5.4 Hz, 2H), 3.53 (t, J = 11.4 Hz, 1H), 3.40 (d, J = 11.9 Hz, 2H), 3.21 (t, J = 12.3 Hz, 2H), 3.09 – 2.90 (m, 3H), 2.82 (d, J = 11.2 Hz, 1H), 2.54 (s, 2H), 1.17 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 724.3 [M+H]+. EXAMPLE 45 Synthesis of 2-(6-(4-(1H-pyrrolo[2,3-c]pyridine-7-carbonyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 45
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1H-pyrrolo[2,3-c]pyridine-7-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 11.64 (s, 1H), 10.39 (s, 1H), 8.18 – 8.09 (m, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 7.65 (d, J = 5.4 Hz, 1H), 7.59 (t, J = 2.6 Hz, 1H), 6.83 (s, 1H), 6.58 (d, J = 2.8 Hz, 1H), 5.32 (s, 2H), 4.68 (d, J = 12.4 Hz, 1H), 4.23 (d, J = 14.1 Hz, 3H), 3.80 (t, J = 5.6 Hz, 2H), 3.54 (dt, J = 32.3, 11.0 Hz, 3H), 3.04 (dq, J = 23.7, 8.7 Hz, 3H), 2.86 (d, J = 11.3 Hz, 1H), 2.66 (d, J = 10.8 Hz, 1H), 2.54 (d, J = 1.6 Hz, 2H), 1.20 (t, J = 7.5 Hz, 3H).LCMS observed m/z = 710.3 [M+H]+. EXAMPLE 46 Synthesis of 2-(6-(4-(1H-pyrrolo[3,2-b]pyridine-7-carbonyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1H-pyrrolo[3,2-b]pyridine-7-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 10.88 (s, 1H), 10.81 (s, 1H), 10.39 (s, 1H), 8.05 (d, J = 8.7 Hz, 1H), 7.96 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 7.05 – 6.89 (m, 3H), 6.82 (s, 1H), 5.31 (s, 2H), 4.50 (s, 1H), 4.25 (s, 2H), 3.79 (d, J = 5.9 Hz, 2H), 3.37-3.33 (m, 3H), 2.86 – 2.59 (m, 3H), 1.19 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 710.3 [M+H]+. EXAMPLE 47
Synthesis of 2-(6-(4-(1H-pyrrolo[3,2-c]pyridine-7-carbonyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 47
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1H-pyrrolo[3,2-c]pyridine-7-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 11.71 (s, 1H), 10.37 (s, 1H), 8.89 (s, 1H), 8.20 (d, J = 5.9 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 7.49 (d, J = 3.0 Hz, 1H), 6.82 (s, 1H), 6.67 (d, J = 3.0 Hz, 1H), 5.31 (s, 2H), 4.25 (s, 2H), 3.80 (s, 2H), 3.55-3.33 (m, 4H), 2.99 (d, J = 7.9 Hz, 3H), 2.74-2.62 (m, 3H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 710.3 [M+H]+. EXAMPLE 48 Synthesis of 2-(6-(4-(3H-[1,2,3]triazolo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)- 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 48
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1H-[1,2,3]triazolo[4,5-c]pyridine-4-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 10.37 (s, 1H), 8.50 (d, J = 5.8 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 2H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 6.88 – 6.78 (m, 1H), 5.32 (s, 2H), 4.67 (d, J = 12.4 Hz, 1H), 4.25 (q, J = 2.9 Hz, 2H), 3.80 (t, J = 5.5 Hz, 2H), 3.67 – 3.53 (m,
1H), 3.49 (t, J = 11.5 Hz, 1H), 3.19 – 3.06 (m, 1H), 3.00 (d, J = 7.7 Hz, 2H), 2.88 (d, J = 11.2 Hz, 1H), 2.70 – 2.57 (m, 1H), 2.51 (s, 2H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 712.3 [M+H]+. EXAMPLE 49 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(1-methyl-1H-benzo[d][1,2,3]triazole-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 49
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1-methyl-1H-benzo[d][1,2,3]triazole-4-carboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 8.01 – 7.89 (m, 2H), 7.70 (dd, J = 8.8, 2.1 Hz, 1H), 7.64 (t, J = 7.7 Hz, 1H), 7.44 (d, J = 7.0 Hz, 1H), 6.82 (s, 1H), 5.31 (s, 2H), 4.67 (d, J = 12.5 Hz, 1H), 4.35 (s, 3H), 4.25 (q, J = 2.9 Hz, 2H), 3.80 (t, J = 5.3 Hz, 2H), 3.56 (t, J = 11.3 Hz, 1H), 3.41 (d, J = 13.4 Hz, 3H), 3.28 (d, J = 7.7 Hz, 3H), 3.15 – 3.04 (m, 1H), 2.99 (d, J = 8.0 Hz, 2H), 2.86 (d, J = 11.2 Hz, 1H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 725.3 [M+H]+. EXAMPLE 50 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(imidazo[1,2-a]pyridine-8-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 50
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with imidazo[1,2-a]pyridine-8-carboxylic acid.1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.62 (d, J = 6.8 Hz, 1H), 8.11 – 8.00 (m, 2H), 7.96 (d, J = 2.1 Hz, 1H), 7.71 (d, J = 8.4 Hz, 1H), 7.63 (s, 1H), 7.28 (d, J = 6.8 Hz, 1H), 6.97 (t, J = 6.8 Hz, 1H), 6.82 (s, 1H), 5.31 (s, 2H), 4.61 (d, J = 12.5 Hz, 1H), 4.25 (q, J = 3.0 Hz, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.60 – 3.37 (m, 4H), 3.00 (dq, J = 21.3, 8.2 Hz, 3H), 2.83 (d, J = 11.3 Hz, 1H), 2.62 – 2.51 (m, 3H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 710.3 [M+H]+. EXAMPLE 51 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(2-oxo-2,3-dihydro-1H-benzo[d]imidazole-4-carbonyl)piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 51
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-oxo-2,3-dihydro-1H-benzo[d]imidazole-4-carboxylic acid.1H NMR (400MHz, DMSO-d6) δ 11.57 (s, 1H), 10.40 (s, 1H), 8.39 (d, J = 4.8 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.96 (s, 1H), 7.77 – 7.64 (m, 2H), 7.11 (d, J = 4.8 Hz, 1H), 6.82 (s, 1H), 6.63 (d, J = 3.1 Hz, 1H), 5.31 (s, 2H), 4.62 (d, J = 12.2 Hz, 1H), 4.25 (s, 2H), 3.80 (t, J = 5.5 Hz, 2H), 3.55 (s, 4H), 3.07 (d, J = 19.0 Hz, 2H), 2.98 (q, J = 7.5 Hz, 2H), 2.94 – 2.75 (m, 2H), 2.64 (d, J = 17.7 Hz, 1H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 726.2 [M+H]+.
EXAMPLE 52 Synthesis of 2-(6-(4-acetylpiperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with acetic acid.1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (s, 1H), 7.71 (d, J = 8.6 Hz, 1H), 6.83 (s, 1H), 5.31 (s, 2H), 4.40 (d, J = 12.4 Hz, 1H), 4.25 (s, 2H), 3.80 (t, J = 5.9 Hz, 2H), 3.46 (t, J = 11.8 Hz, 2H), 3.21 (t, J = 12.2 Hz, 2H), 2.98 (d, J = 8.2 Hz, 3H), 2.69 (s, 4H), 2.05 (s, 3H), 1.18 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 608.3 [M+H]+. EXAMPLE 53 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(2-hydroxyacetyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with glycolic acid.1H NMR (400MHz, DMSO-d6) δ 10.37 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.96 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 6.82 (s, 1H), 5.31 (s, 2H),
4.61 (t, J = 5.5 Hz, 1H), 4.36 (d, J = 12.6 Hz, 1H), 4.21 (d, J = 25.9 Hz, 3H), 4.08 (q, J = 9.8 Hz, 1H), 3.88 – 3.75 (m, 2H), 3.71 (d, J = 12.6 Hz, 1H), 3.42 (dt, J = 21.7, 11.2 Hz, 4H), 3.15 (t, J = 12.1 Hz, 1H), 2.97 (q, J = 7.6 Hz, 2H), 2.79 (t, J = 12.4 Hz, 1H), 2.69 (t, J = 10.6 Hz, 2H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 624.2 [M+H]+. EXAMPLE 54 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(2-hydroxypropanoyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with lactic acid.1H NMR (400MHz, DMSO-d6) δ 10.37 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.2 Hz, 1H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 6.83 (s, 1H), 5.31 (s, 2H), 5.03 – 4.87 (m, 1H), 4.56 – 4.31 (m, 2H), 4.25 (q, J = 2.8 Hz, 2H), 4.04 (t, J = 12.9 Hz, 1H), 3.80 (t, J = 5.5 Hz, 2H), 3.43 (dd, J = 27.5, 12.5 Hz, 4H), 3.19 (t, J = 12.7 Hz, 1H), 3.08 – 2.92 (m, 2H), 2.73 (dt, J = 22.3, 11.6 Hz, 3H), 1.43 – 1.05 (m, 6H). LCMS observed m/z = 638.3 [M+H]+. EXAMPLE 55 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(1-(hydroxymethyl)cyclopropane-1-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1-(hydroxymethyl)cyclopropanecarboxylic acid. 1H NMR (400MHz, DMSO-d6) δ 10.39 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (s, 1H), 7.79 – 7.62 (m, 1H), 6.83 (s, 1H), 5.32 (s, 2H), 4.86 (s, 1H), 4.32 (d, J = 12.4 Hz, 2H), 4.25 (d, J = 3.2 Hz, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.49 (s, 2H), 3.39 (t, J = 11.9 Hz, 4H), 2.98 (d, J = 7.6 Hz, 4H), 2.70 (d, J = 11.1 Hz, 2H), 1.19 (t, J = 7.4 Hz, 3H), 0.72 (d, J = 9.2 Hz, 4H). LCMS observed m/z = 664.2 [M+H]+. EXAMPLE 56 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2,2-difluoro-3- hydroxypropanoyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 56
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2,2-difluoro-3-hydroxypropanoic acid. 1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 6.83 (s, 1H), 5.68 (d, J = 6.6 Hz, 1H), 5.32 (s, 2H), 4.36 (d, J = 12.6 Hz, 1H), 4.25 (d, J = 3.3 Hz, 2H), 4.17 (d, J = 12.8 Hz, 1H), 3.97 – 3.74 (m, 4H), 3.44 (dt, J = 23.7, 11.2 Hz, 3H), 2.97 (dd, J = 14.7, 7.5 Hz, 3H), 2.78 (d, J = 11.1 Hz, 2H), 1.19 (t, J = 7.4 Hz, 3H).LCMS observed m/z = 674.2 [M+H]+. EXAMPLE 57 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2,2-difluoro-2-(1- hydroxycyclobutyl)acetyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 57
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2,2-difluoro-2-(1-hydroxycyclobutyl)acetic acid. 1H NMR (400MHz, DMSO-d6) δ 10.39 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.79 – 7.66 (m, 1H), 6.83 (s, 1H), 6.10 (s, 1H), 5.32 (s, 2H), 4.39 (d, J = 12.2 Hz, 1H), 4.35 – 4.17 (m, 3H), 3.80 (t, J = 5.4 Hz, 2H), 3.44 (t, J = 12.7 Hz, 3H), 3.31 – 3.11 (m, 2H), 3.10 – 2.83 (m, 3H), 2.74 (s, 2H), 2.49 – 2.40 (m, 2H), 2.04 (s, 2H), 1.84 (tq, J = 10.4, 4.9 Hz, 1H), 1.65 (h, J = 9.1 Hz, 1H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 714.2 [M+H]+. EXAMPLE 58 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(2-hydroxycyclopentane-1-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 58
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-hydroxycyclopentane-1-carboxylic acid.1H NMR (400MHz, DMSO-d6) δ 10.39 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 6.83 (s, 1H), 5.32 (s, 2H), 4.80 (d, J = 22.6 Hz, 1H), 4.44 (d, J = 12.3 Hz, 1H), 4.25 (s, 2H), 4.20 (s, 1H), 4.15 – 4.04 (m, 1H), 3.80 (s, 2H), 3.54 – 3.36 (m, 3H), 3.19 (d, J = 19.0 Hz, 2H), 2.98 (q, J = 7.5 Hz, 2H), 2.89 (d, J = 7.1 Hz, 1H), 2.70 (d, J = 10.6 Hz, 3H), 1.90 (s, 1H), 1.81 – 1.55 (m, 4H), 1.55 – 1.40 (m, 1H), 1.19 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 678.3 [M+H]+.
EXAMPLE 59 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(methylsulfonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 59
To a mixture of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (30 mg, 0.053 mmol, Intermediate K) and DIPEA (0.047 mL, 0.265 mmol) in DMF (0.5 mL) was added methanesulfonyl chloride (6.07 mg, 0.053 mmol). The mixture was stirred at 25 °C for 30 minutes. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (5.7 mg, 16% yield) of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-6-(4-(methylsulfonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide as a white solid.1H NMR (400MHz, DMSO-d6) δ 10.31 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.90 (s, 1H), 7.65 (d, J = 8.6 Hz, 1H), 6.77 (s, 1H), 5.25 (s, 2H), 4.19 (d, J = 3.2 Hz, 2H), 3.74 (t, J = 5.6 Hz, 2H), 3.47 (d, J = 10.6 Hz, 5H), 2.85 (d, J = 16.5 Hz, 8H), 2.71 (d, J = 11.1 Hz, 2H), 1.10 (q, J = 6.5 Hz, 3H). LCMS observed m/z = 644.2 [M+H]+. EXAMPLE 60 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4- (cyclopropylsulfonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 60
The title compound was prepared using similar procedure as Example 59, replacing methanesulfonyl chloride with cyclopropanesulfonyl chloride.1H NMR (400MHz, DMSO- d6) δ 10.36 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 6.83 (s, 1H), 5.31 (s, 2H), 4.25 (s, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.67 – 3.49 (m, 4H), 3.09 – 2.88 (m, 4H), 2.76 (d, J = 11.1 Hz, 2H), 2.66 (s, 2H), 1.17 (t, J = 7.4 Hz, 4H), 1.03 (d, J = 8.0 Hz, 2H), 0.96 (d, J = 5.0 Hz, 2H).LCMS observed m/z = 670.2 [M+H]+. EXAMPLE 61 Synthesis of 2-(6-(4-(2-(1H-1,2,4-triazol-1-yl)acetyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 61
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 1,2,4-triazole-1-acetic acid. 1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.48 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (s, 2H), 7.72 (d, J = 8.7 Hz, 1H), 6.83 (s, 1H), 5.44 (d, J = 16.8 Hz, 1H), 5.32 (s, 2H), 5.26 (d, J = 16.7 Hz, 1H), 4.34 (d, J = 12.5 Hz, 1H), 4.26 (d, J = 3.6 Hz, 2H), 3.94 (d, J = 12.9 Hz, 1H), 3.80 (t, J = 5.5 Hz, 2H), 3.57 (t, J = 11.3 Hz, 1H), 3.40 (t, J = 11.2 Hz, 2H), 3.27 (d, J = 12.0 Hz, 2H), 3.00 (d, J = 7.6 Hz, 2H), 2.83 (t, J = 12.2 Hz, 1H), 2.74 (s, 2H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 675.3 [M+H]+. EXAMPLE 62
Synthesis of 2-(6-(4-(2-(1H-1,2,4-triazol-1-yl)propanoyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 62
The title compound was prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 2-(1H-1,2,4-triazol-1-yl)propanoic acid. 1H NMR (400MHz, DMSO-d6) δ 10.38 (s, 1H), 8.67 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (q, J = 2.1 Hz, 2H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 6.83 (s, 1H), 5.80 (dd, J = 14.7, 7.2 Hz, 1H), 5.31 (s, 2H), 4.36 (d, J = 12.5 Hz, 1H), 4.25 (q, J = 2.8 Hz, 2H), 4.08 (t, J = 12.7 Hz, 1H), 3.80 (t, J = 5.4 Hz, 2H), 3.51 (t, J = 11.5 Hz, 1H), 3.37-3.33 (m, 3H), 3.17 (d, J = 12.2 Hz, 1H), 2.98 (d, J = 8.3 Hz, 2H), 2.76 (dt, J = 25.1, 10.9 Hz, 3H), 1.65 (dd, J = 12.5, 6.9 Hz, 3H), 1.19 (dt, J = 7.8, 3.8 Hz, 3H). LCMS observed m/z = 689.3 [M+H]+. EXAMPLE 63 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(5-hydroxy-1-methyl-1H-imidazole-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 63
The title compound can be prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with 5-(benzyloxy)-1-methyl-1H-imidazole-4-carboxylic acid.
EXAMPLE 64 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(5-hydroxythiazole-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 64
The title compound can be prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with 5-(benzyloxy)thiazole-4-carboxylic acid. EXAMPLE 65 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(4-hydroxyisothiazole-3-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 65
The title compound can be prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with 4-(benzyloxy)isothiazole-3-carboxylic acid. EXAMPLE 66 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(pyrimidin-4-yl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide.
Compound 66
Step 1: Preparation of tert-butyl 4-(pyrimidin-4-yl)piperazine-1-carboxylate. Compound 66.1
A solution of tert-butyl 4-(2-chloropyrimidin-4-yl)piperazine-1-carboxylate (1.0 g, 3.3 mmol) in MeOH (20 mL) was treated with Pd/C (100 mg, 0.940 mmol) at 25 °C under H2 for 2 hours. The mixture was filtered and the filter cake was washed with MeOH (3 x 3 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 100% CH2Cl2) to afford (820 mg, 93% yield) of tert-butyl 4- (pyrimidin-4-yl)piperazine-1-carboxylate as a colorless oil. LCMS observed m/z = 265.1 [M+H]+. Step 2: Preparation of 4-(piperazin-1-yl)pyrimidine. Compound 66.2
A solution of tert-butyl 4-(pyrimidin-4-yl)piperazine-1-carboxylate (820 mg, 3.1 mmol) in dioxane (3 mL, 0.023 mmol) was treated with HCl (4 M in dioxane, 20 mL) solution at 25 °C for 3 hours. The mixture was concentrated in vacuo to afford (500 mg, crude) of 4-(piperazin-1-yl)pyrimidine as a colorless oil. The crude product was used directly in the next step without further purification. LCMS observed m/z = 165.2 [M+H]+. Step 3: Preparation of methyl 3-oxo-2-[4-(pyrimidin-4-yl)piperazin-1- yl]pentanoate. Compound 66.3
A solution of 4-(piperazin-1-yl)pyrimidine hydrochloride (500 mg, 2.5 mmol) and methyl 2-chloro-3-oxopentanoate (451 mg, 2.7 mmol) in MeCN (10 mL) was treated with K2CO3 (1.03 g, 7.5 mmol) in one portion at 25 ℃ under N2. The mixture was heated to 60 ℃ and stirred at 60 ℃ for 16 hours under N2. The mixture was cooled to 25 ℃, quenched with H2O (30 mL) and extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were washed with brine and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-7% MeOH in CH2Cl2) to afford (350 mg, 48% yield) of methyl 3-oxo-2-[4-(pyrimidin-4-yl)piperazin-1-yl]pentanoate as a brown yellow oil. LCMS observed m/z = 293.1 [M+H]+. Step 4: Preparation of 2-bromo-5-ethyl-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyrimidin-7-one. Compound 66.4
A solution of methyl 3-oxo-2-[4-(pyrimidin-4-yl)piperazin-1-yl]pentanoate (340 mg, 1.2 mmol) and 5-bromo-2H-1,2,4-triazol-3-amine (190 mg, 1.2 mmol) in EtOH (3 mL) was treated with H3PO4 (114 mg, 1.2 mmol) in one portion at 25 ℃ under N2. The mixture was heated to 80 ℃ and stirred at 80 ℃ for 16 hours under N2. The mixture was cooled to 25 ℃, acidified to pH 8 with saturated Na2CO3 aqueous solution and concentrated in vacuo. The residue was dissolved in EtOH (2 mL) and purified by reversed phase C18 silica gel column chromatography (mobile phase: 10-30% MeOH in H2O) to afford (95 mg, 20% yield) of 2-bromo-5-ethyl-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyrimidin-7-one as a brown yellow solid. LCMS observed m/z = 407.2 [M+H]+. Step 5: Preparation of 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(pyrimidin-4- yl)piperazin-1-yl]-4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one. Compound 66.5
A solution of 2-bromo-5-ethyl-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyrimidin-7-one (90 mg, 0.2 mmol) and 2-(3,6-dihydro-2H-pyran-4- yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (70 mg, 0.3 mmol) in mixed solvent of dioxane (1.5 mL) and H2O (0.5 mL) was treated with Pd(dppf)Cl2.CH2Cl2 (18 mg, 0.02 mmol) and Na2CO3 (71 mg, 0.6 mmol) in one portion at 25 ℃ under N2. The mixture was heated to 100 ℃ and stirred at 100 ℃ for 16 hours under N2. The mixture was cooled to 25 ℃ and concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 10-40% MeOH in H2O) to afford (30 mg, 33% yield) of 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyrimidin-7-one) as a white solid. LCMS observed m/z = 409.2 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 66
A solution of 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(pyrimidin-4- yl)piperazin-1-yl]-4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one (24 mg, 0.059 mmol) and N- [2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (23.49 mg, 0.065 mmol) in DMF (3 mL) was treated with DIPEA (22.78 mg, 0.177 mmol) in one portion at 25 °C under N2 for 2 hours. The residue was purified by reversed phase flash chromatography (Column: YMC Triart C18 ExRs 5 m, 19 mm * 250 mm; mobile phase: 52-72% MeCN in H2O) to afford (7.9 mg, 20.65% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-[4-(pyrimidin-4-yl)piperazin-1-yl]-
[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.33 (s, 1H), 8.52 (s, 1H), 8.21 (d, J = 6.2 Hz, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.2 Hz, 1H), 7.77 – 7.70 (m, 1H), 6.89 (d, J = 6.2 Hz, 1H), 6.84 (s, 1H), 5.34 (s, 2H), 4.49 – 4.41 (m, 2H), 4.26 (d, J = 3.0 Hz, 2H), 3.85 – 3.77 (m, 2H), 3.59 – 3.44 (m, 3H), 3.15 – 2.93 (m, 5H), 2.81 – 2.75 (m, 2H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 644.20 [M+H]+. EXAMPLE 67 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-{4-[(2Z)-3-hydroxybut-2-enoyl]piperazin-1-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 67
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl]acetamide (30 mg, 0.05 mmol, Intermediate K) and diketene (5 mg, 0.06 mmol) in DMF (2 mL) was stirred at 25 °C for 1 hour. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: YMC-Triart C85 m, 20 mm x 150 mm; mobile phase: 45- 60% MeCN in H2O) to afford (11.6 mg, 31% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-{4-[(2Z)-3- hydroxybut-2-enoyl]piperazin-1-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, CDCl3) δ 9.03 (s, 1H), 8.50 (d, J = 8.7 Hz, 1H), 7.66 (d, J = 2.0 Hz, 1H), 7.55 (d, J = 8.7 Hz, 1H), 6.99 (s, 1H), 5.13 (s, 2H), 4.65 - 4.62 (m, 1H), 4.43-4.36 (m, 2H), 3.94 (t, J = 5.5 Hz, 2H), 3.82 – 3.56 (m, 5H), 3.35 - 3.29 (m, 1H), 3.20- 3.10 (m, 2H), 2.88 (t, J = 12.5 Hz, 1H), 2.75 (d, J = 7.4 Hz, 4H), 2.33 (s, 3H), 1.38-1.25 (m, 3H). LCMS observed m/z = 650.30 [M+H]+. EXAMPLE 68
Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 68
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetyl chloride. Compound 68.1
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetic acid (50 mg, 0.0813 mmol, Intermediate S) in CH2Cl2 (1.0 mL) at 0 °C under N2 was added 1-chloro-N,N,2-trimethyl-1-propenylamine (0.0215 mL, 0.163 mmol). The mixture stirred at 0 °C for 2 hours. The mixture was concentrated in vacuo and the crude product was used directly in the next step without further purification. Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 68.2
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetyl chloride (50 mg, 0.079 mmol) in THF (0.4 mL) at 0 °C was added 3- (trifluoromethyl)bicyclo[1.1.1]pentan-1-amine (14.3 mL, 0.0948 mmol). The mixture stirred at 0 °C and was warmed to 25 °C and stirred at 25 °C for 3 hours. The mixture was concentrated in vacuo and the crude product was used directly in the next step without further purification. Step 3: Preparation of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 68
To a mixture of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-yl)acetamide (59.1 mg, 0.079 mmol) in CH2Cl2 (0.79 mL) was added dropwise a 1 molar solution of boron trichloride in dichloromethane (0.395 mL, 0.395 mmol). The mixture was stirred at -40 °C under N2 for 15 minutes. The mixture was quenched with MeOH (1 mL) and warmed to 25 °C. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (24.8 mg, 45% yield) of 2-(2-(3,6-dihydro-
2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (trifluoromethyl)bicyclo[1.1.1]pentan-1-yl)acetamide as a white solid.1H NMR (400MHz, DMSO-d6) δ 9.18 (s, 1H), 8.54 (s, 1H), 6.81 (t, J = 1.9 Hz, 1H), 4.90 (s, 2H), 4.51 (d, J = 12.3 Hz, 1H), 4.26 (d, J = 2.9 Hz, 2H), 3.81 (t, J = 5.4 Hz, 2H), 3.57 – 3.39 (m, 4H), 3.31 – 3.17 (m, 3H), 2.97 (td, J = 12.5, 3.4 Hz, 1H), 2.92 – 2.81 (m, 2H), 2.78 (d, J = 11.4 Hz, 1H), 2.61 (d, J = 10.3 Hz, 1H), 2.43 (s, 3H), 2.23 (s, 6H), 1.13 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 658.3 [M+H]+. EXAMPLE 69 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 69
The title compound was prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-amine. 1H NMR (400MHz, DMSO-d6) δ 9.20 (s, 1H), 8.55 (s, 1H), 6.81 (s, 1H), 4.90 (s, 2H), 4.51 (d, J = 12.5 Hz, 1H), 4.32 – 4.18 (m, 2H), 3.81 (t, J = 5.4 Hz, 2H), 3.46 (t, J = 11.2 Hz, 3H), 3.33 (s, 2H), 3.30 – 3.16 (m, 2H), 2.97 (d, J = 3.4 Hz, 1H), 2.82 (dd, J = 28.6, 9.8 Hz, 3H), 2.61 (d, J = 11.0 Hz, 1H), 2.43 (s, 3H), 2.26 (s, 6H), 1.13 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 708.3 [M+H]+. EXAMPLE 70 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(1-(trifluoromethyl)-2-oxabicyclo[2.1.1]hexan-4-yl)acetamide. Compound 70
The title compound was prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 1-(trifluoromethyl)-2- oxabicyclo[2.1.1]hexan-4-amine. 1H NMR (400MHz, DMSO-d6) δ 9.34 (s, 1H), 8.48 (s, 1H), 6.81 (s, 1H), 6.62 (s, 1H), 4.95 (s, 2H), 4.51 (d, J = 12.4 Hz, 1H), 4.26 (s, 2H), 3.83 (d, J = 21.3 Hz, 4H), 3.47 (q, J = 11.4 Hz, 4H), 3.22 (t, J = 12.5 Hz, 2H), 2.91 (dd, J = 24.6, 9.7 Hz, 3H), 2.78 (d, J = 11.3 Hz, 1H), 2.60 (d, J = 10.8 Hz, 1H), 2.41 (s, 3H), 2.37 (d, J = 4.4 Hz, 2H), 2.04 (d, J = 4.3 Hz, 2H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 674.3 [M+H]+. EXAMPLE 71 Synthesis of N-(4-(2-chloro-1,1,2,2-tetrafluoroethyl)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 71
The title compound was prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4-(2-chloro-1,1,2,2- tetrafluoroethyl)aniline.1H NMR (400MHz, DMSO-d6) δ 10.91 (s, 1H), 8.55 (s, 1H), 7.79 (d, J = 8.5 Hz, 2H), 7.64 (d, J = 8.5 Hz, 2H), 6.79 (s, 1H), 5.19 (s, 2H), 4.52 (d, J = 12.5 Hz, 1H), 4.23 (q, J = 2.9 Hz, 2H), 3.79 (t, J = 5.5 Hz, 2H), 3.60 – 3.44 (m, 3H), 3.23 (dd, J = 29.9, 17.1 Hz, 2H), 2.97 (d, J = 9.7 Hz, 3H), 2.81 (d, J = 11.2 Hz, 1H), 2.65 (s, 1H), 2.43 (s, 3H), 1.17 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 734.2 [M+H]+.
EXAMPLE 72 Synthesis of N-(4-(chlorodifluoromethoxy)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 72
The title compound was prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4-(chlorodifluoromethoxy)aniline. 1H NMR (400MHz, DMSO-d6) δ 10.72 (s, 1H), 8.56 (s, 1H), 7.75 – 7.61 (m, 2H), 7.33 (d, J = 8.5 Hz, 2H), 6.80 (s, 1H), 5.16 (s, 2H), 4.52 (d, J = 12.6 Hz, 1H), 4.23 (d, J = 3.7 Hz, 2H), 3.79 (t, J = 5.5 Hz, 2H), 3.64 – 3.41 (m, 4H), 3.29 – 3.17 (m, 2H), 2.96 (d, J = 10.5 Hz, 3H), 2.81 (d, J = 11.3 Hz, 1H), 2.63 (d, J = 10.9 Hz, 1H), 2.43 (s, 3H), 1.17 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 700.3 [M+H]+. EXAMPLE 73 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-(trifluoromethyl)-2-oxabicyclo[2.1.1]hexan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4-(trifluoromethyl)-2- oxabicyclo[2.1.1]hexan-1-amine. EXAMPLE 74
Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-(trifluoromethyl)bicyclo[2.1.1]hexan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4- (trifluoromethyl)bicyclo[2.1.1]hexan-1-amine. EXAMPLE 75 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-(trifluoromethyl)bicyclo[2.2.1]heptan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4- (trifluoromethyl)bicyclo[2.2.1]heptan-1-amine. EXAMPLE 76 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-(trifluoromethyl)bicyclo[2.2.2]octan-1-yl)acetamide. Compound 76
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 4- (trifluoromethyl)bicyclo[2.2.2]octan-1-amine. EXAMPLE 77 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide.
Step 1: Preparation of 2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one. Compound 77.1
A solution of tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)piperazine-1- carboxylate (1.0 g, 3.19 mmol, Intermediate C) and 5-amino-3-(dimethylamino)-1H-1,2,4- triazole (609 mg, 4.79 mmol) in n-butanol (8 mL) was treated with p-toluenesulfonic acid monohydrate (182 mg, 10.957 mmol). The mixture was heated to 120 ℃ and stirred at 120 ℃ for 16 hours under N2. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-50% of 20% MeOH in CH2Cl2) to
afford (265 mg, 20% yield) of 2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one. LCMS observed m/z = 393.3 [M+H]+. Step 2: Preparation of tert-butyl 4-(2-(dimethylamino)-5-ethyl-4-(2-((2-methyl-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 77.2
A solution of 2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one (265 mg, 0.677 mmol) and N-[4-(trifluoromethyl)-2-tolyl]iodoacetamide (348 mg, 1.02 mmol) in DMF (3 mL) was treated with DIPEA (0.354 mL, 2.03 mmol) and the mixture heated to 45 °C under N2 for 2 hours. The mixture was concentrated in vacuo and purified by silica gel column chromatography (mobile phase: 0-40% of 20% MeOH in CH2Cl2) to afford (320 mg, 74.0% yield) of tert-butyl 4-(2-(dimethylamino)-5-ethyl-4-(2-((2-methyl-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate as yellow solid. LCMS observed m/z = 607.3 [M+H]+. Step 3: Preparation of 2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide. Compound 77.3
A mixture of tert-butyl 4-(2-(dimethylamino)-5-ethyl-4-(2-((2-methyl-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (320 mg, 0.527 mmol) in CH2Cl2 (5.27 mL) was added TFA (0.605 mL) stirred at 0 °C for 1 hour. The mixture was concentrated in vacuo to afford (150 mg, crude) of 2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide. LCMS observed m/z = 507.3 [M+H]+. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-methyl-4-(trifluoromethyl)phenyl)acetamide. Compound 77.4
A mixture of 2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide (125 mg, 0.247 mmol), HATU (141 mg, 0.247 mmol) and DIPEA (141 g, 0.370 mmol) in DMF (2.47 mL) was stirred at 25 °C for 30 minutes. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-70% of 20% MeOH in CH2Cl2) to afford (141 mg, 66% yield) of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide as a brown solid. LCMS observed m/z = 733.3 [M+H]+. Step 5: Preparation of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-methyl-4-(trifluoromethyl)phenyl)acetamide. Compound 77
To a mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- methyl-4-(trifluoromethyl)phenyl)acetamide (141.1 mg, 0.192 mmol) in CH2Cl2 (1.15 mL) was added dropwise a 1 molar solution of boron trichloride in dichloromethane (1.15 mL, 1.15 mmol). The mixture was stirred at -40 °C under N2 for 15 minutes. The mixture was quenched with MeOH (1 mL) and warmed to 25 °C. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (13.2 mg, 10% yield) of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-methyl-4-(trifluoromethyl)phenyl)acetamide as a white solid. 1H NMR (400MHz, DMSO-d6) δ 10.01 (s, 1H), 8.55 (s, 1H), 7.70 (d, J = 8.4 Hz, 1H), 7.62 (d, J = 2.1 Hz, 1H), 7.58 – 7.48 (m, 1H), 5.14 (s, 2H), 4.50 (d, J = 12.5 Hz, 1H), 3.49 (qd, J = 11.1, 3.8 Hz, 4H), 3.27 – 3.14 (m, 2H), 2.96 (s, 6H), 2.95 – 2.88 (m, 2H), 2.76 (d, J = 11.1 Hz, 1H), 2.59 (d, J = 10.8 Hz, 1H), 2.43 (s, 3H), 2.34 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 643.3 [M+H]+. EXAMPLE 78 Synthesis of 2-(2-cyclopropyl-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide. Compound 78
The title compound was prepared using similar procedure as Example 77, replacing 5-amino-3-(dimethylamino)-1H-1,2,4-triazole with 5-cyclopropyl-4h-1,2,4-triazol-3- amine.1H NMR (400MHz, DMSO-d6) δ 9.98 (s, 1H), 8.55 (s, 1H), 7.69 (d, J = 8.4 Hz, 1H), 7.63 (s, 1H), 7.53 (d, J = 8.5 Hz, 1H), 5.17 (s, 2H), 4.51 (d, J = 12.5 Hz, 1H), 3.56 – 3.41 (m, 3H), 3.24 (d, J = 13.1 Hz, 2H), 2.98 (d, J = 11.6 Hz, 3H), 2.78 (d, J = 11.4 Hz, 1H), 2.60 (d, J = 11.1 Hz, 1H), 2.43 (s, 3H), 2.34 (s, 3H), 2.05 (tt, J = 9.1, 4.9 Hz, 1H), 1.17 (t, J = 7.5 Hz, 3H), 1.07 – 0.95 (m, 2H), 0.93 – 0.80 (m, 2H). LCMS observed m/z = 640.3 [M+H]+. EXAMPLE 79 Synthesis of 2-(2-cyclobutyl-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-methyl-4- (trifluoromethyl)phenyl)acetamide. Compound 79
The title compound was prepared using similar procedure as Example 77, replacing 5-amino-3-(dimethylamino)-1H-1,2,4-triazole with 5-cyclobutyl-1H-1,2,4-triazol-3-amine. 1H NMR (400MHz, DMSO-d6) δ 10.01 (s, 1H), 8.54 (s, 1H), 7.70 (d, J = 8.4 Hz, 1H), 7.63 (s, 1H), 7.53 (d, J = 8.5 Hz, 1H), 5.23 (s, 2H), 4.51 (d, J = 12.5 Hz, 1H), 3.60 (p, J = 8.5 Hz, 1H), 3.49 (d, J = 12.2 Hz, 3H), 3.24 (t, J = 13.5 Hz, 2H), 2.98 (d, J = 10.1 Hz, 3H), 2.79 (d, J = 11.1 Hz, 1H), 2.61 (d, J = 11.0 Hz, 1H), 2.43 (s, 3H), 2.35 (s, 3H), 2.29 (td, J = 8.7, 6.5 Hz, 4H), 2.08 – 1.99 (m, 1H), 1.90 (dq, J = 11.5, 6.3 Hz, 1H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 654.3 [M+H]+. EXAMPLE 80 Synthesis of 2-(6-(4-(3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 80
A solution of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5- ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (90 mg, 0.2 mmol, Intermediate N), DIPEA (44 mg, 0.3 mmol), HATU (71 mg, 0.2 mmol) and 3H- imidazo[4,5-c]pyridine-4-carboxylic acid (34 mg, 0.2 mmol) in DMF (10 mL) was stirred at 25 °C for 1 hour under N2. The mixture was poured into H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: Xbridge BEH Phenyl 5 m, 30 mm * 150 mm; mobile phase: 42-50% MeCN in H2O) to afford (40.1 mg, 35% yield) of 2-(6-(4- (3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a white solid.1H NMR (400 MHz, CDCl3) δ 9.08 (s, 1H), 8.52 (s, 2H), 8.46 (d, J = 8.7 Hz, 1H), 8.02 (s, 1H), 7.69 – 7.62 (m, 1H), 7.56 (s, 1H), 5.10 (s, 2H), 4.80 (d, J = 12.7 Hz, 2H), 3.81 – 3.57 (m, 3H), 3.11 (s, 9H), 2.86 – 2.81 (m, 1H), 2.71 – 2.65 (m, 1H), 1.30 (q, J = 6.5, 5.6 Hz, 4H). LCMS observed m/z = 672.25 [M+H]+. EXAMPLE 81 Synthesis of 2-(6-(4-acetylpiperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 81
The title compound was prepared using similar procedure as Example 80, replacing 3H-imidazo[4,5-c]pyridine-4-carboxylic acid with acetic acid.1H NMR (400 MHz, DMSO) δ 10.37 (s, 1H), 8.04 (d, J = 8.6 Hz, 1H), 7.95 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 6.72 (s, 1H), 5.20 (s, 2H), 4.38 (d, J = 13.1 Hz, 1H), 3.82 (d, J = 12.8 Hz, 1H), 3.46 (s, 1H), 3.24 – 3.12 (m, 2H), 2.95 (s, 6H), 2.91 (s, 1H), 2.66 (d, J = 9.2 Hz, 3H), 2.04 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 569.3 [M+H]+. EXAMPLE 82 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl- 6-(4-(2-(methylsulfonamido)benzoyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 82
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(dimethylamino)-5- ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (50 mg, 0.1 mmol, Intermediate N), TCFH (27 mg, 0.1 mmol), NMI (8 mg, 0.1 mmol) and 2- methanesulfonamidobenzoic acid (31 mg, 0.15 mmol) in DMF (5 mL) was stirred at 25 °C for 2 hours. The mixture was quenched with brine (20 mL) and exacted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine and dried over anhydrous Na2SO4. After filtration, the filtrate was purified by preparatory HPLC (Column: X-select CSH C185 m, 30 mm * 150 mm; mobile phase: 47-55% MeCN in H2O) to afford (6.1 mg, 9% yield) of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2- (dimethylamino)-5-ethyl-6-(4-(2(methylsulfonamido)benzoyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a] pyrimidin-4(7H)-yl)acetamide as white solid. LCMS observed m/z = 724.20 [M+H]+. EXAMPLE 83
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl- 6-(4-(3-hydroxy-1-methyl-1H-pyrazole-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 80, replacing 3H-imidazo[4,5-c]pyridine-4-carboxylic acid with 3-hydroxy-1-methylpyrazole-4- carboxylic acid.1H NMR (400 MHz, CDCl3) δ 9.07 (s, 1H), 8.49 (d, J = 8.7 Hz, 1H), 7.66 (s, 1H), 7.56 (d, J = 8.7 Hz, 1H), 7.43 (s, 1H), 5.04 (s, 2H), 4.43 – 4.37 (m, 2H), 3.81 – 3.75 (s, 5H), 3.23 (s, 2H), 3.15 – 3.11 (m, 8H), 2.81 – 2.76 (m, 2H), 1.31 (t, J = 7.6 Hz, 3H )LCMS observed m/z = 651.35 [M+H]+. EXAMPLE 84 Synthesis of 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1- c][1,4]oxazine-2-carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
The title compound was prepared using similar procedure as Example 32, replacing 3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (Intermediate L) with N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(dimethylamino)-5- ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (Intermediate N).1H NMR (400 MHz, DMSO-d6) δ 9.95 (s, 2H), 8.30 – 7.86 (m, 2H), 7.73 (s, 1H), 5.21 (s, 2H), 4.59 (s, 2H), 4.45 (d, J = 12.0 Hz, 1H), 4.04 (s, 2H), 3.86 (s, 2H), 3.70
– 3.41 (m, 3H), 3.23 – 3.09 (m, 2H), 3.09 – 2.79 (m, 8H), 2.78 – 2.68 (m, 1H), 2.64 – 2.57 (m, 1H), 1.16 (s, 3H). LCMS observed m/z = 721.30 [M+H]+. EXAMPLE 85 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as Example 80, replacing 3H-imidazo[4,5-c]pyridine-4-carboxylic acid with 2-cyclopropanesulfonamidobenzoic acid.1H NMR (400 MHz, DMSO-d6) δ 10.35 (s, 1H), 9.12 (s, 1H), 8.05 (d, J = 8.6 Hz, 2H), 7.97 (s, 2H), 7.77 – 7.70 (m, 2H), 7.53 – 7.41 (m, 4H), 7.34 (d, J = 7.6 Hz, 2H), 7.30 (s, 1H), 5.22 (s, 3H), 4.52 (s, 1H), 3.47 (s, 2H), 2.97 (s, 4H), 1.17 (t, J = 7.5 Hz, 5H), 0.99 (s, 2H), 0.93 (s, 3H). LCMS observed m/z = 750.25 [M+H]+. EXAMPLE 86 Synthesis of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(3-hydroxy-4-oxo- 4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 86
Step 1: Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-6-(4-(3-(benzyloxy)-4-oxo- 4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 86.1
To a stirred solution of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (100 mg, 0.2 mmol, Intermediate T) and 3-(benzyloxy)- 4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carboxylic acid (50 mg, 0.2 mmol, Intermediate L) in MeCN (3 mL) were added NMI (41 mg, 0.5 mmol) and TCFH (46 mg, 0.2 mmol) in portions at 25 °C. The mixture was heated to 50 °C and stirred at 50 °C under N2 for 1 hour. The mixture was cooled to 25 °C, quenched with H2O (20 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (90 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase C18 silica gel column chromatography (mobile phase: 0-100% MeCN in H2O) to afford (115 mg, 71% yield) of 2-[2-(2H-1,3-benzodioxol- 5-yl)-6-{4-[3-(benzyloxy)-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 886.5 [M+H]+. Step 2: Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(3-hydroxy-4- oxo-4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Example 86
To a solution of 2-[2-(2H-1,3-benzodioxol-5-yl)-6-{4-[3-(benzyloxy)-4-oxo- 6H,7H,9H-pyrimido[2,1-c][1,4]oxazine-2-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (110 mg, 0.12 mmol) in CH2Cl2 (5 mL) was added boron trichloride (5 mL) dropwise at 0 °C. The mixture was stirred at 25 °C under N2 for 1 hour. The reaction was quenched with H2O (10 mL) and extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: Xbridge BEH Phenyl 5 m, 30 mm * 150 mm; mobile phase: 35-53% MeCN in H2O) to afford (1.7 mg, 2% yield) of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(3-hydroxy-4- oxo-4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.98 (s, 1H), 7.72 (d, J = 8.6 Hz, 1H), 7.69 – 7.64 (m, 1H), 7.54 (d, J = 1.7 Hz, 1H), 7.06 (d, J = 8.2 Hz, 1H), 6.12 (s, 2H), 5.38 (s, 2H), 4.60 (s, 2H), 4.51 – 4.46 (m, 1H), 4.04 (t, J = 5.4 Hz, 2H), 3.87 (d, J = 5.5 Hz, 2H), 3.61 – 3.57 (m, 1H), 3.45 (s, 2H), 3.07 – 2.91 (m, 3H), 2.83 – 2.78 (m, 2H), 2.65 (s, 1H), 1.21 (t, J = 7.5 Hz, 3H).LCMS observed m/z = 798.40 [M+H]+. EXAMPLE 87 Synthesis of 2-[2-(2H-1,3-benzodioxol-5-yl)-6-[4-(2- cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 87
To a mixture of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (85 mg, 0.1 mmol, Intermediate T) and 2-cyclopropanesulfonamidobenzoic acid (37 mg, 0.1 mmol) in DMF (6 mL) were added TCFH (47 mg, 0.1 mmol) and NMI (58 mg, 0.7 mmol) in one portion at 25 °C. The mixture was heated to 40 °C and stirred at 40 °C for 3 hours under N2. The mixture was cooled to 25 °C, quenched with H2O (10 mL) and extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase flash chromatography (Column: YMC Triart C18 ExRs 5 m, 19 mm * 250 mm; mobile phase: 70-90% MeCN in H2O) to afford (25.2 mg, 21% yield) of 2-[2-(2H-1,3-benzodioxol-5-yl)-6-[4-(2- cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 9.22 – 9.07 (m, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.74 – 7.74 (m, 1H), 7.68 –7.66 (m, 1H), 7.55 (d, J = 1.6 Hz, 1H), 7.51 – 7.46 (m, 2H), 7.38 – 7.35 (m, 2H), 7.07 (d, J = 8.1 Hz, 1H), 6.12 (s, 2H), 5.38 (s, 2H), 4.55 (s, 1H), 3.50 (s, 3H), 3.02 (d, J = 8.1 Hz, 3H), 2.84 (s, 1H), 2.79 – 2.53 (m, 2H), 2.04 – 1.97 (m, 1H), 1.25 – 1.20 (m, 3H), 1.02 – 0.98 (m, 2H), 0.94 – 0.86 (m, 2H). LCMS observed m/z = 827.20 [M+H]+. EXAMPLE 88 Synthesis of 2-(6-(4-(3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-2- (benzo[d][1,3]dioxol-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 88
The title compound was prepared using similar procedure as Example 80, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate N) with 2-(6-(4-(5- (benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid (Intermediate T).1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 8.52 (d, J = 5.5 Hz, 1H), 8.38 (dd, J = 5.7, 3.0 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.79 – 7.61 (m, 3H), 7.55 (d, J = 1.7 Hz, 1H), 7.06 (d, J = 8.1 Hz, 1H), 6.12 (s, 2H), 5.38 (s, 2H), 4.70 – 4.66 (m, 1H), 3.63 – 3.57 (m, 3H), 3.14 – 3.00 (m, 3H), 2.91 – 2.87 (m, 1H), 2.66 – 2.63 (m, 2H), 1.22 (t, J = 7.6 Hz, 3H ). LCMS observed m/z = 749.35 [M+H]+. EXAMPLE 89 Synthesis of 2-(6-(4-acetylpiperazin-1-yl)-2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
To a mixture of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (85 mg, 0.1 mmol, Intermediate T) and DIPEA (32.1 mg, 0.248 mmol) in DMF (0.5 mL) was added acetyl chloride (6.5 mg, 0.0828 mmol) at 25 °C and stirred for 30 minutes. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (4.91 mg, 9% yield) of 2-(6-(4-acetylpiperazin-1-
yl)-2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)- N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.41 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.79 – 7.70 (m, 1H), 7.66 (dd, J = 8.1, 1.7 Hz, 1H), 7.54 (d, J = 1.6 Hz, 1H), 7.05 (d, J = 8.1 Hz, 1H), 6.11 (s, 2H), 5.37 (s, 2H), 4.41 (d, J = 12.1 Hz, 1H), 3.85 (d, J = 12.6 Hz, 1H), 3.50 (d, J = 11.3 Hz, 1H), 3.27 – 3.13 (m, 2H), 3.01 (d, J = 8.4 Hz, 2H), 2.70 (d, J = 7.5 Hz, 3H), 2.05 (s, 3H), 1.21 (d, J = 7.4 Hz, 3H). LCMS observed m/z = 646.2 [M+H]+. EXAMPLE 90 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-1H-2- benzopyran-6-yl)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,9H-pyrimido[2,1-c][1,4]oxazine- 2-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 90
The title compound was prepared using similar procedure as Example 86, replacing 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (Intermediate T) with N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl- 7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (Intermediate U). 1H NMR (400 MHz, CDCl3) δ 10.38 (s, 1H), 9.18 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 8.09 (d, J = 10.2 Hz, 2H), 7.62 (s, 1H), 7.55 (d, J = 8.5 Hz, 1H), 7.11 (d, J = 7.9 Hz, 1H), 5.21 (s, 2H), 4.85 (s, 4H), 4.65 (s, 2H), 4.14 – 4.01 (m, 6H), 3.84 (s, 2H), 3.39 (s, 1H), 3.23 (d, J = 7.8 Hz, 3H), 2.96 (t, J = 5.6 Hz, 2H), 2.80 (s, 2H), 1.37 (t, J = 7.1 Hz, 3H). LCMS observed m/z = 810.45 [M+H]+. EXAMPLE 91 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{6-[4-(2- cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-2-(3,4-dihydro-1H-2-benzopyran-6-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 91
The title compound was prepared using similar procedure as Example 87, replacing 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (Intermediate T) with N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(isochroman-6-yl)-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate U). 1H NMR (400 MHz, DMSO-d6) δ 10.43 (s, 1H), 9.15 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.90 (dt, J = 4.0, 2.3 Hz, 2H), 7.73 (dd, J = 8.7, 2.2 Hz, 1H), 7.54 – 7.42 (m, 2H), 7.36 (d, J = 7.3 Hz, 1H), 7.31 (t, J = 7.4 Hz, 1H), 7.19 (d, J = 8.4 Hz, 1H), 5.40 (s, 2H), 4.74 (s, 2H), 4.56 (s, 1H), 3.92 (t, J = 5.7 Hz, 2H), 3.52 – 3.48 (m, 3H), 3.34 – 3.30 (m, 1H), 3.03 (d, J = 7.8 Hz, 3H), 2.88 (t, J = 5.8 Hz, 3H), 2.79 – 2.69 (m, 2H), 1.23 (t, J = 7.4 Hz, 3H), 1.00 (s, 2H), 0.94 (d, J = 4.7 Hz, 2H). LCMS observed m/z = 839.25 [M+H]+. EXAMPLE 92 Synthesis of 2-(6-(4-(3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-5- ethyl-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
The title compound was prepared using similar procedure as Example 80, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate N) with N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(isochroman-6-yl)-7-oxo-6-(piperazin-1-
yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate U). 1H NMR (400 MHz, CDCl3) δ 9.19 (s, 1H), 8.54 – 8.39 (m, 3H), 8.03 (d, J = 8.6 Hz, 2H), 7.95 (s, 1H), 7.60 (s, 1H), 7.52 (s, 1H), 7.09 (d, J = 7.9 Hz, 1H), 5.22 (s, 3H), 4.83 (s, 3H), 4.02 (t, J = 5.7 Hz, 2H), 3.86 – 3.74 (m, 2H), 3.61 (s, 1H), 3.23 (d, J = 9.0 Hz, 3H), 2.93 (d, J = 6.2 Hz, 3H), 2.75 (m, 1H), 1.37 (t, J = 7.5 Hz, 4H). LCMS observed m/z = 761.20 [M+H]+. EXAMPLE 93 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(3-hydroxy-4- oxo-4,6,7,9-tetrahydropyrimido[2,1-c][1,4]oxazine-2-carbonyl)piperazin-1-yl)-7-oxo-2- ((3aR,6aS)-tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 93
The title compound was prepared using similar procedure as Example 32, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate K) with 2-{2-[(3aR,6aS)-hexahydrofuro[3,4-c]pyrrol-5-yl]-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (Intermediate V).1H NMR (400 MHz, Acetonitrile-d3) δ 8.80 (s, 1H), 8.32 (d, J = 8.7 Hz, 1H), 7.84 (d, J = 2.1 Hz, 1H), 7.64 (dd, J = 8.7, 2.1 Hz, 1H), 7.49 – 7.36 (m, 1H), 5.11 (s, 2H), 4.61 (d, J = 4.5 Hz, 3H), 4.56 (s, 1H), 4.06 (t, J = 5.4 Hz, 2H), 3.94 – 3.84 (m, 4H), 3.64 – 3.53 (m, 6H), 3.42 – 3.36 (m, 2H), 3.32 (s, 1H), 3.07 – 2.93 (m, 5H), 2.79 – 2.73 (m, 2H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 789.20 [M+H]+. EXAMPLE 94 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 94
The title compound was prepared using similar procedure as Example 87, replacing 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (Intermediate T) with N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (Intermediate V). 1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 9.14 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.2 Hz, 1H), 7.73 (dd, J = 9.0, 2.1 Hz, 1H), 7.52 – 7.42 (m, 2H), 7.37 – 7.28 (m, 2H), 5.22 (s, 2H), 4.52 (s, 1H), 3.82 (dd, J = 8.7, 6.5 Hz, 2H), 3.57 – 3.40 (m, 7H), 3.31 – 3.27 (m, 2H), 2.95 (m, 5H), 2.85 – 2.56 (m, 4H), 1.17 (t, J = 7.4 Hz, 3H), 0.99 (s, 2H), 0.93 (d, J = 4.8 Hz, 2H). LCMS observed m/z = 818.25 [M+H]+. EXAMPLE 95 Synthesis of 2-(6-(4-(3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-5- ethyl-7-oxo-2-((3aR,6aS)-tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide.
The title compound was prepared using similar procedure as Example 80, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate N) with N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2-
(cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-2-((3aR,6aS)- tetrahydro-1H-furo[3,4-c]pyrrol-5(3H)-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (Intermediate V).1H NMR (400 MHz, CDCl3) δ 9.10 (s, 1H), 8.51 – 8.39 (m, 3H), 7.91 (d, J = 5.6 Hz, 1H), 7.67 (d, J = 2.1 Hz, 1H), 7.55 (dd, J = 9.0, 2.0 Hz, 1H), 5.09 (s, 2H), 4.82 (d, J = 12.6 Hz, 2H), 3.98 (dd, J = 8.8, 6.3 Hz, 3H), 3.85 – 3.62 (m, 5H), 3.56 – 3.49 (m, 3H), 3.14 (d, J = 7.8 Hz, 3H), 3.05 (q, J = 3.5 Hz, 2H), 2.86 – 2.82 (m, 1H), 2.70 – 2.66 (m, 1H), 1.31 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 740.25 [M+H]+. EXAMPLE 96 Synthesis of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
Step 1: Preparation of 2-[2-(2H-1,3-benzodioxol-5-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 96.1
To a solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0] octa-1(6),2,4-trien-2-yl]acetamide (100 mg, 0.1 mmol,
Intermediate R) and 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (38 mg, 0.2 mmol) in dioxane (9 mL) and H2O (2 mL) were added Pd(dppf)Cl2.CH2Cl2 (9 mg, 0.01 mmol) and KOAc (31 mg, 0.3 mmol) in portions at 25 °C under N2. The mixture was heated to 90 °C and stirred for 2 hours at 90 °C under N2. The mixture was cooled to 25 °C and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in CH2Cl2) to afford (100 mg, 94% yield) of 2-[2- (2H-1,3-benzodioxol-5-yl)-6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a white solid. LCMS observed m/z = 822.29 [M+H]+. Step 2: Preparation of 2-[2-(2H-1,3-benzodioxol-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 96
A solution of 2-[2-(2H-1,3-benzodioxol-5-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl) bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (94 mg, 0.1 mmol) in boron trichloride (5 mL) and CH2Cl2 (5 mL) was stirred at 25 °C under N2 for 4 hours. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: X bridge BEH C18 OBD prep column 130, 5 m, 30 mm * 150 mm; mobile phase: 37-55% MeCN in H2O) to afford (14.6 mg, 17% yield) of 2-[2-(2H-1,3- benzodioxol-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa- 1(6),2,4-trien-2-yl]acetamide as a white solid.1H NMR (400 MHz, CDCl3) δ 11.97 (s, 1H), 9.07 (s, 1H), 8.63 (s, 1H), 7.82 (d, J = 8.1 Hz, 1H), 7.72 (s, 1H), 7.51 (d, J = 8.6 Hz, 1H), 7.35 (d, J = 8.6 Hz, 1H), 6.92 (d, J = 8.0 Hz, 1H), 6.07 (s, 2H), 5.78 – 5.61 (m, 1H), 5.11 (s,
2H), 4.91 – 4.78 (m, 1H), 3.87 – 3.79 (m, 2H), 3.52 (s, 1H), 3.29 – 3.09 (m, 7H), 2.88 – 2.79 (m, 2H), 2.62 (s, 3H), 1.35 (d, J = 7.7 Hz, 3H). LCMS observed m/z = 732.35 [M+H]+. EXAMPLE 97 Synthesis of 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. Compound 97.1
To a solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (100 mg, 0.1 mmol, Intermediate R), 2,3-dihydro-1,4-benzodioxin-5-ylboronic acid (25 mg, 0.1 mmol) and K2CO3 (53 mg, 0.4 mmol) in dioxane (5 mL) and H2O (0.5 mL) was added Pd(PPh3)4 (15 mg, 0.1 mmol) in portions at 25 °C under N2. The mixture was heated to 100 °C and stirred at 100 °C for 16 hours under N2. The mixture was cooled to 25 °C, quenched with H2O (10
mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over Na2SO4. After filtration, the filtrate was concentrated in vacuo to afford (140 mg, crude) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. The crude product was used directly in the next step without further purification. LCMS observed m/z = 836.3 [M+H]+. Step 2: Preparation of 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 97
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (140 mg, 0.2 mmol) in TFA (10 mL) was stirred at 25 °C for 16 hours under N2. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: Xbridge BEH Phenyl 5 μm, 30 *150 mm; mobile phase: 31-40% MeCN in H2O) to afford (15.1 mg, 10% yield) of 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.73 (s, 1H), 10.13 (s, 1H), 8.58 (s, 1H), 7.46 (d, J = 1.9 Hz, 2H), 7.31 (dd, J = 7.7, 1.7 Hz, 1H), 7.00 (dd, J = 8.0, 1.7 Hz, 1H), 6.92 (t, J = 7.9 Hz, 1H), 5.25 (s, 2H), 4.57 – 4.53 (m, 1H), 4.34 – 4.24 (m, 4H), 3.61 – 3.52 (m, 2H), 3.51 (s, 1H), 3.27 (dd, J = 5.4, 3.0 Hz, 3H), 3.17 (s, 2H), 3.00 (d, J = 9.3 Hz, 2H), 2.88 – 2.82 (m, 1H), 2.68 (d, J = 9.4 Hz, 1H), 2.45 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 746.40 [M+H]+. EXAMPLE 98
Synthesis of 2-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
The title compound was prepared using similar procedure as Example 96, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2-(isochroman- 6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, DMSO-d6) δ 10.77 (s, 1H), 8.57 (s, 1H), 7.89 (d, J = 6.8 Hz, 2H), 7.52 – 7.44 (m, 2H), 7.18 (d, J = 8.5 Hz, 1H), 5.28 (s, 2H), 4.74 (s, 2H), 4.58 – 4.52 (m, 2H), 3.91 (t, J = 5.7 Hz, 2H), 3.58 – 3.49 (m, 3H), 3.28 (d, J = 4.4 Hz, 3H), 3.18 (s, 2H), 3.04 – 2.98 (m, 3H), 2.88 (t, J = 5.6 Hz, 3H), 2.45 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 744.25 [M+H]+. EXAMPLE 99 Synthesis of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(2,3-dihydro-1-benzofuran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
The title compound was prepared using similar procedure as Example 96, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2-(2,3- dihydrobenzofuran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400 MHz,
DMSO-d6) δ 10.76 (s, 1H), 10.25 (s, 1H), 8.57 (s, 1H), 7.60 (dd, J = 7.7, 1.5 Hz, 1H), 7.54 – 7.43 (m, 2H), 7.40 – 7.33 (m, 2H), 5.26 (s, 2H), 4.64 – 4.49 (m, 3H), 3.61 – 3.47 (m, 3H), 3.22 (d, J = 8.7 Hz, 6H), 3.17 (s, 2H), 2.99 (d, J = 9.4 Hz, 3H), 2.87 – 2.81 (m, 1H), 2.45 (s, 3H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 730.25 [M+H]+. EXAMPLE 100 Synthesis of 2-[2-(2,3-dihydro-1-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 100
The title compound was prepared using similar procedure as Example 96, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2-(2,3-dihydro- 1-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO- d6) δ 10.76 (s, 1H), 10.26 (s, 1H), 8.57 (s, 1H), 7.95 (s, 1H), 7.85 (d, J = 8.4 Hz, 1H), 7.47 (q, J = 8.9, 8.5 Hz, 2H), 6.87 (d, J = 8.4 Hz, 1H), 5.25 (s, 2H), 4.64 – 4.47 (m, 3H), 3.61 – 3.45 (m, 3H), 3.26 (s, 3H), 3.17 (s, 3H), 3.11 - 2.89 (m, 3H), 2.87 – 2.81 (m, 1H), 2.71 – 2.63 (m, 2H), 2.45 (s, 3H), 1.20 (q, J = 9.9, 7.7 Hz, 3H). LCMS observed m/z = 730.25 [M+H]+. EXAMPLE 101 Synthesis of 2-[2-(1,3-dihydro-2-benzofuran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl) bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 101
The title compound was prepared using similar procedure as Example 96, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2-(1,3-dihydro- 2-benzofuran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 8.81 (s, 1H), 8.62 (s, 1H), 8.18 (d, J = 7.3 Hz, 1H), 7.50 -7.35 (m, 5H), 5.69 – 5.62 (m, 1H), 5.55 (s, 2H), 5.29 – 5.06 (m, 4H), 4.85 – 4.79 (m, 1H), 3.89 – 3.83 (m, 2H), 3.53 (s, 1H), 3.25 - 3.12 (m, 7H), 2.89 – 2.83 (m, 2H), 2.61 (s, 3H), 1.36 (d, J = 7.5 Hz, 3H). LCMS observed m/z = 730.35 [M+H]+. EXAMPLE 102 Synthesis of 2-[2-(3,4-dihydro-1H-2-benzopyran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
The title compound was prepared using similar procedure as Example 97, replacing 2,3-dihydro-1,4-benzodioxin-5-ylboronic acid with 2-(3,4-dihydro-1H-2-benzopyran-5-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, CDCl3) δ 11.94 (s, 1H), 8.92 (s, 1H), 8.62 (s, 1H), 8.04 – 7.95 (m, 1H), 7.47 (d, J = 8.7 Hz, 1H), 7.38 – 7.30 (m, 2H), 7.17 – 7.11 (m, 1H), 5.67 (m, 1H), 5.12 (s, 2H), 4.86 (s, 3H), 3.97 (t, J = 5.8 Hz, 2H), 3.91 – 3.82 (m, 2H), 3.53 (s, 1H), 3.34 (t, J = 5.8 Hz, 2H), 3.21 (d, J = 5.3 Hz, 4H), 3.12 (d, J = 4.3 Hz, 3H), 2.93 - 2.75 (m, 2H), 2.62 (s, 3H), 1.37 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 744.45 [M+H]+. EXAMPLE 103
Synthesis of 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(isochroman-8-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide.
The title compound was prepared using similar procedure as Example 97, replacing 2,3-dihydro-1,4-benzodioxin-5-ylboronic acid with 2-(isochroman-8-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, DMSO-d6) δ 10.76 (s, 1H), 8.56 (s, 1H), 7.89 (d, J = 7.2 Hz, 1H), 7.51 – 7.44 (m, 2H), 7.36 – 7.27 (m, 2H), 5.26 (s, 2H), 5.05 (s, 2H), 4.58 – 4.52 (m, 1H), 3.88 (t, J = 5.7 Hz, 2H), 3.61 – 3.47 (m, 3H), 3.29 – 3.25 (m, 3H), 3.17 (s, 2H), 3.03 (d, J = 8.9 Hz, 3H), 2.89 (t, J = 5.8 Hz, 3H), 2.69 – 2.66 (m, 1H), 2.45 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H ). LCMS observed m/z = 744.25 [M+H]+. EXAMPLE 104 Synthesis of 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 104.1
To a stirred mixture of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid (80 mg, 0.1 mmol, Intermediate O) in DMF (5 mL) were added 5-(trifluoromethyl)bicyclo[4.2.0]octa-1,3,5-trien-2-amine (26 mg, 0.1 mmol), TCFH (58 mg, 0.2 mmol) and NMI (45 mg, 0.6 mmol) in portions at 25 °C. The mixture was heated to 40 °C and stirred at 40 °C for 16 hours under N2. The reaction was cooled to 25 °C, quenched with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by reversed phase flash chromatography (mobile phase: 0-85% MeOH in H2O) to afford (24 mg, 23% yield) of 2- (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(dimethylamino)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a white solid. LCMS observed m/z = 745.3 [M+H]+. Step 2: Preparation of 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 104
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (70 mg, 0.1 mmol) in boron trichloride (1 mL) and CH2Cl2 (2 mL) was stirred at 25 °C under N2 for 16 hours. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: YMC Triart C18 5 m, 30 mm * 150 mm; mobile phase: 33-56% MeCN in H2O) to afford (8.9 mg, 14% yield) of 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.69 (s, 1H), 8.55 (s, 1H), 7.46 (d, J = 2.0 Hz, 2H), 5.09 (s, 2H), 4.55 – 4.49 (m, 1H), 3.56 – 3.44 (m, 4H), 3.29 – 3.16 (m, 6H), 2.97 – 2.93 (m, 8H), 2.81 – 2.75 (m, 1H), 2.63 -2.59 (m, 1H), 2.44 (s, 3H), 1.15 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 655.25 [M+H]+. EXAMPLE 105 Synthesis of 2-{6-[4-(2-cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide.
To a solution of 2-cyclopropanesulfonamidobenzoic acid (39 mg, 0.2 mmol) and NMI (55 mg, 0.7 mmol) in MeCN (2 mL) were added 2-[2-(dimethylamino)-5-ethyl-7-oxo- 6-(piperazin-1-yl)- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl]-N-[5-(trifluoromethyl) bicyclo [4.2.0] octa-1(6),2,4-trien-2-yl] acetamide (70 mg, 0.1 mmol, Intermediate W) and TCFH (75 mg, 0.2 mmol) in portions at 25 ℃ under N2. The mixture was heated to 50 ℃ and stirred at 50 ℃ for 2 hours. The mixture was quenched with H2O (10 mL) and extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo.
The residue was purified by preparatory HPLC (Column: YMC Triart C18 ExRs 5 m, 19 mm * 250 mm; mobile phase: 63-80% MeCN in H2O) to afford (2.1 mg, 2% yield) 2-{6-[4- (2-cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.68 (s, 1H), 9.11 (s, 1H), 7.47 (p, J = 9.3, 8.7 Hz, 4H), 7.35 – 7.29 (m, 2H), 5.10 (s, 2H), 4.53 (s, 1H), 3.48 (s, 4H), 3.29 – 3.22 (m, 3H), 3.18 (s, 2H), 3.01 – 2.88 (m, 9H), 2.83 – 2.66 (m, 2H), 1.16 (t, J = 7.5 Hz, 3H), 1.07 – 0.85 (m, 4H). LCMS observed m/z = 742.15 [M+H]+. EXAMPLE 106 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(2- (methylsulfonamido)benzoyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 106
The title compound was prepared using similar procedure as Example 105, replacing 2-cyclopropanesulfonamidobenzoic acid with 2-methanesulfonamidobenzoic acid. 1H NMR (400 MHz, CDCl3) δ 8.98 (s, 1H), 7.96 (s, 1H), 7.66 (d, J = 8.2 Hz, 1H), 7.49 (d, J = 7.8 Hz, 2H), 7.36 (d, J = 8.2 Hz, 2H), 7.22 (t, J = 7.5 Hz, 1H), 5.10 (s, 2H), 3.77 (s, 3H), 3.35 – 3.23 (m, 6H), 3.19 – 3.07 (m, 11H), 2.80 (s, 3H), 1.29 (s, 3H). LCMS observed m/z = 716.35 [M+H]+. EXAMPLE 107 Synthesis of 2-[2-(dimethylamino)-5-ethyl-6-[4-(3-hydroxy-1-methylpyrazole-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 107
The title compound was prepared using similar procedure as Example 33, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate K) with 2-[2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (Intermediate W).1H NMR (400 MHz, DMSO-d6) δ 10.69 (s, 1H), 10.57 (s, 1H), 7.89 (s, 1H), 7.51 – 7.43 (m, 2H), 5.10 (s, 2H), 4.21 (s, 2H), 3.67 (s, 3H), 3.46 (s, 2H), 3.27 (s, 2H), 3.18 (s, 2H), 3.08 (s, 2H), 2.95 (s, 6H), 2.91 (s, 2H), 2.73 – 2.67 (m, 2H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 643.20 [M+H]+. EXAMPLE 108 Synthesis of 2-(6-(4-(3H-imidazo[4,5-c]pyridine-4-carbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide.
The title compound was prepared using similar procedure as Example 80, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate N) with 2-[2- (dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N- [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (Intermediate W). 1H NMR (400 MHz, DMSO-d6) δ 13.04 (s, 1H), 10.69 (s, 1H), 8.45 (s, 1H), 8.34 (d, J = 5.5
Hz, 1H), 7.70 (s, 1H), 7.50 – 7.42 (m, 2H), 5.10 (s, 2H), 4.66 (d, J = 12.4 Hz, 1H), 3.66 – 3.55 (m, 1H), 3.49 (s, 1H), 3.27 (dd, J = 5.4, 2.9 Hz, 3H), 3.20 – 3.15 (m, 2H), 3.14 – 2.99 (m, 2H), 2.95 (s, 6H), 2.92 (d, J = 7.9 Hz, 2H), 2.83 (m, 1H), 2.58 (s, 1H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 664.35 [M+H]+. EXAMPLE 109 Synthesis of 2-[2-(dimethylamino)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,9H- pyrimido[2,1-c][1,4]oxazine-2-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 109
The title compound was prepared using similar procedure as Example 84, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl-7-oxo-6-(piperazin- 1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (Intermediate N) with 2-[2- (dimethylamino)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N- [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (Intermediate W). 1H NMR (400 MHz, DMSO-d6) δ 10.70 (s, 1H), 9.69 (s, 1H), 7.52 – 7.41 (m, 2H), 5.10 (s, 2H), 4.65 – 4.54 (m, 2H), 4.53 – 4.41 (m, 1H), 4.04 (t, J = 5.4 Hz, 2H), 3.88 (d, J = 5.7 Hz, 2H), 3.57 (m, 1H), 3.42 (m, 2H), 3.29 – 3.24 (m, 2H), 3.24 – 3.13 (m, 3H), 2.95 (s, 6H), 2.90 (d, J = 7.8 Hz, 2H), 2.77 – 2.71 (m, 1H), 2.63 – 2.59 (m, 1H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 713.25 [M+H]+. EXAMPLE 110 Synthesis of 2-{6-[4-(2-cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-2-(3,4- dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 110
Step 1: Preparation of tert-butyl 4-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl- 7-oxo-4-({[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate. Compound 110.1
To a solution of tert-butyl 4-[2-bromo-5-ethyl-7-oxo-4-({[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (120 mg, 0.2 mmol, Intermediate Q), 2-(3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (52 mg, 0.2 mmol) and K2CO3 (76 mg, 0.5 mmol) in dioxane (5 mL) and H2O (1 mL) was added Pd(dppf)Cl2.CH2Cl2 (13 mg, 0.1 mmol) in one portion at 25 °C under N2. The mixture was heated to 60 °C and stirred at 60 °C for 16 hours under N2. The mixture was cooled to 25 °C, quenched with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 33% EtOAc in petroleum ether) to afford (57 mg, 43% yield) tert-butyl 4-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl- 7-oxo-4-({[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a yellow solid. LCMS observed m/z = 708.3 [M+H]+.
Step 2: Preparation of 2-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 110.2
To a solution of tert-butyl 4-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo- 4-({[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (57 mg, 0.1 mmol) in CH2Cl2 (6 mL) was added TFA (3 mL) in one portion at 25 °C. The mixture was stirred at 25 °C for 20 minutes. The mixture was concentrated in vacuo to afford (40 mg, crude) of 2-[2-(3,4- dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a yellow solid. The crude product was used directly in the next step without further purification. LCMS observed m/z = 608.2 [M+H]+. Step 3: Preparation of 2-{6-[4-(2-cyclopropanesulfonamidobenzoyl)piperazin-1- yl]-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl}-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 110
To a solution of 2-[2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (100 mg, 0.2 mmol), 2-
cyclopropanesulfonamidobenzoic acid (44 mg, 0.2 mmol) and TCFH (69 mg, 0.2 mmol) in DMF (3 mL) was added NMI (68 mg, 0.8 mmol) in one portion at 25 °C under N2. The mixture was stirred at 25 °C under N2 for 3 hours. The mixture was quenched with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by preparatory HPLC (Column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 60-75% MeCN in H2O) to afford (6.4 mg, 4% yield) of 2-{6-[4-(2-cyclopropanesulfonamidobenzoyl)piperazin-1-yl]- 2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.76 (s, 1H), 9.06 (s, 1H), 7.88 (d, J = 7.1 Hz, 2H), 7.53 – 7.41 (m, 5H), 7.39 – 7.33 (m, 1H), 7.33 – 7.27 (m, 1H), 7.19 (d, J = 8.5 Hz, 1H), 5.28 (s, 2H), 4.74 (s, 2H), 4.55 (s, 1H), 3.91 (t, J = 5.7 Hz, 2H), 3.50 (s, 2H), 3.28 (s, 2H), 3.27 (s, 1H), 3.18 (s, 2H), 2.99 (d, J = 7.6 Hz, 3H), 2.88 (s, 3H), 1.22 (q, J = 8.1, 7.5 Hz, 4H), 1.00 (s, 2H), 0.94 (d, J = 4.6 Hz, 3H). LCMS observed m/z = 831.20 [M+H]+. EXAMPLE 111 Synthesis of 2-(2-(benzo[d][1,3]dioxol-5-yl)-6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2- yl)acetamide. Compound 111
The title compound was prepared using similar procedure as Example 110, replacing 2-(3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (benzo[d][1,3]dioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO-d6) δ 10.76 (s, 1H), 9.11 (s, 1H), 7.66 – 7.63 (m, 1H), 7.53 – 7.43 (m, 5H), 7.37 – 7.29 (m, 2H), 7.06 (d, J = 8.1 Hz, 1H), 6.12 (s, 2H), 5.26 (s, 2H), 4.55 (s, 1H), 3.50 (s, 3H),
3.27 (d, J = 4.5 Hz, 3H), 3.18 (s, 2H), 2.99 (d, J = 7.9 Hz, 3H), 2.84 (s, 1H), 2.77 – 2.63 (m, 2H), 1.20 (t, J = 7.5 Hz, 3H), 1.00 (s, 2H), 0.94 (d, J = 4.7 Hz, 2H). LCMS observed m/z = 819.25 [M+H]+. EXAMPLE 112 Synthesis of 2-{2-[(3aR,6aS)-hexahydrofuro[3,4-c]pyrrol-5-yl]-6-[4-(2- cyclopropanesulfonamidobenzoyl)piperazin-1-yl]-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 112
The title compound was prepared using similar procedure as Example 94, replacing tert-butyl 4-(2-bromo-4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-5- ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate F) with tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Q). 1H NMR (400 MHz, DMSO-d6) δ 10.68 (s, 1H), 9.09 (s, 1H), 7.47 (d, J = 8.1 Hz, 4H), 7.32 (q, J = 7.3 Hz, 2H), 5.09 (s, 2H), 4.51 (s, 1H), 3.80 (dd, J = 8.8, 6.2 Hz, 2H), 3.55 – 3.41 (m, 7H), 3.35 – 3.21 (m, 5H), 3.18 (d, J = 4.0 Hz, 2H), 3.02 – 2.85 (m, 5H), 2.81 – 2.67 (m, 2H), 2.61 (s, 1H), 1.14 (t, J = 7.4 Hz, 3H), 0.98 (s, 2H), 0.92 (d, J = 4.8 Hz, 2H). LCMS observed m/z = 810.25 [M+H]+. EXAMPLE 113 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 113
Step 1: Preparation of tert-butyl 4-(4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carbonyl)-7,8- dihydropyrido[3,2-d]pyrimidine-5(6H)-carboxylate. Compound 113.1
The title compound can be prepared using similar procedure as Example 34, replacing 3-aminopicolinic acid with 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2- d]pyrimidine-4-carboxylic acid. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-6-(4-(5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 113
The title compound can be prepared using similar procedure as Example 14, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(5,7- dihydrofuro[3,4-b]pyridin-3-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)- N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide with tert-butyl 4-(4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carbonyl)-7,8- dihydropyrido[3,2-d]pyrimidine-5(6H)-carboxylate. EXAMPLE 114 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(1,2,3,4-tetrahydro-1,7-naphthyridine-8-carbonyl)piperazin-1- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 1-(tert-butoxycarbonyl)-1,2,3,4-tetrahydro-1,7-naphthyridine-8-carboxylic acid. EXAMPLE 115 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(3,4-dihydro-2H- benzo[b][1,4]oxazine-5-carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 115
The title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 4-(tert-butoxycarbonyl)-3,4-dihydro-2H-benzo[b][1,4]oxazine-5-carboxylic acid. EXAMPLE 116 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(1,2,3,4-tetrahydroimidazo[1,5-a]pyrimidine-8- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 1-(tert-butoxycarbonyl)-1,2,3,4-tetrahydroimidazo[1,5-a]pyrimidine-8- carboxylic acid. EXAMPLE 117 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(1-methyl-4,5,6,7-tetrahydro-1H-pyrazolo[4,3-b]pyridine-3- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic
acid with 4-(tert-butoxycarbonyl)-1-methyl-4,5,6,7-tetrahydro-1H-pyrazolo[4,3- b]pyridine-3-carboxylic acid. EXAMPLE 118 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(4,5,6,7-tetrahydroisothiazolo[4,5-b]pyridine-3- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 4-(tert-butoxycarbonyl)-4,5,6,7-tetrahydroisothiazolo[4,5-b]pyridine-3- carboxylic acid. EXAMPLE 119 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-6-(4-(4,5,6,7-tetrahydroisoxazolo[4,5-b]pyridine-3- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Example 113, replacing 5-(tert-butoxycarbonyl)-5,6,7,8-tetrahydropyrido[3,2-d]pyrimidine-4-carboxylic acid with 4-(tert-butoxycarbonyl)-4,5,6,7-tetrahydroisoxazolo[4,5-b]pyridine-3-carboxylic acid.
EXAMPLE 120 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 7- (trifluoromethyl)benzo[d]thiazol-4-amine. EXAMPLE 121 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(1-methyl-7-(trifluoromethyl)-1H-benzo[d]imidazol-4-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 1-methyl-7- (trifluoromethyl)-1H-benzo[d]imidazol-4-amine. EXAMPLE 122
Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(1-methyl-2-oxo-6-(trifluoromethyl)-1,2-dihydropyridin-3-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 3-amino-1-methyl-6- (trifluoromethyl)pyridin-2(1H)-one. EXAMPLE 123 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-(trifluoromethyl)pyridin-2-yl)acetamide.
The title compound can be prepared using similar procedure as Example 68, replacing 3-(trifluoromethyl)bicyclo[1.1.1]pentan-1-amine with 5-(trifluoromethyl)pyridin- 2-amine. EXAMPLE 124 Synthesis of N-(1-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)- 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin- 6-yl)azetidin-3-yl)-5-hydroxy-N,6-dimethylpyrimidine-4-carboxamide. Compound 124
Step 1: Preparation of methyl 2-(3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1- yl)-3-oxopentanoate. Compound 124.1
The title compound can be prepared using similar procedure as Intermediate C, replacing tert-butyl piperazine-1-carboxylate with tert-butyl azetidin-3- yl(methyl)carbamate. Step 2: Preparation of tert-butyl (1-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate. Compound 124.2
The title compound can be prepared using similar procedure as Intermediate E, replacing tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)piperazine-1-carboxylate (Intermediate C) with methyl 2-(3-((tert-butoxycarbonyl)(methyl)amino)azetidin-1-yl)-3- oxopentanoate. Step 3: Preparation of tert-butyl (1-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate. Compound 124.3
The title compound can be prepared using similar procedure as Intermediate I, replacing tert-butyl 4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl}piperazine-1-carboxylate (Intermediate E) with tert-butyl (1-(2-bromo-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate. Step 4: Preparation of tert-butyl (1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate. Compound 124.4
The title compound can be prepared using similar procedure as Intermediate J, replacing tert-butyl 4-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate I) with tert- butyl (1-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(3-(methylamino)azetidin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 124.5
The title compound can be prepared using similar procedure as Intermediate K, replacing tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (Intermediate J) with tert-butyl (1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)(methyl)carbamate. Step 6: Preparation of 5-(benzyloxy)-N-(1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)-N,6- dimethylpyrimidine-4-carboxamide. Compound 124.6
The title compound can be prepared using similar procedure as Intermediate H, replacing 2-(2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (Intermediate G) with N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(3- (methylamino)azetidin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Step 7: Preparation of N-(1-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)azetidin-3-yl)-5-hydroxy-N,6-dimethylpyrimidine-4-carboxamide. Compound 124
The title compound can be prepared using similar procedure as Example 1, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2,3- dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide with 5-(benzyloxy)-N-(1-(4-(2-((2- chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)azetidin-3-yl)-N,6- dimethylpyrimidine-4-carboxamide. EXAMPLE 125 Synthesis of N-(1-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)- 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin- 6-yl)pyrrolidin-3-yl)-5-hydroxy-N,6-dimethylpyrimidine-4-carboxamide.
The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl methyl(pyrrolidin-3- yl)carbamate. EXAMPLE 126 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-((1R,6R)-7-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,7- diazabicyclo[4.2.0]octan-3-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 126
The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl (1R,6R)-3,7- diazabicyclo[4.2.0]octane-7-carboxylate. EXAMPLE 127 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-((1S,5R)-6-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,6- diazabicyclo[3.2.0]heptan-3-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide.
The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl (1S,5R)-3,6- diazabicyclo[3.2.0]heptane-6-carboxylate. EXAMPLE 128 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-6-(5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 128
The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with tert-butyl 5,6-dihydroimidazo[1,5- a]pyrazine-7(8H)-carboxylate. EXAMPLE 129 Synthesis of 2-(6-(3-acetyl-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide.
The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with 1-(5,6,7,8-tetrahydroimidazo[1,5- a]pyrazin-3-yl)ethan-1-one. EXAMPLE 130 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(3-(5-hydroxypyrimidin-4-yl)-5,6-dihydroimidazo[1,5-a]pyrazin-7(8H)- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 130
The title compound can be prepared using similar procedure as Example 124, replacing tert-butyl azetidin-3-yl(methyl)carbamate with 3-(5-methoxypyrimidin-4-yl)- 5,6,7,8-tetrahydroimidazo[1,5-a]pyrazine. EXAMPLE 131 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-2,2-difluoroacetamide.
Step 1: Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-1,1-difluoro-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 131.1
The title compound can be prepared using similar procedure as Intermediate J, replacing N-(2-chloro-4-(trifluoromethyl)phenyl)-2-iodoacetamide (Intermediate D) with N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoro-2-iodoacetamide. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-2,2-difluoroacetamide. Compound 131.2
The title compound can be prepared using similar procedure as Intermediate K, replacing tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (Intermediate J) with tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-1,1-difluoro-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoroacetamide. Compound 131.3
The title compound can be prepared using similar procedure as Intermediate H, replacing 2-(2-bromo-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-
4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (Intermediate G) with N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-2,2-difluoroacetamide. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoroacetamide. Compound 131
The title compound can be prepared using similar procedure as Compound 1, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2,3- dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide with 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2- difluoroacetamide. EXAMPLE 132 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(isochroman-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-2,2-difluoroacetamide. Compound 132
The title compound can be prepared using similar procedure as Compound 131, replacing 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (isochroman-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. EXAMPLE 133 Synthesis of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2,2-difluoroacetamide.
The title compound can be prepared using similar procedure as Compound 131, replacing 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. EXAMPLE 134 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(dimethylamino)-5-ethyl- 6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-2,2-difluoroacetamide.
The title compound can be prepared using similar procedure as Compound 25, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (Intermediate H) with tert-butyl 4-(2-bromo-4-(2-((2-
chloro-4-(trifluoromethyl)phenyl)amino)-1,1-difluoro-2-oxoethyl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate X). EXAMPLE 135 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(3-hydroxy-1-methyl-1H-pyrazole-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-2,2-difluoroacetamide.
The title compound can be prepared using similar procedure as Compound 131, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)-1-methyl-1H-pyrazole-4-carboxylic acid. EXAMPLE 136 Synthesis of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 136.1
The title compound can be prepared using similar procedure as Intermediate R, replacing tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Q) with tert- butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((3-(perfluoroethyl)bicyclo[1.1.1]pentan-1- yl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Y). Step 2: Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 136.2
The title compound can be prepared using similar procedure as Compound 10, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (Intermediate H) with 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
Step 3: Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 136
The title compound can be prepared using similar procedure as Compound 1, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2,3- dihydrobenzo[b][1,4]dioxin-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide with 2-(2-(benzo[d][1,3]dioxol-5-yl)- 6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1- yl)acetamide. EXAMPLE 137 Synthesis of 2-(2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 137
The title compound can be prepared using similar procedure as Compound 136, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (2,3-dihydrobenzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. EXAMPLE 138
Synthesis of 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 136, replacing 2-(2H-1,3-benzodioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane with 2- (isochroman-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. EXAMPLE 139 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 134, replacing 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)-2,2-difluoroacetamide with 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(dimethylamino)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan-1- yl)acetamide. EXAMPLE 140
Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(3-hydroxypicolinoyl)piperazin-1- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 139, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)picolinic acid. EXAMPLE 141 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(3-hydroxy-1-methyl-1H-pyrazole- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 139, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)-1-methyl-1H-pyrazole-4-carboxylic acid. EXAMPLE 142 Synthesis of 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-morpholino-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)acetamide. Compound 142
The title compound can be prepared using similar procedure as Compound 139, replacing dimethylamine with morpholine. EXAMPLE 143 Synthesis of 2-(5-ethyl-6-(4-(3-hydroxypicolinoyl)piperazin-1-yl)-2-morpholino-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-(perfluoroethyl)bicyclo[1.1.1]pentan- 1-yl)acetamide. Compound 143
The title compound can be prepared using similar procedure as Compound 140, replacing dimethylamine with morpholine. EXAMPLE 144 Synthesis of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 131, replacing tert-butyl 4-(2-bromo-4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-1,1- difluoro-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (Intermediate X) with tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- ((trifluoromethyl)thio)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Z). EXAMPLE 145 Synthesis of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(4-(3-hydroxypicolinoyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 144, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)picolinic acid. EXAMPLE 146 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-((trifluoromethyl)thio)phenyl)acetamide. Compound 146
The title compound can be prepared using similar procedure as Compound 131, replacing tert-butyl 4-(2-bromo-4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-1,1-
difluoro-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (Intermediate X) with tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4- (2-oxo-2-((4-((trifluoromethyl)thio)phenyl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate AA). EXAMPLE 147 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(3- hydroxypicolinoyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(4- ((trifluoromethyl)thio)phenyl)acetamide.
The title compound can be prepared using similar procedure as Compound 146, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)picolinic acid. EXAMPLE 148 Synthesis of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(dimethylamino)-5- ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 148
The title compound can be prepared using similar procedure as Compound 139, replacing tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Y) with tert-butyl 4-(2-bromo-4-
(2-((2-chloro-4-((trifluoromethyl)thio)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Z). EXAMPLE 149 Synthesis of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(dimethylamino)-5- ethyl-6-(4-(3-hydroxypicolinoyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 148, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)picolinic acid. EXAMPLE 150 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(4- ((trifluoromethyl)thio)phenyl)acetamide. Compound 150
The title compound can be prepared using similar procedure as Compound 139, replacing tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-(2-oxo-2-((3- (perfluoroethyl)bicyclo[1.1.1]pentan-1-yl)amino)ethyl)-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate Y) with tert-butyl 4-(2-bromo-5- ethyl-7-oxo-4-(2-oxo-2-((4-((trifluoromethyl)thio)phenyl)amino)ethyl)-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (Intermediate AA).
EXAMPLE 151 Synthesis of 2-(2-(dimethylamino)-5-ethyl-6-(4-(3-hydroxypicolinoyl)piperazin-1- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(4- ((trifluoromethyl)thio)phenyl)acetamide.
The title compound can be prepared using similar procedure as Compound 150, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)picolinic acid. EXAMPLE 152 Synthesis of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(4-(3-hydroxy-1-methyl-1H-pyrazole-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound can be prepared using similar procedure as Compound 145, replacing 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (Intermediate A) with 3- (benzyloxy)-1-methyl-1H-pyrazole-4-carboxylic acid. EXAMPLE 153 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro-8H- imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
Compound 153
Step 1: Preparation of 2,3-dibromo-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine. Compound 153.1
To a solution of 5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine (1.0 g, 0.8 mmol, Intermediate) in ACN (10 mL) was added NBS (2.9 g, 1.6 mmol) in portions at 0 °C, and then stirred for additional 15 min at 0 °C. The resulting mixture was quenched by water (20 mL) at 0 °C, extracted with EA (2 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by neutral alumina 200-300 mesh column chromatography, eluted with DCM/MeOH (10/1) to afford 2,3-dibromo-5,6-dihydro-8H-imidazo[2,1- c][1,4]oxazine (1.2 g, 56% yield) as a white solid. LCMS observed m/z = 281.93[M+H] +. Step 2: Preparation of 2-bromo-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine. Compound 153.2
To a solution of 2,3-dibromo-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine (1.2 g, 4.3 mmol) in THF (15 mL) was added isopropyl magnesium chloride - Lithium chloride complex (700 mg, 6.8 mmol, 1.3 M solution in THF) dropwise at 0 °C under nitrogen atmosphere, and stirred at 0 °C for additional 2 h. The resulting mixture was quenched by water (15 mL), extracted with EA (2 x 15 mL). The combined organic phases were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 2-bromo-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine (120 mg, crude) as a yellow solid. LCMS observed m/z = 203.05[M+H] +.
Step 3: Preparation of 2-(tributylstannyl)-5,6-dihydro-8H-imidazo[2,1- c][1,4]oxazine. Compound 153.3
To a solution of 2-bromo-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine (100 mg, 0.5 mmol) in THF (2 mL) was added n-BuLi (32 mg, 1.6 M in hexane, 0.6 mmol) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was stirred at -78 °C for 30 min under nitrogen atmosphere. To the above mixture was added tributyl(chloro)stannane (208 mg, 0.6 mmol) in portions over 10 min at -78 °C. The resulting mixture was stirred for additional 30 min at -78 °C. The reaction was quenched by the addition of water (5 mL) at 25 °C. The aqueous layer was extracted with EA (2x5 mL). The combined organic phases were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE:(DCM/MeOH=10:1) = (0-100) to afford 2- (tributylstannyl)-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine (170 mg, crude) as a yellow oil. LCMS observed m/z = 413.10[M+H] +. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 153.4
To a solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide, Intermediate H (120 mg, 0.2 mmol, Intermediate) in DMF (2 mL) were added 2-(tributylstannyl)-5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazine
(157 mg, 0.4 mmol) and Pd (PPh3)4 (18 mg, 0.02 mmol) at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 4 hours under nitrogen atmosphere. The resulting mixture was quenched by water (5 mL), extracted with EA (2 x 5 mL). The combined organic layers were washed with brine (2 x 30 mL), dried over anhydrous Na2SO4, concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 813.10[M+H] +. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro- 8H-imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 153
To a solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(5,6-dihydro-8H-imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (100 mg, 0.1 mmol) in DCM (2 mL) was added boron trichloride (56 mg, 0.5 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C for 1 h under nitrogen atmosphere. The reaction was quenched with water at 25 °C. The aqueous layer was extracted with DCM (2 x 4 mL). The organic layers were concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column: SunFire C185 m, 30 mm X 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 28% B to 65% B in 8 min; Wave Length: 254 nm/220 nm; RT1(min): 6.55/8.00 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro-8H- imidazo[2,1-c][1,4]oxazin-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (15 mg, 16% yield ) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.43 (s, 1H), 8.53 (s,
1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.2 Hz, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.56 (s, 1H), 5.37 (s, 2H), 4.82 (s, 2H), 4.52 (s, 1H), 4.39 (t, J = 5.3 Hz, 2H), 4.08 (t, J = 5.3 Hz, 2H), 3.58 – 3.44 (m, 4H), 3.09 – 2.94 (m, 3H), 2.86 – 2.80 (m, 1H), 2.71 – 2.62 (m, 1H), 2.44 (s, 3H), 1.22 (q, J = 7.4, 6.7 Hz, 3H). LCMS observed m/z = 742.25[M+H] +. Note: One exchangeable proton was not visible in NMR spectra. EXAMPLE 154 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{4H,6H,7H-pyrazolo[3,2- c][1,4]oxazin-2-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 154
Step 1: Preparation of methyl 3-bromo-1-(2-bromoethyl)-1H-pyrazole-5- carboxylate. Compound 154.1
To a stirred mixture of methyl 5-bromo-2H-pyrazole-3-carboxylate (2.0 g, 9.8 mmol) and K2CO3 (4.0 g, 29.3 mmol) in ACN (10 mL) was added dibromoethane (9.2 g, 48.8 mmol) dropwise at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 80 °C for 2 hours under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to 25 °C. The resulting mixture was filtered, the filter cake was washed with ACN (3 x 50 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford methyl 3-bromo-1-(2-bromoethyl)-1H-pyrazole-5-carboxylate (980 mg, 32.1% yield) as a white solid. LCMS observed m/z = 310.90 [M+H]+. Step 2: Preparation of (3-bromo-1-(2-bromoethyl)-1H-pyrazol-5-yl)methanol. Compound 154.2
To a stirred mixture of methyl 5-bromo-2-(2-bromoethyl)pyrazole-3-carboxylate (500 mg, 1.6 mmol) in THF (5 mL) was added DIBAL-H (8 mL, 8.0 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 25 °C for 1 hour under nitrogen atmosphere. After completion of the reaction, the reaction was quenched by the addition of water/ice (10 mL) at 0 °C. The aqueous layer was extracted with EA (3 x 20 mL). The combined organic layers were concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 35% gradient in 20 minutes; detector, UV 220 nm) to give (3-bromo-1-(2-bromoethyl)-1H-pyrazol-5- yl)methanol (360 mg, 79.1% yield) as a white solid. LCMS observed m/z = 282.90 [M+H]+. Step 3: Preparation of 2-bromo-6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazine. Compound 154.3
To a stirred solution of methyl 3-oxopentanoate (30.0 g, 230.5 mmol) in DCM (300 mL) was added SO2Cl2 (22 mL, 276.6 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The reaction was quenched with water (500 mL) at room temperature and extracted with DCM (3 x 300 mL). The organic layers were washed with brine (500 mL) and dried over anhydrous Na2SO4. After filtration the filtrate was concentrated under reduced pressure to afford 2-bromo-6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazine (34.2 g, 91.2% yield) as a colorless oil. LCMS observed m/z = 202.97 [M+H]+. Step 4: Preparation of 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-6,7-dihydro- 4H-pyrazolo[5,1-c][1,4]oxazine. Compound 154.4
To a stirred mixture of 2-bromo-4H,6H,7H-pyrazolo[3,2-c][1,4]oxazine (100 mg, 0.5 mmol), KOAc (145 mg, 1.5 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (250 mg, 1.0 mmol) in dioxane (1 mL) was added Pd(dppf)Cl2 (22 mg, 0.03 mmol) in portions at 25 °C under nitrogen atmosphere. The
resulting mixture was stirred at 100 °C for 2 hours under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to 25 °C. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 0% to 20% gradient in 20 minutes; detector, UV 254 nm) to give 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-6,7-dihydro-4H-pyrazolo[5,1- c][1,4]oxazine (35 mg, 28.3% yield) as a colorless oil. LCMS observed m/z = 251.15 [M+H]+. Step 5: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(6,7-dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 154.5
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol), 2- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4H,6H,7H-pyrazolo[3,2-c][1,4]oxazine (32 mg, 0.2 mmol) and K2CO3 (53 mg, 0.4 mmol) in dioxane (1 mL) and H2O (0.2 mL) was added Pd(dppf)Cl2 (9 mg, 0.01 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 90 °C for 2 hours under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to 25 °C. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 40% to 60% gradient in 20 minutes; detector, UV 254
nm) to give 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(6,7- dihydro-4H-pyrazolo[5,1-c][1,4]oxazin-2-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (42 mg, 40.1% yield) as a white solid. LCMS observed m/z = 832.26 [M+H]+. Step 6: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(6,7-dihydro- 4H-pyrazolo[5,1-c][1,4]oxazin-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 154
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{4H,6H,7H-pyrazolo[3,2-c][1,4]oxazin-2-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (40 mg, 0.05 mmol) in DCM (2 mL) was added boron trichloride (56 mg, 0.5 mmol) in portions at 0 °C. The resulting mixture was stirred at 0 °C for 1 hour. After completion of the reaction, the reaction mixture was quenched with MeOH (1 mL) and concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 20% to 40% gradient in 20 minutes; detector, UV 254 nm) to give N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-{4H,6H,7H-pyrazolo[3,2-c][1,4]oxazin-2-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (4.0 mg, 10.5% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.80 (s, 1H), 8.82 (s, 1H), 8.62 (s, 1H), 8.48 (d, J = 8.6 Hz, 1H), 7.66 (s, 1H), 7.56 (d, J = 8.8 Hz, 1H), 7.04 (s, 1H), 5.87 (d, J = 15.2 Hz, 1H), 5.70 (d, J = 12.4 Hz, 1H), 5.19 (s, 2H), 4.95 (d, J = 8.7 Hz, 1H), 4.89 – 4.68 (m, 2H), 3.92 – 3.74 (m, 2H), 3.61 – 3.44 (m, 1H), 3.29 – 3.17 (m, 2H), 3.17 – 3.04 (m, 1H), 2.95 – 2.76 (m, 2H), 2.60 (s, 3H), 2.21 (s, 1H), 1.43 – 1.33 (m, 3H), 1.33 – 1.22 (m, 2H). LCMS observed m/z = 742.30 [M+H]+.
EXAMPLE 155 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylbenzo[d]oxazol-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 155
Step 1: Preparation of 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3-benzoxazole. Compound 155.1
To a stirred mixture of 4-bromo-2-methyl-1,3-benzoxazole (300 mg, 1.4 mmol) and bis(pinacolato)diboron (719 mg, 2.8 mmol) in dioxane (5 mL) were added KOAc (417 mg, 4.2 mmol) and Pd(dppf)Cl2.CH2Cl2 (114 mg, 0.1 mmol) in portions at 25 °C. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 1 h under nitrogen atmosphere. The reaction was quenched with water (5 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 8 mL). The combined organic layers were dried over anhydrous Na2SO4. The mixture was concentrated in vacuo to afford (365 mg, crude) of 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzoxazole as a white solid. LCMS observed m/z =260.14 [M+H]+. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(2-methyl-1,3-benzoxazol-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 155.2
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzoxazole (66 mg, 0.3 mmol) in dioxane (1 mL)/H2O (100 uL) were added K3PO4 (81 mg, 0.4 mmol) and Pd(dppf)Cl2.CH2Cl2 (10 mg, 0.01 mmol) in portions at 25 °C. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 16 h under nitrogen atmosphere. The reaction was quenched with water (5 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 9% MeOH in DCM) to afford (20 mg, 15% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(2-methyl-1,3-benzoxazol-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)- N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a brown oil. LCMS observed m/z =841.25 [M+H]+. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylbenzo[d]oxazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 155
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(2-methyl-1,3-benzoxazol-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)- N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (20 mg, 0.02 mmol) in TFA (1 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 31% B to 50%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.33) to afford (13.9 mg, 75% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-1,3-benzoxazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.57 (s, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 7.5 Hz, 2H), 7.83 (d, J = 8.1 Hz, 1H), 7.72 (dd, J = 8.6, 2.1 Hz, 1H), 7.48 (t, J = 7.9 Hz, 1H), 5.43 (s, 2H), 4.56 (m, 1H), 3.55 (m, 3H), 3.26 (m, 1H), 3.09 – 3.01 (m, 3H), 2.87 (m, 1H), 2.68 (s, 4H), 2.45 (s, 3H), 1.24 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z =751.20 [M+H]+. EXAMPLE 156 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-1,3-benzothiazol-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 156
Step 1: Preparation of 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3-benzothiazole. Compound 156.1
To a stirred solution of 4-bromo-2-methyl-1,3-benzothiazole (600 mg, 2.6 mmol) and bis(pinacolato)diboron (1.3 g, 5.3 mmol) in DMF (10 mL) were added KOAc (0.8 g, 7.9 mmol) and Pd(dppf)Cl2.CH2Cl2 (214 mg, 0.3 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred overnight at 80 °C under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (10:1) to afford 2-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzothiazole (610 mg, 84% yield) as a white solid. LCMS observed m/z = 275.20 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-1,3-benzothiazol-4- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 156
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzothiazole (521 mg, 0.2 mmol) in dioxane (1.5 mL) and H2O (0.3 mL) were added Pd(dppf)Cl2CH2Cl2 (10 mg, 0.1 mmol) and K2CO3 (52 mg, 0.4 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred overnight at 90 °C under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18, 5 μm, 30 *150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 42% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.53) to afford N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-(2-methyl-1,3-benzothiazol-4-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}acetamide (3.1 mg, 3% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.58 (s, 1H), 8.21 (d, J = 8.1 Hz, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 10.5 Hz, 2H), 7.71 (d, J = 8.6 Hz, 1H), 7.52 (t, J = 7.8 Hz, 1H), 5.42 (s, 2H), 4.55 (m, 1H), 3.04 (d, J = 9.5 Hz, 4H), 2.90 – 2.84 (m, 2H), 2.82 (s, 3H), 2.71 (s, 1H), 2.45 (s, 2H), 1.45 (s, 1H), 1.24 (d, J = 6.9 Hz, 5H). LCMS observed m/z = 767.30 [M+H]+. Note: One exchangeable proton was not visible in NMR spectra. EXAMPLE 157 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{2H,3H-furo[2,3- b]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide. Compound 157
Step 1: Preparation of 5-bromo-2-(but-3-yn-1-yloxy)pyrimidine. Compound 157.1
A solution of NaH (1.2 g, 29.0 mmol, 60%) and 3-butyn-1-ol (2.0 g, 29.0 mmol) in THF (10 mL) was stirred for 30 min at 0 °C. To the above mixture was added 5-bromo-2- chloropyrimidine (4.0 g, 20.7 mmol) dropwise over 20 min at 0 °C. The resulting mixture was stirred overnight at room temperature. The reaction was quenched with water (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (3.3 g, 70% yield) of 5-bromo-2-(but-3-yn-1-yloxy)pyrimidine as a white solid. LCMS observed m/z =224.97 [M-H]-. Step 2: Preparation of 5-bromo-2H,3H-furo[2,3-b]pyridine. Compound 157.2
A solution of 5-bromo-2-(but-3-yn-1-yloxy)pyrimidine (500 mg, 2.2 mmol) in nitrobenzene (5 mL) was stirred for 2 h at 230 °C. The reaction mixture was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (165 mg, 37% yield) of 5-bromo-2H,3H-furo[2,3-b]pyridine as a white solid. LCMS observed m/z =199.96 [M+H]+. Step 3: Preparation of 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H,3H- furo[2,3-b]pyridine.
Compound 157.3
To a stirred mixture of 5-bromo-2H,3H-furo[2,3-b]pyridine (165 mg, 0.8 mmol) and bis(pinacolato)diboron (251 mg, 0.9 mmol) in dioxane (2 mL) were added KOAc (243 mg, 2.5 mmol) and Pd(dppf)Cl2CH2Cl2 (65 mg, 0.08 mmol) in portions at 25 °C. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 16 h under nitrogen atmosphere. The reaction was quenched with water (3 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (92 mg, 45% yield) of 5-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-2H,3H-furo[2,3-b]pyridine as a light-yellow oil. LCMS observed m/z =248.14 [M+H]+. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrofuro[2,3-b]pyridin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 157.4
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (150 mg, 0.2 mmol) and 5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H,3H-furo[2,3-b]pyridine (94 mg, 0.4 mmol) in dioxane (800 uL) were added K3PO4 (56 mg, 0.6 mmol), H2O (200 uL) and Pd(PPh3)4 (22 mg, 0.02 mmol) in portions at 25 °C. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 16 h under nitrogen atmosphere. The reaction was
quenched with water (2 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (16 mg, 10% yield) of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrofuro[2,3-b]pyridin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide as a yellow oil. LCMS observed m/z =829.25 [M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2- {2H,3H-furo[2,3-b]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide. Compound 157
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-{2H,3H-furo[2,3-b]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (16 mg, 0.02 mmol) in TFA (1 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min mL/min; Gradient: 33% B to 42%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.22) to afford (5 mg, 34% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{2H,3H- furo[2,3-b]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.65 – 8.62 (m, 1H), 8.56 (s, 1H), 8.25 (d, J = 2.0 Hz, 1H), 8.06 (d, J = 8.5 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.73 (d, J = 8.6 Hz, 1H), 5.39 (s, 2H), 4.66 (t, J = 8.6 Hz, 2H), 4.54 (m, 1H), 3.53 (m, 3H), 3.28 (m, 3H), 3.03 (m, 3H), 2.84 (m, 1H), 2.67
(m, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z =739.20 [M+H]+. EXAMPLE 158 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-7-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 158
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1-benzofuran-7-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide Compound 158.1
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]-acetamide, Intermediate H (120 mg, 0.2 mmol) and 2-(2,3-dihydro-1-benzofuran-7-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane
(45 mg, 0.2 mmol) in dioxane (2 mL) and H2O (0.5 mL) were added Pd(dppf)Cl2CH2Cl2 (12 mg, 0.1 mmol) and K2CO3 (63 mg, 0.5 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 100 ℃. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL) at room temperature. The resulting mixture was extracted with EA (3 x 5 mL). The combined organic layers were washed with brine (2 x 3 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (125 mg) was used in the next step directly without further purification. LCMS observed m/z = 828.25 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-7-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 158
A mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(2,3-dihydro-1-benzofuran-7-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (125 mg, 0.2 mmol, 1.0 equiv.) in TFA (5 mL) was stirred for one hour at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18 5μm, 30*150mm; Mobile Phase A: Water(10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 34% B to 90% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 6.80/9.98) to afford (9.7 mg, 8.6% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-7-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400
MHz, DMSO-d6) δ 10.36 (s, 1H), 8.53 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.96 (d, J = 2.1 Hz, 1H), 7.79 – 7.69 (m, 2H), 7.35 (d, J = 7.2 Hz, 1H), 6.93 (t, J = 7.6 Hz, 1H), 5.37 (s, 2H), 4.64 (t, J = 8.8 Hz, 2H), 4.53 (m, 1H), 3.52 (m, 3H), 3.24 (t, J = 8.6 Hz, 3H), 3.01 (d, J = 9.2 Hz, 3H), 2.83 (m, 1H), 2.67 (m, 1H), 2.43 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 738.20 [M+H]+. Note: One exchangeable proton was not visible in NMR spectra. EXAMPLE 159 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-2H-1- benzopyran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 159
Step 1: Preparation of 2-(3,4-dihydro-2H-1-benzopyran-5-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane Compound 159.1
To a stirred mixture of 5-bromo-3,4-dihydro-2H-1-benzopyran (40 mg, 0.2 mmol) and bis(pinacolato)diboron (57 mg, 0.2 mmol) in dioxane (2 mL) were added Pd(dppf)Cl2CH2Cl2 (15 mg, 0.1 mmol) and KOAc (55 mg, 0.6 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 100 ℃. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with EA (3 x 2 mL). The filtrate was concentrated under
reduced pressure. The crude product (49 mg) was used in the next step directly without further purification. LCMS observed m/z = 261.14 [M+H]+. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-2H-1-benzopyran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide Compound 159.2
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- (3,4-dihydro-2H-1-benzopyran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (49 mg, 0.2 mmol) in dioxane (2 mL) and H2O (0.5 mL) were added Pd(dppf)Cl2CH2Cl2 (10 mg, 0.1 mmol) and K2CO3 (53 mg, 0.4 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 100 ℃. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL) at room temperature. The resulting mixture was extracted with EA (3 x 5 mL). The combined organic layers dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (120 mg) was used in the next step directly without further purification. LCMS observed m/z = 842.28 [M+H]+. Step 3: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 2H-1-benzopyran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 159
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,4-dihydro-2H-1-benzopyran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (120 mg, 0.1 mmol) in TFA (5 mL) was stirred for 1 hour at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C185 μm, 30*150mm; Mobile Phase A: Water (10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 34% B to 90% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 6.80/9.98) to afford (12.1 mg, 11.1% yield) of N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-2H-1-benzopyran-5-yl)-5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.36 (s, 2H), 8.55 (s, 1H), 8.05 (d, J = 8.5 Hz, 1H), 7.99 – 7.95 (m, 1H), 7.72 (dd, J = 9.3, 2.1 Hz, 1H), 7.47 (dd, J = 7.7, 1.3 Hz, 1H), 7.19 (t, J = 7.9 Hz, 1H), 6.88 (m, 1H), 5.37 (s, 2H), 4.54 (m, 1H), 4.18 – 4.09 (m, 2H), 3.53 (m, 3H), 3.04 (m, 4H), 2.85 (m, 1H), 2.67 (s, 1H), 2.44 (s, 3H), 1.85 (m, 2H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.20 [M+H]+. Note: Two exchangeable protons were not visible in NMR spectra. EXAMPLE 160 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-2H-1- benzopyran-8-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 160
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-2H-1-benzopyran-8-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 160.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (80 mg, 0.1 mmol) and 3,4- dihydro-2H-1-benzopyran-8-ylboronic acid (27 mg, 0.1 mmol) in dioxane (2 mL) and H2O (0.2 mL) were added K2CO3 (42 mg, 0.3 mmol) and Pd(dppf)Cl2CH2Cl2 (16 mg, 0.1 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 80 °C and stirred at 80 °C for 24 hours under nitrogen atmosphere. The mixture was allowed to cool down to room temperature, diluted with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with CH2Cl2/MeOH (10:1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-2H-1-benzopyran-8-yl)-5-ethyl-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (80 mg, 93% yield) as a yellow solid. LCMS observed m/z = 842.28 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 2H-1-benzopyran-8-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 160
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-2H-1-benzopyran-8-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.1 mmol) in THF (3 mL) was added BCl3 (1 mL) in one portion at room temperature. The reaction mixture was stirred overnight at room temperature under nitrogen atmosphere. Then the resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18, 5 μm, 30 * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 35% B to 52% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.07) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 2H-1-benzopyran-8-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (10.1 mg, 22% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.09 (s, 1H), 8.56 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.70 – 7.74 (m, 1H), 7.53 (d, J = 7.5 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 6.91 (t, J = 7.6 Hz, 1H), 5.37 (s, 2H), 4.54 (m, 1H), 4.17 (t, J = 5.1 Hz, 2H), 3.60 – 3.48 (m, 3H), 2.98 – 3.05 (m, 2H), 2.83 (m, 3H), 2.71 – 2.63 (m, 4H), 2.45 (s, 3H), 1.94 – 1.99 (m, 2H), 1.22 (q, J = 7.5, 5.7 Hz, 3H). LCMS observed m/z = 752.15 [M+H]+. EXAMPLE 161
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyri midine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{1H,3H,4H-pyrano[4,3-c]pyridin- 8-yl}-[1,2,4]triazolo[1,5-a]pyri midin-4-yl}acetamide. Compound 161
omo-5-[(E)-2-ethoxyethenyl]pyridine-4- carboxylate. Compound 161.1
To a mixture of methyl 3,5-dibromopyridine-4-carboxylate (1.8 g, 6.1 mmol) and 1- [(E)-2-ethoxyethenyl]-3,3,4,4-tetramethyl-1lambda3,2,5-bromadioxolane (2 mL, 6.1 mmol) in dioxane (40 mL) and H2O (10 mL) was added Pd(dppf)Cl2 (446 mg, 0.6 mmol) and K2CO3 (1.7 g, 12.2 mmol) at 25 °C. The mixture was heated to 80°C and stirred at 80°C for 2h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25°C. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford methyl 3-bromo-5-[(E)-2-ethoxyethenyl]pyridine-4-carboxylate (760 mg, 43% yield) as a light yellow oil. LCMS observed m/z = 286.00 [M+H] +. Step 2: Preparation of methyl 3-bromo-5-(2-oxoethyl)pyridine-4-carboxylate. Compound 161.2
To a solution of methyl 3-bromo-5-[(E)-2-ethoxyethenyl]pyridine-4-carboxylate (760 mg, 2.6 mmol) in THF (10 mL) was added conc. HCl (10 mL) drop wise at 25 °C. The mixture was stirred at 25 °C for 2 h. Then the reaction was basified to pH 9 with sat. aq. NaHCO3 at 25 °C. The resulting mixture was extracted with EA (3 x 20 mL). The combined
organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford methyl 3-bromo-5-(2-oxoethyl)pyridine-4- carboxylate (650 mg, 94% yield) as a brown oil. LCMS observed m/z = 257.97 [M+H] +. Step 3: Preparation of 2-[5-bromo-4-(hydroxymethyl)pyridin-3-yl]ethanol. Compound 161.3
To a solution of methyl 3-bromo-5-(2-oxoethyl)pyridine-4-carboxylate (650 mg, 2.5 mmol) in MeOH (20 mL) was added NaBH4 (381 mg, 10.1 mmol) at 25 °C. The mixture was stirred at 25°C for 2 h under nitrogen atmosphere. Then the reaction was quenched by the addition of H2O (20 mL) at 25°C. The resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (0/1) to afford 2-[5-bromo-4- (hydroxymethyl)pyridin-3-yl]ethanol (330 mg, 56% yield) as a white solid. LCMS observed m/z = 231.99 [M+H] +. Step 4: Preparation of 8-bromo-1H,3H,4H-pyrano[4,3-c]pyridine. Compound 161.4
To a solution of 2-[5-bromo-4-(hydroxymethyl)pyridin-3-yl]ethanol (30 mg, 0.1 mmol) in ACN (5 mL) was added TsOH (44 mg, 0.2 mmol) at 25 °C and the mixture was heated to 110 °C and stirred at 110 °C for 36 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 8-bromo-1H,3H,4H-pyrano[4,3-c]pyridine (9 mg, 32% yield) as a yellow oil. LCMS observed m/z = 213.98 [M+H]+. Step 5: Preparation of 8-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H,3H,4H- pyrano[4,3-c]pyridine. Compound 161.5
To a mixture of 8-bromo-1H,3H,4H-pyrano[4,3-c]pyridine (9 mg, 0.04 mmol) and bis(pinacolato)diboron (14 mg, 0.05 mmol) in dioxane (1 mL) was added Pd(dppf)Cl2 (3 mg, 0.004 mmol) and KOAc (8 mg, 0.08 mmol, 2.0 equiv.) in one portions under nitrogen atmosphere. Then heated the reaction mixture to 100 °C and stirred at 100 °C for 16 h under nitrogen atmosphere. Then the resulting mixture was used for next step directly without further purification. LCMS observed m/z = 213.98 [M+H] +. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{1H,3H,4H-pyrano[4,3-c]pyridin-8-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoro methyl) phenyl]acetamide. Compound 161.6
To the resulting mixture from last step was added 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifle oromethyl)phenyl]acetamide, Intermediate H (15 mg, 0.02 mmol), Pd(dppf)Cl2 (1 mg, 0.002 mmol), K2CO3 (5 mg, 0.04 mmol), dioxane (0.4 mL) and H2O (0.1 mL) at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for additional 1h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25°C. The resulting mixture was filtered, the filter cake was washed with EA (3 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA = 0/1) to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{1H,3H,4H-pyrano[4,3- c]pyridin-8-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl] acetamide (6 mg, 39% yield) as a brown oil. LCMS observed m/z = 843.27 [M+H] +.
Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyri midine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{1H,3H,4H-pyrano[4,3- c]pyridin-8-yl}-[1,2,4]triazolo[1,5-a]pyri midin-4-yl}acetamide. Compound 161
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-5-ethyl-7-oxo-2-{1H,3H,4H-pyrano[4,3-c]pyridin-8-yl}-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide (5 mg, 0.006 mmol) in DCM (1 mL) was added boron trichloride (0.7 mg, 0.006 mmol) at 25°C under nitrogen atmosphere. The mixture was stirred at 25 °C for 30 min. Then the reaction was quenched with MeOH at 25°C. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X- Bridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 27% B to 50% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 7.8) to afford N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-{1H,3H,4H-pyrano[4,3-c]pyridin-8-yl}-[1,2,4] triazolo[1,5-a]pyrimidin-4-yl}acetamide (0.7 mg, 15% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.80 (s, 1H), 9.28 (s, 1H), 8.86 (s, 1H), 8.62 (s, 1H), 8.48 (d, J = 8.4 Hz, 2H), 7.64 (d, J = 2.0 Hz, 1H), 7.55 (d, J = 9.9 Hz, 1H), 5.71 (d, J = 13.1 Hz, 1H), 5.29 (s, 2H), 5.23 (s, 2H), 4.83 (d, J = 12.8 Hz, 1H), 4.04 (t, J = 5.6 Hz, 2H), 3.86 (t, J = 11.5 Hz, 2H), 3.54 (s, 1H), 3.23 (d, J = 7.9 Hz, 2H), 3.12 (s, 1H), 3.00 (t, J = 5.6 Hz, 2H), 2.83 (s, 2H), 2.60 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 753.22 [M+H] +. EXAMPLE 162 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{5H,6H,8H-pyrano[3,4-b]pyridin- 3-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 162
Step 1: Preparation of methyl 5-bromo-2-(bromomethyl)pyridine-3-carboxylate. Compound 162.1
A solution of methyl 5-bromo-2-methylpyridine-3-carboxylate (4.0 g, 17.3 mmol), AIBN (0.3 g, 1.7 mmol) and NBS (3.0 g, 17.3 mmol) in CCl4 (40 mL) was stirred overnight at 80 °C. The resulting mixture was concentrated under reduced pressure. To the above mixture was added ethylbis(propan-2-yl)amine (1.8 mL, 13.9 mmol) and diethyl phosphonate (1.4 mL, 10.1 mmol) in THF (50 mL) at room temperature. The resulting mixture was stirred for additional 16 hours at room temperature. The reaction was quenched with H2O (50 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (3.4 g, 63% yield) of methyl 5-bromo-2-(bromomethyl)pyridine- 3-carboxylate as a yellow oil. LCMS observed m/z = 309.90 [M+H]+. Step 2: Preparation of methyl 3-bromo-5-hydroxy-8H-pyrano[3,4-b]pyridine-6- carboxylate. Compound 162.2
NaH (807 mg, 33.6 mmol) was added to a mixture of methyl 5-bromo-2- (bromomethyl)pyridine-3-carboxylate (5.2 g, 16.8 mmol) and methyl 2-hydroxyacetate (1.5 g, 16.8 mmol) in DMF (50 mL) at 0 °C. The mixture was stirred at 25 °C for 1 h. The reaction was quenched by the addition of sat. NH4Cl (aq.) (100 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 100 mL) and the combined organics were washed with brine (100 mL), dried over Na2SO4. The resulting mixture was concentrated under reduced pressure. This resulted in (4.4 g, 91% yield) of methyl 3-bromo- 5-hydroxy-8H-pyrano[3,4-b]pyridine-6-carboxylate as a brown solid. LCMS observed m/z = 288.00 [M+H]+. Step 3: Preparation of 3-bromo-6H,8H-pyrano[3,4-b]pyridin-5-one. Compound 162.3 A solution of methyl 3-bromo-5-hydroxy-8H-pyrano[3,4-b]pyridine-6-carboxylate (4.4 g, 15.3 mmol) and conc. HCl (10.4 mL, 344.8 mmol) in EtOH (10 mL) was stirred for 1 h at 120 °C .The mixture was allowed to cool down to room temperature. The reaction was quenched with water (50 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 50 mL). The resulting mixture was dried over Na2SO4 and concentrated under reduced pressure. This resulted in (3.7 g, crude) of 3-bromo-6H,8H-pyrano[3,4- b]pyridin-5-one as a brown solid. LCMS observed m/z = 228.05 [M+H]+. Step 4: Preparation of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridin-5-ol. Compound 162.4 A solution of 3-bromo-6H,8H-pyrano[3,4-b]pyridin-5-one (3.7 g, 16.2 mmol) and NaBH4 (3.0 g, 81.1 mmol) in MeOH (3 mL) was stirred for 3 h at room temperature .The reaction was quenched with water (20 mL) at room temperature. The resulting mixture was extracted with CH2Cl2 (3 x 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (1.7 g, 45%
yield) of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridin-5-ol as a light yellow solid. LCMS observed m/z = 230.05 [M+H]+. Step 5: Preparation of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridin-5-yl methanesulfonate. Compound 162.5
To a stirred solution of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridin-5-ol (1.6 g, 7.2 mmol) and Et3N (3.0 mL, 21.7 mmol) in DCM (20 mL) was added MsCl (1.1 mL, 14.5 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 0 °C under nitrogen atmosphere. The reaction was quenched with water (20 mL) at 0 °C. The aqueous layer was extracted with CH2Cl2 (3 x 20 mL), dried over anhydrous Na2SO4. The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (300 mg, 13% yield) of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridin-5-yl methanesulfonate as a white solid. LCMS observed m/z = 307.95 [M+H]+ . Step 6: Preparation of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridine. Compound 162.6
To a stirred solution of Zn (11.5 g, 176.5 mmol) and 3-bromo-5H,6H,8H- pyrano[3,4-b]pyridin-5-yl methanesulfonate (1.7 g, 5.5 mmol) in DMF (100 mL) was added NaI (26.4 g, 176.54 mmol) at room temperature. The resulting mixture was stirred overnight at room temperature. To the above mixture was added AcOH (6.3 mL, 110.3 mmol) and acetone (50 mL) at room temperature. The resulting mixture was stirred for additional 2 min at room temperature. The resulting mixture was filtered, the filter cake was washed with EtOAc (3 x 10 mL). The reaction was quenched with water (500 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with brine (2 x 500 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (570 mg,
48%) of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridine as a yellow solid. LCMS observed m/z = 214.05 [M+H]+. Step 7: Preparation of 5H,6H,8H-pyrano[3,4-b]pyridin-3-ylboronic acid. Compound 162.7
To a stirred solution of 3-bromo-5H,6H,8H-pyrano[3,4-b]pyridine (330 mg, 1.5 mmol) and Pd(dppf)Cl2CH2Cl2 (135 mg, 0.2 mmol) in 2-methyloxolane (3 mL) were added 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (782 mg, 3.0 mmol), MeOH (1 mL) and potassium 2,2-dimethylpropanoate (324 mg, 2.3 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 90 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10 % MeOH in CH2Cl2) to afford (200 mg, 72% yield) of 5H,6H,8H-pyrano[3,4- b]pyridin-3-ylboronic acid as a pink oil. LCMS observed m/z = 180.15 [M+H]+. Step 8: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{5H,6H,8H-pyrano[3,4-b]pyridin-3-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 162.8
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (50 mg, 0.06 mmol), H2O (0.2 mL) and 5H,6H,8H-pyrano[3,4-b]pyridin-3-ylboronic acid (13 mg, 0.07 mmol) in dioxane (1 mL) were added Pd(PPh3)4 (7.3 mg, 0.006 mmol) and K2CO3 (17.5 mg, 0.1 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred
overnight at 90 °C under nitrogen atmosphere. The reaction mixture was purified by reversed phase C18 silica gel column chromatography to afford (7 mg, 13% yield) of 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2- {5H,6H,8H-pyrano[3,4-b]pyridin-3-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide as a colorless solid. LCMS observed m/z = 843.25 [M+H]+. Step 9: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{5H,6H,8H-pyrano[3,4- b]pyridin-3-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 162
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-{5H,6H,8H-pyrano[3,4-b]pyridin-3-yl}-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (7 mg, 0.008 mmol) in TFA (2 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C185 μm, 30*150mm; Mobile Phase A: water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 25% B to 40%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.63) to afford (3.0 mg, 46% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{5H,6H,8H-pyrano[3,4-b]pyridin- 3-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO- d6) δ 9.04 (s, 1H), 8.24 (s, 1H), 8.06 (d, J = 8.2 Hz, 2H), 7.94 (s, 1H), 7.68 (d, J = 8.4 Hz, 1H), 6.06 (s, 2H), 5.37 (s, 2H), 4.72 (s, 2H), 4.53 (m, 1H), 3.94 (t, J = 5.7 Hz, 2H), 3.49 (m, 3H), 3.04 (s, 2H), 2.94 (d, J = 6.0 Hz, 3H), 2.80 (m, 2H), 2.64 (m, 1H), 2.27 (s, 3H), 1.24 (s, 3H). LCMS observed m/z = 753.20 [M+H]+.
EXAMPLE 163 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,4-dihydro-1H- pyrano[3,4-c]pyridin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyri midin-4(7H)-yl)acetamide. Compound 163
Step 1: Preparation of methyl 2-(3,5-dibromopyridin-4-yl)acetate. Compound 163.1
To a solution of 3,5-dibromo-4-methylpyridine (3.0 g, 11.9 mmol) in THF (15 mL) was added LiHMDS (24.0 mL, 24.0 mmol) drop wise at 0 °C and the mixture was stirred at 0°C for 3 h under nitrogen atmosphere followed by the addition of dimethyl carbonate (1.0 mL, 11.9 mmol) dropwise at room temperature. The resulting mixture was stirred at room temperature for 3h under nitrogen atmosphere. The reaction was quenched by the addition of sat. aq. NH4Cl (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10/1) to afford methyl 2-(3,5-dibromopyridin-4- yl)acetate (3.2 g, 86% yield) as a colorless oil. LCMS observed m/z = 309.88. [M+H] +. Step 2: Preparation of methyl 5-bromo-4-(2-methoxy-2-oxoethyl)nicotinate. Compound 163.2
A mixture of methyl 2-(3,5-dibromopyridin-4-yl)acetate (3.2 g, 10.3 mmol), TEA (5.2 g, 51.7 mmol) and Pd(dppf)Cl2 (0.7 g, 1.0 mmol, 0.1 equiv.) in MeOH (100 mL) was heated to 100 °C and stirred at 100 °C for 3 h under carbon monoxide atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford methyl 5-bromo-4-(2-methoxy- 2-oxoethyl) nicotinate (1.1 g, 36.4% yield) as a yellow oil. LCMS observed m/z = 287.98 [M+H]+. Step 3: Preparation of 2-(3-bromo-5-(hydroxymethyl)pyridin-4-yl)ethan-1-ol. Compound 163.3
To a stirred mixture of methyl 5-bromo-4-(2-methoxy-2-oxoethyl)pyridine-3- carboxylate (1.0 g, 3.4 mmol) in THF (20 mL) was added lithium aluminum hydride(1.0M in THF) (3.4 mL, 3.4 mmol) drop wise at 0°C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1h under nitrogen atmosphere. The reaction was quenched by the addition of water (0.2 mL) at 0°C. Na2SO4(5 g) was added with stirring. The mixture was filtrated and the filtrate cake was washed with MeOH (3 x 10 mL). The filtrate was concentrated and the resulting mixture was purified by silica gel column chromatography, eluted with CH2Cl2 / MeOH (10/1) to afford 2-[3-bromo-5- (hydroxymethyl)pyridin-4-yl]ethanol (400 mg, 49% yield) as a yellow solid. LCMS observed m/z = 231.99 [M+H]+. Step 4: Preparation of 5-bromo-3,4-dihydro-1H-pyrano[3,4-c]pyridine. Compound 163.4
To a stirred solution of 2-[3-bromo-5-(hydroxymethyl)pyridin-4-yl]ethanol (400 mg, 1.7 mmol) in DCE (10 mL) was added H2SO4 (0.4 mL, 8.6 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 80°C for 4h under nitrogen atmosphere. The reaction was quenched by the addition of sat. aq. Na2CO3 (10mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 10mL). The combined organic layers dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography,
eluted with PE / EA (5/1) to afford 5-bromo-1H,3H,4H-pyrano[3,4-c]pyridine (120 mg, 32% yield) as a yellow solid. LCMS observed m/z = 213.98 [M+H] +. Step 5: Preparation of 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3,4-dihydro- 1H-pyrano[3,4-c]pyridine. Compound 163.5
To a stirred mixture of 5-bromo-1H,3H,4H-pyrano[3,4-c]pyridine (120 mg, 0.5 mmol), bis(pinacolato)diboron (170 mg, 0.6 mmol) and KOAc(138 mg, 1.4 mmol) in dioxane (10 mL) was added Pd(dppf)Cl2 (82 mg, 0.1 mmol) in portions at 25 °C. The mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. The residue was concentrated and the residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1H,3H,4H-pyrano[3,4-c]pyridine (80 mg, 54% yield) as a yellow oil. LCMS observed m/z = 262.15 [M+H]+. Step 6: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,4-dihydro-1H-pyrano[3,4-c]pyridin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(tri fluoromethyl)phenyl)acetamide. Compound 163.6
To a stirred mixture of 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H,3H,4H- pyrano[3,4-c]pyridine (40 mg, 0.1 mmol) in dioxane (10 mL) and H2O (1 mL) were added K2CO3 (63 mg, 0.4 mmol), Pd(dppf)Cl2 (11 mg, 0.01 mmol) and 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-
a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide, Intermediate H (145 mg, 0.2 mmol) in turns at 25 °C under nitrogen atmosphere. The mixture was heated to 100 °C and stirred at 100 °C for 2 hours under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 40% to 90% gradient in 15 min; detector, UV 254 nm to give 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3,4-dihydro- 1H-pyrano[3,4-c]pyridin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (2-chloro-4-(trifluoromethyl)phenyl)acetamide (80 mg, 62% yield) as a yellow solid. LCMS observed m/z = 843.27 [M+H]+. Step 7: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,4-dihydro- 1H-pyrano[3,4-c]pyridin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyri midin-4(7H)-yl)acetamide. Compound 163
A solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(3,4-dihydro-1H-pyrano[3,4-c]pyridin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoro methyl)phenyl)acetamide (80 mg, 0.09 mmol) in TFA (3 mL) was heated to 100 °C and stirred at 80 °C for 30 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (80 mg) was purified by Prep-HPLC with the following conditions (Column: X- select CSH C185 m, 30 mm * 150 mm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 ml/min; Gradient: 40% B to 55% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 5.68) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,4- dihydro-1H-pyrano[3,4-c]pyridin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (8.7 mg, 11% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.43 (s, 1H), 10.24 (s,
1H), 8.99 (s, 1H), 8.59 (s, 1H), 8.40 (s, 1H), 8.06 (d, J = 8.5 Hz, 1H), 8.00 – 7.96 (m, 1H), 7.73 (dd, J = 9.0, 2.1 Hz, 1H), 5.41 (s, 2H), 4.81 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 3.88 (t, J = 5.8 Hz, 2H), 3.61 – 3.46 (m, 3H), 3.25 (s, 1H), 3.18 (t, J = 5.9 Hz, 2H), 3.04 (m, 3H), 2.86 (d, J = 11.3 Hz, 1H), 2.68 (d, J = 10.1 Hz, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 753.15 [M+H] + EXAMPLE 164 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 164
In a 2-dram vial equipped with a stir bar was added 2-(2-bromo-5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide, Intermediate AC (60 mg, 86 µmol), (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid (25 mg, 100 µmol), K3PO4 (55 mg, 260 µmol), Pd(dppf)Cl2 (7.0 mg, 8.6 µmol), anhydrous 1,4-dioxane (400 µL), and deionized milliQ water (125 µL). The reaction mixture was degassed by sparging with argon for 10 minutes, then sealed with phenolic cap and heated to 80 °C via aluminum heating block. The reaction mixture was allowed to stir at 80 °C for 4 hours, then allowed to cool to room temperature and concentrated in vacuo. The crude product was purified by reverse phase column chromatography to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,1-dimethyl-1,3-dihydro-5-isobenzofuranyl)-6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide (40 mg, 63% yield) as a white solid. 1H NMR (400 MHz, DMSO) δ 10.38 (s, 1H), 8.47 (s, 1H), 7.98 (t, J = 7.5 Hz, 2H), 7.94 – 7.88 (m, 2H), 7.68 – 7.61 (m, 1H), 7.34 (d, J = 7.9 Hz, 1H), 5.33 (s, 2H), 4.95 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.47 - 3.41 (m, 3H), 3.20 – 3.16 (m, 1H), 2.98 - 2.88 (m, 3H), 2.77 (d, J = 11.1 Hz, 1H), 2.59 (d, J = 10.6 Hz, 1H), 2.37 (s, 3H), 1.38 (s, 6H), 1.14 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 766.3 [M+H]+.
EXAMPLE 165 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(3,3-dimethyl-1,3-dihydro- 5-isobenzofuranyl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(3,3-dimethyl-1,3-dihydro-5-isobenzofuranyl)- 6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo- 1,3,3a,7-tetraaza-7-indenyl] acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.41 (s, 1H), 8.45 (s, 1H), 7.98 (t, J = 7.8 Hz, 2H), 7.93 – 7.82 (m, 2H), 7.68 – 7.61 (m, 1H), 7.34 (d, J = 7.9 Hz, 1H), 5.33 (s, 2H), 4.94 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.50 – 3.41 (m, 3H), 3.20 - 3.16 (m, 1H), 2.99 - 2.89 (m, 3H), 2.77 (d, J = 11.2 Hz, 1H), 2.60 (d, J = 10.4 Hz, 1H), 2.36 (s, 3H), 1.39 (s, 6H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 766.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 166 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-3-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 166
The title compound was prepared using similar procedure as compound 164 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)- 5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.56 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.90 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 5.33 (s, 2H), 4.52 (d, J = 12.5 Hz, 1H), 4.13 (t, J = 7.3 Hz, 2H), 3.50 (t, J = 10.3 Hz, 3H), 3.27 (d, J = 14.0 Hz, 2H), 3.02 (dt, J = 26.7, 8.6 Hz, 5H), 2.82 (d, J = 11.3 Hz, 1H), 2.66 (s, 3H), 2.44 (s, 3H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 726.3 [M+H]+. EXAMPLE 167 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(4,5,6,7-tetrahydropyrazolo[1,5- a]pyridin-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 167
The title compound was prepared using similar procedure as compound 164 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-2-(4,5,6,7-tetrahydropyrazolo[1,5-a]pyridin-3-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.55 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.86 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 5.33 (s, 2H), 4.52 (d, J = 12.5 Hz, 1H), 4.10 (t, J = 6.0 Hz, 2H), 3.49 (t, J = 11.2 Hz, 3H), 3.25 (s, 2H), 3.11 – 2.92 (m, 5H), 2.82 (d, J = 11.2 Hz, 1H), 2.64 (d, J = 11.6 Hz, 1H), 2.43 (s, 3H), 1.98 (s, 2H), 1.81 (d, J = 7.6 Hz, 2H), 1.19 (t, J = 7.4 Hz, 3H) . LCMS observed m/z = 740.3 [M+H]+. EXAMPLE 168
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-2,3-dihydro-1-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 168
Step 1: Preparation of ethyl 2-(4-bromo-2-formylphenoxy)propanoate. Compound 168.1
To a stirred solution of 5-bromo-2-hydroxybenzaldehyde (2.0 g, 10.0 mmol) and ethyl a-bromopropionate (2.2 g, 11.9 mmol) in ACN (25 mL) was added K2CO3 (4.1 g, 29.8 mmol) in one portion at 25 °C. The mixture was heated to 70 °C and stirred at 70 °C for 4 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10/1) to afford ethyl 2-(4-bromo- 2-formylphenoxy)propanoate (2.7 g, 90 % yield) as a white solid. LCMS observed m/z = 301.01;303.01 [M+H] +. Step 2: Preparation of (1-(2-methoxyethyl)piperidin-4-yl)methanamine: Compound 168.2
To a stirred solution of ethyl 2-(4-bromo-2-formylphenoxy)propanoate (2.7 g, 9.0 mmol) in THF (15 mL) and H2O (3 mL) was added LiOH.H2O (0.7 g, 17.9 mmol) in portions at 25 °C. The mixture was stirred at 25 °C for 2 h. The resulting mixture was concentrated under reduced pressure to remove THF. The aqueous phase was acidified to
pH 4 with conc. HCl. The aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over Na2SO4, filtered and concentrated to give 2-(4- bromo-2-formylphenoxy)propanoic acid (2.2 g, 90 % yield) as a yellow oil. LCMS observed m/z = 270.97, 272.97 [M-H] - Step 3: Preparation of 5-bromo-2-methylbenzofuran: Compound 168.3
To a stirred solution/mixture of 2-(4-bromo-2-formylphenoxy)propanoic acid (500 mg, 1.8 mmol) in Ac2O (5 mL) was added NaOAc (300 mg, 3.6 mmol) in portions at 25 °C. The mixture was heated to 140 °C and stirred at 140 °C for 16 h. The brown reaction solution was allowed to cool to room temperature, and was diluted with toluene (10 mL). The solution was treated with 1 N NaOH (10 mL) and stirred at 25 °C for 10 min. The mixture was extracted with EtOAc (3 x 10 mL). The combined organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (100/1) to afford 5-bromo-2-methyl-1-benzofuran (90 mg, 23% yield) as a colorless oil. LCMS observed m/z = 208.97, 210.97 [M+H] +: Step 4: Preparation of 4,4,5,5-tetramethyl-2-(2-methyl-1-benzofuran-5-yl)-1,3,2- dioxaborolane Compound 168.4
To a solution of 5-bromo-2-methyl-1-benzofuran (80 mg, 0.3 mmol) and bis(pina colato)diboron (144 mg, 0.5 mmol) in dioxane (3 mL) and KOAc (111 mg, 1.1 mmol) were added Pd(dppf)Cl2 (27 mg, 0.04 mmol) at 25 °C under a nitrogen atmosphere. The mixture was heated to 90 °C and stirred at 90 °C for 5 h. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (30/1) to afford 4,4,5,5-tetramethyl-2-(2- methyl-1-benzofuran-5-yl)-1,3,2-dioxaborolane (68 mg, 69 % yield) as a yellow oil. LCMS observed m/z = 259.14 [M+H] +: Step 5: Preparation of 4,4,5,5-tetramethyl-2-(2-methyl-2,3-dihydro-1-benzofuran-5- yl)-1,3,2-dioxaborolane
Compound 168.5
To a stirred solution of 4,4,5,5-tetramethyl-2-(2-methyl-1-benzofuran-5-yl)-1,3,2- dioxaborolane (80 mg, 0.3 mmol) in EtOH (5 mL) was added Pd/C (15 mg) in one portion. The mixture was heated to 70 °C and stirred at 70 °C for 8 h under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOH (5 mL) (3 x 10 mL). The filtrate was concentrated under reduced pressure to give 4,4,5,5-tetramethyl-2-(2- methyl-2,3-dihydro-1-benzofuran-5-yl)-1,3,2-dioxaborolane (70 mg, 87% yield) as a yellow oil. LCMS observed m/z = 261.16 [M+H] + Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-2,3-dihydro-1- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 168
To a solution of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (60 mg, 0.09 mmol), K2CO3 (35 mg, 0.3 mmol) and 4,4,5,5-tetramethyl-2-(2-methyl-2,3-dihydro-1-benzofuran-5-yl)-1,3,2-dioxaborolane (27 mg, 0.1 mmol) in dioxane (2 mL) and H2O (0.4 mL) were added Pd(dppf)Cl2 (6 mg, 0.009 mmol) at 25 °C under a nitrogen atmosphere. The mixture was heated to 90 °C and stirred at 90 °C for 4 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm to give crude product (35 mg). The crude product was purified by reversed-phase flash chromatography with the following conditions: Column: X Bridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10 mmol/L
NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 50%B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 6.58 to give N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-(2-methyl-2,3-dihydro-1-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (9.2 mg, 98.3% purity) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.97 (s, 1H), 9.23 (s, 1H), 8.64 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.13 (s, 1H), 8.08 (dd, J = 8.4, 1.9 Hz, 1H), 7.61 (d, J = 2.0 Hz, 1H), 7.57 – 7.52 (m, 1H), 6.83 (d, J = 8.4 Hz, 1H), 5.70 (s, 1H), 5.18 (s, 2H), 5.04 (q, J = 7.4 Hz, 1H), 4.83 (d, J = 13.0 Hz, 1H), 3.86 (t, J = 11.6 Hz, 2H), 3.53 (s, 1H), 3.40 (dd, J = 15.6, 8.8 Hz, 1H), 3.24 (d, J = 7.9 Hz, 2H), 3.11 (s, 1H), 2.95 – 2.75 (m, 3H), 2.63 (s, 3H), 1.53 (d, J = 6.2 Hz, 3H), 1.38 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 752.35 [M+H] +. EXAMPLE 169 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][6-ethyl-2-(6-fluoro-2,3- dihydro-1-benzofuran-5-yl)-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] Compound 169
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl][6-ethyl-2-(6-fluoro-2,3-dihydro-1-benzofuran-5- yl)-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.36 (s, 1H), 8.46 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.90 (d, J = 2.1 Hz, 1H), 7.81 (d, J = 7.5 Hz, 1H), 7.68 – 7.61 (m, 1H), 6.77 (d, J = 11.3 Hz, 1H), 5.31 (s, 2H), 4.60 (t, J = 8.7 Hz, 2H), 4.46 (d, J = 12.4 Hz, 1H), 3.49 – 3.40 (m, 2H), 3.21 – 2.12 (m, 4H), 2.96 - 2..88 (m, 3H), 2.76 (d, J = 11.1 Hz, 1H), 2.36 (s, 3H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 756.3 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 170
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(3-methyl-2,3-dihydro-1-benzofuran-5- yl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-2-(3-methyl-2,3-dihydro-1-benzofuran-5-yl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.40 (s, 1H), 8.42 (s, 1H), 7.98 (s, 1H), 7.89 (d, J = 14.5 Hz, 2H), 7.82 (d, J = 8.4 Hz, 1H), 7.64 (d, J = 8.7 Hz, 1H), 6.81 (d, J = 8.3 Hz, 1H), 5.31 (s, 2H), 4.68 (t, J = 8.9 Hz, 1H), 4.46 (d, J = 12.4 Hz, 1H), 4.05 (t, J = 8.1 Hz, 1H), 3.56 – 3.40 (m, 3H), 3.28 - 3.15 (m, 2H), 2.97 - 2.89 (m, 3H), 2.76 (d, J = 11.2 Hz, 1H), 2.59 (d, J = 11.2 Hz, 1H), 2.35 (s, 3H), 1.24 (d, J = 6.8 Hz, 3H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 752.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 171 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(7-methyl-2,3-dihydrobenzofuran-5-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
Step 1: Preparation of 5-bromo-7-methyl-2,3-dihydrobenzofuran.
Compound 171.1
To a solution of 4-bromo-2-iodo-1-methoxybenzene (2.0 g, 6.4 mmol) in NMP (20 mL) were added TBAB (10.3 g, 32.0 mmol), Pd(AcO)2 (0.14 g, 0.6 mmol) and KOAc (2.5 g, 25.6 mmol) at -78 ℃ under nitrogen atmosphere. Dimethyl carbonate (6.9 g, 77.0 mmol) was added dropwise, keeping the temperature below -65 ℃. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 12 hours. The resulting mixture was cooled down to room temperature and quenched with water (10 mL), and extracted with EA (2 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10/1) to afford 5-bromo-7-methyl-2,3- dihydrobenzofuran (500 mg, 36% yield) as a yellow oil. LCMS observed m/z = No signal. Step 2: Preparation of (7-methyl-2,3-dihydrobenzofuran-5-yl)boronic acid. Compound 171.2
To a mixture of 5-bromo-7-methyl-2,3-dihydrobenzofuran (200 mg, 1.0 mmol) in THF (5 mL) was added n-BuLi (0.2 mL, 2.1 mmol, 1.6 M in hexane) dropwise at -78°C under nitrogen atmosphere. The resulting mixture was stirred for 1h at -78°C under nitrogen atmosphere. To the above mixture was added triisopropyl borate (0.6 mL) in portions at -78 °C. The reaction mixture was then allowed to warm to room temperature and sit at room temperature for 2 h. After completed, the residue was dissolved in EA/water. The mixture was acidified to pH 2 with aq. HCl and extracted with EA (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with n-hexane/EA (5/1) to afford (7-methyl-2,3- dihydrobenzofuran-5-yl)boronic acid (120 mg, 71% yield) as a white solid. LCMS observed m/z = 177.65 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(7-methyl-2,3-dihydrobenzofuran-5-yl)-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 171.3
To a solution of 2-(6-(4-(5-(benzyloxy)pyrimidine-4-carbonyl)piperazin-1-yl)-2- bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (50 mg, 0.1 mmol, Intermediate) in Dioxane/H2O (1 mL/0.3 mL) were added (7-methyl-2,3-dihydrobenzofuran-5-yl)boronic acid (13.5 mg, 0.1 mmol), K2CO3 (22.0 mg, 0.2 mmol) and Pd(dppf)Cl2 (4.7 mg, 0.01 mmol) at 25 ℃ under N2 atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours. The resulting mixture was cooled down to room temperature and quenched with water (5 mL), and extracted with EA (2 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (5/1) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-5- ethyl-2-(7-methyl-2,3-dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (50 mg, 93% yield) as a brown oil. LCMS observed m/z = 840.45 [M+H]+. Step 4: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(7-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 171
A solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-5-ethyl-2-(7-methyl-2,3-dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (45 mg, 0.1 mmol) in TFA (2 mL) was heated to 80 ℃ and stirred at 80 ℃ for 0.5 hours. The resulting mixture was cooled down to room temperature and quenched with water (5 mL), and extracted with DCM (2 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in Water (10 mmol/L NH4HCO3), 35% to 50% gradient in 10 min; detector, UV 254 nm to afford N-(2-chloro-4-(trifluoromethyl)phenyl)- 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(7-methyl- 2,3-dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (11.1 mg, 26% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.03 (s, 1H), 9.26 (s, 1H), 8.64 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.00 (s, 1H), 7.95 (s, 1H), 7.61 (d, J = 2.1 Hz, 1H), 7.55 (d, J = 8.3 Hz, 1H), 5.73 – 5.62 (m, 1H), 5.18 (s, 2H), 4.89 – 4.78 (m, 1H), 4.67 (t, J = 8.7 Hz, 2H), 3.95 – 3.80 (m, 2H), 3.54 (s, 1H), 3.36 – 3.18 (m, 4H), 3.19 – 3.08 (m, 1H), 2.93 – 2.77 (m, 2H), 2.64 (s, 3H), 2.28 (s, 3H), 1.38 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 752.25 [M+H]+. EXAMPLE 172 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-2-(1,1,3,3-tetramethyl-1,3-dihydro- 5-isobenzofuranyl)-1,3,3a,7-tetraaza-7-indenyl)acetamide Compound 172
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-2-(1,1,3,3-tetramethyl-1,3-dihydro-5- isobenzofuranyl)-1,3,3a,7-tetraaza-7-indenyl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.40 (s, 1H), 8.45 (s, 1H), 7.98 (d, J = 8.3 Hz, 2H), 7.91 (s, 1H), 7.83 (s, 1H), 7.65 (d, J = 8.7 Hz, 1H), 7.31 (d, J = 7.9 Hz, 1H), 5.33 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.50 - 3.41 (m, 3H), 3.21 – 3.10 (m, 1H), 2.99 - 2.89 (m, 3H), 2.78 (d, J = 11.2 Hz, 1H), 2.60 (d, J = 10.2 Hz, 1H), 2.36 (s, 3H), 1.40 (d, J = 5.8 Hz, 12H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 794.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 173 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(7-fluoro-2,3- dihydrobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 173
Step 1: Preparation of 7-fluoro-2,3-dihydro-1-benzofuran. Compound 173.1
To a solution of 7-fluoro-1-benzofuran (500 mg, 3.6 mmol) in MeOH (7.5 mL) was added Pd/C (10%, 100 mg) in a 50 mL round-bottom flask. The mixture was hydrogenated at room temperature for 3 h under hydrogen atmosphere using a hydrogen balloon, filtered through a Celite pad and concentrated under reduce pressure to afford 7-fluoro-2,3-dihydro- 1-benzofuran (480 mg, 91% yield) as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ 7.11 – 6.97 (m, 2H), 6.87 – 6.77 (m, 1H), 4.63 (t, J = 8.8 Hz, 2H), 3.25 (t, J = 8.7 Hz, 2H). Step 2: Preparation of 7-fluoro-5-nitro-2,3-dihydro-1-benzofuran. Compound 173.2
A stirred solution of 7-fluoro-2,3-dihydro-1-benzofuran (300 mg, 2.2 mmol) and added nitric acid (3 mL) was stirred for 2 h at -10 ℃. After completion of reaction, the reaction was quenched by the addition of water (20 mL) at 0 ℃ and was extracted with EA (3 x 20 mL). The combined organic layers were washed with water (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 7-fluoro-5-nitro-2,3-dihydro-1-benzofuran (260 mg, 63% yield) as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ 8.11 – 8.04 (m, 2H), 4.85 (t, J = 8.9 Hz, 2H), 3.41 – 3.34 (m, 2H). Step 3: Preparation of 7-fluoro-2,3-dihydro-1-benzofuran-5-amine. Compound 173.3
To a solution of 7-fluoro-5-nitro-2,3-dihydro-1-benzofuran (200 mg, 1.1 mmol) in EA (6 mL) was added Pd/C (10%, 40 mg) in a 25 mL round-bottom flask. The mixture was hydrogenated at room temperature for overnight under hydrogen atmosphere using a hydrogen balloon, filtered through a Celite pad and concentrated under reduce pressure to afford 7-fluoro-2,3-dihydro-1-benzofuran-5-amine (180 mg, 97% yield) as a light brown solid. LCMS observed m/z = 154.15 [M+H]+. Step 4: Preparation of 5-bromo-7-fluoro-2,3-dihydro-1-benzofuran. Compound 173.4
To a stirred solution of 7-fluoro-2,3-dihydro-1-benzofuran-5-amine (150 mg, 0.98 mmol) and TBAB (600 mg, 1.86 mmol) in ACN (5 mL) were added PTSA (202 mg, 1.18 mmol), CuBr2 (22 mg, 0.098 mmol) and (3-methylbutyl) nitrite (138 mg, 1.18 mmol) in portions at room temperature, after that, the mixture was stirred for 2 h at room temperature. After completion of reaction, the mixture was concentrated to dryness under reduce pressure, then the residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 5-bromo-7-fluoro-2,3-dihydro-1-benzofuran (195 mg, 83% yield) as a light yellow solid. LCMS observed m/z = 217.22 [M+H]+. Step 5: Preparation of 2-(7-fluoro-2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 173.5
To a stirred solution of 5-bromo-7-fluoro-2,3-dihydro-1-benzofuran (80 mg, 0.37 mmol) and AcOK (109 mg, 1.1 mmol) in dioxane (5 mL) were added 4,4,5,5-tetramethyl- 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (140 mg, 0.55 mmol) and Pd(dppf)Cl2 (27 mg, 0.037 mmol) in portions at room temperature under nitrogen atmosphere, after that, the resulting mixture was heated to 100 ℃ and stirred for overnight at 100 ℃ under nitrogen atmosphere. After completion of reaction, the resulting mixture was filtered, the filter cake was washed with dioxane (5 mL). The filtrate was concentrated to dryness under reduced pressure. This resulted in 2-(7-fluoro-2,3-dihydro-1-benzofuran- 5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (90 mg, crude) as a brown oil. LCMS observed m/z = 265.25 [M+H]+. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(7-fluoro-2,3-dihydro-1-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 173.6
To a stirred solution of 2-(7-fluoro-2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (90 mg, 0.34 mmol) and 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (403 mg, 0.5 mmol) in dioxane (5 mL) and H2O (1 mL) were added K2CO3 (141 mg, 1.02 mmol) and Pd(dppf)Cl2 (25 mg, 0.034 mmol) in portions at room temperature under nitrogen atmosphere, after that, the resulting mixture was heated to 80 ℃ and stirred for 3 h at 80 ℃ under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature and filtered, the filter cake was washed with EA (5 mL). The filtrate was concentrated to dryness under reduced pressure. This resulted in 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(7-fluoro-2,3- dihydro-1-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (150 mg, crude) as a yellow solid. LCMS observed m/z = 846.25 [M+H]+. Step 7: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(7- fluoro-2,3-dihydrobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(7-fluoro-2,3-dihydro-1-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.06 mmol) in trifluoroacetaldehyde (0.5 mL) was stirred for 2 h at 80 ℃. After completion of reaction, the resulting mixture was concentrated to dryness under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 19 mm * 250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 60% B to 68% B in 8 min; Wave Length: 254 nm/220 nm; RT1(min): 7.58) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(7-fluoro- 2,3-dihydrobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (11.5 mg, 25% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.45 (s, 1H), 8.54 (s, 1H), 8.06 (d, J = 8.5 Hz, 1H), 7.98 (s, 1H), 7.83 (s, 1H), 7.76 – 7.65 (m, 2H), 5.38 (s, 2H), 4.73 (t, J = 8.7 Hz, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.64 – 3.46 (m, 4H), 3.01 (d, J = 10.2 Hz, 3H), 2.84 (d, J = 11.3 Hz, 1H), 2.66 (d, J = 11.0 Hz, 1H), 2.44 (s, 3H), 1.36 – 1.11 (m, 6H). LCMS observed m/z = 756.20 [M+H]+. EXAMPLE 174 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(6-methyl-2,3-dihydrobenzofuran-5-yl)-7- oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Step 1: Preparation of 1-bromo-2-(2-bromoethoxy)-4-methylbenzene. Compound 174.1
To a stirred mixture of dibromoethane (15.0 g, 80.1 mmol) in ACN (150 mL) were added K2CO3 (11.0 g, 80.1 mmol) and 2-bromo-5-methylphenol (5 g, 26.7 mmol) in portions at room temperature under nitrogen atmosphere. The mixture was heated to 80 °C and stirred at 80 °C for 10 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was filtered, the filter cake was washed with ACN (3x10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 40% to 80% gradient in 15 min; detector, UV 254 nm to give 1-bromo-2-(2-bromoethoxy)-4-methylbenzene (4.0 g, 50% yield) as an off-white oil. 1H NMR (400 MHz, Chloroform-d) δ 7.43 (d, J = 7.9 Hz, 1H), 6.78 – 6.70 (m, 2H), 4.34 (t, J = 6.6 Hz, 2H), 3.70 (t, J = 6.6 Hz, 2H), 2.33 (s, 3H). Step 2: Preparation of 6-methyl-2,3-dihydrobenzofuran. Compound 174.2
To a stirred mixture of 1-bromo-2-(2-bromoethoxy)-4-methylbenzene (3.4 g, 11.5 mmol) in THF (25 mL) was added n-BuLi (5.6 mL, 2M in THF, 13.8 mmol) dropwise at - 78 °C under nitrogen atmosphere. The resulting mixture was stirred at -78 °C for 1.5 h under nitrogen atmosphere. The reaction was quenched with aq. NH4Cl (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10/1) to afford 6-methyl-2,3-dihydrobenzofuran (810 mg, 52% yield) as a colorless oil.1H NMR (400 MHz, Chloroform-d) δ 7.10 (d, J = 7.4 Hz, 1H), 6.73 – 6.64 (m, 2H), 4.58 (t, J = 8.7 Hz, 2H), 3.19 (t, J = 8.6 Hz, 2H), 2.34 (s, 3H). Step 3: Preparation of 5-bromo-6-methyl-2,3-dihydrobenzofuran. Compound 174.3
To a stirred mixture of 6-methyl-2,3-dihydro-1-benzofuran (450 mg, 3.3 mmol) in ACN (15 mL) was added NBS (596 mg, 3.3 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue
was purified by silica gel column chromatography, eluted with PE / EA (30/1) to afford 5- bromo-6-methyl-2,3-dihydro-1-benzofuran (600 mg, 83% yield) as a white solid. LCMS observed m/z = No signal. Step 4: Preparation of 4,4,5,5-tetramethyl-2-(6-methyl-2,3-dihydrobenzofuran-5- yl)-1,3,2-dioxaborolane. Compound 174.4
To a stirred mixture of 5-bromo-6-methyl-2,3-dihydro-1-benzofuran (200 mg, 0.9 mmol), KOAc (276 mg, 2.8 mmol) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2- dioxaborolane) (286 mg, 1.1 mmol) in dioxane (10 mL) was added Pd(dppf)Cl2 (137 mg, 0.2 mmol) in one portion at room temperature under nitrogen atmosphere. The mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10/1) to afford 4,4,5,5-tetramethyl-2-(6- methyl-2,3-dihydrobenzofuran-5-yl)-1,3,2-dioxaborolane (140 mg, 83% yield) as a yellow solid. LCMS observed m/z = 261.16 [M+H]+ Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(6-methyl-2,3-dihydrobenzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl) phenyl)acetamide, Intermediate H (100 mg, 0.1 mmol), K2CO3 (54 mg, 0.4 mmol) and 4,4,5,5-tetramethyl-2-(6-methyl-2,3-dihydrobenzofuran-5-
yl)-1,3,2-dioxaborolane (28 mg, 0.2 mmol) in dioxane (15 mL) and H2O (1.5 mL) was added Pd(dppf)Cl2 (18 mg, 0.03 mmol) in one portion at room temperature under nitrogen atmosphere. The mixture was heated to 100 °C and stirred at 100 °C for 3 h under nitrogen atmosphere. The reaction was quenched with water (15 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 20% to 60% gradient in 10 min; detector, UV 254 nm to give 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-2-(6- methyl-2,3-dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (2-chloro-4-(trifluoromethyl) phenyl)acetamide (60 mg, 55.3% yield) as a yellow solid. LCMS observed m/z = 842.28 [M+H]+ Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(6-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Into a 4 mL sealed tube were added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(6-methyl-2,3-dihydrobenzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (60 mg, 0.07 mmol) and TFA (2 mL) at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 20 min. The resulting mixture was concentrated under reduced pressure. The crude product (60 mg) was purified by Prep-HPLC with the following conditions (Column: YMC-Actus Triart C185 m, 30 mm*150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 45%B in 8 min; Wave Length: 254 nm/220
nm; RT1(min): 7.97) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(6-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)aceta mide (18.5 mg, 31% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.24 (s, 1H), 9.21 (s, 1H), 8.69 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 7.98 (s, 1H), 7.61 (d, J = 2.0 Hz, 1H), 7.59 – 7.52 (m, 1H), 6.74 (s, 1H), 5.63 (d, J = 12.9 Hz, 1H), 5.18 (s, 2H), 4.83 (d, J = 12.6 Hz, 1H), 4.65 (t, J = 8.7 Hz, 2H), 3.88 (t, J = 11.6 Hz, 2H), 3.56 (s, 1H), 3.25 (t, J = 8.5 Hz, 4H), 3.14 (s, 1H), 2.95 – 2.80 (m, 2H), 2.71 (s, 3H), 2.69 (s, 3H), 1.39 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 752.40 [M+H] + EXAMPLE 175 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dimethyl-2-oxo-1,3- dihydro-1,3-benzimidazol-5-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide Compound 175
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dimethyl-2-oxo-1,3-dihydro-1,3- benzimidazol-5-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.41 (s, 1H), 8.43 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.91 (s, 1H), 7.82 (d, J = 8.2 Hz, 1H), 7.71 (s, 1H), 7.64 (d, J = 8.7 Hz, 1H), 7.21 (d, J = 8.2 Hz, 1H), 5.33 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.51 – 3.41 (m, 3H), 3.33 (s, 6H), 3.20 - 3.16 (m, 1H), 2.98 - 2.88 (m, 3H), 2.77 (d, J = 11.2 Hz, 1H), 2.60 (d, J = 10.0 Hz, 1H), 2.36 (s, 3H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 780.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 176
Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3a,7,7a'-hexaaza-2,5'- biindenyl-7-yl)acetamide Compound 176
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3a,7,7a'-hexaaza-2,5'-biindenyl-7- yl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.39 (s, 1H), 8.73 (d, J = 7.3 Hz, 1H), 8.42 (d, J = 46.5 Hz, 2H), 8.01 (d, J = 8.1 Hz, 2H), 7.92 (s, 1H), 7.65 (d, J = 8.7 Hz, 1H), 7.44 (d, J = 7.3 Hz, 1H), 6.76 (s, 1H), 5.35 (s, 2H), 4.47 (d, J = 12.5 Hz, 1H), 3.51 - 3.42 (m, 3H), 3.21 - 3.17 (m, 1H), 3.00 – 2.91 (m, 3H), 2.78 (d, J = 11.3 Hz, 1H), 2.60 (d, J = 10.6 Hz, 1H), 2.37 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 736.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 177 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-2H-indazol-6-yl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-2H-indazol-6-yl)-4-oxo-1,3,3a,7-
tetraaza-7-indenyl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.41 (s, 1H), 8.34 (s, 1H), 8.27 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.91 (s, 1H), 7.74 (q, J = 8.8 Hz, 2H), 7.64 (d, J = 8.7 Hz, 1H), 5.35 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 4.13 (s, 3H), 3.50 - 3.40 (m, 3H), 3.20 - 3.15 (m, 1H), 3.00 - 3.92 (m, 3H), 2.77 (d, J = 11.2 Hz, 1H), 2.59 (d, J = 10.2 Hz, 1H), 2.35 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 178 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(3,3-difluoro-5-indanyl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide.
The title compound was prepared using a similar procedure as compound 164, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-isoindolinone with 2-(3,3-difluoro-5-indanyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.45 (s, 1H), 10.32 (s, 1H), 8.55 (s, 1H), 8.26 (d, J = 8.0 Hz, 1H), 8.21 (s, 1H), 8.04 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.71 (d, J = 8.6 Hz, 1H), 7.58 (d, J = 8.1 Hz, 1H), 5.41 (s, 2H), 4.53 (d, J = 12.7 Hz, 1H), 3.52 (q, J = 11.7 Hz, 3H), 3.28 – 3.21 (m, 1H), 3.11 (s, 2H), 3.07 – 2.94 (m, 3H), 2.84 (d, J = 11.4 Hz, 1H), 2.73 – 2.58 (m, 3H), 2.43 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 772.2 [M+H]+. EXAMPLE 179 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,2- difluorobenzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 179
The title compound was prepared using similar procedure as compound 164 using intermediate AC to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,2- difluorobenzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 8.55 (s, 1H), 8.10 – 7.94 (m, 4H), 7.71 (d, J = 8.7 Hz, 1H), 7.57 (d, J = 8.4 Hz, 1H), 5.39 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 3.59 – 3.44 (m, 3H), 3.26 (t, J = 12.4 Hz, 2H), 3.00 (dd, J = 14.9, 9.6 Hz, 3H), 2.83 (d, J = 11.2 Hz, 1H), 2.67 (s, 1H), 2.43 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 776.2 [M+H]+. EXAMPLE 180 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4-fluoro-2,3- dihydro-1-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 180
Step 1: Preparation of 2-bromo-1-(2-bromoethoxy)-3-fluorobenzene. Compound 180.1
To a stirred solution of 2-bromo-3-fluorophenol (5.0 g, 26.2 mmol) and K2CO3 (10.9 g, 78.5 mmol) in ACN (50 mL) was added dibromoethane (9.8 g, 52.4 mmol) in portions at 25 °C. Then the resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h. After completion of reaction, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (3.2 g, 41% yield) of 2-bromo-1-(2-bromoethoxy)-3- fluorobenzene as a colorless oil. LCMS observed m/z = 298.88 [M+H]+ Step 2: Preparation of 4-fluoro-2,3-dihydro-1-benzofuran. Compound 180.2
To a stirred solution of 2-bromo-1-(2-bromoethoxy)-3-fluorobenzene (2.0 g, 6.7 mmol) in THF (20 mL) was added n-BuLi (3.2 mL, 8.0 mmol, 2.5 M) dropwise at -78 °C under nitrogen atmosphere. Then the resulting mixture was stirred at -78 °C for 2 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched with sat. NH4Cl (aq.) at 0 °C. The aqueous layer extracted with EA (3 x 15 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (660 mg, 71% yield) of 4-fluoro-2,3-dihydro-1-benzofuran as a yellow oil. Step 3: Preparation of 5-bromo-4-fluoro-2,3-dihydro-1-benzofuran. Compound 180.3
To a stirred solution of 4-fluoro-2,3-dihydro-1-benzofuran (250 mg, 1.8 mmol) in ACN (3 mL) was added NBS (354 mg, 2.0 mmol) in portion at 25 °C. Then the resulting mixture was stirred at 25 °C for 2 h. After completion of reaction, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column
chromatography (eluent: with 5% EtOAc in petroleum ether) to afford (50 mg, 12% yield) of 5-bromo-4-fluoro-2,3-dihydro-1-benzofuran as a white oil. Step 4: Preparation of 2-(4-fluoro-2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 180.4
To a stirred solution of 5-bromo-4-fluoro-2,3-dihydro-1-benzofuran (160 mg, 0.7 mmol) and KOAc (217 mg, 2.2 mmol) in dioxane (16 mL) were added Pd(dppf)Cl2 (54 mg, 0.07 mmol) and B2Pin2(281 mg, 1.1 mmol) in portions at 25 °C under nitrogen atmosphere. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 18 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to 25 °C. The resulting solution was used in the next step directly without further purification. LCMS observed m/z = 265.13 [M+H]+ Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-2,3-dihydro-1-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
To a stirred solution of 2-(4-fluoro-2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (crude) in dioxane (5 mL) were added 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl] acetamide, Intermediate H (200 mg, 0.3 mmol), K2CO3 (105 mg, 0.8 mmol), Pd(dppf)Cl2 (19 mg, 0.03 mmol) and H2O (1 mL) in portions at 25 °C under nitrogen atmosphere. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 18 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to 25°C and
concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (150 mg, 70% yield) of 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro- 2,3-dihydro-1-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 846.25 [M+H]+ Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4- fluoro-2,3-dihydro-1-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 180
Into a 8 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-2,3-dihydro-1-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (60 mg, 0.07 mmol) and TFA (2 mL) at 25 °C. Then the resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. After completion of reaction, the mixture was allowed to cool down to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep-HPLC (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 28-51% MeCN in H2O) to afford (6.9 mg, 11% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[5-ethyl-2-(4-fluoro-2,3-dihydro-1-benzofuran-5-yl)-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)pipera zin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 2H), 8.56 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.84 (t, J = 8.0 Hz, 1H), 7.72 (dd, J = 8.7, 2.2 Hz, 1H), 6.78 (d, J = 8.4 Hz, 1H), 5.38 (s, 2H), 4.71 (t, J = 8.7 Hz, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.53 (q, J = 11.3, 10.1 Hz, 3H), 3.31 – 3.20 (m, 3H), 3.02
(d, J = 9.6 Hz, 3H), 2.84 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 10.5 Hz, 1H), 2.44 (s, 3H), 1.26 – 1.19 (m, 3H). LCMS observed m/z = 756.10 [M+H]+. EXAMPLE 181 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2R)-2-methoxy-2,3-dihydro-1H-inden- 5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 181
Step 1: Preparation of 5-bromo-2-methoxy-2,3-dihydro-1H-indene. Compound 181.1
To a stirred mixture of 5-bromo-2,3-dihydro-1H-inden-2-ol (1.0 g, 4.6 mmol) and KOH (0.5 g, 9.3 mmol) in DMSO (10 mL) was added MeI (0.3 mL, 4.6 mmol) at room temperature. The resulting mixture was stirred for 2 h at room temperature. The reaction was quenched with water (100 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (800 mg, 75% yield) of 5-bromo-2-methoxy-2,3-dihydro-1H-indene as a colorless oil. Step 2: Preparation of (2R)-5-bromo-2-methoxy-2,3-dihydro-1H-indene. As used herein in connection with chemical structures, “assumed” refers to the assumed stereochemistry of the structure. Such assumed stereochemistry is not intended to be limiting as to the actual stereochemistry of the compositions prepared according to the methods described herein. Compound 181.2
The racemic (500 mg) resulting from the last step was purified by Prep-HPLC with the following conditions (Column: CHIRALPAK IH 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: IPA: HEX=1: 2(0.1% 2mMNH3); Flow rate: 90 mL/min; Gradient: isocratic 15% B; Column Temperature (℃): 35; Back Pressure(bar): 100; Wave Length: 220 nm; RT1(min): 3.03; RT2(min): 4.38; Sample Solvent: IPA; Injection Volume: 1 mL) to afford (170 mg, 34% yield) of (2R)-5-bromo-2-methoxy-2,3-dihydro-1H-indene as a colorless oil . Step 3: Preparation of 2-[(2R)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 181.3
To a stirred solution of (2R)-5-bromo-2-methoxy-2,3-dihydro-1H-indene (100 mg, 0.4 mmol) and Pd(dppf)Cl2CH2Cl2 (32 mg, 0.04 mmol) in dioxane (1 mL) were added AcOK (129 mg, 1.3 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3,2-dioxaborolane (167 mg, 0.7 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 100 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 8% EtOAc in petroleum ether) to afford (78 mg, 64% yield) of 2-[(2R)-2-methoxy-2,3-dihydro-1H-inden-5-yl]- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane as a white solid. LCMS observed m/z = 275.25 [M+H]+. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-[(2R)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 181.4
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- [(2R)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (38 mg, 0.1 mmol) in dioxane (1 mL) were added Pd(dppf)Cl2CH2Cl2 (9 mg, 0.01 mmol), H2O (0.2 mL) and K2CO3 (43 mg, 0.3 mmol) at room temperature. The resulting mixture was stirred for 3 h at 95 °C under nitrogen atmosphere. The reaction was quenched with water (50 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 25% EtOAc in petroleum ether) to afford (78 mg, 71% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-[(2R)-2-methoxy-2,3-dihydro-1H- inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 856.30 [M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2R)-2-methoxy-2,3-dihydro- 1H-inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-[(2R)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (70 mg, 0.08 mmol) in TFA (1.0 mL) was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC Triart C185 m, 30 mm * 150 mm; Mobile Phase A: water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 55% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 6.27) to afford (6.5 mg, 10% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2R)-2-methoxy-2,3- dihydro-1H-inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 8.53 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 8.01 – 7.94 (m, 2H), 7.92 (d, J = 8.0 Hz, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.36 (d, J = 7.9 Hz, 1H), 5.39 (s, 2H), 4.54 (m, 1H), 4.24 (m, 1H), 3.53 (m, 3H), 3.27 (s, 4H), 3.16 (m, 2H), 3.07 – 2.89 (m, 5H), 2.84 (m, 1H), 2.67 (m, 1H), 2.44 (s, 3H), 1.22 (t, 3H). LCMS observed m/z = 766.35 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 182 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2S)-2-methoxy-2,3-dihydro-1H-inden-5- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 182
Step 1: Preparation of (2S)-5-bromo-2-methoxy-2,3-dihydro-1H-indene. Compound 182.1
The racemic (500 mg) resulting from the last step was purified by Prep-HPLC with the following conditions (Column: CHIRALPAK IH 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: IPA: HEX=1: 2(0.1% 2mMNH3); Flow rate: 90 mL/min; Gradient: isocratic 15% B; Column Temperature(℃): 35; Back Pressure(bar): 100; Wave Length: 220 nm; RT1(min): 3.03; RT2(min): 4. 38; Sample Solvent: IPA; Injection Volume: 1 mL) to afford (180 mg, 36% yield) of (2S)-5-bromo-2-methoxy-2,3-dihydro-1H-indene as a colorless oil. Step 2: Preparation of 2-[(2S)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 182.2
To a stirred solution of (2S)-5-bromo-2-methoxy-2,3-dihydro-1H-indene (100 mg, 0.4 mmol) and Pd(dppf)Cl2CH2Cl2 (32 mg, 0.04 mmol) in dioxane (1 mL) were added AcOK (129 mg, 1.3 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3,2-dioxaborolane (167 mg, 0.6 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 100 °C under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with DCM (3 x 10 mL). The filtrate was concentrated under reduced pressure. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 8% EtOAc in petroleum ether) to afford (110 mg, 91% yield) of 2-[(2S)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-4,4,5,5-tetramethyl-1,3,2-dioxaborolane as a yellow solid. LCMS observed m/z = 275.25 [M+H]+. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-[(2S)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 182.3
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- [(2S)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (52 mg, 0.2 mmol) in dioxane (1 mL) were added Pd(dppf)Cl2CH2Cl2 (9 mg, 0.01 mmol), H2O (0.2 mL) and K2CO3 (43 mg, 0.3 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 95 °C under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3 x 10 mL). The filtrate was concentrated under reduced pressure. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (78 mg, 71% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-[(2S)-2-methoxy-2,3-dihydro-1H- inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 856.30 [M+H]+. Step 4: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2S)-2-methoxy-2,3-dihydro- 1H-inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-[(2S)-2-methoxy-2,3-dihydro-1H-inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in TFA (10 mL) was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 5 m, 30 mm * 150 mm; Mobile Phase A: water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 55% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 6.27) to afford (14.9 mg, 16% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2S)-2-methoxy-2,3- dihydro-1H-inden-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.31 (s, 1H), 8.56 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 8.00 – 7.94 (m, 2H), 7.92 (d, J = 7.8 Hz, 1H), 7.72 (dd, J = 8.7, 2.2 Hz, 1H), 7.36 (d, J = 7.9 Hz, 1H), 5.40 (s, 2H), 4.54 (m, 1H), 4.24 (m, 1H), 3.53 (m, 3H), 3.27 (s, 4H), 3.16 (m, 2H), 3.08 – 2.89 (m, 5H), 2.85 (m, 1H), 2.67 (m, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 766.35 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 183 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro-4H- pyrrolo[1,2-b]pyrazol-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Step 1: Preparation of 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-yl trifluoromethanesulfonate. Compound 183.1
To a stirred mixture of 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-ol (200 mg, 1.6 mmol) and TEA (326 mg, 3.2 mmol) in DCM (5 mL) was added trifluoromethanesulfonic anhydride (682 mg, 2.4 mmol) dropwise at 0 °C. The resulting mixture was stirred at 0 °C for 1 h under nitrogen atmosphere. The reaction was quenched with water (20 mL) at room temperature and extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 20% gradient in 10 min; detector, UV 254 nm) to afford 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-yl trifluoromethanesulfonate (290 mg, 70% yield) as a yellow oil. LCMS observed m/z = 257.01 [M+H] +. Step 2: Preparation of 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,6-dihydro- 4H-pyrrolo[1,2-b]pyrazole. Compound 183.2
To a stirred mixture of 5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-yl trifluoromethanesulfonate (150 mg, 0.6 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (156 mg, 0.6 mmol) in dioxane (3 mL) were added Pd(dppf)Cl2 (21 mg, 0.03 mmol), dppf (16 mg, 0.03 mmol) and KOAc (172 mg, 1.8 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 100 °C for 12 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. This resulted in 2-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazole (60 mg, crude) as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 235.15 [M+H] +. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl) phenyl)acetamide.
Compound 183.3
To a stirred mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl) acetamide, Intermediate H (170 mg, 0.2 mmol, Intermediate) and 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,6-dihydro-4H- pyrrolo[1,2-b]pyrazole (60 mg, 0.2 mmol) in dioxane (2 mL) and H2O (0.4 mL) were added Pd(dppf)Cl2 (16 mg, 0.02 mmol) and K2CO3 (89 mg, 0.6 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 85 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 50% to 60% gradient in 10 min; detector, UV 254 nm) to afford 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-2- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl) acetamide (35 mg, 17% yield) as a brown solid. LCMS observed m/z = 816.27 [M+H] +. Step 4: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro- 4H-pyrrolo[1,2-b]pyrazol-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 183
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-{4H,5H,6H-pyrrolo[1,2-b]pyrazol-2-yl}-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (35 mg, 0.04 mmol) in TFA (2 mL) was stirred at 80 °C for 1 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 30% B to 40% B in 8 min; Wave Length: 254 nm/220 nm; RT 1(min): 7.75) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,6-dihydro-4H-pyrrolo[1,2- b]pyrazol-2-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (14.5 mg, 45% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.39 (s, 1H), 8.52 (s, 1H), 8.10 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.73 – 7.71 (m, 1H), 6.52 (s, 1H), 5.38 (s, 2H), 4.55 – 4.52 (m, 1H), 4.13 (t, J = 7.2 Hz, 2H), 3.53 – 3.50 (m, 3H), 3.24 (s, 1H), 3.03 – 2.96 (m, 3H), 2.89 (t, J = 7.3 Hz, 2H), 2.84 – 2.82 (m, 1H), 2.69 – 2.62 (m, 1H), 2.59 – 2.57 (m, 2H), 2.43 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 726.15 [M+H] +. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 184 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methyl pyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{4H,5H,6H,7H-pyrazolo[1,5- a]pyridin-2-yl}-[1,2,4]triazolo [1,5-a]pyrimidin-4-yl}acetamide. Compound 184
Step 1: Preparation of ethyl 7-chloro-3-oxoheptanoate. Compound 184.1
To a stirred solution of ethyl acetoacetate (5.0 g, 38.4 mmol) in THF (50 mL) was added LDA (2M in THF) (38.4 mL, 76.8 mmol) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was stirred at -78 °C for 1 h under nitrogen atmosphere. The resulting mixture was warmed to 0 °C. To above mixture was added a solution of 1- chloro-3-iodopropane (7.8 g, 38.4 mmol) in THF dropwise at 0 °C. Then the resulting mixture was heated to 25 °C and stirred at 25 °C for 1 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 0 °C and quenched by the addition of sat. NH4Cl (aq.) (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford ethyl 7-chloro-3-oxoheptanoate (3.4 g, 43% yield) as a light-yellow oil. LCMS observed m/z = 207.07 [M+H] +. Step 2: Preparation of 4H,5H,6H,7H-pyrazolo[1,5-a]pyridin-2-ol. Compound 184.2
A mixture of ethyl 7-chloro-3-oxoheptanoate (2.7 g, 13.1 mmol) and NH2NH2.H2O (1.9 mL, 39.2 mmol) in EtOH (30 mL) was heated to 100 °C and stirred at 100 °C for 4 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2 / MeOH (10/1) to afford 4H,5H,6H,7H-
pyrazolo[1,5-a]pyridin-2-ol (530 mg, 29% yield) as a yellow solid. LCMS observed m/z = 139.08 [M+H] +. Step 3: Preparation of 4H,5H,6H,7H-pyrazolo[1,5-a]pyridin-2- yltrifluoromethanesulfonate. Compound 184.3
To a stirred mixture of 4H,5H,6H,7H-pyrazolo[1,5-a]pyridin-2-ol (500 mg, 3.6 mmol) and DIEA (935 mg, 7.2 mmol) in DCM (5 mL) was added trifluoromethanesulfonic anhydride (1.5 g, 5.4 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 25 °C for 2 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 0 °C and quenched by the addition of ice water (30 mL) at 0 °C. The resulting mixture was extracted with CH2Cl2 (3 x 50 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/1) to afford 4H,5H,6H,7H- pyrazolo[1,5-a]pyridin-2-yltrifluoromethanesulfonate (700 mg, 72% yield) as a light brown oil. LCMS observed m/z = 271.03 [M+H]+. Step 4: Preparation of 2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 4H,5H,6H,7H-pyrazolo[1,5-a]pyri dine. Compound 184.4
To a solution of 4H,5H,6H,7H-pyrazolo[1,5-a]pyridin-2- yltrifluoromethanesulfonate (100 mg, 0.4 mmol) and bis(pinacolato)diboron (188 mg, 0.7 mmol) in dioxane (3 mL) were added potassium acetate (109 mg, 1.1 mmol) and Pd(dppf)Cl2 (10 mg, 0.01 mmol) at 25 ℃ under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 3 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The crude product was used for next step directly. LCMS observed m/z = 249.17 [M+H]+.
Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{4H,5H,6H,7H- pyrazolo[1,5-a]pyridin-2-yl}-[1,2,4]triazolo [1,5-a]pyrimidin-4-yl}acetamide. Compound 184
To a solution of 2-(6-{4-[5-(benzyloxy)pyrimidine-4-carbonyl]piperazin-1-yl}-2- bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) and 2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-4H,5H,6H,7H-pyrazolo[1,5-a]pyridine (48 mg, 0.2 mmol) in dioxane (4 mL) and H2O (0.5 mL) were added K2CO3 (53 mg, 0.4 mmol) and Pd(dppf)Cl2 (9 mg, 0.01 mmol) at 25 ℃ under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 25 °C. The reaction mixture was poured into water (5 mL) and extracted with EtOAc (5 x 10 mL). The combined organic layers were concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X-select CSH C185 m, 30 mm X 150 mm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 60% B to 77% B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 5.13) to afford N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-{4H,5H,6H,7H-pyrazolo[1,5-a]pyridin-2-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (20.0 mg, 19% yield) as a yellow solid.1H NMR (400 MHz, Chloroform-d) δ 11.95 (s, 1H), 9.29 (s, 1H), 8.64 (s, 1H), 8.40 (d, J = 8.8 Hz, 1H), 7.59 (s, 1H), 7.45 (d, J = 8.8 Hz, 1H), 6.81 (s, 1H), 5.95 – 5.59 (m, 1H), 5.40 (s, 2H), 4.83 (d, J = 12.4 Hz, 1H), 4.28 (s, 2H), 4.01 – 3.76 (m, 2H), 3.67 – 3.43 (m, 1H), 3.33 – 3.03 (m, 3H), 3.03 – 2.74 (m, 4H), 2.63 (s, 3H), 2.06 (s, 1H), 1.94 (s, 3H), 1.43 – 1.25 (m, 3H). LCMS observed m/z = 740.40 [M+H] +. EXAMPLE 185
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-1-oxo-5-isoindolinyl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide.
Step 1: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5- (benzyloxy)-6-methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-6-ethyl-2-(2-methyl-1-oxo- 5-isoindolinyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide.
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6- methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-2-bromo-6-ethyl-4-oxo-4,7-dihydro- 1,3,3a,7-tetraaza-7-indenyl]acetamide (60 mg, 76 µmol, Intermediate H), 2-methyl-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-isoindolinone (20.8 mg, 76 µmol) and K3PO4 (48.4 mg, 228 µmol) in a mixture of dioxane (1.5 mL) and H2O (0.5 mL) was added Pd(dppf)Cl2.CH2Cl2 (5.56 mg, 7.6 µmol). The mixture was stirred at 80 °C under N2 for 16 hours. The mixture was concentrated in vacuo. The residue was dissolved in CH2Cl2 and filtered through a pad of Celite and the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-20% MeOH in CH2Cl2) to
afford (47.2 mg, 73% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5- (benzyloxy)-6-methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-6-ethyl-2-(2-methyl-1-oxo- 5-isoindolinyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide as a yellow film. LCMS observed m/z = 855.4 [M+H]+. Step 2: N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-1-oxo-5-isoindolinyl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 185
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6- methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-6-ethyl-2-(2-methyl-1-oxo-5- isoindolinyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide (47.2 mg, 55.2 µmol) in CH2Cl2 (2.21 mL) was added dropwise a 1 molar solution of boron trichloride in CH2Cl2 (276 µL, 276 µmol). The mixture was stirred at -40 °C under N2 for 1 hour. The mixture was quenched with MeOH (1.0 mL) and warmed to 25 °C. The mixture was concentrated in vacuo and the residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (24.0 mg, 57% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5- {4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-1-oxo-5- isoindolinyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide as an off white solid.1H NMR (400MHz, DMSO-d6) δ 10.13 (s, 1H), 8.23 (s, 1H), 8.00 (s, 1H), 7.90 (d, J = 8.0 Hz, 1H), 7.75 (d, J = 8.6 Hz, 1H), 7.66 (s, 1H), 7.48 (d, J = 7.9 Hz, 1H), 7.39 (d, J = 8.7 Hz, 1H), 5.10 (s, 2H), 4.23 (s, 2H), 4.20 (d, J = 11.5 Hz, 1H), 3.21 (q, J = 11.8 Hz, 3H), 3.01 – 2.86 (m, 1H), 2.78 (s, 3H), 2.70 (h, J = 8.9 Hz, 3H), 2.53 (d, J = 11.1 Hz, 1H), 2.36 (d, J = 10.6 Hz, 1H), 2.12 (s, 3H), 0.90 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 765.3 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 186
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-3-oxo-5-isoindolinyl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 186
The title compound was prepared using a similar procedure as compound 164 using intermediate AC, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1- isoindolinone with 4,4,5,5-tetramethyl-2-(2-methyl-5-isoindolinyl)-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO) δ 10.45 (s, 1H), 10.24 (s, 1H), 8.58 (s, 1H), 8.38 – 8.29 (m, 2H), 8.04 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (dd, J = 13.8, 8.3 Hz, 2H), 5.43 (s, 2H), 4.64 – 4.46 (m, 3H), 3.60 – 3.44 (m, 3H), 3.31 – 3.21 (m, 1H), 3.09 (s, 3H), 3.07 – 2.96 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.67 (d, J = 10.0 Hz, 1H), 2.45 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 765.3 [M+H]+. EXAMPLE 187 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-difluoro-5-indanyl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide. Compound 187
The title compound was prepared using a similar procedure as compound 164 using intermediate AC, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1- isoindolinone with 2-(1,1-difluoro-5-indanyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400MHz, MeOD) δ 8.54 (s, 1H), 8.43 (s, 1H), 8.24 – 8.19 (m, 2H), 8.17 (d, J = 8.6 Hz, 1H), 7.83 (s, 1H), 7.61 (d, J = 8.4 Hz, 2H), 5.47 (s, 2H), 4.72 (d, J = 12.8 Hz, 1H), 3.95 (s, 1H), 3.76 (d, J = 11.6 Hz, 2H), 3.54 – 3.40 (m, 1H), 3.18 – 3.09 (m, 5H), 2.96 (d, J = 11.3 Hz, 1H), 2.81 (d, J = 11.6 Hz, 1H), 2.62 (tt, J = 13.9, 6.7 Hz, 2H), 2.50 (s, 3H), 1.34 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 772.3 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 188 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-2-(3-oxo-5-isoindolinyl)-1,3,3a,7- tetraaza-7-indenyl)acetamide. Compound 188
The title compound was prepared using a similar procedure as compound 164 using intermediate AC, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1- isoindolinone with 6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-isoindolinone. 1H
NMR (400MHz, DMSO-d6) δ 10.45 (s, 1H), 10.24 (s, 1H), 8.71 (s, 1H), 8.58 (s, 1H), 8.37 (s, 1H), 8.33 (d, J = 8.0 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.76 – 7.67 (m, 2H), 5.42 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 4.46 (s, 2H), 3.64 – 3.45 (m, 3H), 3.28 – 3.22 (m, 1H), 3.14 – 2.93 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.67 (d, J = 10.1 Hz, 1H), 2.45 (s, 3H), 1.21 (t, J = 7.3 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. EXAMPLE 189 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(3S)-3-methyl-1,3-dihydro-2-benzofuran- 5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 189
Step 1: Preparation of N'-[(1E)-(4-bromo-2- hydroxyphenyl)methylidene]acetohydrazide. Compound 189.1
A solution of 4-bromo-2-hydroxybenzaldehyde (15.0 g, 74.620 mmol) in EtOH (150 mL) was treated with acetohydrazide (11.1 g, 149.157 mmol) for one hour at 85 °C. The mixture was allowed to cool down to 0 °C. The precipitated solids were collected by filtration and washed with cold EtOH (50 mL). This resulted in N'-[(1E)-(4-bromo-2- hydroxyphenyl)methylidene]acetohydrazide (17.1 g, 89% yield) as a white solid. LCMS observed m/z = 257.00 [M+H]+. Step 2: Preparation of N'-[(1E)-(4-bromo-2- hydroxyphenyl)methylidene]acetohydrazide. Compound 189.2
To a stirred solution of N'-[(1E)-(4-bromo-2- hydroxyphenyl)methylidene]acetohydrazide (2.3 g, 8.946 mmol) in THF (30 mL) was added Pb(OAc)4 (8.1 g, 18.268 mmol) in portions at rt under N2 atmosphere. The resulting mixture was stirred for 4 hours at rt under N2 atmosphere. The reaction was quenched by the addition of H2O (50 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 16% EtOAc in petroleum ether) to afford (1.4 g, 70% yield) of 2-acetyl-4-bromobenzaldehyde as a light-brown solid. LCMS observed m/z = 227.00 [M+H]+. Step 3: Preparation of 1-[5-bromo-2-(hydroxymethyl)phenyl]ethanol. Compound 189.3
To a stirred solution of 2-acetyl-4-bromobenzaldehyde (1.1 g, 4.986 mmol) in MeOH (20 mL) was added NaBH4 (750 mg, 19.944 mmol) in portions at 0 °C under N2 atmosphere. The resulting mixture was stirred for 16 hours at room temperature under N2 atmosphere. The reaction was quenched by the addition of H2O (50 mL) at 0 °C. The resulting mixture was extracted with DCM (3 x 50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (1.0 g, 91% yield) of 1-[5-bromo-2- (hydroxymethyl)phenyl]ethanol as a white solid. LCMS observed m/z = 231.00 [M+H]+. Step 4: Preparation of 6-bromo-1-methyl-1,3-dihydro-2-benzofuran. Compound 189.4
A solution of 1-[5-bromo-2-(hydroxymethyl)phenyl]ethanol (1.0 g, 4.506 mmol) in DMF (20 mL) was treated with TsCl (1.0 g, 5.408 mmol) at 0 °C under nitrogen atmosphere
followed by the addition of NaH (450 mg, 11.258 mmol, 60%) in portions at this temperature. The resulting mixture was stirred for 16 hours at room temperature under N2 atmosphere. The reaction was quenched by the addition of H2O (30 mL) at 0 °C. The resulting mixture was extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (577 mg, 60% yield) of 6-bromo-1-methyl-1,3-dihydro-2-benzofuran as a light-yellow oil. Step 5: Preparation of 4,4,5,5-tetramethyl-2-(3-methyl-1,3-dihydro-2-benzofuran-5- yl)-1,3,2-dioxaborolane. Compound 189.5
A solution of 6-bromo-1-methyl-1,3-dihydro-2-benzofuran (536 mg, 2.516 mmol) in dioxane (10 mL) was treated with 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (957 mg, 3.772 mmol) at room temperature under nitrogen atmosphere followed by the addition of Pd(dppf)Cl2CH2Cl2 (206 mg, 0.253 mmol) at this temperature. To the above mixture was added AcOK (741 mg, 7.550 mmol) in one portion. The resulting mixture was stirred for additional 3 hours at 100 °C. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered, the filter cake was washed with DCM (10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford 4,4,5,5-tetramethyl-2-(3-methyl-1,3-dihydro-2- benzofuran-5-yl)-1,3,2-dioxaborolane (620 mg, 94% yield) as a light-yellow oil. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(3-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 189.6
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (203 mg, 0.257 mmol) and 4,4,5,5-tetramethyl-2-(3-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2-dioxaborolane (100 mg, 0.384 mmol) in dioxane (10 mL) was added a solution of K2CO3 (106 mg, 0.767 mmol) in H2O (2 mL) at room temperature under N2 atmosphere. To the above mixture was added Pd(dppf)Cl2CH2Cl2 (21 mg, 0.026 mmol) in one portion. The resulting mixture was stirred for additional 16 hours at 95 °C. The reaction mixture was purified by reversed phase C18 silica gel column chromatography to afford (50 mg, 23% yield) of 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(3-methyl-1,3- dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a light-brown solid. LCMS observed m/z = 842.27 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(3S)-3-methyl-1,3-dihydro-2- benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 189
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(3-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.059 mmol) in TFA (5 mL) was stirred at 25 °C for 16 hours. The resulting mixture was concentrated under vacuum. The residue was purified by preparatory HPLC (Column: Xselect CSH C185 m, 30 mm * 150 mm; mobile phase: 52-62% MeCN in H2O). The resulting racemic mixture 189.7 was purified by chiral SFC to afford (5.8 mg, 13% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(3S)-3-methyl-1,3-dihydro-2-benzofuran- 5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.44 (s, 1H), 8.55 (s, 1H), 8.09 – 8.03 (m, 2H), 8.01 – 7.94 (m, 2H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 7.44 (d, J = 7.9 Hz, 1H), 5.41 (s, 2H), 5.30 (d, J = 6.6 Hz, 1H), 5.08 (m, 1H), 4.97 (m, 1H), 4.55 (m, 1H), 3.60 – 3.47 (m, 3H), 3.27 (s, 2H), 3.01 (m, 3H), 2.85 (m, 1H), 2.68 (d, J = 9.6 Hz, 1H), 2.44 (s, 3H), 1.45 (d, J = 6.3 Hz, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.20 [M+H]+. EXAMPLE 190 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(3R)-3-methyl-1,3-dihydro-2- benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide
The racemic 189.7 was purified by chiral SFC to afford (4.6 mg, 22.7% yield) of N- [2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-[(3R)-3-methyl-1,3-dihydro-2-benzofuran-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.44 (s, 1H), 8.54 (s, 1H), 8.06 (dd, J = 8.3, 3.0 Hz, 2H), 8.00 – 7.91 (m, 2H),
7.72 (dd, J = 8.6, 2.1 Hz, 1H), 7.44 (d, J = 7.9 Hz, 1H), 5.41 (s, 2H), 5.30 (d, J = 6.7 Hz, 1H), 5.08 (m, 1H), 4.97 (m, 1H), 4.55 (m, 1H), 3.53 (m, 3H), 3.01 (m, 3H), 2.85 (m, 1H), 2.68 (d, J = 9.1 Hz, 1H), 2.44 (s, 3H), 1.45 (d, J = 6.3 Hz, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.20 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 191 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(1R)-1-methyl-1,3-dihydro-2- benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 191
Step 1: Preparation of N'-[(1E)-(5-bromo-2- hydroxyphenyl)methylidene]acetohydrazide Compound 191.1
A solution of 5-bromo-2-hydroxybenzaldehyde (3.0 g, 14.9 mmol) and acetohydrazide (2.2 g, 30.0 mmol) in EtOH (30 mL) was stirred for 1 hour at 85 ℃ under N2 atmosphere. The precipitated solids were collected by filtration and washed with EtOH (20 mL x 3). This resulted in (3.5 g, 89.9% yield) of N'-[(1E) -(5-bromo-2- hydroxyphenyl)methylidene]acetohydrazide as a white solid. LCMS observed m/z = 257.09 [M+H]+. Step 2: Preparation of 2-acetyl-5-bromobenzaldehyde Compound 191.2
To a stirred mixture of N'-[(1E)-(5-bromo-2- hydroxyphenyl)methylidene]acetohydrazide (3.5 g, 13.6 mmol) in THF (40 mL) were added Pb(OAc)4 (7.2 g, 16.4 mmol) in portions at 0 ℃ under N2 atmosphere. The resulting mixture was stirred for additional 4 hours at 0 ℃. Desired product could be detected by LCMS. The reaction was quenched with H2O (50 mL) at room temperature. The resulting mixture was extracted with EA (50 mL x 3). The combined organic layers were washed with brine (40 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 227.06 [M+H]+. Step 3: Preparation of 1-[4-bromo-2-(hydroxymethyl)phenyl] ethanol Compound 191.3
To a stirred solution of 2-acetyl-5-bromobenzaldehyde (3.6 g, 15.9 mmol) in MeOH (70 mL) was added NaBH4 (2.4 g, 63.4 mmol) in portions at 0 ℃ under N2 atmosphere. The resulting mixture was stirred for 16 hours at room temperature under N2 atmosphere. The reaction was quenched by the addition of H2O (50 mL). The resulting mixture was extracted with DCM (50 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (3.1 g, 84.6% yield) of 1-[4-bromo-2-(hydroxymethyl)phenyl]ethanol as a white solid. LCMS observed m/z = 231.09 [M+H]+. Step 4: Preparation of 5-bromo-1-methyl-1,3-dihydro-2-benzofuran Compound 191.4
A solution of 1-[4-bromo-2-(hydroxymethyl)phenyl]ethanol (400 mg, 1.7 mmol) in DMF (20 mL) was treated with TsCl (396 mg, 2.1 mmol) at 0 ℃ under nitrogen atmosphere followed by the addition of NaH (60%, 173 mg, 4.3 mmol) in portions at this temperature. The resulting mixture was stirred for 16 hours at room temperature under N2 atmosphere. The reaction was quenched by the addition of H2O (30 mL) at 0℃. The resulting mixture was extracted with EA (50 mL x 3). The combined organic layers were washed with brine (50 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (205 mg, 55.6% yield) of 5-bromo-1-methyl-1,3-dihydro-2-benzofuran as a light-yellow oil. LCMS observed m/z = 213.07 [M+H]+. Step 5: Preparation of 4,4,5,5-tetramethyl-2-(1-methyl-1,3-dihydro-2-benzofuran-5- yl)-1,3,2-dioxaborolane Compound 191.5
A solution of 5-bromo-1-methyl-1,3-dihydro-2-benzofuran (400 mg, 1.9 mmol) in dioxane (8 mL) was treated with 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3,2-dioxaborolane (715 mg, 2.8 mmol) at room temperature under nitrogen atmosphere followed by the addition of Pd(dppf)Cl2CH2Cl2 (153 mg, 0.2 mmol) at this temperature. To the above mixture was added AcOK (553mg, 5.6 mmol) in one portion at room temperature. The resulting mixture was stirred for additional 3 hours at 100 ℃. The mixture was allowed to cool down to room temperature. The resulting mixture was filtered, the filter cake was washed with DCM. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (362 mg, 74.1% yield) of 4,4,5,5- tetramethyl-2-(1-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2-dioxaborolane as a light- yellow oil. LCMS observed m/z = 261.16 [M+H]+. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(1-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide Compound 191.6
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (400 mg, 0.5 mmol) and 4,4,5,5-tetramethyl-2-(1-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2- dioxaborolane (198 mg, 0.8 mmol) in dioxane (8 mL) and H2O (2 mL) were added Pd(dppf)Cl2CH2Cl2 (41 mg, 0.1 mmol) and K2CO3 (210 mg, 1.5 mmol) in portions atroom temperatureunder N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 95 ℃. Desired product could be detected by LCMS. The reaction was quenched with H2O (10 mL). The resulting mixture was extracted with EA (10 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in DCM) to afford (145 mg, 34.0% yield) of 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(1-methyl- 1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 842.27 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(1-methyl-1,3-dihydro-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 191.7
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(1-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (135 mg, 0.2 mmol) in TFA (3 mL) was stirred for 15 min at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product (150 mg) was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Phenyl 5 m, 30 mm * 150 mm; Mobile Phase A: Water(10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 40% B in 9min; Wave Length: 254nm/220nm nm; RT1(min): 7.83) to afford (102 mg, 84.6% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(1-methyl-1,3-dihydro-2-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a light yellow solid. LCMS observed m/z = 752.35 [M+H]+. Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(1R)-1-methyl-1,3-dihydro-2- benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(1-methyl-1,3-dihydro-2-benzofuran-5-
yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (65 mg, 0.1 mmol) was purified by SFC to afford (19.2 mg, 29.3% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5- ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(1R)-1-methyl- 1,3-dihydro-2-benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 10.24 (s, 1H), 8.58 (s, 1H), 8.09 – 8.00 (m, 3H), 7.97 (d, J = 2.1 Hz, 1H), 7.71 (m, 1H), 7.41 (d, J = 7.8 Hz, 1H), 5.40 (s, 2H), 5.27 (q, J = 6.6 Hz, 1H), 5.09 (m, 1H), 4.98 (m, 1H), 4.54 (m, 1H), 3.52 (m, 3H), 3.27 (m, 1H), 3.07 – 2.96 (m, 3H), 2.84 (m, 1H), 2.67 (d, J = 9.5 Hz, 1H), 2.45 (s, 3H), 1.43 (d, J = 6.3 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 752.35 [M+H]+. EXAMPLE 192 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(1S)-1-methyl-1,3-dihydro-2-benzofuran- 5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 192
Step 1: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(1S)-1-methyl-1,3-dihydro-2- benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 192
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(1-methyl-1,3-dihydro-2-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (65 mg, 0.1 mmol) was purified by SFC to afford (22.7 mg, 34.6% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5- ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(1S)-1-methyl- 1,3-dihydro-2-benzofuran-5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 10.24 (s, 1H), 8.58 (s, 1H), 8.08 – 8.01 (m, 3H), 7.97 (d, J = 2.1 Hz, 1H), 7.71 (m, 1H), 7.41 (d, J = 7.8 Hz, 1H), 5.39 (s, 2H), 5.27 (d, J = 6.7 Hz, 1H), 5.12 – 5.05 (m, 1H), 4.98 (m, 1H), 4.54 (m, 1H), 3.52 (m, 3H), 3.28 (m, 1H), 3.02 (m, 3H), 2.84 (m, 1H), 2.67 (d, J = 9.3 Hz, 1H), 2.45 (s, 3H), 1.43 (d, J = 6.3 Hz, 3H), 1.21 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 752.35 [M+H]+. EXAMPLE 193 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(3,3-dimethyl-2,3-dihydro- 1-benzofuran-5-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide Compound 193
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl][2-(3,3-dimethyl-2,3-dihydro-1-benzofuran-5-yl)- 6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo- 1,3,3a,7-tetraaza-7-indenyl]acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.39 (s, 1H), 8.47 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.91 (s, 1H), 7.84 (d, J = 7.9 Hz, 2H), 7.65 (d, J = 8.7 Hz, 1H), 6.83 (d, J = 8.3 Hz, 1H), 5.32 (s, 2H), 4.46 (d, J = 12.4 Hz, 1H), 4.23 (s, 2H), 3.50 - 3.41 (m, 3H), 3.23 - 3.16 (m, 1H), 2.96 - 2.88 (m, 3H), 2.77 (d, J = 11.2 Hz, 1H), 2.59 (d, J = 10.8 Hz, 1H), 2.37 (s, 3H), 1.27 (s, 6H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 766.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 194
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(2,2-dimethyl-2,3-dihydro- 1-benzofuran-5-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide Compound 194
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl][2-(2,2-dimethyl-2,3-dihydro-1-benzofuran-5-yl)- 6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo- 1,3,3a,7-tetraaza-7-indenyl]acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.36 (s, 1H), 8.46 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.89 (d, J = 18.8 Hz, 2H), 7.80 (d, J = 8.4 Hz, 1H), 7.65 (d, J = 8.7 Hz, 1H), 6.74 (d, J = 8.3 Hz, 1H), 5.30 (s, 2H), 4.46 (d, J = 12.4 Hz, 1H), 3.49 - 3.41 (m, 3H), 3.19 - 3.15 (m, 1H), 3.01 (s, 2H), 2.95 - 2.92 (m, 3H), 2.76 (d, J = 11.2 Hz, 1H), 2.58 (d, J = 11.4 Hz, 1H), 2.36 (s, 3H), 1.37 (s, 6H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 766.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 195 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,1-dimethyl-6- isochromanyl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide Compound 195
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,1-dimethyl-6-isochromanyl)-6-ethyl-5-{4- [(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7- indenyl]acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 10.25 (s,
1H), 8.59 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.99 (s, 1H), 7.90 (d, J = 8.2 Hz, 1H), 7.85 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 8.2 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.87 (t, J = 5.5 Hz, 2H), 3.63 – 3.43 (m, 3H), 3.31 – 3.24 (m, 1H), 3.09 – 2.95 (m, 3H), 2.88 – 2.76 (m, 3H), 2.67 (d, J = 10.1 Hz, 1H), 2.45 (s, 3H), 1.47 (s, 6H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 780.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 196 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimi dine-4-carbonyl)pipera zin-1-yl]-2-(4-methyl-1,3-dihydro-2-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyr imidin-4-yl}acetamide. Compound 196
Step 1: Preparation of [3-bromo-6-(hydroxymethyl)-2-methylphenyl]methanol. Compound 196.1
To a stirred mixture of 5-bromo-4-methyl-3H-2-benzofuran-1-one (400 mg, 1.7 mmol, 1.0 equiv.) in DCM (20 mL) was added Diisobutylaluminium hydride (2.5 g, 17.6 mmol, 10.0 equiv.) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was stirred for additional 1 h at -78 °C under nitrogen atmosphere. After completion of the reaction, the mixture was warmed to 0 ℃, quenched with ice water (10 mL) at 0 ℃ and extracted with CH2Cl2 (3 x 20 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (3:1) to afford [3-bromo-6-(hydroxymethyl)-2-methylphenyl]methanol (250 mg, 61% yield) as a white solid. LCMS observed m/z= 230.99[M+H]+.
Step 2: Preparation of 5-bromo-4-methyl-1,3-dihydro-2-benzofuran. Compound 196.2
To a stirred solution of [3-bromo-6-(hydroxymethyl)-2-methylphenyl]methanol (150 mg, 0.6 mmol, 1.0 equiv.) and TsCl (148 mg, 0.8 mmol, 1.2 equiv.) in DMF (2 mL) was added NaH (39 mg, 1.6 mmol, 2.5 equiv.) in portions at 0 °C. The resulting mixture was stirred for additional 1 h at 25 °C under nitrogen atmosphere. After completion of the reaction, the resulting mixture was quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (10:1) to afford 5-bromo-4-methyl-1,3-dihydro-2-benzofuran (105 mg, 45% yield) as a colorless oil. LCMS observed m/z = 212.98[M+H]+. Step 3: Preparation of 4,4,5,5-tetramethyl-2-(4-methyl-1,3-dihydro-2-benzofuran-5- yl)-1,3,2-dioxaborolane. Compound 196.3
To a stirred solution of 5-bromo-4-methyl-1,3-dihydro-2-benzofuran (105 mg, 0.5 mmol, 1.0 equiv.) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3,2-dioxaborolane (625 mg, 2.5 mmol, 5.0 equiv.) in dioxane (5 mL) were added AcOK (145 mg, 1.5 mmol, 3.0 equiv.) and Pd(dppf)Cl2 (36 mg, 0.05 mmol, 0.1 equiv.) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred for additional 2 h at 100 °C under nitrogen atmosphere. After completion of the reaction, the resulting mixture was quenched with H2O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 4,4,5,5-tetramethyl-2-(4-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2- dioxaborolane (28.0 mg, crude) as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 261.15[M+H]+.
Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]aceta mide.
To a stirred solution of 2-(6-{4-[5-(benzyloxy)pyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (27 mg, 0.03 mmol, 1.0 equiv.) and 4,4,5,5-tetramethyl-2-(4-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2-dioxaborolane (9 mg, 0.03 mmol, 1.0 equiv.) in dioxane (1 mL) and H2O (0.1 mL) were added Pd(dppf)Cl2 (3 mg, 0.004 mmol, 0.1 equiv.) and K2CO3 (10 mg, 0.07 mmol, 2.0 equiv.) in portions at 25°C under nitrogen atmosphere. The resulting mixture was heated to 60 °C and stirred for additional 12 h at 60 °C under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature, quenched with H2O (5 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with DCM/MeOH (20:1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(4- methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide (8 mg, 27% yield) as a white solid. LCMS observed m/z = 842.27[M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimi dine-4-carbonyl)pipera zin-1-yl]-2-(4-methyl-1,3-dihydro-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyr imidin-4-yl}acetamide. Compound 196
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(4-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoro methyl)phenyl]acetamide (8 mg, 0.009 mmol, 1.0 equiv.) in THF (0.5 mL) was stirred for 1 h at 80 °C under air atmosphere. Then the resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 33% B to 56% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 5.83) to afford N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-(4-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (0.8 mg, 10% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.28 (s, 1H), 8.52 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.88 (d, J = 7.9 Hz, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.26 (d, J = 7.9 Hz, 1H), 5.39 (s, 2H), 5.09 (s, 4H), 4.55 (m, 1H), 3.54 (m, 3H), 3.03 (m, 3H), 2.85 (m, 1H), 2.68 (s, 1H), 2.49 (s, 3H), 2.43 (s, 2H), 1.31 – 1.16 (m, 5H). LCMS observed m/z = 752.50[M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 197 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-5-isoindolinyl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 197
The title compound was prepared using a similar procedure as compound 164 using intermediate AC, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1- isoindolinone with 4,4,5,5-tetramethyl-2-(2-methyl-5-isoindolinyl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 8.56 (s, 1H), 8.32 (s, 1H), 8.27 – 8.12 (m, 3H), 7.82 (s, 1H), 7.61 (d, J = 8.7 Hz, 1H), 7.50 (d, J = 7.9 Hz, 1H), 5.47 (s, 2H), 4.72 (d, J = 12.8 Hz, 1H), 4.64 (s, 4H), 4.09 (d, J = 13.0 Hz, 1H), 3.76 (t, J = 12.2 Hz, 2H), 3.45 (t, J = 12.3 Hz, 1H), 3.22 – 3.09 (m, 3H), 3.07 (s, 3H), 2.96 (d, J = 11.8 Hz, 1H), 2.87 – 2.74 (m, 1H), 2.66 (s, 1H), 2.52 (s, 3H), 1.33 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. EXMPLE 198 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(4-methyl-3-oxo-2,4-dihydro-1,4- benzoxazin-6-yl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 198
The title compound was prepared using a similar procedure as compound 164 using intermediate AC, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1- isoindolinone with 4-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,4-dihydro- 1,4-benzoxazin-3-one. 1H NMR (400MHz, DMSO-d6) δ 10.44 (s, 1H), 8.56 (s, 1H), 8.06
(d, J = 8.6 Hz, 1H), 7.98 (s, 1H), 7.81 – 7.68 (m, 3H), 7.14 (d, J = 8.2 Hz, 1H), 5.40 (s, 2H), 4.74 (s, 2H), 4.54 (d, J = 12.2 Hz, 1H), 3.63 – 3.42 (m, 3H), 3.34 (s, 3H), 3.29 – 3.19 (m, 1H), 3.11 – 2.91 (m, 3H), 2.84 (d, J = 11.4 Hz, 1H), 2.66 (d, J = 11.3 Hz, 1H), 2.44 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 781.3 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 199 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(1-methyl-5-indolinyl)-4-oxo-1,3,3a,7- tetraaza-7-indenyl)acetamide. Compound 199
The title compound was prepared using a similar procedure as compound 164, replacing 2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-isoindolinone with 4,4,5,5-tetramethyl-2-(1-methyl-5-indolinyl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.40 (s, 1H), 10.27 (br s, 1H), 8.57 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.80 (d, J = 8.3 Hz, 1H), 7.75 (s, 1H), 7.71 (d, J = 8.7 Hz, 1H), 6.55 (d, J = 8.2 Hz, 1H), 5.35 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 3.61 – 3.44 (m, 3H), 3.41 – 3.35 (m, 2H), 3.28 – 3.17 (m, 1H), 3.05 – 2.91 (m, 5H), 2.82 (d, J = 11.2 Hz, 1H), 2.77 (s, 3H), 2.64 (d, J = 11.7 Hz, 1H), 2.44 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 773.2 [M+Na]+. EXAMPLE 200 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4-fluoro-1,3- dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 200
Step 1: Preparation of 1,2-dimethyl 4-bromo-3-fluorophthalate. Compound 200.1
To a stirred solution of 4-bromo-3-fluorobenzene-1,2-dicarboxylic acid (350 mg, 1.3 mmol, 1.0 equiv.) and Cs2CO3 (867 mg, 2.6 mmol, 2.0 equiv.) in DMF (5 mL) was added MeI (1.1 g, 7.9 mmol, 6.0 equiv.) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was heated to 50 °C and stirred for 24 h at 50 °C under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to room temperature, quenched with H2O (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5:1) to afford 1,2-dimethyl 4-bromo-3- fluorophthalate (362 mg, 93% yield) as a colorless oil. LCMS observed m/z = 290.95 [M+H]+. Step 2: Preparation of [3-bromo-2-fluoro-6-(hydroxymethyl)phenyl]methanol. Compound 200.2
To a stirred solution of 1,2-dimethyl 4-bromo-3-fluorophthalate (360 mg, 1.2 mmol, 1.0 equiv.) and CaCl2 (823 mg, 7.4 mmol, 6.0 equiv.) in MeOH (5 mL) was added NaBH4 (561 mg, 14.8 mmol, 12.0 equiv.) in portions at 0 °C. The resulting mixture was heated to 50 °C and stirred for additional 3 h at 50 °C under nitrogen atmosphere. After completion
of the reaction, the reaction was quenched with NH4Cl (sat., 20 mL) at 0 °C and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford [3-bromo-2-fluoro-6-(hydroxymethyl)phenyl]methanol (225 mg, 77% yield) as a white solid. LCMS observed m/z = 234.96 [M+H]+. Step 3: Preparation of 5-bromo-4-fluoro-1,3-dihydro-2-benzofuran. Compound 200.3
To a stirred solution of [3-bromo-2-fluoro-6-(hydroxymethyl)phenyl]methanol (185 mg, 0.8 mmol, 1.0 equiv) in DCE (2 mL) was added MnO2 (342 mg, 4.0 mmol, 5.0 equiv.) in portions at room temperature. The resulting mixture was stirred for additional 1 h at room temperature under air atmosphere. Then the resulting mixture was filtered and the filter cake was washed with DCE (2 mL). To the above filtrate was added Et3SiH (333 mg, 2.8 mmol, 3.5 equiv.) dropwise at 25 °C. The mixture was stirred for additional 30 min at room temperature, and then TFA (0.3 mL, 4.7 mmol, 5.8 equiv.) was added dropwise at 25 °C. The resulting mixture was stirred for additional 25 h at room temperature. After completion of the reaction, the resulting mixture was quenched with H2O (2 mL) and extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were washed with water (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 5-bromo-4-fluoro-1,3- dihydro-2-benzofuran (66 mg, 31% yield) as a white solid. LCMS observed m/z = 216.95[M+H]+. Step 4: Preparation of 2-(4-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 200.4
To a stirred solution of 5-bromo-4-fluoro-1,3-dihydro-2-benzofuran (66 mg, 0.3 mmol, 1.0 equiv.) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-
1,3,2-dioxaborolane (386 mg, 1.5 mmol, 5.0 equiv.) in dioxane (1 mL) were added AcOK (89 mg, 0.9 mmol, 3.0 equiv.) and Pd(dppf)Cl2.CH2Cl2 (24 mg, 0.03 mmol, 0.1 equiv.) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 70 °C and stirred for additional 12 h at 70 °C under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to room temperature, quenched with H2O (5 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 2-(4-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (60 mg, 74% yield) as a yellow solid. LCMS observed m/z = 265.13 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 200.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (60 mg, 0.07 mmol, 1.0 equiv.) and 2-(4-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (20 mg, 0.07 mmol, 1.0 equiv.) in dioxane (1 mL) and H2O (0.1 mL) were added Pd(dppf)Cl2CH2Cl2 (6 mg, 0.008 mmol, 0.1 equiv.) and K2CO3 (21 mg, 0.1 mmol, 1.4 equiv.) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 60 °C and stirred for additional 12 h at 60 °C under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature, quenched with H2O (2 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL)
and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with CH2Cl2 / MeOH (20:1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoro methyl)phenyl]acetamide (20 mg, 31% yield) as a white solid. LCMS observed m/z = 846.24 [M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4- fluoro-1,3-dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 200
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(4-fluoro-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluorom ethyl)phenyl]acetamide (20 mg, 0.02 mmol, 1.0 equiv.) in TFA (0.5 mL) was stirred for 1 h at 25 °C under air atmosphere. Then the resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 19 mm * 250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 58% B to 83% B in 8min; Wave Length: 254 nm/220 nm; RT1 (min): 5.98) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4-fluoro-1,3-dihydro-2- benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]tri azolo[1,5-a]pyrimidin-4-yl]acetamide (1.1 mg, 5.9% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.52 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 8.02 – 7.89 (m, 2H), 7.71 (d, J = 8.6 Hz, 1H), 7.30 (d, J = 7.8 Hz, 1H), 5.39 (s, 2H), 5.17 (s, 2H), 5.10 (s, 2H), 4.54 (m, 1H), 3.57 - 3.48 (m, 3H), 3.06 - 3.00 (m, 3H), 2.91 – 2.79 (m, 1H), 2.67 (s, 1H), 2.43 (s, 3H), 1.26 - 1.20 (m, 3H), 1.17 (s, 1H). LCMS observed m/z = 756.15[M+H]+. Note: One exchangeable proton was not observed in NMR spectra.
EXAMPLE 201 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-1,3- dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 201
Step 1: Preparation of 1,2-dimethyl 4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl) phthalate. Compound 201.1
To a stirred mixture of bis(pinacolato)diboron (538 mg, 2.1 mmol) and Di-mu- methoxobis(1,5-cyclooctadiene)diiridium(I) (18 mg, 0.03 mmol) in 2-methoxy-2- methylpropane (0.6 mL) were added 1,2-dimethyl 4-fluorophthalate (300 mg, 1.4 mmol) and 4-tert-butyl-2-(4-tert-butylpyridin-2-yl)pyridine (15 mg, 0.05 mmol) in portions at 25 °C. The resulting mixture was heated to 100 °C and stirred at 100 °C for 4 h under nitrogen atmosphere. After the reaction was completed, the mixture was allowed to cool down to 25 °C and poured into water (20 mL). The reaction mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give 1,2-dimethyl 4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phthalate (350 mg, crude) as a brown yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 339.13 [M+H] +. Step 2: Preparation of 1,2-dimethyl 4-bromo-5-fluorophthalate.
Compound 201.2
To a stirred solution of 1,2-dimethyl 4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phthalate (1.0 g, 2.9 mmol) in MeOH (20 mL) and H2O (20 mL) was added CuBr2 (800 mg, 5.9 mmol) in portions at 25 °C. The resulting mixture was heated to 65 °C and stirred at 65 °C for 2 h. After the reaction was completed, the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 1,2-dimethyl 4-bromo-5-fluorophthalate (350 mg, 40.5% yield) as a white solid. LCMS observed m/z = 290.96 [M+H] +. Step 3: Preparation of [5-bromo-4-fluoro-2-(hydroxymethyl)phenyl]methanol. Compound 201.3
To a solution of NaBH4 (332 mg, 8.7 mmol) in THF (15 mL) was added CaCl2 (488 mg, 4.4 mmol) at 0 °C under nitrogen atmosphere followed by the addition of 1,2-dimethyl 4-bromo-5-fluorophthalate (320 mg, 1.1 mmol) in portions at 0 °C. The resulting mixture was heated to 25 °C and stirred at 25 °C for 8 h. After the reaction was completed, the reaction was quenched by the addition of MeOH (10 mL) at 25 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford [5-bromo-4-fluoro-2- (hydroxymethyl)phenyl]methanol (160 mg, 61.4% yield) as a white solid. LCMS observed m/z = 234.97 [M+H] +. Step 4: Preparation of 5-bromo-6-fluoro-1,3-dihydro-2-benzofuran. Compound 201.4
To a stirred mixture of [5-bromo-4-fluoro-2-(hydroxymethyl)phenyl]methanol (200 mg, 0.9 mmol) in DCM (8 mL) was added MnO2 (739 mg, 8.5 mmol) in portions at 25 °C. The resulting mixture was stirred at 25 °C for 2 h. After the reaction was completed, the
resulting mixture was filtered, the filter cake was washed with DCM (3 x 10 mL). The filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. To a stirred solution of 6-bromo-5-fluoro-1,3- dihydro-2-benzofuran-1-ol (50 mg, 0.2 mmol) and Et3SiH (74 mg, 0.6 mmol) in DCM (10 mL) was added TFA (122 mg, 1.0 mmol) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 2 h. After the reaction was completed, the resulting mixture was quenched with water and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford 5-bromo-6- fluoro-1,3-dihydro-2-benzofuran (30 mg, 64.3% yield) as a white solid. LCMS observed m/z = 216.96 [M+H] +. Step 5: Preparation of 2-(6-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 201.5
To a stirred mixture of 5-bromo-6-fluoro-1,3-dihydro-2-benzofuran (50 mg, 0.2 mmol) and bis(pinacolato)diboron (70 mg, 0.2 mmol) in dioxane (3 mL) were added Pd(dppf)Cl2 (16 mg, 0.02 mmol) and KOAc (45 mg, 0.4 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 8 h under nitrogen atmosphere. After the reaction was completed, the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure. The crude product(20 mg) was used in the next step directly without further purification. LCMS observed m/z = 245.13 [M+H] +. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(6-fluoro-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl] acetamide. Compound 201.6
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- (6-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (40 mg, 0.1 mmol) in dioxane (3 mL) and H2O (0.3 mL) were added K2CO3 (43 mg, 0.3 mmol) and Pd(dppf)Cl2 (9 mg, 0.01 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h. After the reaction was completed, the mixture was allowed to cool down to room temperature and poured into water (15 mL). The reaction mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/1) to afford 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(6-fluoro-1,3- dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (50 mg, 46.2% yield) as a white solid. LCMS observed m/z = 846.25 [M+H] +. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6- fluoro-1,3-dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 201
Into a 20 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(6-fluoro-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoro methyl)phenyl]acetamide (50 mg, 0.05 mmol) and TFA (5 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 30 min. After the reaction was completed, the mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-1,3- dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (8.4 mg, 17.2% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.12 (s, 1H), 8.62 (s, 1H), 8.49 (d, J = 8.7 Hz, 1H), 8.06 (d, J = 6.3 Hz, 1H), 7.62 (d, J = 2.0 Hz, 1H), 7.55 (d, J = 8.4 Hz, 1H), 7.10 (d, J = 10.0 Hz, 1H), 5.71 (d, J = 13.1 Hz, 1H), 5.20 (s, 2H), 5.15 (d, J = 8.9 Hz, 4H), 4.83 (d, J = 12.7 Hz, 1H), 3.86 (t, J = 11.7 Hz, 2H), 3.60 – 3.46 (m, 1H), 3.25 (d, J = 7.9 Hz, 2H), 3.10 (s, 1H), 2.92 – 2.85 (m, 1H), 2.83 – 2.76 (m, 1H), 2.59 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 756.15 [M+H] +. EXAMPLE 202 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(7-fluoro-1,3- dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 202
Step 1: Preparation of [4-bromo-2-fluoro-6-(hydroxymethyl)phenyl]methanol. Compound 202.1
To a stirred mixture of 5-bromo-3-fluorobenzene-1,2-dicarboxylic acid (370 mg, 1.4 mmol) and ZnCl2 (192 mg, 1.4 mmol) in THF (5 mL) was added NaBH4 (64 mg, 1.7 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 70 °C and stirred at 70 °C for 6 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 0 °C and quenched with ice water (10 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give [4-bromo-2-fluoro-6- (hydroxymethyl)phenyl]methanol (130 mg, 39% yield) as a white solid. LCMS observed m/z = 232.97 [M-H]-. Step 2: Preparation of 6-bromo-4-fluoro-1,3-dihydro-2-benzofuran-1-ol. Compound 202.2
To a stirred solution of [4-bromo-2-fluoro-6-(hydroxymethyl)phenyl]methanol (140 mg, 0.6 mmol) in DCM (3 mL) was added MnO2 (259 mg, 3.0 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was filtered, the filter cake was washed with DCM (3 mL). The filtrate was concentrated under reduced
pressure to give 6-bromo-4-fluoro-1,3-dihydro-2-benzofuran-1-ol (130 mg, 94% yield) as a light-yellow oil. LCMS observed m/z = 230.95 [M-H]-. Step 3: Preparation of 6-bromo-4-fluoro-1,3-dihydro-2-benzofuran. Compound 202.3
To a stirred solution of 6-bromo-4-fluoro-1,3-dihydro-2-benzofuran-1-ol (130 mg, 0.6 mmol) in DCM (2 mL) were added Et3SiH (195 mg, 1.7 mmol) and TFA (318 mg, 2.8 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. After completion of reaction, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford 6-bromo- 4-fluoro-1,3-dihydro-2-benzofuran (40 mg, 33% yield) as a light-yellow solid. LCMS observed m/z = 214.96 [M-H]-. Step 4: Preparation of 2-(7-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 202.4
To a stirred mixture of 6-bromo-4-fluoro-1,3-dihydro-2-benzofuran (30 mg, 0.1 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (53 mg, 0.2 mmol) in dioxane (1 mL) were added Pd(dppf)Cl2 (11 mg, 0.01 mmol) and KOAc (41 mg, 0.4 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 12 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The crude product was used for next step directly. LCMS observed m/z = 265.15 [M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(7- fluoro-1,3-dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 202
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (90 mg, 0.1 mmol) and 2- (7-fluoro-1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (30 mg, 0.1 mmol) in dioxane (2 mL) and H2O (0.2 mL) were added Pd(dppf)Cl2 (9 mg, 0.01 mmol) and K2CO3 (47 mg, 0.3 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 25 °C and diluted with water (10 mL). The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (30 mg) was purified by Prep-HPLC with the following conditions (Column: X- bridge BEH Phenyl 5 m, 19 mm * 250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: MeOH; Flow rate: 25 mL/min; Gradient: 65% B to 75% B in 8 min; Wave Length: 254 nm/220 nm; RT1(min): 7.42) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]- 2-[5-ethyl-2-(7-fluoro-1,3-dihydro-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (6.3 mg, 91.1% purity) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.32 (s, 1H), 8.49 (s, 1H), 8.05 (d, J = 8.4 Hz, 1H), 8.00 – 7.94 (m, 1H), 7.92 (s, 1H), 7.80 – 7.64 (m, 2H), 5.39 (s, 2H), 5.13 (d, J = 11.2 Hz, 4H), 4.81 – 4.40 (m, 1H), 3.64 – 3.46 (m, 3H), 3.32 (s, 1H), 3.02 (s, 3H), 2.84 (d, J = 11.2 Hz, 1H), 2.67 (s, 2H), 2.42 (s, 2H), 1.30 – 1.15 (m, 3H). LCMS observed m/z = 756.15 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 203
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(6-methyl-1,3-dihydro-2-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 203
Step 1: Preparation of 4-bromo-5-methylbenzene-1,2-dicarboxylic acid Compound 203.1
A solution of 4-methylbenzene-1,2-dicarboxylic acid (25.0 g, 138.8 mmol) in H2O (139 mL) was treated with NaOH (16.7 g, 416.3 mmol) at 0 ℃ under N2 atmosphere followed by the addition of Br2 (22.2 g, 138.8 mmol) dropwise at 0 ℃. The resulting mixture was stirred for 1.5 hours at 80 ℃ under N2 atmosphere. The mixture was neutralized to pH=7 with HCl (aq.). The resulting mixture was filtered, the filter cake was washed with water (100 mL x 3). The filtrate was concentrated under reduced pressure. This resulted in (16.0 g, 44.5% yield) of 4-bromo-5-methylbenzene-1,2-dicarboxylic acid as a white solid. LCMS observed m/z = 256.95 [M-H]-. Step 2: Preparation of [4-bromo-2-(hydroxymethyl)-5-methylphenyl]methanol Compound 203.2
A solution of 4-bromo-5-methylbenzene-1,2-dicarboxylic acid (8.0 g, 30.9 mmol) in THF (154 mL) was treated with BH3-Me2S (46.0 mL, 92.0 mmol) at 0 ℃ under N2 atmosphere. The resulting mixture was stirred for 16 hours at 55
℃ under N2 atmosphere. To the above mixture was added NaOH (2 M, 140 mL) dropwise at 0 ℃. The resulting mixture was stirred for additional 1 hour at room temperature. The mixture was neutralized to pH=7 with HCl (aq.). The resulting mixture was extracted with EA (100 mL x 3). The combined organic layers were washed with brine (100 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (4.4 g, 61.7% yield) of [4-bromo-2-(hydroxymethyl)-5-methylphenyl]methanol as a white solid. LCMS observed m/z = 228.99 [M-H]-. Step 3: Preparation of 5-bromo-6-methyl-1,3-dihydro-2-benzofuran Compound 203.3
A solution of [4-bromo-2-(hydroxymethyl)-5-methylphenyl]methanol (1.5 g, 6.5 mmol) in DMF (20 mL) was treated with TsCl (1.5 g, 7.8 mmol) at 0 ℃ under N2 atmosphere followed by the addition of NaH (60%, 650 mg, 16.2 mmol) in portions at this temperature. The resulting mixture was stirred for 16 hours at room temperature under N2 atmosphere. The reaction was quenched by the addition of H2O (30 mL) at 0 ℃. The resulting mixture was extracted with EA (50 mL x 3). The combined organic layers were washed with brine (50 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (1.1 g, 80.0% yield) of 5-bromo-6-methyl-1,3-dihydro-2-benzofuran as a light-yellow oil. LCMS observed m/z = 212.98 [M+H]+. Step 4: Preparation of 4,4,5,5-tetramethyl-2-(6-methyl-1,3-dihydro-2-benzofuran-5- yl)-1,3,2-dioxaborolane Compound 203.4
To a stirred solution of 5-bromo-6-methyl-1,3-dihydro-2-benzofuran (300 mg, 1.4 mmol) and bis(pinacolato)diboron (536 mg, 2.1 mmol) in dioxane (5 mL) were added Pd(dppf)Cl2CH2Cl2 (115 mg, 0.1 mmol) and AcOK (415 mg, 4.2 mmol) at room
temperature under N2 atmosphere. The resulting mixture was stirred for additional 1 hour at 100 ℃. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with EA (5 mL x 3). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (190 mg, 51.9% yield) of 4,4,5,5-tetramethyl- 2-(6-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2-dioxaborolane as a yellow solid. LCMS observed m/z = 261.16 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(6-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide Compound 203.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 4,4,5,5-tetramethyl-2-(6-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2- dioxaborolane (59 mg, 0.2 mmol) in dioxane (5 mL) and H2O (1 mL) were added Pd(dppf)Cl2CH2Cl2 (10 mg, 0.1 mmol) and K2CO3 (53 mg, 0.4 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 95 ℃. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with MeOH (2 mL x 3). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography to afford (65 mg, 60.9% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(6-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 842.27 [M+H]+.
Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(6-methyl-1,3-dihydro-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 203
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(6-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.1 mmol) in TFA (1 mL) was stirred for 15 min at 85 ℃ under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: Water(10 mmol/ L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/ min; Gradient: 34% B to 57% B in 8min; Wave Length: 254nm/ 220nm nm; RT1(min): 6.68) to afford (18.5 mg, 41.1% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(6-methyl-1,3-dihydro-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.47 (s, 1H), 8.02 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 2.1 Hz, 1H), 7.85 (s, 1H), 7.70 (m, 1H), 7.27 (s, 1H), 5.35 (s, 2H), 5.00 (s, 4H), 4.51 (m, 1H), 3.55 – 3.41 (m, 3H), 3.28 (m, 1H), 3.01 (d, J = 8.7 Hz, 3H), 2.84 (m, 1H), 2.67 (p, J = 1.7 Hz, 1H), 2.58 (s, 3H), 2.40 (s, 3H), 1.20 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.25 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 204 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1H-inden-6-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide.
Step 1: Preparation of 5-bromo-1-methoxy-2,3-dihydro-1H-indene. Compound 204.1
To a stirred solution of 5-bromo-2,3-dihydro-1H-inden-1-ol (1.3 g, 6.2 mmol) and NaOH (371 mg, 9.3 mmol) in THF (10 mL) was added MeI (1.0 g, 7.4 mmol) dropwise at 0 °C under nitrogen atmosphere. The reaction mixture was heated to 40 °C and stirred at 40 °C for 12 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and extracted with DCM (3 x 50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 40% to 60% gradient in 20 minutes; detector, UV 254 nm) to give 5- bromo-1-methoxy-2,3-dihydro-1H-indene (1.1 g, 75.9% yield) as a brown oil. LCMS observed m/z = 227.00 [M+H]+. Step 2: Preparation of 2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 204.2
To a solution of 5-bromo-1-methoxy-2,3-dihydro-1H-indene (100 mg, 0.4 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (113 mg, 0.4 mmol) in dioxane (3 mL) were added Pd(dppf)Cl2 (32 mg, 0.04 mmol) and KOAc (108 mg, 1.1 mmol) in one portion at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 90 °C and stirred at 90 °C for 2 hours. After completion of reaction, the resulting mixture was cooled down to 25 °C and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 minutes; detector, UV 254 nm) to give 2-(1-methoxy-2,3-dihydro- 1H-inden-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (60 mg, 44.7% yield) as a white solid. LCMS observed m/z = 275.17 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (150 mg, 0.2 mmol), K2CO3 (66 mg, 0.5 mmol) and 2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane (68 mg, 0.2 mmol) in dioxane (2 mL ) and H2O (0.2 mL) was added Pd(dppf)Cl2 (14 mg, 0.02 mmol) in one portion at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 100 °C and stirred at 100 °C for 2 hours. After completion of reaction, the resulting mixture was cooled down to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (20/1) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-
carbonyl)piperazin-1-yl)-5-ethyl-2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (75 mg, 39.2% yield) a light yellow solid. LCMS observed m/z = 856.29 [M+H]+. Step 4: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1H-inden-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 204
Into a 8 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-[(1S)-1-methoxy-2,3-dihydro-1H-inden-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (20 mg, 0.02 mmol, 1.0 equiv.) and TFA (1 mL) dropwise at 25 °C. The reaction mixture was heated to 80 °C and stirred at 80 °C for 30 minutes. After completion of reaction, the resulting mixture was cooled down to 25 °C and concentrated under reduced pressure, collected the residue. The crude product was purified by prep-HPLC with the following conditions ((Mobile Phase A: water (10 mmol/L NH4HCO3); Mobile Phase B: ACN; Flow Rate: 60 min; Wave Length: 254 nm / 220 nm) to give N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-(3H-inden-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide (1.0 mg, 5.6% yield) as a white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.49 (s, 1H), 8.33 (s, 1H), 8.22 – 8.04 (m, 2H), 7.84 (d, J = 2.1 Hz, 1H), 7.68 – 7.45 (m, 2H), 6.97 (d, J = 5.6 Hz, 1H), 6.78 – 6.66 (m, 1H), 5.49 (s, 2H), 4.79 – 4.68 (m, 1H), 4.11 – 3.95 (m, 1H), 3.89 – 3.70 (m, 2H), 3.57 – 3.45 (m, 3H), 3.25 – 3.09 (m, 3H), 3.05 – 2.92 (m, 1H), 2.90 – 2.72 (m, 1H), 2.53 (s, 3H), 1.44 – 1.33 (m, 3H). LCMS observed m/z = 734.20 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra.
EXAMPLE 205 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(4-difluoromethylene-1- piperidyl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4- oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 205
Step 1: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5- (benzyloxy)-6-methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-2-(4-difluoromethylene-1- piperidyl)-6-ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 205.1
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6- methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-2-bromo-6-ethyl-4-oxo-4,7-dihydro- 1,3,3a,7-tetraaza-7-indenyl]acetamide, Intermediate H (0.1 g, 127 µmol, Intermediate H), 4-difluoromethylenepiperidine—hydrogen chloride (1/1) (64.5 mg, 380 µmol), and KOAc (104 mg, 760 µmol) was added DMF (375 µL) and DMSO (375 µL). The mixture was allowed to stir at 100 °C for 16 hours, then allowed to cool to room temperature and
concentrated in vacuo. The concentrated residue was diluted with EtOAc (5 mL) and water (3 mL) and the phases were partitioned. The aqueous phase was further extracted with EtOAc (2 x 5 mL). The combined organic extracts were dried over Na2SO4, filtered, and concentrated in vacuo. The crude product was purified by silica gel column chromatography (mobile phase: 0-20% MeOH in CH2Cl2) to afford N-[2-chloro-4- (trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6-methyl-4-pyrimidinyl]carbonyl}-1- piperazinyl)-2-(4-difluoromethylene-1-piperidyl)-6-ethyl-4-oxo-1,3,3a,7-tetraaza-7- indenyl]acetamide (63 mg, 47% yield) as an amber oil. LCMS observed m/z = 841.3 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(4- difluoromethylene-1-piperidyl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 205
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6- methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-2-(4-difluoromethylene-1-piperidyl)-6- ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide (51 mg, 61 µmol) in CH2Cl2 (300 µL) was added dropwise a 1 molar solution of boron trichloride in CH2Cl2 (300 µL, 300 µmol). The mixture was stirred at -40 °C under N2 for 2 hours. The mixture was quenched with MeOH (1.0 mL) and warmed to 25 °C. The residue was purified by reversed phase flash chromatography (Column: Xbridge C185 µm, 19 mm X 100 mm; mobile phase: 5-95% MeCN in H2O with 0.1% NH4OH modifier) to afford (8.2 mg, 18% yield) of N-[2-chloro- 4-(trifluoromethyl)phenyl][2-(4-difluoromethylene-1-piperidyl)-6-ethyl-5-{4-[(5-hydroxy- 6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7- indenyl]acetamide as a white solid.1H NMR (400MHz, DMSO-d6) δ 10.35 (s, 1H), 10.23 (s, 1H), 8.57 (s, 1H), 8.03 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.73 (d, J = 8.6 Hz, 1H), 5.22 (s,
2H), 4.51 (d, J = 12.4 Hz, 1H), 3.59 – 3.38 (m, 7H), 3.28 – 3.15 (m, 1H), 3.04 – 2.84 (m, 3H), 2.78 (d, J = 11.4 Hz, 1H), 2.60 (d, J = 10.8 Hz, 1H), 2.44 (s, 3H), 2.24 – 2.10 (m, 4H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. EXAMPLE 206 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(7-methyl-1,3-dihydro-2-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide
Step 1: Preparation of 5-bromo-7-methyl-3H-2-benzofuran-1-one Compound 206.1
To a stirred solution of 4-bromo-2,6-dimethylbenzoic acid (5.0 g, 21.8 mmol) and NBS (9.7 g, 54.6 mmol) in chlorobenzene (100 mL) was added benzoyl benzenecarboperoxoate (0.5 g, 2.2 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 70 ℃ for 2.5 hours. Desired product could be detected by LCMS. The reaction was quenched with 40% Na2SO3 (aq.) (150 mL) at room temperature. The aqueous layer was extracted with EA (100 mL x 2).The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was dissolved in N, N- dimethylacetamide (100 mL) in N, N-dimethylacetamide (100 mL) dropwise over 10 min at 0 ℃. The resulting mixture was stirred at room temperature for additional 1.5 hours. Desired product could be detected by LCMS. The reaction was quenched by the addition of H2O (120 mL). The resulting mixture was extracted with EA (150 mL x 3). The
combined organic layers were washed with brine (100 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford (2.6 g, 52.5% yield) of 5-bromo-7-methyl-3H-2-benzofuran-1-one as a white solid. LCMS observed m/z = 226.96 [M+H]+. Step 2: Preparation of [5-bromo-2-(hydroxymethyl)-3-methylphenyl]methanol Compound 206.2
To a stirred solution of 5-bromo-7-methyl-3H-2-benzofuran-1-one (2.5 g, 11.0 mmol) in DCM (100 mL) was added diisobutylaluminum hydride (44.0 mL, 44.0 mmol) dropwise at 0 ℃ under N2 atmosphere. The resulting mixture was stirred at room temperature for additional 2 hours. Desired product could be detected by LCMS. The reaction was quenched with saturated potassium sodium tartrate solution (150 mL) at room temperature. The resulting mixture was extracted with EA (200 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 80% EtOAc in petroleum ether) to afford (2.0 g, 78.6% yield) of [5-bromo-2-(hydroxymethyl)-3-methylphenyl] methanol as a white solid. LCMS observed m/z = 228.99 [M-H]-. Step 3: Preparation of 6-bromo-4-methyl-1,3-dihydro-2-benzofuran Compound 206.3
To a stirred solution of [5-bromo-2-(hydroxymethyl)-3-methylphenyl]methanol (2.0 g, 8.7 mmol) and TsCl (3.3 g, 17.3 mmol) in DMF (30 mL) was added NaH (60%, 0.4 g, 10.4 mmol) in portions at 0 ℃ under N2 atmosphere. The resulting mixture was stirred at room temperature for additional 4 hours. Desired product could be detected by LCMS. The reaction was quenched with H2O (50 mL), the resulting mixture was extracted with EA (60 mL x 3). The combined organic layers were washed with brine (50 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 20%
EtOAc in petroleum ether) to afford (650 mg, 35.3% yield) of 6-bromo-4-methyl-1,3- dihydro-2-benzofuran as a white solid. LCMS observed m/z = 212.98 [M+H]+. Step 4: Preparation of 4,4,5,5-tetramethyl-2-(7-methyl-1,3-dihydro-2-benzofuran-5- yl)-1,3,2-dioxaborolane Compound 206.4
To a stirred mixture of 6-bromo-4-methyl-1,3-dihydro-2-benzofuran (500 mg, 2.3 mmol) and bis(pinacolato)diboron (894 mg, 3.5 mmol) in dioxane (10 mL) were added Pd(dppf)Cl2CH2Cl2 (191 mg, 0.2 mmol) and AcOK (691 mg, 7.0 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 1 hour at 100 ℃. Desired product could be detected by LCMS. The reaction was quenched with H2O (15 mL) at room temperature. The resulting mixture was extracted with EA (20 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (290 mg, 47.5% yield) of 4,4,5,5-tetramethyl-2-(7-methyl-1,3-dihydro-2- benzofuran-5-yl)-1,3,2-dioxaborolane as a yellow solid. LCMS observed m/z = 261.12 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(7-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide Compound 206.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-
[2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (150 mg, 0.2 mmol) and 4,4,5,5-tetramethyl-2-(7-methyl-1,3-dihydro-2-benzofuran-5-yl)-1,3,2- dioxaborolane (89 mg, 0.3 mmol) in dioxane (5 mL) and H2O (1 mL) were added Pd(dppf)Cl2CH2Cl2 (15 mg, 0.1 mmol) and K2CO3 (79 mg, 0.6 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 95 ℃. Desired product could be detected by LCMS. The reaction was quenched by the addition of H2O (5 mL) at room temperature. The resulting mixture was extracted with EA (10 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in DCM) to afford (65 mg, 40.6% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(7-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 842.27 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(7-methyl-1,3-dihydro-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 206
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(6-methyl-1,3-dihydro-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.1 mmol) in TFA (1.2 mL) was stirred for 15 min at 85 ℃ under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: Water(10 mmol/ L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient:
34% B to 57% B in 8min; Wave Length: 254nm/220nm nm; RT1(min): 6.68) to afford (16.2 mg, 35.7% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(6-methyl-1,3-dihydro-2-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.35 (s, 1H), 8.56 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.85 (d, J = 3.6 Hz, 2H), 7.72 (m, 1H), 5.39 (s, 2H), 5.14 – 4.98 (m, 4H), 4.54 (m, 1H), 3.52 (m, 3H), 3.26 (s, 1H), 3.02 (d, J = 9.8 Hz, 3H), 2.84 (m, 1H), 2.67 (d, J = 8.9 Hz, 1H), 2.44 (s, 3H), 2.28 (s, 3H), 1.21 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 752.20 [M+H]+. One exchangeable proton not visible in NMR. EXAMPLE 207 Synthesis of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H- inden-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 207
Step 1: Preparation of 5-bromo-1-methoxy-2,3-dihydro-1H-indene. Compound 207.1
To a solution of 5-bromo-2,3-dihydro-1H-inden-1-ol (1.3 g, 6.2 mmol) and NaOH (371 mg, 9.3 mmol) in THF (0.5 mL) was stirred for 30 min at 0℃. To the above mixture was added MeI (1.05 g, 7.4 mmol) dropwise at 0℃. The reaction mixture was heated to 40℃ and stirred at 40℃ for 16 hours. The resulting mixture was cooled down to room temperature and filtered, the filter cake was washed with EA (2x2 mL), The filtrate was concentrated under reduced pressure. The residue was dissolved in water, extracted with EA
(2 x 10 mL) and the combined organic extracts were dried over Na2SO4, and concentrated. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10mmol/L NH4HCO3), 40% to 60% gradient in 10 min; detector, UV 254 nm. This resulted in 5-bromo-1-methoxy- 2,3-dihydro-1H-indene (1.09 g, 75% yield) as a brown liquid.1H NMR (400 MHz, DMSO- d6) δ 8.85 (t, J = 1.3 Hz, 1H), 8.34 (s, 1H), 7.95 – 7.84 (m, 4H), 7.52 – 7.45 (m, 1H), 7.38 – 7.30 (m, 1H), 3.84 (s, 3H). Step 2: Preparation of 2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 207.2
To a solution of 5-bromo-1-methoxy-2,3-dihydro-1H-indene (100 mg, 0.44 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (112 mg, 0.44 mmol) in dioxane (3 mL) were added AcOK (108 mg, 1.1 mmol) and Pd(dppf)Cl2 (32 mL, 0.04 mmol) at 25℃ under N2 atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure, collected the residue. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 220 nm. This resulted in 2-(1-methoxy-2,3- dihydro-1H-inden-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (60 mg, 44% yield) as a white solid. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H- inden-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 207.3
To a solution of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (183 mg, 0.26 mmol, Intermediate) and 2-(1-methoxy- 2,3-dihydro-1H-inden-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (72 mg, 0.26 mmol) in 1,4-dioxane/H2O (2 mL/0.2 mL) were added K2CO3 (90 mg, 0.66 mmol) and Pd(dppf)Cl2 (19 mg, 0.026 mmol) at 25℃ under N2 atmosphere. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 16 hours. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure, collected the residue. The crude productwas purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Phenyl 5 m, 19 mm * 250 mm; Mobile Phase A: Water(0.1% FAl), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 70% B to 80% B in 7min; Wave Length: 254nm/220nm; RT1(min): 6.42) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(1-methoxy-2,3-dihydro-1H- inden-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (34 mg, 17% yield) as a white solid. LCMS observed m/z = 766.20 [M+H]+. Step 4: Preparation of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro- 1H-inden-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 207
The N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (34 mg) was purified by Prep- HPLC with the following conditions (Column: CHIRAL ART Amylose-SA, 2*25 cm, 5 μm; Mobile Phase A: MeOH: DCM=1: 1--HPLC, Mobile Phase B: Hex(0.1% TFA)-- HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1(min): 8.5; RT2(min): 15.5; Sample Solvent: MeOH; Injection Volume: 0.5 mL; Number Of Runs: 2) to afford (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (8.1 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 10.25 (s, 1H), 8.59 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 8.02 (s, 1H), 8.01 – 7.95 (m, 2H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.51 (d, J = 7.8 Hz, 1H), 5.41 (s, 2H), 4.83 (dd, J = 6.6, 4.3 Hz, 1H), 4.55 (d, J = 12.5 Hz, 1H), 3.60 – 3.48 (m, 3H), 3.47 – 3.37 (m, 2H), 3.33 – 3.22 (m, 2H), 3.09 – 2.96 (m, 4H), 2.91 – 2.79 (m, 2H), 2.71 – 2.64 (m, 1H), 2.46 (s, 3H), 2.42 – 2.29 (m, 1H), 2.05 – 1.94 (m, 1H), 1.26 – 1.18 (m, 3H). LCMS observed m/z = 766.20. EXAMPLE 208 Synthesis of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H- inden-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 208
The N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (34 mg) was purified by Prep- HPLC with the following conditions (Column: CHIRAL ART Amylose-SA, 2*25 cm, 5 μm; Mobile Phase A: MeOH: DCM=1: 1--HPLC, Mobile Phase B: Hex(0.1% TFA)-- HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1(min): 8.5; RT2(min): 15.5; Sample Solvent: MEOH; Injection Volume: 0.5 mL; Number Of Runs: 2) to afford (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methoxy-2,3-dihydro-1H-inden-5-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (3.2 mg) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 2H), 8.54 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 8.02 (s, 1H), 8.01 – 7.95 (m, 2H), 7.75 – 7.70 (m, 1H), 7.51 (d, J = 7.9 Hz, 1H), 5.40 (s, 2H), 4.85 – 4.80 (m, 1H), 4.55 (d, J = 12.5 Hz, 1H), 3.59 – 3.48 (m, 3H), 3.35 – 3.34 (m, 2H), 3.31 – 3.23 (m, 2H), 3.07 – 2.96 (m, 4H), 2.89 – 2.81 (m, 2H), 2.70 – 2.65 (m, 1H), 2.44 (s, 3H), 2.39 – 2.35 (m, 1H), 2.03 – 1.96 (m, 1H), 1.22 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 766.15. EXAMPLE 209 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-3'H-spiro[azetidine-3,1'- isobenzofuran]-5'-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 209
Step 1: Preparation of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 209.1
Into a 8 mL sealed tube was added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) and BCl3 (1 M, dissolved in DCM, 2.5 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for overnight. After completion of reaction, the resulting mixture was quenched with water (5 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 2-(2-bromo-5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (70 mg, crude) as a yellow solid. LCMS observed m/z = 698.05 [M+H]+. Step 2: Preparation of 5'-bromo-3'H-spiro[azetidine-3,1'-isobenzofuran]. Compound 209.2
Into a 8 mL sealed tube was added tert-butyl 5'-bromo-3'H-spiro[azetidine-3,1'- [2]benzofuran]-1-carboxylate (200 mg, 0.6 mmol) and HCl (4 M, dissolved in 1,4-dioxane, 2 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 3 h. After completion of reaction, the mixture was concentrated under reduced pressure to afford 5'-bromo-3'H-spiro[azetidine-3,1'-isobenzofuran] (130 mg, crude) as a white solid. LCMS observed m/z = 239.99 [M+H]+. Step 3: Preparation of 5'-bromo-1-methyl-3'H-spiro[azetidine-3,1'-isobenzofuran]. Compound 209.3
To a stirred solution of 5'-bromo-3'H-spiro[azetidine-3,1'-isobenzofuran] (90 mg, 0.4 mmol) in MeOH (5 mL) was added polyformaldehyde (83 mg, 2.0 mmol) in portions at room temperature. Then the mixture was heated to 40 ℃ and stirred at 40 ℃ for 30 min. After that, the resulting mixture was allowed to cool down to room temperature, and NaBH3CN (118 mg, 2.0 mmol) was added in portions at room temperature. The resulting mixture was heated to 40 ℃ and stirred at 40 ℃ for 1 h. After completion of reaction, the resulting mixture was quenched with water (5 mL) and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, MeCN in water (10 mmol/L NH4HCO3), 70% to 80% gradient in 10 min; detector, UV 220 nm) to afford 5'-bromo-1-methyl-3'H- spiro[azetidine-3,1'-isobenzofuran] (20 mg, 21% yield) as a light yellow oil. LCMS observed m/z = 254.25 [M+H]+. Step 4: Preparation of 1-methyl-5'-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 3'H-spiro[azetidine-3,1'-isobenzofuran]. Compound 209.4
To a stirred solution of 5'-bromo-1-methyl-3'H-spiro[azetidine-3,1'-isobenzofuran] (15 mg, 0.06 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (30 mg, 0.1 mmol) in dioxane (2 mL) were added AcOK (17 mg, 0.2 mmol) and Pd(dppf)Cl2 (4 mg, 0.006 mmol) in one portion at room temperature. After that, the resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 h under nitrogen atmosphere.
After completion of reaction, the mixture was allowed to cool down to room temperature and filtered, the filter cake was washed with dioxane (3 x 2 mL). The filtrate was concentrated under reduced pressure to afford 1-methyl-5'-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3'H-spiro[azetidine-3,1'-isobenzofuran] (35 mg, crude) as a brown solid. LCMS observed m/z = 302.05 [M+H]+. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-3'H-spiro[azetidine- 3,1'-isobenzofuran]-5'-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 209
To a stirred solution of 1-methyl-5'-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 3'H-spiro[azetidine-3,1'-isobenzofuran] (30 mg, 0.1 mmol) and 2-{2-bromo-5-ethyl-6-[4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate AC (84 mg, 0.1 mmol) in dioxane (3 mL) and H2O (0.6 mL) were added Pd(dppf)Cl2 (7 mg, 0.01 mmol) and K2CO3 (41 mg, 0.3 mmol) in portions at room temperature. After that, the resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature, quenched with water (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 27% B to 41% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.92) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl- 6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-3'H- spiro[azetidine-3,1'-isobenzofuran]-5'-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-
yl)acetamide (5.1 mg, 6% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.43 (s, 1H), 8.53 (s, 1H), 8.15 (d, J = 7.9 Hz, 1H), 8.06 (d, J = 8.6 Hz, 1H), 8.03 – 7.94 (m, 2H), 7.77 (d, J = 8.0 Hz, 1H), 7.74 – 7.68 (m, 1H), 5.40 (s, 2H), 5.07 (s, 2H), 4.59 – 4.50 (m, 1H), 3.57 – 3.50 (m, 6H), 3.28 – 3.18 (m, 5H), 3.12 – 2.80 (m, 3H), 2.91 – 2.80 (m, 1H), 2.43 (s, 2H), 2.36 (s, 3H), 1.31 – 1.16 (m, 3H). LCMS observed m/z = 793.25 [M+H]+. EXAMPLE 210 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(2-{4H,5H,6H- cyclopenta[d][1,3]thiazol-2-yl}-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide Compound 210
Step 1: Preparation of 2-bromo-4H,5H,6H-cyclopenta[d][1,3]thiazole Compound 210.1
To a stirred solution of CuBr2 (3.8 g, 17.1 mmol) in ACN (40 mL) was added tert- butyl nitrite (2.4 mL, 20.0 mmol) in portions at room temperature, the resulting mixture was stirred for additional 40 min at 40 ℃. Then the 4H,5H,6H-cyclopenta[d] [1,3] thiazol-2- amine (2.0 g, 14.3 mmol) in ACN (30 mL) was added to the mixture under N2 atmosphere. The resulting mixture was stirred for additional 2 hours at 40 ℃. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 30% EtOAc in petroleum ether) to afford (1.4 g, 46.4% yield) of 2-bromo-4H,5H,6H-cyclopenta[d] [1,3] thiazole as a yellow solid. LCMS observed m/z = 203.94 [M+H]+. Step 2: Preparation of 2-(tributylstannyl)-4H,5H,6H-cyclopenta[d][1,3]thiazole Compound 210.2
To a stirred solution of 2-bromo-4H,5H,6H-cyclopenta[d][1,3]thiazole (100 mg, 0.5 mmol) in THF (5 mL) was added n-BuLi (0.2 mL, 0.5 mmol, 2.5 M in hexane) dropwise at -78 ℃ under N2 atmosphere, the resulting mixture was stirred for additional 30 min at - 78 ℃. Then to the above mixture was added Bu3SnCl (159 mg, 0.5 mmol) dropwise over 5 min at -78 ℃. The resulting mixture was stirred for additional 40 min at -78 ℃ and 16 hours at room temperature. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure, diluted with n-hexane (5 mL), the resulting mixture was filtered, the filter cake was washed with n-hexane (2 mL x 3). The filtrate was concentrated under reduced pressure to afford (220 mg, 97.6% yield) of 2-(tributylstannyl)- 4H,5H,6H-cyclopenta[d][1,3]thiazole, the crude product was used in the next step directly without further purification. LCMS observed m/z = 416.14 [M+H]+. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-{4H,5H,6H-cyclopenta[d][1,3]thiazol-2-yl}-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide Compound 210.3
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (120 mg, 0.2 mmol) and 2- (tributylstannyl)-4H,5H,6H-cyclopenta[d][1,3]thiazole (220 mg, 0.5 mmol) in DMF (1.5 mL) was added Pd(PPh3)4 (53 mg, 0.1 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred at 130 ℃ for additional 1 hour. Desired product could be detected by LCMS. The reaction was quenched with H2O (2 mL) at room temperature. The resulting mixture was extracted with EA (3 mL x 3). The combined organic layers were washed with brine (3 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was
concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 7% MeOH in DCM) to afford (50 mg, 35.5% yield) of 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-{4H,5H,6H- cyclopenta[d][1,3]thiazol-2-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 833.23 [M+H]+. Step 4: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(2-{4H,5H,6H- cyclopenta[d][1,3]thiazol-2-yl}-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide Compound 210
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-{4H,5H,6H-cyclopenta[d][1,3]thiazol-2-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.1 mmol) in TFA (1 mL) was stirred at 85 ℃ for 10 min under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 52% B to 70% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 5.47) to afford (6.8 mg, 13.9% yield) of N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-(2-{4H,5H,6H-cyclopenta[d][1,3]thiazol-2-yl}-5-ethyl-6-[4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetamide as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 8.50 (s, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.0 Hz, 1H), 7.75 – 7.68 (m, 1H), 5.39 (s, 2H), 4.54 (m, 1H), 3.50 (m, 3H), 3.29 – 3.21 (m, 2H), 3.18 – 2.94 (m, 5H), 2.93 – 2.72 (m, 4H), 2.68 (p, J = 1.9 Hz, 1H), 2.42 (s, 2H), 2.37 – 2.31 (m, 1H), 1.22 (m, 3H). LCMS observed m/z = 743.35 [M+H]+. One exchangeable proton not visible in NMR.
EXAMPLE 211 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-1,1- dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]ace tamide. Compound 211
Step 1: Preparation of 5-bromo-6-fluoro-3H-2-benzofuran-1-one. Compound 211.1
To a stirred solution of 4-bromo-5-fluoro-2-methylbenzoic acid (2.0 g, 8.5 mmol, 1.0 equiv.) and sodium bromate (3.9 g, 25.7 mmol, 3.0 equiv.) in EA (25 mL) and H2O (20 mL) was added NaHSO3 (2.7 g, 25.7 mmol, 3.0 equiv.) in portions at 0 °C. The resulting mixture was stirred for additional 12 h at room temperature under nitrogen atmosphere. After completion of the reaction, the resulting mixture was quenched with H2O (10 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 5-bromo-6-fluoro-3H- 2-benzofuran-1-one (1.2 g, 60% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.13 (d, J = 5.9 Hz, 1H), 7.88 (d, J = 7.4 Hz, 1H), 5.39 (d, J = 1.3 Hz, 2H). Step 2: Preparation of 2-[4-bromo-5-fluoro-2-(hydroxymethyl)phenyl]propan-2-ol. Compound 211.2
To a stirred solution of 5-bromo-6-fluoro-3H-2-benzofuran-1-one (1.2 g, 5.2 mmol, 1.0 equiv.) in THF (10 mL) was added MeMgBr (1.9 g, 15.6 mmol, 3.0 equiv.) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for additional 12 h at room temperature under nitrogen atmosphere. After completion of the reaction, the reaction was quenched with H2O (5 mL) and extracted with EtOAc (3 x 40 mL). The combined organic layers were washed with brine (40 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 2-[4-bromo-5-fluoro-2-(hydroxymethyl)phenyl]propan-2-ol (725 mg, 53% yield) as a yellow oil. LCMS observed m/z = 263.00[M+H]+. Step 3: Preparation of 5-bromo-6-fluoro-1,1-dimethyl-3H-2-benzofuran. Compound 211.3
To a stirred solution of 2-[4-bromo-5-fluoro-2-(hydroxymethyl)phenyl]propan-2-ol (720 mg, 2.7 mmol, 1.0 equiv.) in toluene (5 mL) was added H3PO4 (4.0 g, 41.0 mmol, 15.0 equiv.) dropwise at 25 °C. The resulting mixture was heated to 80 °C and stirred for additional 3 h at 80 °C under air atmosphere. After completion of the reaction, the resulting mixture was cooled to room temperature, quenched with H2O (5 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 5-bromo-6-fluoro-1,1-dimethyl- 3H-2-benzofuran (368 mg, 54 % yield) as a yellow solid. LCMS observed m/z = 244.99[M+H]+. Step 4: Preparation of 2-(6-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 211.4
To a stirred solution of 5-bromo-6-fluoro-1,1-dimethyl-3H-2-benzofuran (70 mg, 0.2 mmol, 1.0 equiv.) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-1,3,2-dioxaborolane (108 mg, 0.4 mmol, 2.0 equiv.) in dioxane (1 mL) were added AcOK (84 mg, 0.8 mmol, 4.0 equiv.) tricyclohexylphosphane (16 mg, 0.06 mmol, 0.3 equiv.) and Pd(dba)2 (16 mg, 0.02 mmol, 0.1 equiv.) in portions at 25 °C under nitrogen atmosphere. The reaction solution was heated to 100 °C and stirred for additional 2 h at 100 °C under nitrogen atmosphere. After completion of the reaction, the resulting mixture was cooled to room temperature, quenched with H2O (10 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 293.16[M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(6-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl] acetamide. Compound 211.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (120 mg, 0.1 mmol, 1.0 equiv.) and 2-(6-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (66 mg, 0.2 mmol, 2.0 equiv.) in dioxane (1 mL) were added K2CO3 (63 mg, 0.4 mmol, 4.0 equiv.) and Pd(dppf)Cl2CH2Cl2 (12 mg, 0.01 mmol, 0.1 equiv.) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 60 °C and stirred for additional 2 h at 60 °C under nitrogen atmosphere. After completion of the reaction, the
resulting mixture was cooled to room temperature, quenched with H2O (5 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(6-fluoro-1,1-dimethyl-3H-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]aceta mide (65 mg, 48% yield) as a white solid. LCMS observed m/z = 874.27[M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6- fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]ace tamide. Compound 211
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(6-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (60 mg, 0.07 mmol, 1.0 equiv.) in TFA (1 mL) was heated to 80 °C for 0.5 h under air atmosphere. After completion of the reaction, the resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C185 μm, 30*150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 50% B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 6.08) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-1,1- dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)pipera zin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (24.4 mg, 44% yield) as a
white solid.1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H),δ 8.38 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 8.0 -7.86 (m, 2H), 7.71 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 10.3 Hz, 1H), 5.38 (s, 2H), 4.99 (s, 2H), 4.54 (m, 1H), 3.51 (m, 3H), 3.11 – 2.93 (m, 3H), 2.83 (m, 1H), 2.72 – 2.63 (m, 1H), 2.43 – 2.31 (m, 2H), 1.46 (s, 6H), 1.30 – 1.14 (m, 5H). LCMS observed m/z = 784.45[M+H]+. Note: One exchangeable proton not visible in NMR. EXAMPLE 212 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl]-2-[5-ethyl-2-(7-fluoro-1,1- dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide. Compound 212
Step 1: Preparation of 5-bromo-7-fluoro-3H-2-benzofuran-1-one. Compound 212.1
A solution of 4-bromo-2-fluoro-6-methylbenzoic acid (3.0 g, 12.9 mmol) in EA (50 mL) and H2O (40 mL) was treated with sodium bromate (5.8 g, 38.6 mmol) and NaHSO3 (4.0 g, 38.6 mmol) in one portion at 25 °C. The reaction mixture was stirred at 25 °C for 24 hours under nitrogen atmosphere. Then the resulting mixture was extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (1.3 g, 43% yield) of 5-bromo-7-fluoro-3H-2-benzofuran-1- one as a white solid. Step 2: Preparation of 2-[4-bromo-2-fluoro-6-(hydroxymethyl) phenyl] propan-2-ol. Compound 212.2
A solution of 5-bromo-7-fluoro-3H-2-benzofuran-1-one (700 mg, 3.0 mmol) in THF (10 mL) was treated with methyl magnesium bromide (7.6 mL, 7.6 mmol, 1.0 M in THF) dropwise at 0 ℃. The reaction mixture was stirred at 0 ℃ for 4 hours under nitrogen atmosphere. The reaction was quenched with H2O (10 mL). Then the resulting mixture was extracted with EA (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the mixture was concentrated in vacuo to afford (700 mg, crude) of 2-[4-bromo-2-fluoro-6-(hydroxymethyl) phenyl] propan-2-ol as a brown yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 263.05 [M-H]-. Step 3: Preparation of 5-bromo-7-fluoro-1,1-dimethyl-3H-2-benzofuran. Compound 212.3
A solution of 2-[4-bromo-2-fluoro-6-(hydroxymethyl) phenyl] propan-2-ol (700 mg, 2.7 mmol) in toluene (15 mL) was treated with H3PO4 (6.1 g, 53.2 mmol, 85% in water) in one portion at 25 °C. The reaction mixture was heated to 80 ℃ and stirred at 80 ℃ for 3 hours under N2 atmosphere. The resulting mixture was diluted with H2O (20 mL). The mixture was allowed to cool down to room temperature and extracted with EA (3 x 20 mL). The combined organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 5% EtOAc in petroleum ether) to afford (330 mg, 50% yield) of 5-bromo-7-fluoro-1,1-dimethyl-3H-2-benzofuran as a brown yellow oil. Step 4: Preparation of 2-(7-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 212.4
To a stirred solution of 5-bromo-7-fluoro-1,1-dimethyl-3H-2-benzofuran (150 mg, 0.6 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (466 mg, 1.8 mmol) in dioxane (5 mL) were added potassium acetate (180 mg, 1.8 mmol) and Pd(dppf)Cl2.CH2Cl2 (49 mg, 0.06 mmol) dropwise at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 100 °C for additional 2 hours. Then the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (80 mg, 44.7% yield) of 2-(7-fluoro- 1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolaneas a white solid. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-5-ethyl-2-(7-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl] acetamide. Compound 212.5
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (162 mg, 0.2 mmol) and 2-(7-fluoro- 1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (60 mg, 0.2 mmol) in dioxane (3 mL) and H2O (1 mL) was treated with K2CO3 (85 mg, 0.6 mmol) and Pd(dppf)Cl2.CH2Cl2 (15 mg, 0.02 mmol) in one portion at 25 ℃. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 2 hours under N2 atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (130 mg, 72% yield) of 2- (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-5-ethyl-2-(7-fluoro- 1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl)-N-[2-
chloro-4-(trifluoromethyl) phenyl] acetamide as a white solid. LCMS observed m/z = 874.20 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl) phenyl]-2-[5-ethyl-2-(7- fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide. Compound 212
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1- yl}-5-ethyl-2-(7-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl] acetamide (125 mg, 0.1 mmol) in TFA (3 mL) was stirred at 80 ℃ for 20 min under N2 atmosphere. The resulting mixture was concentrated under vacuum and purified by preparatory HPLC (Column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 35-50% ACN in H2O) to afford (31 mg, 27% yield) of N-[2-chloro-4-(trifluoromethyl) phenyl]-2- [5-ethyl-2-(7-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4- yl] acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 2H), 8.57 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.2 Hz, 1H), 7.87 (s, 1H), 7.78 – 7.70 (m, 2H), 5.40 (s, 2H), 5.09 (s, 2H), 4.54 (d, 1H), 3.52 (d, 3H), 3.03 (d, 3H), 2.85 (d, 1H), 2.69 (s, 1H), 2.45 (s, 3H), 1.54 (s, 6H), 1.22 (t, 3H). LCMS observed m/z = 784.20 [M+H]+. Note: One exchangeable proton not visible in NMR. EXAMPLE 213 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-(1,1,6-trimethyl-3H-2-benzofuran- 5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 213
Step 1: Preparation of 5-bromo-6-methyl-3H-2-benzofuran-1-one. Compound 213.1
A solution of 4-bromo-3-methylbenzoic acid (2.3 g, 10.6 mmol) in dibromomethane (50 mL) was treated with K2HPO4 (5.5 g, 31.9 mmol) at room temperature under nitrogen atmosphere followed by the addition of Pd(AcO)2 (240 mg, 1.1 mmol) in portions at room temperature. The resulting mixture was stirred for 48 h at 140 °C under nitrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with CH2Cl2 (3 x 50 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10 % EtOAc in petroleum ether) to afford (1.2 g, 49% yield) of 5-bromo-6-methyl-3H-2-benzofuran-1-one as a white solid. Step 2: Preparation of 2-[4-bromo-2-(hydroxymethyl)-5-methylphenyl]propan-2-ol. Compound 213.2
A solution of 5-bromo-6-methyl-3H-2-benzofuran-1-one (1.1 g, 4.8 mmol) in THF (20 mL) was treated with MeMgBr (14.5 mL, 14.5 mmol, 1.0 M in THF) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The reaction was quenched with sat. NH4Cl (aq.) (50 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (700 mg, 55% yield) of 2-[4-bromo-2-(hydroxymethyl)-5- methylphenyl]propan-2-ol as a white solid. LCMS observed m/z = 223.20 [M+H]+.
Step 3: Preparation of 5-bromo-1,1,6-trimethyl-3H-2-benzofuran. Compound 213.3
A solution of 2-[4-bromo-2-(hydroxymethyl)-5-methylphenyl]propan-2-ol (400 mg, 1.5 mmol) and H3PO4 (1.6 mL, 28.8 mmol) in toluene (4 mL) was stirred for 3 h at 80 °C under nitrogen atmosphere. The reaction was quenched with water (50 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (270 mg, 72% yield) of 5-bromo-1,1,6-trimethyl-3H-2- benzofuran as a yellow solid. Step 4: Preparation of 4,4,5,5-tetramethyl-2-(1,1,6-trimethyl-3H-2-benzofuran-5- yl)-1,3,2-dioxaborolane. Compound 213.4
To a stirred solution of 5-bromo-1,1,6-trimethyl-3H-2-benzofuran (100 mg, 0.4 mmol) and Pd(dppf)Cl2CH2Cl2 (30 mg, 0.04 mmol) in 1,4-dioxane were added AcOK (122 mg, 1.2 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3,2-dioxaborolane (116 mg, 0.4 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 90 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 2% CH2Cl2 in petroleum ether) to afford (150 mg, crude) of 4,4,5,5-tetramethyl-2-(1,1,6-trimethyl-3H-2-benzofuran-5-yl)-1,3,2- dioxaborolane as a colorless oil. LCMS observed m/z = 289.20 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-(1,1,6-trimethyl-3H-2-benzofuran-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 213.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide,Intermediate H (100 mg, 0.1 mmol) and 4,4,5,5-tetramethyl-2-(1,1,6-trimethyl-3H-2-benzofuran-5-yl)-1,3,2-dioxaborolane (37 mg, 0.1 mmol) in dioxane (0.6 mL) and H2O (0.1 mL) were added Pd(dppf)Cl2CH2Cl2 (9 mg, 0.01 mmol) and K2CO3 (53 mg, 0.4 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 60 °C under nitrogen atmosphere. The reaction was quenched with water (50 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (110 mg, 99% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-(1,1,6-trimethyl-3H-2- benzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 870.30 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-(1,1,6-trimethyl-3H-2- benzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 213
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-(1,1,6-trimethyl-3H-2-benzofuran-5-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in TFA (3 mL) was stirred at 80 °C for 15 min. The reaction mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 38% B to 61% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 6.54) to afford (22.9 mg, 24% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-(1,1,6-trimethyl-3H-2-benzofuran- 5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid.1H NMR (400 MHz, Chloroform- d) δ 11.90 (s, 1H), 9.05 (s, 1H), 8.63 (s, 1H), 8.51 (d, J = 8.6 Hz, 1H), 7.94 (s, 1H), 7.62 (d, J = 2.0 Hz, 1H), 7.58 – 7.52 (m, 1H), 7.06 (s, 1H), 5.76 – 5.62 (m, 1H), 5.19 (s, 2H), 5.09 (s, 2H), 4.83 (m, 1H), 3.88 (s, 2H), 3.54 (s, 1H), 3.24 (d, J = 7.8 Hz, 2H), 3.12 (s, 1H), 2.86 (m, 2H), 2.74 (s, 3H), 2.62 (s, 3H), 1.54 (s, 6H), 1.44 – 1.37 (m, 3H). LCMS observed m/z = 780.30 [M+H]+. EXAMPLE 214 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,4-trimethyl-1,3- dihydroisobenzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 214
Step 1: 2-(4-bromo-2-(hydroxymethyl)-3-methylphenyl)propan-2-ol. Compound 214.1
To a stirred solution of 5-bromo-4-methylisobenzofuran-1(3H)-one (100 mg, 0.4 mmol) in THF (2 mL) was added MeMgBr (1 mL, 1.3 mmol) dropwise at 0 °C under nitrogen atmosphere. The reaction mixture was warmed to 25 °C and stirred at 25 °C for 3 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 0 °C, quenched with sat. NH4Cl (aq., 10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-(4-bromo-2- (hydroxymethyl)-3-methylphenyl)propan-2-ol (100 mg, crude) as a light yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 256.90 [M-H]-. Step 2: Preparation of 5-bromo-1,1,4-trimethyl-1,3-dihydroisobenzofuran. Compound 214.2
To a stirred solution of 2-(4-bromo-2-(hydroxymethyl)-3-methylphenyl)propan-2- ol (450 mg, 1.7 mmol) in toluene (1 mL) was added H3PO4 (2 mL, 34.7 mmol) dropwise at 25 °C. The reaction mixture was heated to 90 °C and stirred at 90 °C for 3 hours. The reaction mixture was cooled to 25 °C and neutralized to pH 7 with NaOH (aq. 1 N). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 5-bromo-1,1,4-trimethyl-1,3- dihydroisobenzofuran (350 mg, crude) as a yellow oil. Step 3: Preparation of 4,4,5,5-tetramethyl-2-(1,1,4-trimethyl-1,3- dihydroisobenzofuran-5-yl)-1,3,2-dioxaborolane. Compound 214.3
To a stirred solution of 5-bromo-1,1,4-trimethyl-1,3-dihydroisobenzofuran (150 mg, 0.6 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (474 mg, 1.8 mmol) in dioxane (3 mL) were added AcOK (183 mg, 1.8 mmol) and Pd(dppf)Cl2 (50 mg, 0.06 mmol) in portions at 25 °C under nitrogen atmosphere.
The reaction mixture was heated to 100 °C and stirred at 100 °C for 16 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C. After filtration, the filter cake was washed with EtOAc (3x10 mL) and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (40:1) to afford 4,4,5,5-tetramethyl-2-(1,1,4-trimethyl-1,3-dihydroisobenzofuran-5- yl)-1,3,2-dioxaborolane (140 mg, 78% yield) as a white solid. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-2-(1,1,4-trimethyl-1,3-dihydroisobenzofuran-5- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 214.4
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide, Intermediate H (80 mg, 0.1 mmol) and 4,4,5,5-tetramethyl-2-(1,1,4-trimethyl-1,3-dihydroisobenzofuran-5-yl)-1,3,2- dioxaborolane (35 mg, 0.1 mmol) in dioxane (4 mL) and H2O (0.5 mL) were added Pd(dppf)Cl2 (7 mg, 0.01 mmol) and K2CO3 (28 mg, 0.2 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 100 °C and stirred at 100 °C for 2 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C. After filtration, the filter cake was washed with EtOAc (3 x 10 mL) and the filtrate was concentrated under reduced pressure. This resulted in 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-2-(1,1,4-trimethyl-1,3- dihydroisobenzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (100 mg, crude) as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 870.40 [M+H]+.
Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,4-trimethyl-1,3- dihydroisobenzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 214
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-(1,1,4-trimethyl-3H-2-benzofuran-5-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (70 mg, 0.08 mmol) in TFA (1 mL) was heated to 80 °C and stirred at 80 °C for 30 minutes under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Mobile Phase A: 0.1% FA in water; Mobile Phase B: ACN; Flow rate: 60 mL/min) to afford N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,4-trimethyl-1,3-dihydroisobenzofuran-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (30.9 mg, 49% yield) as an off-white solid.1H NMR (400 MHz, Methanol-d4) δ 8.60 (s, 1H), 8.17 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 7.9 Hz, 1H), 7.83 (d, J = 2.1 Hz, 1H), 7.63 (dd, J = 8.8, 2.1 Hz, 1H), 7.13 (d, J = 7.8 Hz, 1H), 5.47 (s, 2H), 5.11 (s, 2H), 4.74 (m, 1H), 4.15 (m, 1H), 3.90 – 3.74 (m, 2H), 3.48 (m, 1H), 3.17 (q, J = 7.7 Hz, 3H), 2.99 (m, 1H), 2.84 (m, 1H), 2.54 (m, 6H), 1.51 (s, 6H), 1.36 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 780.40 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 215 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{3H-spiro[2-benzofuran-1,1'- cyclobutan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 215
Step 1: Preparation of 2-[(2-bromo-5-chlorophenyl)methoxy]oxane. Compound 215.1
To a stirred solution of (2-bromo-5-chlorophenyl)methanol (2.0 g, 9.0 mmol) and TsOH (155 mg, 0.9 mmol) in DCM (1 mL) was added dihydropyran (988 uL, 10.8 mmol) at room temperature. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (2.4 g, 86% yield) of 2-[(2-bromo-5- chlorophenyl)methoxy]oxane as a colorless oil. Step 2: Preparation of 1-{4-chloro-2-[(oxan-2-yloxy)methyl]phenyl}cyclobutan-1- ol. Compound 215.2
A solution of 2-[(2-bromo-5-chlorophenyl)methoxy]oxane (2.0 g, 6.5 mmol) in THF (20 mL) was treated with n-BuLi (3.9 mL, 9.8 mmol, 2.5 M in n-hexane) for 1 h at -78 °C under nitrogen atmosphere followed by the addition of cyclobutanone (586 uL, 7.9 mmol) dropwise at -78 °C. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The reaction was quenched with sat. Na2CO3 (aq.) (50 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 50 mL), dried over
anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (1.3 g, 66% yield) of 1-{4-chloro-2-[(oxan-2- yloxy)methyl]phenyl}cyclobutan-1-ol as a white solid. LCMS observed m/z = 295.05 [M- H]-. Step 3: Preparation of 1-[4-chloro-2-(hydroxymethyl)phenyl]cyclobutan-1-ol. Compound 215.3
A solution of 1-{4-chloro-2-[(oxan-2-yloxy)methyl]phenyl}cyclobutan-1-ol (1.5 g, 4.9 mmol) in MeOH (0.4 mL) and DCM (20 mL) was treated with TsOH (841 mg, 4.9 mmol) for 2 h at 5 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 10% MeOH in DCM) to afford (0.9 g, 86% yield) of 1-[4-chloro-2-(hydroxymethyl)phenyl]cyclobutan-1- ol as a white solid. LCMS observed m/z = 211.05 [M-H]-. Step 4: Preparation of 5-chloro-3H-spiro[2-benzofuran-1,1'-cyclobutane]. Compound 215.4
A solution of 1-[4-chloro-2-(hydroxymethyl)phenyl]cyclobutan-1-ol (500 mg, 2.3 mmol) in DMF (10 mL) was treated with TsCl (537 mg, 2.8 mmol) for 2 min at 0 °C under nitrogen atmosphere followed by the addition of NaH (235 mg, 5.9 mmol, 60%) at 0 °C. The resulting mixture was stirred overnight at room temperature. The reaction was quenched by the addition of water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (78 mg, 17% yield) of 5-chloro-3H-spiro[2-benzofuran- 1,1'-cyclobutane] as a colorless oil.
Step 5: Preparation of 4,4,5,5-tetramethyl-2-{3H-spiro[2-benzofuran-1,1'- cyclobutan]-5-yl}-1,3,2-dioxaborolane. Compound 215.5
To a stirred solution of 5-chloro-3H-spiro[2-benzofuran-1,1'-cyclobutane] (170 mg, 0.9 mmol) and Pd(dba)2 (50 mg, 0.08 mmol) in dioxane (4 mL) were added AcOK (171 mg, 1.7 mmol), tricyclohexylphosphane (3 mg, 0.01 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (266 mg, 1.0 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 100 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (180 mg, 72% yield) of 4,4,5,5-tetramethyl-2-{3H- spiro[2-benzofuran-1,1'-cyclobutan]-5-yl}-1,3,2-dioxaborolane as a yellow solid. LCMS observed m/z = 328.20 [M+H]+. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{3H-spiro[2-benzofuran-1,1'-cyclobutan]-5-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 215.6
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (160 mg, 0.2 mmol) and 4,4,5,5-tetramethyl-2-{3H-spiro[2-benzofuran-1,1'-cyclobutan]-5-yl}-1,3,2-dioxaborolane (87 mg, 0.3 mmol) in dioxane (1 mL) were added Pd(dppf)Cl2CH2Cl2 (15 mg, 0.02 mmol), H2O (0.4 mL) and K2CO3 (84 mg, 0.6 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 60 °C under nitrogen atmosphere.
The reaction mixture was purified by reversed phase C18 silica gel column chromatography to afford (120 mg, 68% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{3H-spiro[2-benzofuran-1,1'-cyclobutan]-5-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 868.40 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{3H-spiro[2- benzofuran-1,1'-cyclobutan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 215
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-{3H-spiro[2-benzofuran-1,1'-cyclobutan]-5-yl}-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (140 mg, 0.1 mmol) in TFA (10 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Shield RP18, 5μm, 19*250mm; Mobile Phase A: water (10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 34% B to 54%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.95) to afford (49.8 mg, 39% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]- 2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{3H- spiro[2-benzofuran-1,1'-cyclobutan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid.1H NMR (400 MHz, Chloroform- d) δ 12.05 (s, 1H), 9.11 (s, 1H), 8.65 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 8.28 (d, J = 7.9 Hz, 1H), 8.13 (s, 1H), 7.62 (s, 1H), 7.56 (dd, J = 8.7, 4.3 Hz, 2H), 5.68 (s, 1H), 5.21 (s, 2H), 5.12 (s, 2H), 4.87 – 4.78 (m, 1H), 3.87 (m, 2H), 3.55 (s, 1H), 3.25 (d, J = 8.1 Hz, 2H), 3.12 (s, 1H), 2.86 (m, 2H), 2.70 (m, 1H), 2.66 (s, 4H), 2.46 (m, 2H), 2.04 – 1.96 (m, 2H), 1.39 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 778.35 [M+H]+.
EXAMPLE 216 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4-fluoro-1,1- dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 216
Step 1: Preparation of 4-bromo-2-(bromomethyl)-3-fluorobenzoic acid. Compound 216.1
To a stirred solution of 4-bromo-3-fluoro-2-methylbenzoic acid (1 g, 4.3 mmol) and sodium bromate (1.9 g, 12.9 mmol) in EA (12.5 mL) and H2O (10 mL) was added NaHSO3 (1.3 g, 12.9 mmol.) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 16 h. After completion of reaction, the reaction was quenched by the addition of sat. sodium hyposulfite (aq.) (10 mL) at room temperature. The aqueous layer was extracted with EA (3x10 mL). The combined organic phases were washed with brine (10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product (1.2 g, crude) of 4-bromo-2-(bromomethyl)-3- fluorobenzoic acid as a white solid. LCMS observed m/z = 210.86 [M-H]-. Step 2: Preparation of 4-bromo-3-fluoro-2-(hydroxymethyl)benzoic acid. Compound 216.2
To a stirred solution of 4-bromo-2-(bromomethyl)-3-fluorobenzoic acid (1.2 g, 3.8 mmol) in dioxane (30 mL) was added dropwise a solution of 6% Na2CO3 (aq.) (10 mL) at 25 °C. The resulting mixture was heated to 110 °C and stirred at 110 °C for 3 h. After completion of reaction, the reaction mixture was acidified to pH 3 with HCl (aq.). The aqueous layer was extracted with EA (3 x 20 mL). The combined organic phases were washed with brine (3 x10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford (800 mg, 83% yield) of 4-bromo-3-fluoro-2- (hydroxymethyl)benzoic acid as a yellow solid. LCMS observed m/z = 248.95 [M+H]+. Step 3: Preparation of 5-bromo-4-fluoro-3H-2-benzofuran-1-one. Compound 216.3
To a stirred solution of 4-bromo-3-fluoro-2-(hydroxymethyl)benzoic acid (1.2 g, 4.8 mmol) in DCM (20 mL) was added TFA (10 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to afford (1.1 g, crude) of 5- bromo-4-fluoro-3H-2-benzofuran-1-one as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 230.94 [M+H]+. Step 4: Preparation of 2-[4-bromo-3-fluoro-2-(hydroxymethyl)phenyl]propan-2-ol. Compound 216.4
To a stirred solution of 5-bromo-4-fluoro-3H-2-benzofuran-1-one (500 mg, 2.2 mmol) in THF (10 mL) was added MeMgBr (2.2 mL, 6.5 mmol, 3M in 2-MeTHF) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 12 h. After completion of reaction, the reaction mixture was quenched by addition of water (10 mL). The aqueous layer was extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford (500 mg, 87% yield)
of 2-[4-bromo-3-fluoro-2-(hydroxymethyl)phenyl]propan-2-ol as a yellow solid. LCMS observed m/z = 263.01 [M+H]+. Step 5: Preparation of 5-bromo-4-fluoro-1,1-dimethyl-3H-2-benzofuran. Compound 216.5
To a stirred solution of 2-[4-bromo-3-fluoro-2-(hydroxymethyl)phenyl]propan-2-ol (320 mg, 1.2 mmol) in toluene (3 mL) was added H3PO4 (1.8 g, 18.2 mmol) dropwise at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 3 h. After completion of reaction, the reaction mixture was basified to pH 8 with NaOH(aq.). The aqueous layer was extracted with EA (3 x 5 mL). The combined organic phases were washed with brine (3 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by silica gel column chromatography (eluted: with 20% EA in PE) to afford (200 mg, 67% yield) of 5-bromo-4- fluoro-1,1-dimethyl-3H-2-benzofuranas a colorless oil. LCMS observed m/z = 244.99 [M+H] +. Step 6: Preparation of 2-(4-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 216.6
To a stirred solution of 5-bromo-4-fluoro-1,1-dimethyl-3H-2-benzofuran (150 mg, 0.6 mmol) in dioxane (3 mL) were added 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3,2-dioxaborolane (310 mg, 1.2 mmol), AcOK (180 mg, 1.8 mmol), Pd(dba)2 (35 mg, 0.06 mmol) and tricyclohexylphosphane (34 mg, 0.1 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h. After completion of reaction, the reaction mixture was cooled to temperature and used in the next step directly without further purification. LCMS observed m/z = 293.16 [M+H] +.
Step 7: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 216.7
To a stirred solution of 2-(4-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (111 mg, 0.4 mmol) in dioxane (10 mL) were added 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (150 mg, 0.2 mmol), AcOK (151 mg, 1.5 mmol), Pd(dppf)Cl2 (42 mg, 0.05 mmol) and H2O (2 mL) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h. After completion of reaction, the resulting mixture was extracted with EA (3 x 5 mL). The combined organic layers were washed with brine (5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography (Column, C18 silica gel; mobile phase: 0- 50% MeCN in Water (0.1% FA)) to afford (80 mg, 48% yield) of 2-(6-{4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-1,1-dimethyl-3H-2- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a brown solid. LCMS observed m/z = 874.28 [M+H]+. Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(4- fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 216
Into a 8 mL sealed tube were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (80 mg, 0.09 mmol) and TFA (2 mL) at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h. After completion of reaction, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC (Column: Xselect CSH C18 5 m, 19 mm * 250 mm; mobile phase: 25-41% ACN in Water(0.1% FA)) to afford (13.1 mg, 12% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[5-ethyl-2-(4-fluoro-1,1-dimethyl-3H-2-benzofuran-5-yl)-6-[4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.59 (s, 1H), 8.18 (d, J = 8.5 Hz, 1H), 8.13 – 8.08 (m, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.63 (dd, J = 8.5, 2.1 Hz, 1H), 7.16 (d, J = 7.9 Hz, 1H), 5.48 (s, 2H), 5.17 (s, 2H), 4.74 (d, J = 11.0 Hz, 1H), 4.13 (m, 1H), 3.85 – 3.74 (m, 2H), 3.54 – 3.46 (m, 1H), 3.21 – 3.11 (m, 3H), 2.99 (d, J = 11.7 Hz, 1H), 2.84 (d, J = 11.8 Hz, 1H), 2.55 (s, 3H), 1.54 (s, 6H), 1.36 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 788.40 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 217 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(5-fluoro-3,4- dihydro-1H-2-benzopyran-6-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 217
Step 1: Preparation of 1-bromo-2-fluoro-3-[(E)-2-methoxyethenyl]benzene. Compound 217.1
To a stirred solution of 3-bromo-2-fluorobenzaldehyde (2.0 g, 24.6 mmol) and (methoxymethyl)triphenylphosphanium chloride (12.7 g, 36.9 mmol) in THF (50 mL) was added KHMDS (39.4 mL, 39.4 mmol, 1.0 M in THF) in portions at 0 ℃ under nitrogen atmosphere. The resulting mixture was stirred for 1.5 h at 25 ℃ under nitrogen atmosphere. The reaction was quenched with sat. sodium hyposulfite (aq.) (60 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 60 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (1.3 g, 65% yield) of 1-bromo-2-fluoro-3-[(E)-2-methoxyethenyl]benzene as a light yellow oil. LCMS observed m/z =230.97 [M+H]+. Step 2: Preparation of 2-(3-bromo-2-fluorophenyl)ethanol. Compound 217.2
A solution of 1-bromo-2-fluoro-3-[(E)-2-methoxyethenyl]benzene (1.0 g, 0.4 mmol) in HCl (3 mL) and THF (3 mL) was stirred at 25 ℃ for 30 min. The mixture was basified to pH 6 with saturated Na2CO3 (aq.). The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. The mixture was concentrated in vacuo to give a residue (890 mg, crude). To the residue in MeOH (10 mL) were added NaBH4 (620 mg, 16.4 mmol) in portions
at 0 ℃. Then the resulting mixture was stirred at 25 ℃ for 1h under nitrogen atmosphere. The reaction was quenched with water (15 mL) at room temperature. The resulting mixture was extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 25% EtOAc in petroleum ether) to afford (251 mg, 28% yield) of 2-(3-bromo-2- fluorophenyl)ethanol as a yellow oil. LCMS observed m/z =218.97 [M+H]+. Step 3: Preparation of 1-bromo-2-fluoro-3-{2-[(2- methoxyethoxy)methoxy]ethyl}benzene. Compound 217.3
To a stirred mixture of 2-(3-bromo-2-fluorophenyl)ethanol (250 mg, 1.1 mmol) and 1-(chloromethoxy)-2-methoxyethane (213 mg, 1.7 mmol) in DCM (5 mL) were added DIEA (221 mg, 1.7 mmol) in portions at 25 ℃. Then the resulting mixture was stirred at 25 ℃ for 3 h under nitrogen atmosphere. The reaction was quenched with HCl (10 mL, 1.0 M in H2O) at room temperature. The resulting mixture was extracted with CH2Cl2 (3 x 10mL). The combined organic layers were dried over anhydrous Na2SO4. The crude product was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (150 mg, 69% yield) of 1-bromo-2-fluoro-3-{2-[(2- methoxyethoxy)methoxy]ethyl}benzene as a light yellow oil. LCMS observed m/z =307.03 [M+H]+. Step 4: Preparation of 6-bromo-5-fluoro-3,4-dihydro-1H-2-benzopyran. Compound 217.4
To a stirred solution of 1-bromo-2-fluoro-3-{2-[(2- methoxyethoxy)methoxy]ethyl}benzene (150 mg, 0.5 mmol) in DCM (2 mL) was added TiCl4 (93 mg, 0.5 mmol) in portions at 0 ℃ under nitrogen atmosphere. The resulting mixture was stirred for 1.5 h at 0 ℃ under nitrogen atmosphere. The reaction was quenched by the addition of brine (5 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4.
After concentration the crude product was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (100 mg, 56% yield) of 6-bromo-5- fluoro-3,4-dihydro-1H-2-benzopyran as a light yellow oil. LCMS observed m/z =230.97 [M+H]+. Step 5: Preparation of 2-(5-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 217.5
To a stirred solution of 6-bromo-5-fluoro-3,4-dihydro-1H-2-benzopyran (100 mg, 0.4 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (219 mg, 0.9 mmol) in dioxane (1 mL) was added Pd(dppf)Cl2CH2Cl2 (73 mg, 0.09 mmol) and AcOK (127 mg, 1.3 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 100 °C under nitrogen atmosphere. The crude product was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (78 mg, 64% yield) of 2-(5-fluoro-3,4-dihydro-1H-2- benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane as a light yellow solid. LCMS observed m/z =279.13 [M+H]+. Step 6: Preparation of 2-(5-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 217.6
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- (5-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (70 mg, 0.3 mmol) in dioxane (1 mL) /H2O (100 uL) was added Pd(dppf)Cl2CH2Cl2 (8 mg,
0.01mmol) and K2CO3 (53 mg, 0.4 mmol) in portions at 25 ℃ under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 100 ℃ under nitrogen atmosphere. The reaction mixture was purified by reversed phase C18 silica gel column chromatography to afford (51 mg, 46% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(5-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a brown solid. LCMS observed m/z =860.26 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(5- fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 217
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(5-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (40 mg, 0.05 mmol) in TFA (1 mL) was stirred for 15 h at 25 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 16% B to 39% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 4.93) to afford (8.6 mg, 22% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(5- fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.91 (s, 1H), 9.16 (s, 1H), 8.63 (s, 1H), 8.49 (d, J = 8.7 Hz, 1H), 8.00 (t, J = 7.6 Hz, 1H), 7.61 (d, J = 2.1 Hz, 1H), 7.56 – 7.52 (m, 1H), 6.92 (d, J = 8.0 Hz, 1H), 5.70 (m, 1H), 5.20 (s, 2H), 4.82 (s, 3H), 4.03 (t, J = 5.7 Hz, 2H), 3.87 (m, 2H), 3.53 (s, 1H), 3.25 (d, J = 7.8 Hz, 2H), 3.11 (s, 1H), 2.91 (t, J = 5.8 Hz,
3H), 2.62 (s, 3H), 1.39 (t, J = 7.3 Hz, 3H). LCMS observed m/z =770.20 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 218 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-1'-methyl-4-oxo-7H-1,2',3,3a,7,7a'- hexaaza-2,5'-biindenyl-7-yl)acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-1'-methyl-4-oxo-7H-1,2',3,3a,7,7a'-hexaaza-2,5'- biindenyl-7-yl)acetamide as a bright yellow solid.1H NMR (400 MHz, DMSO) δ 10.40 (s, 1H), 8.39 (s, 1H), 8.32 (s, 1H), 8.19 (s, 1H), 8.13 (d, J = 7.5 Hz, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.91 (s, 1H), 7.64 (d, J = 8.7 Hz, 1H), 7.43 (s, 1H), 7.21 (d, J = 7.4 Hz, 1H), 5.33 (s, 2H), 4.46 (d, J = 12.4 Hz, 1H), 3.45 – 3.38 (m, 3H), 3.19 – 3.14 (m, 1H), 2.96 – 2.88 (m, 3H), 2.76 (d, J = 11.2 Hz, 1H), 2.59 (d, J = 10.9 Hz, 1H), 2.56 (s, 3H), 2.34 (s, 3H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 750.4 [M+H]+. EXAMPLE 219 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H-1,2',3,3a,7,7a'-hexaaza-2,5'- biindenyl-7-yl)acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H-1,2',3,3a,7,7a'-hexaaza-2,5'-biindenyl-7- yl)acetamide as a bright yellow solid.1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 10.25 (d, J = 2.0 Hz, 1H), 8.59 (d, J = 2.0 Hz, 1H), 8.53 (s, 1H), 8.45 (d, J = 7.4 Hz, 1H), 8.31 (s, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.99 (s, 1H), 7.73 (d, J = 8.7 Hz, 1H), 7.61 (s, 1H), 7.31 (d, J = 7.4 Hz, 1H), 5.41 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.53 - 3.48 (m, 3H), 3.31 - 3.23 (m, 1H), 3.02 - 2.98 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.67 (d, J = 9.6 Hz, 1H), 2.45 (d, J = 2.0 Hz, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 736.3 [M+H]+. EXAMPLE 220 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3',3a,7,7a'-heptaaza-2,5'- biindenyl-7-yl) acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3',3a,7,7a'-heptaaza-2,5'-biindenyl- 7-yl) acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.47 (s, 1H), 10.25 (s, 1H), 9.10 (d, J = 7.1 Hz, 1H), 8.65 – 8.57 (m, 2H), 8.43 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.99 (s, 1H), 7.83 (d, J = 7.2 Hz, 1H), 7.72 (d, J = 8.7 Hz, 1H), 5.44 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 3.58 – 3.50 (m, 3H), 3.28 - 3.25 (m, 1H), 3.06 - 2.99 (m, 3H), 2.86 (d, J = 11.4 Hz, 1H), 2.70 (d, J = 11.4 Hz, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.0 Hz, 3H). LCMS observed m/z = 737.3 [M+H]+. EXAMPLE 221 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,7-trimethyl-1,3- dihydroisobenzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Compound 221
Step 1: Preparation of 2-(4-bromo-2-(hydroxymethyl)-6-methylphenyl)propan-2-ol. Compound 221.1
To a stirred solution of 5-bromo-7-methylisobenzofuran-1(3H)-one (100 mg, 0.4 mmol) in THF (2 mL) was added MeMgBr (1 mL, 1.3 mmol) dropwise at 0 °C under nitrogen atmosphere. The reaction mixture was warmed to 25 °C and stirred at 25 °C for 3 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 0 °C, quenched with sat. NH4Cl (aq.), and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-(4-bromo-2- (hydroxymethyl)-6-methylphenyl)propan-2-ol (100 mg, crude) as a light yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 256.85 [M-H]-. Step 2: Preparation of 5-bromo-1,1,7-trimethyl-1,3-dihydroisobenzofuran. Compound 221.2
To a stirred solution of 2-(4-bromo-2-(hydroxymethyl)-6-methylphenyl)propan-2- ol (400 mg, 1.5 mmol) in toluene (6 mL) was added H3PO4 (2 mL, 34.7 mmol) dropwise at 25 °C. The reaction mixture was heated to 90 °C and stirred at 90 °C for 3 hours. The reaction mixture was cooled to 25 °C and neutralized to pH 7 with NaOH (aq., 1 N). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were
washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (50:1) to afford 5-bromo-1,1,7-trimethyl-1,3- dihydroisobenzofuran (70 mg, 21% yield) as a yellow oil. Step 3: Preparation of 4,4,5,5-tetramethyl-2-(1,1,7-trimethyl-1,3- dihydroisobenzofuran-5-yl)-1,3,2-dioxaborolane. Compound 221.3
To a stirred solution of 5-bromo-1,1,7-trimethyl-1,3-dihydroisobenzofuran (150 mg, 0.6 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (474 mg, 1.8 mmol) in dioxane (3 mL) were added AcOK (183 mg, 1.8 mmol) and Pd(dppf)Cl2 (50 mg, 0.06 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 100 °C and stirred at 100 °C for 16 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C. After filtration, the filter cake was washed with EtOAc (3 x 10 mL) and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (40:1) to afford 4,4,5,5-tetramethyl-2-(1,1,7-trimethyl-1,3-dihydroisobenzofuran-5- yl)-1,3,2-dioxaborolane (60 mg, 33% yield) as an off-white solid. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-2-(1,1,7-trimethyl-1,3-dihydroisobenzofuran-5- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 221.4
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-
yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide, Intermediate H (80 mg, 0.1 mmol) and 4,4,5,5-tetramethyl-2-(1,1,7-trimethyl-1,3-dihydroisobenzofuran-5-yl)-1,3,2- dioxaborolane (35 mg, 0.1 mmol) in dioxane (4 mL) and H2O (0.5 mL) were added Pd(dppf)Cl2 (7 mg, 0.01 mmol) and K2CO3 (28 mg, 0.2 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 100 °C and stirred at 100 °C for 2 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C. After filtration, the filter cake was washed with EtOAc (3 x 10 mL) and the filtrate was concentrated under reduced pressure. This resulted in 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-2-(1,1,7-trimethyl-1,3- dihydroisobenzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (100 mg, crude) as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 870.35 [M+H]+. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,7-trimethyl-1,3- dihydroisobenzofuran-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 221
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-(1,1,7-trimethyl-3H-2-benzofuran-5-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (70 mg, 0.08 mmol) in TFA (1 mL) was heated to 80 °C and stirred at 80 °C for 30 minutes under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Mobile Phase A: 0.1% FA in water; Mobile Phase B: ACN; Flow rate: 60 mL/min) to afford N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,7-trimethyl-1,3-dihydroisobenzofuran-5-yl)-
[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (21.2 mg, 35% yield) as an off-white solid.1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 8.19 (d, J = 8.6 Hz, 1H), 8.00 – 7.78 (m, 3H), 7.63 (d, J = 8.6 Hz, 1H), 5.48 (s, 2H), 5.05 (s, 2H), 4.74 (m, 1H), 4.13 (m, 1H), 3.79 (s, 2H), 3.46 (m, 1H), 3.24 – 3.10 (m, 3H), 2.98 (m, 1H), 2.83 (m, 1H), 2.55 (s, 3H), 2.44 (s, 3H), 1.59 (s, 6H), 1.34 (t, J = 9.1 Hz, 3H). LCMS observed m/z = 780.35 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 222 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,3-dimethyl-1,4- dihydro-2-benzopyran-8-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 222
Step 1: Preparation of 1-bromo-3-[2-(methoxymethoxy)-2-methylpropyl]benzene. Compound 222.1
To a mixture of 1-(3-bromophenyl)-2-methylpropan-2-ol (1.0 g, 4.4 mmol) in DCM (15 mL) were added methane, bromomethoxy- (0.6 g, 4.4 mmol) and DIEA (1.7 g, 13.1 mmol) in portions at room temperature. The reaction mixture was stirred at room temperature for 5 hours. The resulting mixture was quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 1-bromo-3-[2-(methoxymethoxy)- 2-methylpropyl]benzene (800 mg, 64% yield) as a white solid. Step 2: Preparation of 6-bromo-3,3-dimethyl-1,4-dihydro-2-benzopyran.
Compound 222.2
To a mixture of 1-bromo-3-[2-(methoxymethoxy)-2-methylpropyl]benzene (200 mg, 0.7 mmol) in ACN (5 mL) was added TMSOTf (162 mg, 0.7 mmol) in one portion at 0 ℃ under nitrogen atmosphere. The reaction mixture was stirred at 0 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 6-bromo-3,3-dimethyl-1,4-dihydro-2-benzopyran (70 mg, 37% yield) as a colorless oil. Step 3: Preparation of 2-(3,3-dimethyl-1,4-dihydro-2-benzopyran-6-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 222.3
To a mixture of 6-bromo-3,3-dimethyl-1,4-dihydro-2-benzopyran (50 mg, 0.2 mmol), bis(pinacolato)diboron (79 mg, 0.3 mmol) and AcOK (61 mg, 0.6 mmol) in dioxane (3 mL) was added Pd(dppf)Cl2 (15 mg, 0.02 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 2-(3,3-dimethyl-1,4- dihydro-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (20 mg, 31% yield) as a colorless oil. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,3-dimethyl-1,4-dihydro-2-benzopyran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
Compound 222.4
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (20 mg, 0.03 mmol), 2-(3,3-dimethyl- 1,4-dihydro-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (7.3 mg, 0.03 mmol) and K2CO3 (10 mg, 0.08 mmol) in dioxane (3 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (2 mg, 0.003 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,3-dimethyl-1,4-dihydro-2-benzopyran- 6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (20 mg, 86.1%) as a white solid. LCMS observed m/z = 870.30 [M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,3-dimethyl- 1,4-dihydro-2-benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 222
A mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,3-dimethyl-1,4-dihydro-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.06 mmol) in TFA (5 mL) was stirred at room temperature for 2 hours. The resulting mixture was quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase flash with the following conditions (Column: Sunfire C18, 5 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 65% B to 75% B in 9 min; Wave Length: 254/220 nm; RT1(min): 5.35; 6.02) to afford N- [2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,3-dimethyl-1,4-dihydro-2-benzopyran-6-yl)- 5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (8.9 mg, 20% yield ) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 8.42 (m, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.94 – 7.87 (m, 1H), 7.85 (s, 1H), 7.72 (dd, J = 9.0, 2.1 Hz, 1H), 7.22 (d, J = 8.0 Hz, 1H), 5.40 (s, 2H), 4.74 (s, 2H), 4.54 (m, 1H), 3.57 – 3.47 (m, 3H), 3.26 (s, 1H), 3.06 – 3.00 (m, 3H), 2.85 (m, 1H), 2.74 (s, 2H), 2.71 – 2.61 (m, 1H), 2.43 (s, 3H), 1.22 (s, 9H). LCMS observed m/z = 780.20 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 223 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,3-dimethyl-1,4- dihydro-2-benzopyran-8-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 223
A mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,3-dimethyl-1,4-dihydro-2-benzopyran-8-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (20 mg, 0.02 mmol) in TFA (2 mL) was stirred at room temperature for 1h. The resulting mixture was quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reverse phase flash with the following conditions (Column: Sunfire C18 5 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 65% B to 75% B in 9 min; Wave Length: 254/220 nm; RT1 (min): 5.35; 6.02) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,3-dimethyl-1,4-dihydro-2- benzopyran-8-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (2.7 mg, 14% yield) as a white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.54 (s, 1H), 8.45 (s, 1H), 8.19 (d, J = 8.5 Hz, 1H), 8.01 (d, J = 7.5 Hz, 1H), 7.83 (s, 1H), 7.62 (d, J = 8.5 Hz, 1H), 7.35 – 7.22 (m, 2H), 5.46 (s, 2H), 5.21 (s, 2H), 4.75 (d, J = 13.0 Hz, 1H), 4.14 (s, 1H), 3.78 (d, J = 11.3 Hz, 2H), 3.58 – 3.43 (m, 1H), 3.18 (q, J = 7.9 Hz, 3H), 2.98 (d, J = 11.5 Hz, 1H), 2.81 (s, 3H), 2.53 (s, 3H), 1.40 – 1.28 (m, 3H), 1.27 (s, 6H). LCMS observed m/z = 780.20 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 224 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,2-difluoro-1,3- dihydroinden-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 224
Step 1: Preparation of 5-bromo-2,2-difluoro-1,3-dihydroindene. Compound 224.1
Into a 25 mL round-bottom flask were added 5-bromo-1,3-dihydroinden-2-one (200 mg, 0.9 mmol) and BAST (1.0 g, 4.7 mmol) at 0 °C. Then the resulting mixture was heated to 50 °C and stirred at 50 °C for 2 h. After completion of reaction, the mixture was allowed to cool down to 25 °C. The reaction was quenched with water (5 mL) at 0 °C, extracted with EA (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (100 mg, 45% yield) of 5-bromo-2,2-difluoro-1,3-dihydroindene as a yellow oil. LCMS observed m/z = 232.97 [M+H]+. Step 2: Preparation of 2-(2,2-difluoro-1,3-dihydroinden-5-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane. Compound 224.2
To a stirred solution of 5-bromo-2,2-difluoro-1,3-dihydroindene (80 mg, 0.3 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (174 mg, 0.7 mmol) in dioxane (0.5 mL) were added Pd(dppf)Cl2 (25 mg, 0.03 mmol) and KOAc (101 mg, 1.0 mmol) in portions at 25 °C under nitrogen atmosphere. Then the resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to 25 °C to afford (80 mg, crude) of 2-(2,2-difluoro-1,3-dihydroinden-5-yl)-4,4,5,5-tetramethyl-
1,3,2-dioxaborolane as a brown liquid. The resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 281.14 [M+H]+. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,2-difluoro-1,3-dihydroinden-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) and 2- (2,2-difluoro-1,3-dihydroinden-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (36 mg, 0.1 mmol) in dioxane (2 mL)/ H2O (0.4 mL) were added K2CO3 (53 mg, 0.4 mmol) and Pd(dppf)Cl2 (9mg, 0.01 mmol) in portions at 25 °C under nitrogen atmosphere. Then the resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to 25 °C and concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (80 mg, 73% yield) of 2-(6-{4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,2-difluoro-1,3-dihydroinden-5-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a brown solid. LCMS observed m/z = 862.26 [M+H]+. Step 4: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,2-difluoro- 1,3-dihydroinden-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 224
Into a 10 mL round-bottom flask were added 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,2-difluoro-1,3-dihydroinden-5-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (80 mg, 0.09 mmol) and TFA (1 mL) at 25 °C. Then the resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. After completion of reaction, the mixture was allowed to cool down to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep-HPLC (Column: XBridge BEH C185 μm, 30*150mm; Mobile Phase: 35-50% MeCN in H2O) to afford (38.9 mg, 48% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,2-difluoro-1,3-dihydroinden-5-yl)-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.11 (s, 1H), 8.62 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.19 (d, J = 8.0 Hz, 2H), 7.62 (d, J = 2.1 Hz, 1H), 7.56 (d, J = 8.8 Hz, 1H), 7.35 (d, J = 7.9 Hz, 1H), 5.71 (d, J = 13.0 Hz, 1H), 5.20 (s, 2H), 4.83 (d, J = 12.9 Hz, 1H), 3.86 (t, J = 11.5 Hz, 2H), 3.51 (t, J = 14.3 Hz, 5H), 3.30 – 3.19 (m, 2H), 3.18 – 3.05 (m, 1H), 2.95 – 2.75 (m, 2H), 2.59 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H).LCMS observed m/z = 518.30[M+H]+. EXAMPLE 225 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(3-methyl-2-oxo-1,3-benzoxazol-5-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 225
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(3-methyl-2-oxo-1,3-benzoxazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 225.1
To a solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (200 mg, 0.3 mmol), 3-methyl-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzoxazol-2-one (125 mg, 0.5 mmol) and K2CO3 (105 mg, 0.8 mmol) in Dioxane (5 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (19 mg, 0.03 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 3 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was poured into water (10 mL). The aqueous phase was extracted with EA (3 x 10 mL). The combined organic phases were dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (eluted: with 10% MeOH in DCM) to afford (150 mg, 69% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(3-methyl-2-oxo-1,3-benzoxazol-5-yl)-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 856.25 [M+H] +. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(3-methyl-2-oxo-1,3- benzoxazol-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 225
Into a 25 mL round-bottom flask were added 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(3-methyl-2-oxo-1,3-benzoxazol- 5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) and TFA (5 mL) at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure to give crude product which was further purified by Prep-HPLC (Column: Xselect CSH C18 5 m, 19 mm * 250 mm; mobile phase: 15-35% ACN in water(0.1% FA)) to afford (52.5 mg, 58.0% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(3-methyl-2-oxo-1,3-benzoxazol-5-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, Acetonitrile-d3) δ 11.16 (s, 1H), 8.89 (s, 1H), 8.60 (s, 1H), 8.35 (d, J = 8.6 Hz, 1H), 8.02 (dd, J = 8.4, 1.6 Hz, 1H), 7.91 (d, J = 1.6 Hz, 1H), 7.84 (d, J = 2.1 Hz, 1H), 7.66 – 7.61 (m, 1H), 7.34 (d, J = 8.4 Hz, 1H), 5.31 (s, 3H), 4.77 – 4.61 (m, 1H), 3.78 – 3.62 (m, 2H), 3.49 (s, 1H), 3.43 (s, 3H), 3.22 – 3.05 (m, 3H), 2.99 – 2.81 (m, 2H), 2.52 (s, 3H), 1.31 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 676.30 [M+H]+. EXAMPLE 226 Synthesis of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1- (difluoromethoxy)-2,3-dihydro-1H-inden-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-
methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (assumed). Compound 226
Step 1: Preparation of difluoromethanesulfinylbenzene. Compound 226.1
To a stirred solution of [(difluoromethyl)sulfanyl]benzene (5.0 g, 31.2 mmol) in DCM (50 mL) was added 3-chloroperoxybenzoic acid (5.7 g, 32.8 mmol) in portions at 0 ℃ under nitrogen atmosphere. The resulting mixture was stirred at 25 ℃ for additional 16h. After filtration, the filtrate was washed with cold 1M NaOH solution, dried over Na2SO4. The solvent was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (5.1 g, 85% yield) of difluoromethanesulfinylbenzene as a yellow oil. LCMS observed m/z = 177.01 [M+H]+. Step 2: Preparation of difluoromethyl)(phenyl)(2,4,6-trimethoxyphenyl)sulfanium hexafluorophosphate(V). Compound 226.2
Tf2O (8.0 g, 28.4 mmol) was added dropwise slowly into the stirring solution of difluoromethanesulfinylbenzene (5.0 g, 28.4 mmol) and 1,3,5-trimethoxybenzene (5.3 g,
31.2 mmol) in dry diethyl ether (50 mL) at 0 ℃ under N2 atmosphere. After the reaction was finished, solvent and excess trifluoromethanesulfonic caid anhydride was removed under vacuum. The residue was dissolved in dichloromethane and extracted with saturated aqueous KPF6 (4 x 50 mL). The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure. The crude product was precipitated in Et2O. After filtration the crude product was used in the next step directly without further purification. LCMS observed m/z = 327.37 [M+H]+. Step 3: Preparation of 5-bromo-1-(difluoromethoxy)-2,3-dihydro-1H-indene. Compound 226.3
To a stirred solution of difluoromethyl)(phenyl)(2,4,6-trimethoxyphenyl)sulfanium hexafluorophosphate(V) (5.0 g, 10.6 mmol) in H2O (10 mL) and 1,2-dibromoethane (10 mL) was added 5-bromo-2,3-dihydro-1H-inden-1-ol (0.7 g, 3.2 mmol) and Bu4NBF4 (698 mg, 2.1 mmol) in portions at 25 ℃ under nitrogen atmosphere. The resulting mixture was stirred for 48h at 25 ℃ under nitrogen atmosphere. The reaction was quenched by the addition of H2O (30 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (970 mg, 34% yield) of 5-bromo-1-(difluoromethoxy)-2,3-dihydro-1H-indene as a light- yellow oil. LCMS observed m/z = 262.98 [M+H]+. Step 4: Preparation of 2-[1-(difluoromethoxy)-2,3-dihydro-1H-inden-5-yl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 226.4
To a stirred mixture of 5-bromo-1-(difluoromethoxy)-2,3-dihydro-1H-indene (830 mg, 3.1 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3,2-dioxaborolane (1.2 g, 5.0 mmol) in dioxane (10 mL) were added Pd(dppf)Cl2CH2Cl2 (817 mg, 1.0 mmol) and AcOK (930 mg, 9.0 mmol) in portions at 25 ℃. Then the resulting
mixture was stirred at 100 ℃ for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 5% EtOAc in petroleum ether) to afford (112 mg, 34% yield) of 2-[1-(difluoromethoxy)-2,3-dihydro-1H-inden-5-yl]-4,4,5,5-tetramethyl-1,3,2- dioxaborolane as a white solid. LCMS observed m/z = 311.16 [M+H]+. Step 5: Preparation of 2-[1-(difluoromethoxy)-2,3-dihydro-1H-inden-5-yl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 226.5
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in TFA (1 mL) was stirred at 80 ℃ for 0.5 h under nitrogen atmosphere. The mixture was concentrated in vacuo to afford (120 mg, crude) of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a yellow oil. LCMS observed m/z = 698.08 [M+H]+. Step 6: Preparation of 2-[1-(difluoromethoxy)-2,3-dihydro-1H-inden-5-yl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 226.6
To a stirred mixture of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (120 mg, 0.2 mmol) and 2-[1-(difluoromethoxy)-2,3- dihydro-1H-inden-5-yl]-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (80 mg, 0.3 mmol) in dioxane (1 mL)/H2O (100 uL) were added Pd(dppf)Cl2CH2Cl2 (16 mg, 0.02 mmol) and K2CO3 (71 mg, 0.5 mmol) in portions at 25 °C. Then the resulting mixture was stirred at 100 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The reaction mixture was purified by reversed phase C18 silica gel column chromatography to afford (82 mg, 76% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{2-[1-(difluoromethoxy)-2,3-dihydro-1H-inden-5-yl]-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a brown solid. LCMS observed m/z = 802.22 [M+H]+. Step 7 Preparation of 2-[1-(difluoromethoxy)-2,3-dihydro-1H-inden-5-yl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 226
The racemic (82 mg) resulting from the last step was purified by SFC with follow conditions: Column: CHIRAL ART Amylose-SA, 3*25 cm, 5 μm; Mobile Phase A: MeOH: DCM=1: 1--HPLC, Mobile Phase B: Hex(0.3% FA)--HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1(min): 8; RT2(min): 10.3; Sample Solvent: MEOH; Injection Volume: 0.75 mL; Number Of Runs: 4 to afford (7.1 mg, 8.6% yield) of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1-(difluoromethoxy)-2,3-dihydro-1H- inden-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a brown solid. 1H NMR (400 MHz, Chloroform-d) δ 11.90 (s, 1H), 9.14 (s, 1H), 8.63 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.27 – 8.18 (m, 2H), 7.62 (d, J = 2.1 Hz, 1H), 7.54 (m, 8.3 Hz, 2H), 6.40 (m, 2H), 5.79 –
5.65 (m, 2H), 5.20 (s, 2H), 4.84 (m, 1H), 3.87 (m, 2H), 3.54 (s, 1H), 3.30 – 3.07 (m, 4H), 2.99 – 2.77 (m, 3H), 2.57 (d, J = 6.5 Hz, 1H), 2.29 (p, J = 6.4 Hz, 1H), 1.39 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 802.40 [M+H]+. EXAMPLE 227 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[1-(difluoromethoxy)- 2,3-dihydro-1H-inden-5-yl]-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 227
The racemic (80 mg) resulting from the last step was purified by SFC with follow conditions: Column: CHIRAL ART Amylose-SA, 3*25 cm, 5 μm; Mobile Phase A: MeOH: DCM=1: 1--HPLC, Mobile Phase B: Hex(0.3% FA)--HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1(min): 8; RT2(min): 10.3; Sample Solvent: MEOH; Injection Volume: 0.75 mL; Number Of Runs: 4 to afford (2.0 mg, 2.5% yield) of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1-(difluoromethoxy)-2,3-dihydro-1H- inden-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a brown solid. 1H NMR (400 MHz, Chloroform-d) δ 11.90 (s, 1H), 9.14 (s, 1H), 8.63 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.27 – 8.18 (m, 2H), 7.62 (d, J = 2.1 Hz, 1H), 7.54 (m, 8.3 Hz, 2H), 6.40 (m, 2H), 5.79 – 5.65 (m, 2H), 5.20 (s, 2H), 4.84 (m, 1H), 3.87 (m, 2H), 3.54 (s, 1H), 3.30 – 3.07 (m, 4H), 2.99 – 2.77 (m, 3H), 2.57 (d, J = 6.5 Hz, 1H), 2.29 (p, J = 6.4 Hz, 1H), 1.39 (t, J = 7.4 Hz, 3H). LCMS observed m/z =802.40 [M+H]+. EXAMPLE 228 Synthesis of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (assumed). Compound 228
Step 1: Preparation of 1-bromo-3-[2-(1-ethoxyethoxy)ethyl]benzene. Compound 228.1
To a stirred solution of 2-(3-bromophenyl)ethanol (4.0 g, 19.9 mmol) and molecular sieve (5 Å, 4.0 g) in DCM (40 mL) was added ethyl vinyl ether (7.2 g, 99.5 mmol) in portions at room temperature. The resulting mixture was stirred for 12 h at room temperature under nitrogen atmosphere. Then the resulting mixture was filtered and the filter cake was washed with DCM (2 x 40 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with DCM/MeOH (10:1) to afford 1-bromo-3-[2-(1-ethoxyethoxy)ethyl]benzene (4.0 g, 73% yield) as a colorless oil. LCMS observed
Step 2: Preparation of 6-bromo-1-methyl-3,4-dihydro-1H-2-benzopyran. Compound 228.2
To a stirred solution of 1-bromo-3-[2-(1-ethoxyethoxy)ethyl]benzene (3.0 g, 10.9 mmol) in DCM (30 mL) was added TiCl4 (12.5 g, 65.9 mmol) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was warmed to room temperature and stirred overnight at room temperature under nitrogen atmosphere. Then the mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 6-bromo-1-methyl-3,4-dihydro-1H-2-benzopyran (200 mg, 8% yield) as a yellow oil. LCMS observed m/z = 277.10 [M+H]+.
Step 3: Preparation of 4,4,5,5-tetramethyl-2-(1-methyl-3,4-dihydro-1H-2- benzopyran-6-yl)-1,3,2-dioxaborolane. Compound 228.3
To a stirred solution of 6-bromo-1-methyl-3,4-dihydro-1H-2-benzopyran (200 mg, 0.9 mmol) and AcOK (259 mg, 2.6 mmol) in dioxane (4 mL) were added 4,4,5,5- tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (447 mg, 1.8 mmol) and Pd(dppf)Cl2CH2Cl2 (71 mg, 0.1 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 hour under nitrogen atmosphere. Then the mixture was cooled to room temperature, quenched with water (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 4,4,5,5-tetramethyl-2- (1-methyl-3,4-dihydro-1H-2-benzopyran-6-yl)-1,3,2-dioxaborolane (150 mg, 62% yield) as a light-yellow oil. LCMS observed m/z = 274.17 [M+H]+. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(1-methyl-3,4-dihydro-1H-2-benzopyran-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 228.4
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (300 mg, 0.4 mmol) and 4,4,5,5-tetramethyl-2-(1-methyl-3,4-dihydro-1H-2-benzopyran-6-yl)-1,3,2-dioxaborolane (125 mg, 0.5 mmol) in dioxane (5 mL) and H2O (1 mL) were added K2CO3 (157 mg, 1.1
mmol) and Pd(dppf)Cl2CH2Cl2 (31 mg, 0.1mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 hour under nitrogen atmosphere. Then the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with DCM/MeOH (10:1) to afford 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(1-methyl-3,4- dihydro-1H-2-benzopyran-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide (100 mg, 30% yield) as an off-white solid. LCMS observed m/z = 856.30 [M+H]+. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 228.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(1-methyl-3,4-dihydro-1H-2-benzopyran-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in DCM (1 mL) was added TFA (5 mL) in portions at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 0.5 hour. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18, 5 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 55% B to 67% B in 7 min; Wave Length: 254/220 nm; RT1 (min): 6.08) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (80 mg, 88% yield) as an off-white solid. LCMS observed m/z = 766.35 [M+H]+.
Step 6: Preparation of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 228
The racemic mixture of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (80 mg) was resolved by the following method: Column: CHIRALPAK IF, 2*25 cm, 5 μm; Mobile Phase A: MeOH:DCM = 1:1--HPLC, Mobile Phase B: MtBE (0.1% FA)--HPLC; Flow rate: 20 mL/min; Gradient: isocratic; Wave Length: 220 nm; RT1(min): 7.8; RT2 (min): 11.5; Sample Solvent: DCM/MeOH = 1:1 (0.1% HAc); Injection Volume: 1 mL; Number of Runs: 4. This resulted in (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (19.9 mg, 22% yield, ee: 100%) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.45 (s, 1H), 9.15 (s, 1H), 8.71 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 8.12 – 8.08 (d, 2H), 7.62 (d, J = 2.0 Hz, 1H), 7.60 – 7.51 (m, 1H),7.21 (d, J = 7.9 Hz, 1H), 5.63 (s, 1H), 5.42 – 5.02 (m, 2H), 4.96 – 4.81 (m, 2H), 4.23 – 4.19 (m, 1H), 3.93 – 3.84 (m, 3H), 3.58 (s, 1H), 3.25 (d, J = 7.7 Hz, 2H), 3.18 – 3.03 (m, 2H), 2.95 – 2.68 (m, 6H), 1.72 – 1.49 (m, 3H), 1.42 – 1.33 (m, 3H). LCMS observed m/z = 766.35 [M+H]+. EXAMPLE 229 Synthesis of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 229
The racemic mixture of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (80 mg) was resolved by the following method: Column: CHIRALPAK IF, 2*25 cm, 5 μm; Mobile Phase A: MeOH:DCM = 1:1--HPLC, Mobile Phase B: MtBE (0.1% FA)--HPLC; Flow rate: 20 mL/min; Gradient: isocratic; Wave Length: 220 nm; RT1(min): 7.8; RT2 (min): 11.5; Sample Solvent: DCM/MeOH =1: 1 (0.1% HAc); Injection Volume: 1 mL; Number Of Runs: 4. This resulted in (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methylisochroman-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (21.7 mg, 27% yield, ee: 100%) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.45 (s, 1H), 9.15 (s, 1H), 8.71 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 8.12 – 8.08 (d, 2H), 7.62 (d, J = 2.0 Hz, 1H), 7.60 – 7.51 (m, 1H),7.21 (d, J = 7.9 Hz, 1H), 5.63 (s, 1H), 5.42 – 5.02 (m, 2H), 4.96 – 4.81 (m, 2H), 4.23 – 4.19 (m, 1H), 3.93 – 3.84 (m, 3H), 3.58 (s, 1H), 3.25 (d, J = 7.7 Hz, 2H), 3.18 – 3.03 (m, 2H), 2.95 – 2.68 (m, 6H), 1.72 – 1.49 (m, 3H), 1.42 – 1.33 (m, 3H). LCMS observed m/z = 766.40 [M+H]+. EXAMPLE 230 Synthesis of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (assumed) Compound 230
Step 1: Preparation of 5-bromo-2-methylbenzofuran. Compound 230.1
To a stirred solution of 2-(4-bromo-2-formylphenoxy)propanoic acid (1.7 g, 6.1 mmol) in Ac2O (15 mL) was added NaOAc (1.5 g, 18.3 mmol) in portions at 25 °C. The mixture was heated to 140 °C stirred at 140 °C for 16 h. The brown reaction solution was allowed to cool to room temperature, and was diluted with toluene (10 mL). The solution was added aq. NaOH (10 mL, 1 N) and the mixture was stirred at 25 °C for 10 min. After the reaction mixture was completed, the mixture was extracted with EtOAc (3 x 10 mL). The combined organic phases were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (100/1) to afford 5- bromo-2-methyl-1-benzofuran (810 mg, 56% yield) as a colorless oil. LCMS observed m/z: No signal Step 2: Preparation of 4,4,5,5-tetramethyl-2-(2-methylbenzofuran-5-yl)-1,3,2- dioxaborolane. Compound 230.2
To a solution of 5-bromo-2-methyl-1-benzofuran (400 mg, 1.9 mmol) and bis(pinacolato)diboron (722 mg, 2.8 mmol) and KOAc (558 mg, 5.7 mmol) in dioxane (100 mL) were added Pd(dppf)Cl2 (138 mg, 0.2 mmol) at 25 °C under a nitrogen atmosphere. The reaction mixture was heated to 90 °C and stirred at 90 °C for 5 h. under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (100/1) to afford
4,4,5,5-tetramethyl-2-(2-methyl-1-benzofuran-5-yl)-1,3,2-dioxaborolane (410 mg, 79% yield) as a colorless oil. LCMS observed m/z = 259.14 [M+H] + Step 3: Preparation of 4,4,5,5-tetramethyl-2-(2-methyl-2,3-dihydrobenzofuran-5- yl)-1,3,2-dioxaborol ane: Compound 230.3
To a stirred solution of 4,4,5,5-tetramethyl-2-(2-methyl-1-benzofuran-5-yl)-1,3,2- dioxaborolane (440 mg, 1.7 mmol) in EtOH (15 mL) was added Pd/C (50 mg, 0.470 mmol, 10%Wt%) in one portion and the mixture was heated to 70 °C and stirred at 70 °C for 8 h under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOH(3 x 10 mL). The filtrate was concentrated under reduced pressure to give 4,4,5,5- tetramethyl-2-(2-methyl-2,3-dihydro-1-benzofuran-5-yl)-1,3,2-dioxaborolane(430 mg, 97% yield) as a colorless oil. LCMS observed m/z= :261.16 [M+H] + Step 4: Preparation of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)pipera zin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl) acetamide:
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (400 mg, 0.5 mmol) in TFA (15 mL) was heated to 80 °C and stirred at 80 °C for 30 min. The resulting mixture was concentrated under reduced pressure to give 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoro methyl)phenyl]acetamide (350 mg, 95%purity) as a yellow oil. LCMS observed m/z= 698.08 [M+H] +
Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide: Compound 230.5
To a solution of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (355 mg, 0.5 mmol) and 4,4,5,5-tetramethyl-2-(2- methyl-2,3-dihydro-1-benzofuran-5-yl)-1,3,2-dioxaborolane (264 mg, 1.0 mmol) in dioxane (15 mL) and H2O (3 mL) were added K2CO3 (210 mg, 1.5 mmol) and Pd(dppf)Cl2 (50 mg, 0.07 mmol). The mixture was heated to 100 °C and stirred at 100 °C for 5 h. under a nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure and the residue was purified by silica gel column chromatography, eluted with CH2Cl2 / MeOH (10/1) to afford crude product (510 mg). The crude product was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. to give N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl]-2-(2-methyl-2,3-dihydro-1-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}aceta mide (240 mg, 63%yield) as a white solid. LCMS observed m/z= :752.40 [M+H] + Step 6: Preparation of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (assumed): Compound 230
The racemate of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-2,3-dihydro-1- benzofuran-5-yl)-7-oxo-[1,2,4]tria zolo[1,5-a]pyrimidin-4-yl}acetamide (240 mg, 0.3 mmol) was purified Prep-SFC with the following conditions: Column: CHIRALART Cellulose SB; Mobile Phase A: (Hex: DCM=3: 1)(0.1%FA): (MeOH: DCM=1: 1)=70: 30; Flow rate: 1.0 mL/min; Gradient: isocratic ; Injection Volume: 10 L mL to give N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-[(2S)-2-methyl-2,3-dihydro-1-benzofuran-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (74.2 mg, 97.5%purity) and N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-[(2R)-2-methyl-2,3-dihydro-1-benzofuran-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide(80.8 mg, 99.3% purity) as a yellow solid. 1H NMR (400 MHz, Chloroform-d) δ 11.84 (s, 1H), 9.22 (s, 1H), 8.62 (s, 1H), 8.53 (d, J = 8.5 Hz, 1H), 8.14 – 8.04 (m, 2H), 7.61 (s, 1H), 7.55 (d, J = 8.5 Hz, 1H), 6.83 (d, J = 7.5 Hz, 1H), 5.70 (s, 1H), 5.18 (s, 2H), 5.04 (s, 1H), 4.82 (s, 1H), 3.87 (s, 2H), 3.53 (s, 1H), 3.40 (s, 1H), 3.24 (s, 3H), 3.11 (s, 1H), 2.89 (s, 2H), 2.60 (s, 3H), 1.56 – 1.52 (m, 3H), 1.52 – 1.48 (m, 3H). LCMS observed m/z= :752.40 [M+H] +. EXAMPLE 231 Synthesis of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methyl-2,3- dihydrobenzofuran-5-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 231
The racemate of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl]-2-(2-methyl-2,3-dihydro-1- benzofuran-5-yl)-7-oxo-[1,2,4]tria zolo[1,5-a]pyrimidin-4-yl}acetamide (240 mg, 0.3 mmol, 1.0 equiv.) was purified Prep-SFC with the following conditions: Column: CHIRALART Cellulose SB; Mobile Phase A: (Hex: DCM=3: 1)(0.1%FA): (MeOH: DCM=1:1)=70: 30; Flow rate: 1.0 mL/min; Gradient: isocratic ; Injection Volume: 10 L mL to give N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-[(2S)-2-methyl-2,3-dihydro-1-benzofuran- 5-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (74.2 mg, 97.5%purity) and N- [2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-[(2R)-2-methyl-2,3-dihydro-1-benzofuran-5-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (80.8 mg, 99.3%purity) as a yellow solid. 1H NMR (400 MHz, Chloroform-d) δ 11.84 (s, 1H), 9.22 (s, 1H), 8.62 (s, 1H), 8.53 (d, J = 8.5 Hz, 1H), 8.14 – 8.04 (m, 2H), 7.61 (s, 1H), 7.55 (d, J = 8.5 Hz, 1H), 6.83 (d, J = 7.5 Hz, 1H), 5.70 (s, 1H), 5.18 (s, 2H), 5.04 (s, 1H), 4.82 (s, 1H), 3.87 (s, 2H), 3.53 (s, 1H), 3.40 (s, 1H), 3.24 (s, 3H), 3.11 (s, 1H), 2.89 (s, 2H), 2.60 (s, 3H), 1.56 – 1.52 (m, 3H), 1.52 – 1.48 (m, 3H). LCMS observed m/z= :752.40 [M+H] +. EXAMPLE 232 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,2- difluorobenzo[d][1,3]dioxol-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 232
Step 1: Preparation of 2-(2,2-difluorobenzo[d][1,3]dioxol-4-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane. Compound 232.1
To a solution of 2,2-difluoro-1,3-benzodioxole (1.0 g, 6.3 mmol) was dissolved in THF (15 mL) and cooled to -75° C. Sec-BuLi (1.3 M solution in Hexane, 5.6 mL, 7.3 mmol) was added dropwise, keeping the temperature below -65° C under nitrogen atmosphere. The mixture was stirred at -78 °C for 1 h. To the above mixture was added 2-isopropoxy-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (1.18 g, 6.3 mmol) in THF (5 mL) dropwise at -78°C, keeping the temperature below -65° C. The resulting mixture was stirred at -78°C for additional 2 h. The reaction mixture was then allowed to warm to room temperature and stirred at room temperature for 2 h under nitrogen atmosphere. The reaction was quenched with EtOAc and water at 25°C. The aqueous phase was acidified to pH 5 with aqueous 6 M HCl solution. The reaction mixture was extracted with EtOAc (3 x 10 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (12/1) to afford 2-(2,2-difluoro-1,3-benzodioxol-4-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.18 g, 62% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 7.45 (dd, J = 7.7, 1.5 Hz, 1H), 7.15 (dd, J = 7.9, 1.4 Hz, 1H), 7.08 (t, J = 7.8 Hz, 1H), 1.39 (s, 12H). Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,2- difluorobenzo[d][1,3]dioxol-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Compound 232
To a solution of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide, Intermediate AC (176 mg, 0.25 mmol, Intermediate) and 2-(2,2-difluoro-1,3-benzodioxol-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (71 mg, 0.25 mmol) in 1,4-dioxane/H2O (3 mL/0.3 mL) were added K2CO3 (70 mg, 0.5 mmol) and Pd(dppf)Cl2 (18 mg, 0.025 mmol) at 25°C under N2 atmosphere. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 2 hours. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure, collected the residue. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (8/2) to afford product a white solid. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Phenyl 5 μm, 30*150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29% B to 49%B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 6.55) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,2- difluorobenzo[d][1,3]dioxol-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (57.5 mg, 29% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.55 – 10.16 (m, 2H), 8.56 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 8.00 – 7.95 (m, 1H), 7.87 (dd, J = 8.1, 1.2 Hz, 1H), 7.71 (dd, J = 8.9, 2.1 Hz, 1H), 7.58 (m, 1H), 7.38 (t, J = 8.1 Hz, 1H), 5.40 (s, 2H), 4.55 (d, J = 12.6 Hz, 1H), 3.59 – 3.46 (m, 3H), 3.30 – 3.22 (m, 1H), 3.09 – 2.96 (m, 3H), 2.89 – 2.82 (m, 1H), 2.71 – 2.65 (m, 1H), 2.44 (s, 3H), 1.25 – 1.20 (m, 3H).LCMS observed m/z = 776.10 [M+H]+. EXAMPLE 233
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3-methyl-2-oxo-2,3- dihydrobenzo[d]oxazol-6-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 233
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-(trifluoromethyl) phenyl)acetamide, Intermediate H (240 mg, 0.3 mmol) and 3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzo[d]oxazol-2(3H)-one (100 mg, 0.3 mmol) in dioxane (10 mL) and H2O (1 mL) were added Pd(dppf)Cl2 (44 mg, 0.06 mmol) and K2CO3 (126 mg, 0.9 mmol) in one portion at room temperature under nitrogen atmosphere. The mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate
was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 30% to 60% gradient in 10 min; detector, UV 254 nm to give 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl- 2-(3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-6-yl)-7-oxo-[1,2,4] triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (140 mg, 53.5% yield) as a yellow solid. LCMS observed m/z = 857.25 [M+H] +. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3-methyl-2-oxo-2,3- dihydrobenzo[d]oxazol-6-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 233
Into a 8 mL sealed tube were added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimi din-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (130 mg, 0.1 mmol) and TFA (3 mL) at room temperature. The resulting mixture was stirred at 80 °C for 1 h. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product (130 mg) was purified by Prep-HPLC with the following conditions (Column: X- Bridge BEH C18 5 μm, 19*250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 30% B to 40%B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 8.23) to afford N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyri midine-4- carbonyl)piperazin-1-yl)-2-(3-methyl-2-oxo-2,3-dihydrobenzo[d]oxazol-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (58.5 mg, 50.0% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 2H), 8.50 (s, 1H), 8.09 – 8.01 (m, 2H), 7.97 (m, 2H), 7.75 – 7.68 (m, 1H), 7.41 (d, J = 8.3 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.4
Hz, 1H), 3.53 (d, J = 12.0 Hz, 3H), 3.39 (s, 3H), 3.27 (s, 1H), 3.01 (m, 3H), 2.84 (m, 1H), 2.66 (s, 1H), 2.43 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 767.15 [M+H] +. EXAMPLE 234 Synthesis of 2-(2-(benzo[c][1,2,5]oxadiazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide Compound 234
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (2,1,3- benzofurazan-5-yl) boranediol.1H NMR (400 MHz, DMSO) δ 10.40 (br, 2H), 8.60 (s, 1H), 8.49 (s, 1H), 8.23 (dd, J = 9.4, 1.3 Hz, 1H), 8.17 (d, J = 9.5 Hz, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.92 (d, J = 2.1 Hz, 1H), 7.65 (dd, J = 8.7, 2.1 Hz, 1H), 5.37 (s, 2H), 4.48 (d, J = 12.4 Hz, 1H), 3.46 (td, J = 12.9, 6.9 Hz, 3H), 3.19 (d, J = 12.5 Hz, 1H), 3.02 – 2.93 (m, 3H), 2.79 (d, J = 11.2 Hz, 1H), 2.63 (s, 1H), 2.38 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 738.434 [M+H]+. EXAMPLE 235 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methyl-2H-indazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 235
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (2-methyl-2H- indazol-5-yl) boranediol.1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 8.56 (s, 1H), 8.51 (d, J = 7.4 Hz, 2H), 8.08 (d, J = 8.6 Hz, 1H), 8.04 – 7.91 (m, 2H), 7.71 (dd, J = 8.9, 6.2 Hz, 2H), 7.56 (s, 1H), 5.42 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 4.20 (s, 3H), 3.53 (t, J = 11.1 Hz, 3H), 3.27 (s, 1H), 3.03 (d, J = 10.4 Hz, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.68 (d, J = 10.6 Hz, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 751.20 [M+H]+. EXAMPLE 236 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-1H-benzo[d]imidazol-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 236
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (1-methyl-1H- 1,3-benzimidazol-6-yl)boranediol. 1H NMR (400 MHz, DMSO) δ 10.40 (s, 2H), 8.49 (s, 1H), 8.25 – 8.17 (m, 2H), 8.01 (d, J = 8.6 Hz, 1H), 7.99 – 7.89 (m, 2H), 7.72 – 7.62 (m, 2H), 5.36 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.85 (s, 3H), 3.47 (q, J = 12.0 Hz, 3H), 3.18 (d, J =
12.2 Hz, 1H), 2.96 (d, J = 11.2 Hz, 3H), 2.78 (d, J = 11.2 Hz, 1H), 2.61 (d, J = 9.8 Hz, 1H), 2.37 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.534 [M+H]+. EXAMPLE 237 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylbenzo[d]oxazol-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 237
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (2-methyl-1,3- benzoxazol-6-yl)boranediol. 1H NMR (400 MHz, DMSO) δ 10.38 (br, 2H), 8.49 (s, 1H), 8.22 (d, J = 1.5 Hz, 1H), 8.06 (dd, J = 8.3, 1.5 Hz, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.91 (d, J = 2.1 Hz, 1H), 7.72 (d, J = 8.3 Hz, 1H), 7.65 (dd, J = 8.8, 2.2 Hz, 1H), 5.35 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.53 – 3.40 (m, 3H), 3.18 (d, J = 12.2 Hz, 1H), 2.94 (dd, J = 16.7, 9.4 Hz, 3H), 2.78 (d, J = 11.3 Hz, 1H), 2.62 (s, 1H), 2.59 (s, 3H), 2.37 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 751.189 [M+H]+. EXAMPLE 238 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(4-methyl-2,3-dihydro-1-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 238
Step 1: Preparation of 2-bromo-1-(2-hydroxy-6-methylphenyl)ethanone. Compound 238.1
To a stirred solution of 1-(2-hydroxy-6-methylphenyl)ethanone (1.0 g, 6.7 mmol) in DCM (30 mL) was added CuBr2 (3.0 g, 13.3 mmol) in one portions at room temperature. The resulting mixture was stirred at room temperature for 2 h. Desired product could be detected by LCMS. The reaction was quenched with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (4:1) to afford 2-bromo-1-(2-hydroxy-6-methylphenyl)ethanone (800 mg, 49% yield) as a white solid. LCMS observed m/z =
. [M-H]-. Step 2: Preparation of 4-methyl-2H-1-benzofuran-3-one. Compound 238.2
To a stirred solution of 2-bromo-1-(2-hydroxy-6-methylphenyl)ethanone (800 mg, 3.5 mmol) in DMF (20 mL) was added KF (608 mg, 10.5 mmol) in one portions at room temperature. The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (20 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (4:1) to afford 4-methyl-2H-1- benzofuran-3-one (400 mg, 73% yield) as a colorless oil.
Step 3: Preparation of 4-methyl-2,3-dihydro-1-benzofuran. Compound 238.3
To a stirred solution of 4-methyl-2H-1-benzofuran-3-one (400 mg, 2.7 mmol) in AcOH (15 mL) was added Zn (883 mg, 13.5 mmol) in one portion at room temperature. The resulting mixture was heated to 80 ℃ and stirred for 5 h at 80 ℃. After cooling to room temperature, the resulting mixture was filtered and the filter cake was washed with MeOH (3 x 5 mL). The filtrate was concentrated under reduced pressure to afford 4-methyl-2,3-dihydro-1-benzofuran (250 mg, crude) as a yellow oil. The crude product was used in the next step directly without further purification. Step 4: Preparation of 5-iodo-4-methyl-2,3-dihydro-1-benzofuran. Compound 238.4
To a stirred solution of 4-methyl-2,3-dihydro-1-benzofuran (50 mg, 0.4 mmol) in MeOH (5 mL) were added I2 (95 mg, 0.4 mmol) and AgNO2 (38 mg, 0.4 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (20 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 5-iodo-4-methyl-2,3-dihydro-1-benzofuran (80 mg, 78% yield) as a white solid. Step 5: Preparation of 4,4,5,5-tetramethyl-2-(4-methyl-2,3-dihydro-1-benzofuran-5- yl)-1,3,2-dioxaborolane. Compound 238.5
To a stirred solution of 5-iodo-4-methyl-2,3-dihydro-1-benzofuran (55 mg, 0.2 mmol), bis(pinacolato)diboron (79 mg, 0.3 mmol) and AcOK (61 mg, 0.6 mmol) in dioxane
(3 mL) was added Pd(dppf)Cl2 (15 mg, 0.02 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 4,4,5,5-tetramethyl-2-(4- methyl-2,3-dihydro-1-benzofuran-5-yl)-1,3,2-dioxaborolane (20 mg, 31% yield) as a colorless oil. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(4-methyl-2,3-dihydro-1-benzofuran-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (20 mg, 0.03 mmol), 4,4,5,5- tetramethyl-2-(4-methyl-2,3-dihydro-1-benzofuran-5-yl)-1,3,2-dioxaborolane (7.3 mg, 0.03 mmol) and K2CO3 (10 mg, 0.08 mmol) in dioxane (3 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (2 mg, 0.003 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(4-methyl-2,3-dihydro-1-
benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (20 mg, 86.1%) as a white solid. LCMS observed m/z = 842.27 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(4-methyl-2,3-dihydro-1- benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-(4-methyl-2,3-dihydro-1-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (20 mg, 0.02 mmol) in TFA (2 mL) was heated to 50 °C and stirred at 50 °C for 1 h. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18, 5 m, 30 mm x 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 42% B to 55%B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.2) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-(4-methyl-2,3-dihydro-1-benzofuran-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (3.5 mg, 19% yield ) as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.24 (s, 1H), 8.17 (d, J = 8.6 Hz, 1H), 7.82 (d, J = 2.1 Hz, 1H), 7.77 (d, J = 8.4 Hz, 1H), 7.61 (dd, J = 8.8, 2.1 Hz, 1H), 6.64 (d, J = 8.4 Hz, 1H), 5.43 (s, 2H), 4.73 (d, J = 12.7 Hz, 1H), 4.61 (t, J = 8.7 Hz, 2H), 3.82 – 3.70 (m, 2H), 3.56 – 3.46 (m, 1H), 3.17 (dt, J = 21.7, 7.9 Hz, 5H), 3.04 – 2.93 (m, 1H), 2.81 (d, J = 11.4 Hz, 1H), 2.54 (s, 3H), 2.44 (s, 3H), 1.34 (t, J = 7.5 Hz, 3H), 0.12 (d, J = 2.2 Hz, 1H). LCMS observed m/z = 752.20 [M+H]+. Note: Two exchangeable proton were not observed in NMR spectra. EXAMPLE 239
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methyl pyrimidine-4-carbonyl)piperazin-1-yl]-2-{4-methylpyrazolo[1,5-a]pyridin-5-yl}-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 239
Step 1: Preparation of 4-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine. Compound 239.1
To a stirred solution of 5-bromo-4-methylpyrazolo[1,5-a]pyridine (90 mg, 0.4 mmol) and bis(pinacolato)diboron (130 mg, 0.5 mmol) in dioxane (1 mL) was added Pd(dppf)Cl2 (31 mg, 0.04 mmol) and KOAc (84 mg, 0.8 mmol) in portions at 25 °C. The mixture was heated to 90 °C and stirred at 90 °C for 3 h under nitrogen atmosphere. Then the resulting mixture was cooled down to 25°C and concentrated under reduced pressure. The resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 259.13 [M+H]+. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{4-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoro methyl)phenyl]acetamide. Compound 239.2
To a stirred mixture of 4-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine (36 mg, 0.1 mmol) and 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) in dioxane (4 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (9 mg, 0.01 mmol) and K2CO3 (35 mg, 0.2 mmol) in one portion at 25°C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25°C. The resulting mixture was filtered, the filter cake was washed with EA (3 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH(20/1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{4-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl] acetamide (42 mg, 39 % yield) as a light brown solid. LCMS observed m/z = 840.27 [M-H]-. Step 3: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl]-2-{4-methylpyrazolo[1,5- a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 239
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-{4-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(tri fluoromethyl)phenyl]acetamide (40 mg, 0.05 mmol) in TFA (3 mL) was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure. The crude product (50 mg) was purified by Prep- HPLC with the following conditions (Column: X-Bridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 27% B to 50% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 7.8) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6- [4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{4-methylpyrazolo[1,5- a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyri midin-4-yl}acetamide (17.4 mg, 50% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.82 (s, 1H), 8.96 (s, 1H), 8.62 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 8.42 (d, J = 7.3 Hz, 1H), 8.01 (d, J = 2.3 Hz, 1H), 7.62 (d, J = 2.0 Hz, 1H), 7.56 (dd, J = 7.0, 2.3 Hz, 2H), 6.73 (dd, J = 2.4, 0.9 Hz, 1H), 5.72 (d, J = 13.1 Hz, 1H), 5.22 (s, 2H), 4.84 (d, J = 12.8 Hz, 1H), 3.88 (t, J = 11.6 Hz, 2H), 3.54 (s, 1H), 3.25 (d, J = 7.8 Hz, 2H), 3.14 (d, J = 14.6 Hz, 1H), 2.94 (s, 3H), 2.83 (t, J = 2.8 Hz, 2H), 2.60 (s, 3H), 1.40 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 750.22 [M+H] + . Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 240 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl) piperazin-1-yl]-2-{7-methylpyrazolo[1,5-a]pyridin-5-yl}-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 240
Step 1: Preparation of 1-amino-4-bromo-2-methylpyridin-1-ium 2,4,6- trimethylbenzenesulfonate. Compound 240.1
To a stirred solution of 4-bromo-2-methylpyridine (500 mg, 2.9 mmol) in DCM (12 mL) was added amino 2,4,6-trimethylbenzenesulfonate (2.2 g, 10.2 mmol) in portions at 25 °C. The mixture was stirred at 25 °C for 6 h. Then the resulting mixture was concentrated under vacuum to afford the crude product. The crude product was used in the next step directly without further purification. LCMS observed m/z = 387.03 [M+H] +. Step 2: Preparation of ethyl 5-bromo-7-methylpyrazolo[1,5-a]pyridine-3- carboxylate. Compound 240.2
To a stirred mixture of 1-amino-4-bromo-2-methylpyridin-1-ium 2,4,6- trimethylbenzene sulfonate (1.1 g, 2.9 mmol) and K2CO3 (800 mg, 5.8 mmol) in DMF (11 mL) was added ethyl propiolate (340 mg, 3.5 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 25 °C for 3 h under nitrogen atmosphere. Then the resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (3 x 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford ethyl 5-bromo-7- methylpyrazolo[1,5-a]pyridine-3-carboxylate (104 mg, 12% yield) as a light brown solid. LCMS observed m/z = 283.00 [M+H] +. Step 3: Preparation of 5-bromo-7-methylpyrazolo[1,5-a]pyridine. Compound 240.3
A solution of ethyl 5-bromo-7-methylpyrazolo[1,5-a]pyridine-3-carboxylate (100 mg, 0.3 mmol) and H2SO4 (1.0 g, 10.2 mmol) in H2O (4 mL) was stirred at 50 °C for 16 h
under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The residue was neutralized to pH 6-8 with sat. aq. NaHCO3. The resulting mixture was extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (3 x10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 5-bromo-7-methylpyrazolo[1,5-a]pyridine (52 mg, 69% yield) as a light yellow solid. LCMS observed m/z = 210.98 [M+H] + Step 4: Preparation of 7-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine. Compound 240.4
To a stirred mixture of 5-bromo-7-methylpyrazolo[1,5-a]pyridine (50 mg, 0.2 mmol) and bis(pinacolato)diboron (66 mg, 0.3 mmol) in dioxane (2 mL) was added Pd(dppf)Cl2 (17 mg, 0.02 mmol) and KOAc (46 mg, 0.5 mmol) in one portion at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure to afford the crude product. The crude product was used in the next step directly without further purification. LCMS observed m/z = 259.15 [M+H] +. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{7-methylpyra zolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide. Compound 240.5
To a stirred mixture of 7-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine (33 mg, 0.1 mmol) and 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-
a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide, Intermediate H (102 mg, 0.1 mmol) in dioxane (8 mL) and H2O (2 mL) was added Pd(dppf)Cl2 (9 mg, 0.01 mmol) and K2CO3 (36 mg, 0.3 mmol) in one portion at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The residue was purified by Prep-TLC (PE/EA 0/1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{7-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(tri fluoromethyl)phenyl]aceta mide (43 mg, 39% yield) as a light brown solid. LCMS observed m/z = 840.27 [M+H] +. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl]-2-{7-methylpyrazolo[1,5- a]pyridin-5-yl}-7-oxo-[1,2,4]tria zolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 240
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-{7-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (40 mg, 0.05 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under vacuum. The crude product (50 mg) was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 27% B to 50% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 7.8) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-2-{7-methylpyrazolo[1,5- a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (9.0 mg, 99.2% purity) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.05 (s, 1H), 8.62 (s, 1H), 8.54 (d, J = 8.7 Hz, 1H), 8.44 (t, J = 1.2 Hz, 1H), 8.08 (d, J = 2.3 Hz, 1H), 7.64
– 7.59 (m, 2H), 7.56 (d, J = 9.1 Hz, 1H), 6.74 (d, J = 2.3 Hz, 1H), 5.73 (d, J = 13.0 Hz, 1H), 5.22 (s, 2H), 4.84 (d, J = 12.8 Hz, 1H), 3.87 (t, J = 11.6 Hz, 2H), 3.55 (d, J = 13.5 Hz, 1H), 3.25 (d, J = 7.9 Hz, 2H), 3.13 (d, J = 12.7 Hz, 1H), 2.85 (s, 5H), 2.60 (s, 3H), 1.40 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 750.20 [M+H] +. EXAMPLE 241 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{4- fluoropyrazolo[1,5-a]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide.
Step 1: Preparation of 1-amino-4-bromo-3-fluoropyridin-1-ium 2,4,6- trimethylbenzenesulfonate. Compound 241.1
To a stirred solution of 4-bromo-3-fluoropyridine (20.0 g, 113.6 mmol) in DCM (200 mL) was added amino 2,4,6-trimethylbenzenesulfonate (36.7 g, 170.7 mmol) in portions at 25 ℃ under nitrogen atmosphere. The resulting mixture was stirred for 18 h at 25 ℃ under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in EtOAc. The precipitated solids were collected by filtration and washed with EtOAc. The crude product was used directly in the next step without further purification. LCMS observed m/z =190.96 [M+H]+. Step 2: Preparation of ethyl 5-bromo-4-fluoropyrazolo[1,5-a]pyridine-3- carboxylate. Compound 241.2
To a stirred solution of 1-amino-4-bromo-3-fluoropyridin-1-ium 2,4- dimethylbenzenesulfonate (40.0 g, 106.2 mmol) in DMF (60 mL) was added ethyl propiolate (12.5 g, 127.2 mmol) and K2CO3 (29.3 g, 212.0 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred for 18 h at 25 °C under nitrogen atmosphere. The reaction was quenched with water (80 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 80 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 5% EtOAc in petroleum ether) to afford (15 g, 51% yield) of ethyl 5-bromo-4-fluoropyrazolo[1,5-a]pyridine-3-carboxylate as a white solid. LCMS observed m/z =286.98 [M+H]+. Step 3: Preparation of 5-bromo-4-fluoropyrazolo[1,5-a]pyridine. Compound 241.3
A solution of ethyl 5-bromo-4-fluoropyrazolo[1,5-a]pyridine-3-carboxylate (14.0 g, 48.8 mmol) in H2O (48 mL) was added AcOH (48 mL) and HCl (36mL). The resulting mixture was stirred at 100 °C for 18 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 5% EtOAc in petroleum ether) to afford (9.7 g, 92% yield) of 5-bromo-4-fluoropyrazolo[1,5-a]pyridine as a white solid. LCMS observed m/z =214.95 [M+H]+. Step 4: Preparation of 4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine. Compound 241.4
To a stirred solution of 5-bromo-4-fluoropyrazolo[1,5-a]pyridine (9.0 g, 41.9 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (21.3 g, 83.7 mmol) in dioxane (100 mL) were added Pd(dppf)Cl2 (3.4 g, 4.2 mmol) and AcOK (12.3 g, 125.6 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 90 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z =263.13 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{4-fluoropyrazolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 241.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (180 mg, 0.2 mmol) and 4- fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (90 mg, 0.3 mmol) in dioxane (5 mL) /H2O (500 uL) was added Pd(dppf)Cl2CH2Cl2 (17 mg, 0.02 mmol) and K2CO3 (95 mg, 0.7 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 100 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (95 mg, 49% yield) of 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-{4- fluoropyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide as a brown solid. LCMS observed m/z =844.24 [M+H]+.
Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{4-fluoropyrazolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 241
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-{6-fluoropyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in TFA (1 mL) was stirred at room temperature for 18 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C185 μm, 30*150mm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: MEOH; Flow rate: 60 mL/min; Gradient: 55% B to 70%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 5.52) to afford (43.7 mg, 58% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-(5-ethyl-2-{4-fluoropyrazolo[1,5-a]pyridin-5-yl}-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetamide as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.00 (s, 1H), 8.62 (s, 1H), 8.49 (d, J = 8.7 Hz, 1H), 8.38 (d, J = 7.3 Hz, 1H), 8.03 (d, J = 2.4 Hz, 1H), 7.64 – 7.53 (m, 4H), 6.88 – 6.86 (m, 1H), 5.73 (s, 1H), 5.23 (s, 2H), 4.83 (s, 1H), 3.87 (m, 3H), 3.54 (s, 1H), 3.26 (d, J = 7.8 Hz, 2H), 3.12 (s, 1H), 2.60 (s, 3H), 1.40 (t, J = 7.5 Hz, 3H). LCMS observed m/z =754.35 [M+H]+. EXAMPLE 242 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(6- fluoropyrazolo[1,5-a]pyridin-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 242
Step 1: Preparation of 6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine. Compound 242.1
To a stirred solution of 5-bromo-6-fluoropyrazolo[1,5-a]pyridine (400 mg, 1.8 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (566 mg, 2.2 mmol) in dioxane (10 mL) were added Pd(dppf)Cl2 (272 mg, 0.4 mmol) and KOAc (547 mg, 5.5 mmol) in portions at 25°C under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (30 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The resulting mixture was used in the next step directly without further purification as a yellow oil (180 mg crude). LCMS observed m/z = 263.13 [M+H] +. Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(6-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoro methyl)phenyl)acetamide. Compound 242.2
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (300 mg, 0.4 mmol) and 6- fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (119 mg, 0.4 mmol) in dioxane (10 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (27 mg, 0.03 mmol) and K2CO3 (157 mg, 1.1 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The reaction was quenched with water (30 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2 / MeOH (20 / 1) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(6-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (180 mg, 56 %yield,) as a yellow solid. LCMS observed m/z = 844.25 [M+H] +. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(6-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoro methyl)phenyl)acetamide. Compound 242
Into a 4 mL sealed tube were added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(6-fluoropyrazolo[1,5-a]pyridin-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (180 mg, 0.2 mmol) and TFA (2 mL) at room temperature. The resulting mixture was heated to 80 °C and stirred at 80°C for 20 min. The resulting mixture was concentrated under reduced pressure. The crude product (180 mg) was purified by Prep-HPLC with the following conditions (Column: X-Bridge BEH C185 μm, 30*150mm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 26% B to 46%B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 5.77) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5- ethyl-2-(6-fluoropyrazolo[1,5-a]pyridin-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (58.8 mg, 36% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ10.14(br, 1H) 9.19 (d, J = 6.2 Hz, 1H), 8.59 – 8.41 (m, 2H), 8.15 – 8.04 (m, 2H), 7.98 (s, 1H), 7.72 (d, J = 9.0 Hz, 1H), 6.89 (d, J = 2.4 Hz, 1H), 5.42 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 3.53 (d, J = 12.2 Hz, 3H),3.29 – 3.27(m, 1H), 3.05 – 3.01 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H),2.67 (s, 1H), 2.42 (s, 3H), 1.23 (t, J = 7.4 Hz,3H). LCMS observed m/z = 754.10 [M+H] +. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 243 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{6-methylpyrazolo[1,5-a]pyridin-5-yl}-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 243
Step 1: Preparation of 1-amino-4-bromo-3-methylpyridin-1-ium. Compound 243.1
To a stirred solution of 4-bromo-3-methylpyridine (500 mg, 2.9 mmol) in DCM (30 mL) was added amino 2,4,6-trimethylbenzenesulfonate (625 mg, 2.9 mmol) in one portion at room temperature. The resulting mixture was stirred at room temperature for 2 h. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure to afford 1-amino-4-bromo-3-methylpyridin-1-ium (800 mg, crude) as yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 186.99 [M]+. Step 2: Preparation of ethyl 5-bromo-6-methylpyrazolo[1,5-a]pyridine-3- carboxylate. Compound 243.2
To a stirred solution of 1-amino-4-bromo-3-methylpyridin-1-ium (500 mg, 2.7 mmol) in DMF (20 mL) were added ethylpropiolate (260 mg, 2.7 mmol) and K2CO3 (1.1 g, 8.0 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water (20 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE
/ EA (5:1) to afford ethyl 5-bromo-6-methylpyrazolo[1,5-a]pyridine-3-carboxylate (400 mg, 50% yield) as a white solid. LCMS observed m/z = 283.00 [M+H]+. Step 3: Preparation of methyl 2-chloro-3-oxopentanoate. Compound 243.3
A solution of ethyl 5-bromo-6-methylpyrazolo[1,5-a]pyridine-3-carboxylate (500 mg, 1.8 mmol) in H2SO4 (30 mL) and H2O (30 mL) was heated to 50 ℃ and stirred overnight at 50 ℃. Desired product could be detected by LCMS. The resulting mixture was cooled to room temperature, quenched with water (20 mL) and extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 5-bromo-6-methylpyrazolo[1,5-a]pyridine (300 mg, 80% yield) as a white solid. LCMS observed m/z = 210.98 [M+H]+. Step 4: Preparation of 6-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine. Compound 243.4
To a mixture of 5-bromo-6-methylpyrazolo[1,5-a]pyridine (500 mg, 2.4 mmol), bis(pinacolato)diboron (902 mg, 3.6 mmol) and KOAc (697 mg, 7.1 mmol) in 1,4- dioxane (50 mL) was added Pd(dppf)Cl2 (193 mg, 0.2 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 6-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (200 mg, 31% yield) as a white solid. LCMS observed m/z = 259.15 [M+H]+.
Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{6-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 243.5
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (20 mg, 0.03 mmol), 6-methyl-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (7.3 mg, 0.03 mmol) and K2CO3 (10 mg, 0.08 mmol) in dioxane (3 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (2 mg, 0.003 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 5 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (5:1) to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-{6-methylpyrazolo[1,5-a]pyridin- 5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (20 mg, 86% yield) as a white solid. LCMS observed m/z = 840.27 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{6-methylpyrazolo[1,5- a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 243
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-{6-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (20 mg, 0.02 mmol) in TFA (2 mL) was heated to 50 °C and stirred at 50 °C for 1 h. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18, 5 m, 30 mm x 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 42% B to 55%B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.2) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{6-methylpyrazolo[1,5-a]pyridin-5-yl}-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (3.4 mg, 19% yield ) as a white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.41 (m, 3H), 8.18 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 2.4 Hz, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.66 – 7.59 (m, 1H), 6.68 (s, 1H), 5.48 (s, 2H), 4.73 (m, 1H), 4.08 (s, 1H), 3.80 – 3.75 (m, 2H), 3.56 – 3.44 (m, 1H), 3.23 – 3.13 (m, 3H), 2.99 (m, 1H), 2.83 (m, 1H), 2.65 (s, 3H), 2.51 (s, 3H), 1.37 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 750.15 [M+H]+. Note: Two exchangeable proton were not observed in NMR spectra. EXAMPLE 244 Synthesis of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(6-fluoro-1- methoxy-2,3-dihydro-1H-inden-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 244
Step 1: Preparation of 5-bromo-6-fluoro-2,3-dihydro-1H-inden-1-ol. Compound 244.1
To a stirred mixture of 5-bromo-6-fluoro-2,3-dihydroinden-1-one (2.0 g, 8.7 mmol) in EtOH (30 mL) was added NaBH4 (300 mg, 8.7 mmol) in portions at 0 °C. The resulting mixture was stirred at room temperature for 1 h under nitrogen atmosphere. The mixture was concentrated under reduced pressure to remain about one-third volume. The residue was poured into water (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 5-bromo-6-fluoro-2,3-dihydro-1H-inden-1-ol (2.0 g, 99.5% yield) as a colorless oil. LCMS observed m/z = 230.97 [M-H]-. Step 2: Preparation of 5-bromo-6-fluoro-1-methoxy-2,3-dihydro-1H-indene. Compound 244.2
To a stirred solution of 5-bromo-6-fluoro-2,3-dihydro-1H-inden-1-ol (2.0 g, 8.6 mmol) in THF (8 mL) was added NaH (500 mg, 21.5 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C for 30 min under nitrogen atmosphere. To the above mixture was added CH3I (1.4 g, 10.3 mmol) dropwise at 0 °C. The resulting mixture was stirred at room temperature for additional 8 h under nitrogen atmosphere. The reaction was quenched with water (10 mL) at 0 °C. The resulting mixture
was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 5-bromo-6-fluoro-1-methoxy-2,3-dihydro-1H-indene (1.4 g, 66.5% yield) as a colorless oil. LCMS observed m/z = 244.99 [M+H]+. Step 3: Preparation of 2-(6-fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 244.3
To a stirred solution of 5-bromo-6-fluoro-1-methoxy-2,3-dihydro-1H-indene (500 mg, 2.0 mmol) and B2Pin2 (621 mg, 2.4 mmol) in dioxane (15 mL) were added KOAc (500 mg, 5.0 mmol) and Pd(dppf)Cl2 (149 mg, 0.2 mmol) in portions at room temperature. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. The resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 2-(6-fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (450 mg, 75.0% yield) as a colorless oil. LCMS observed m/z = 293.16 [M+H] +. Step 4: Preparation of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Compound 244.4
Into a 20 mL sealed tube were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine- 4-carbonyl] piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-
N-[2-chloro-4-(trifluoromethyl)phenyl] acetamide (1.0 g, 1.3 mmol) and TFA (10 mL) dropwise at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. Then the mixture was cooled down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4] riazolo[1,5-a]pyrimidin-4-yl}- N-[2-chloro-4-(trifluoromethyl) phenyl] acetamide (760 mg, 85.2% yield) as an off-white solid. LCMS observed m/z = 698.08 [M+H] +. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6- fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 244.5
To a stirred mixture of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate AC (760 mg, 1.0 mmol) and 2-(6-fluoro- 1-methoxy-2,3-dihydro-1H-inden-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (476 mg, 1.5 mmol) in dioxane (10 mL) and H2O (1 mL) were added K2CO3 (375 mg, 2.5 mmol) and Pd(dppf)Cl2 (79 mg, 0.1 mmol) in one portion at room temperature. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-1-methoxy- 2,3-dihydro-1H-inden-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (500 mg, 58.6% yield) as a brown oil. LCMS observed m/z = 784.23 [M+H] +.
Step 6: Preparation of (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(6- fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 244
The crude product N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro- 1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (50 mg, 0.1 mmol) was purified by Prep-SFC with the following conditions (Column: CHIRAL ART amylose-SA, 2 * 25 cm, 5 μm; mobile phase A: hexane (0.1% TFA)--HPLC, mobile phase B: MeOH/DCM = 1/1--HPLC; Flow rate: 20 mL/min; Gradient: isocratic; wave length: 220 nm; RT1 (min): 5.5; RT2 (min): 8.5; Sample solvent: MeOH/DCM=1/1--HPLC; Injection volume: 2 mL; Number of runs: 3) to afford (S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2- (5-ethyl-2-(6-fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (29.8 mg, 59.5% yield) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.59 (s, 1H), 8.07 (d, J = 8.4 Hz, 1H), 7.99 – 7.90 (m, 2H), 7.72 (d, J = 8.6 Hz, 1H), 7.40 – 7.29 (m, 1H), 5.40 (s, 2H), 4.98 – 4.73 (m, 1H), 4.55 (d, J = 12.5 Hz, 1H), 3.63 – 3.48 (m, 3H), 3.34 – 3.20 (m, 4H), 3.10 – 2.94 (m, 4H), 2.88 – 2.76 (m, 2H), 2.72 – 2.65 (m, 1H), 2.45 (s, 3H), 2.41 – 2.32 (m, 1H), 2.05 – 1.92 (m, 1H), 1.44 – 1.20 (m, 3H), 1.19 (s, 1H). LCMS observed m/z = 784.20 [M+H] +. EXAMPLE 245 Synthesis of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(6-fluoro-1- methoxy-2,3-dihydro-1H-inden-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 245
Step 1: Preparation of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(6- fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The crude product N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro- 1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (90 mg, 0.1 mmol) was purified by Prep-SFC with the following conditions (Column: CHIRAL ART amylose-SA, 2 * 25 cm, 5 μm; Mobile phase A: hex (0.1% TFA)--HPLC, mobile phase B: MeOH/DCM=1/1--HPLC; Flowrate: 20 mL/min; Gradient: isocratic; Wave length: 220 nm; RT1 (min): 5.5; RT2 (min): 8.5; Sample solvent: MeOH/DCM=1/1--HPLC; Injection volume: 2 mL; Number of runs: 3) to afford (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2- (5-ethyl-2-(6-fluoro-1-methoxy-2,3-dihydro-1H-inden-5-yl)-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (53.3 mg, 59.2% yield) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 10.24 (s, 1H), 8.59 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.93 (d, J = 6.6 Hz, 1H), 7.72 (d, J = 8.7, 2.1 Hz, 1H), 7.38 – 7.32 (m, 1H), 5.40 (s, 2H), 4.97 – 4.80 (m, 1H), 4.59 – 4.49 (m, 1H), 3.59 – 3.47 (m, 3H), 3.42 – 3.26 (m,
5H), 3.10 – 2.91 (m, 4H), 2.88 – 2.75 (m, 2H), 2.45 (s, 3H), 2.42 – 2.32 (m, 1H), 2.06 – 1.93 (m, 1H), 1.28 – 1.20 (m, 3H). LCMS observed m/z = 784.40 [M+H] +. EXAMPLE 246 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-1'-methyl-4-oxo-7H-1,2',3,3',3a,7,7a'- heptaaza-2,5'-biindenyl-7-yl) acetamide. Compound 246
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-1'-methyl-4-oxo-7H-1,2',3,3',3a,7,7a'-heptaaza-2,5'- biindenyl-7-yl) acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.40 (s, 1H), 8.50 – 8.40 (m, 2H), 8.24 (s, 1H), 7.98 (d, J = 8.6 Hz, 1H), 7.91 (d, J = 2.1 Hz, 1H), 7.68 – 7.61 (m, 1H), 7.55 – 7.50 (m, 1H), 5.36 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.53 – 3.40 (m, 3H), 3.2 - 3.16 (m, 1H), 3.03 – 2.89 (m, 3H), 2.78 (d, J = 11.2 Hz, 1H), 2.66 (s, 3H), 2.63 – 2.58 (m, 1H), 2.37 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 751.2 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 247 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-2'-methyl-4-oxo-7H-1,1',3,3a,7,7a'- hexaaza-2,5'-biindenyl-7-yl) acetamide. Compound 247
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-2'-methyl-4-oxo-7H-1,1',3,3a,7,7a'-hexaaza-2,5'- biindenyl-7-yl) acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.38 (s, 1H), 8.59 (d, J = 7.3 Hz, 1H), 8.44 (s, 1H), 8.24 – 8.20 (m, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.91 (d, J = 2.1 Hz, 1H), 7.68 – 7.60 (m, 1H), 7.36 – 7.30 (m, 1H), 6.52 (s, 1H), 5.34 (s, 2H), 4.47 (d, J = 12.3 Hz, 1H), 3.52 – 3.41 (m, 3H), 3.21 – 3.16 (m, 1H), 2.98 – 2.89 (m, 3H), 2.77 (d, J = 11.2 Hz, 1H), 2.60 (d, J = 9.1 Hz, 1H), 2.36 (s, 3H), 2.34 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.2 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 248 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] {2-[2-(difluoromethyl)-2H- indazol-5-yl]-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}- 4-oxo-1,3,3a,7-tetraaza-7-indenyl} acetamide. Compound 248
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] {2-[2-(difluoromethyl)-2H-indazol-5-yl]-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl} acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.98 (s, 1H), 8.55 (s, 1H), 8.37 (s, 1H), 8.28 (d, J = 11.0 Hz, 1H), 8.07 – 8.03 (m, 1H), 8.03 – 7.96 (m, 1H), 7.89 (s, 1H), 7.78 (d, J = 9.2 Hz, 1H), 7.66 – 7.59 (m, 1H), 5.34 (s, 2H), 4.46 (d, J = 12.5 Hz, 1H), 3.49 – 3.40 (m, 3H), 3.19 - 3.14 (m, 1H), 2.98 – 2.92 (m, 3H), 2.92 – 2.86 (m, 1H), 2.76 (d, J = 11.0 Hz, 1H), 2.63 – 2.56 (m, 1H), 2.31 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 786.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 249
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-2-[(2S)-6-fluoro-2- methyl-2,3-dihydro-1-benzofuran-5-yl]-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 249
Step 1: Preparation of 2-(4-bromo-5-fluoro-2-formylphenoxy)propanoic acid. Compound 249.1
A solution of 5-bromo-4-fluoro-2-hydroxybenzaldehyde (10.0 g, 45.6 mmol) in ACN (50 mL) was treated with K2CO3 (18.9 g, 136.9 mmol) at room temperature for under nitrogen atmosphere followed by the addition of methyl 2-bromopropanoate (9.1 g, 54.7 mmol) dropwise at room temperature. The resulting mixture was stirred at 85 °C for 3 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. To the above mixture was added NaOH (5.5 g, 136.9 mmol) and H2O (50 mL) dropwise at room temperature. The resulting mixture was stirred at 85 °C for additional 2 h. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 291.07 [M+H]+. Step 2: Preparation of 5-bromo-6-fluoro-2-methyl-1-benzofuran. Compound 249.2
A solution of 2-(4-bromo-5-fluoro-2-formylphenoxy)propanoic acid (13.0 g, 44.6 mmol) in acetic anhydride (60 mL) was treated with AcONa (10.9 g, 133.9 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 140 °C for 16 h under nitrogen atmosphere. The mixture was basified to pH 8 with NaOH. The reaction was quenched with water (100 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 80 mL). The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 9% EtOAc in petroleum ether) to afford (679 mg, 7%yield) of 5-bromo-6-fluoro-2-methyl-1-benzofuran as a white solid. LCMS observed m/z = 229.07 [M+H]+. Step 3: Preparation of 2-(6-fluoro-2-methyl-1-benzofuran-5-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane. Compound 249.3
A solution of 5-bromo-6-fluoro-2-methyl-1-benzofuran (647 mg, 2.8 mmol) in dioxane (20 mL) was treated with Pd2(dba)3 (258 mg, 0.3 mmol) and AcOK (554 mg, 5.6 mmol) at room temperature under nitrogen atmosphere followed by the addition of 4,4,5,5- tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.1 g, 8.5 mmol) and PCy3 (158 mg, 0.5 mmol) in portions at room temperature. The resulting mixture was stirred at 100 °C for 16 h under nitrogen atmosphere. The reaction was quenched with water (40 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 40 mL). The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 26% EtOAc in petroleum ether) to afford (600 mg, 77%yield) of 2-(6-fluoro-2-methyl-1-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane as a yellow solid. LCMS observed m/z = 229.07 [M+H]+. Step 4: Preparation of 2-(6-fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane Compound 249.4
A solution of 2-(6-fluoro-2-methyl-1-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (500 mg, 1.8 mmol) in MeOH (20 mL) was treated with Pd/C (500 mg, 0.5 mmol, 10%) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 70 °C for 16 h under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with MeOH (3 x 40 mL). The filtrate was concentrated under reduced pressure. This resulted in (460 mg, 91% yield) of 2-(6-fluoro-2-methyl-2,3-dihydro-1- benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane as a yellow solid. LCMS observed m/z = 278.15 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(6-fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide Compound 249.5
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (300 mg, 0.3 mmol) in dioxane (15 mL) and H2O (3 mL) was treated with Pd(dppf)Cl2 (28 mg, 0.03 mmol) and K2CO3 (157 mg, 1.1 mmol) at room temperature under nitrogen atmosphere followed by the addition of 2-(6- fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (158 mg, 0.5 mmol) in portions at room temperature. The resulting mixture was stirred at 100 °C for 16 h under nitrogen atmosphere. The reaction was quenched with water (50 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 50 mL). The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 50% EtOAc in petroleum ether) to afford (75 mg,
23% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5- ethyl-2-(6-fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 860.27 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6- fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2- (6-fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (70 mg, 0.1 mmol) was treated with TFA (5 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 80 °C for 1 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. This resulted in (50 mg, 80%yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-2-methyl-2,3-dihydro-1-benzofuran-5-yl)- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a yellow solid. LCMS observed m/z = 770.20 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{7-hydroxy-2H,3H,4H-pyrano[3,2-b]pyridine-6- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 249
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-2-methyl-2,3- dihydro-1-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (50 mg, 0.1 mmol) was purified by SFC with the following conditions (Column: CHIRAL ART Cellulose-SB, 2*25 cm, 5 μm; Mobile Phase A: MeOH: DCM=1: 1--HPLC, Mobile Phase B: Hex: DCM=2: 1(0.1% FA)--HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 254 nm; RT1(min): 6.2; RT2(min): 7.7; Sample Solvent: MEOH: DCM=3: 1; Injection Volume: 0.5 mL; Number Of Runs: 8) to afford (11.5 mg, 23% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-2-[(2S)-6-fluoro-2-methyl-2,3-dihydro-1-benzofuran- 5-yl]-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.57 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.85 (d, J = 7.5 Hz, 1H), 7.76 – 7.69 (m, 1H), 6.80 (m, 1H), 5.38 (s, 1H), 5.07 (q, J = 7.3 Hz, 1H), 4.54 (m, 1H), 3.56 (m, 1H), 3.51 (s, 2H), 3.42 – 3.33 (m, 2H), 3.25 (m, 1H), 3.02 (d, J = 9.2 Hz, 3H), 2.82 (m, 2H), 2.65 (s, 1H), 2.45 (s, 3H), 1.42 (d, J = 6.2 Hz, 3H), 1.25 – 1.20 (m, 3H). LCMS observed m/z = 770.20 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 250 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-2-[(2R)-6-fluoro-2- methyl-2,3-dihydro-1-benzofuran-5-yl]-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 250
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(6-fluoro-2-methyl-2,3- dihydro-1-benzofuran-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (50 mg, 0.1 mmol) was purified by SFC with the following conditions (Column: CHIRAL ART Cellulose-SB, 2*25 cm, 5 μm; Mobile Phase A: MeOH: DCM=1: 1--HPLC, Mobile Phase B: Hex: DCM=2: 1(0.1% FA)--HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 254 nm; RT1(min): 6.2; RT2(min): 7.7; Sample Solvent: MEOH: DCM=3: 1; Injection Volume: 0.5 mL; Number Of Runs: 8) to afford (13.9 mg, 28%yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-2-[(2R)-6-fluoro-2-methyl-2,3-dihydro-1-benzofuran- 5-yl]-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H), 8.57 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.85 (d, J = 7.5 Hz, 1H), 7.76 – 7.69 (m, 1H), 6.80 (m, 1H), 5.38 (s, 2H), 5.07 (q, J = 7.2 Hz, 1H), 4.54 (m, 1H), 3.53 (m, 3H), 3.42 – 3.33 (m, 1H), 3.25 (m, 1H), 3.02 (d, J = 9.2 Hz, 3H), 2.84 (m, 3H), 2.45 (s, 3H), 1.42 (d, J = 6.2 Hz, 4H), 1.15 (t, J = 7.2 Hz, 3H). LCMS observed m/z = 770.15 [M+H]+. EXAMPLE 251 Synthesis of of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{2- fluoropyrazolo[1,5-a]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide. Compound 251
Step 1: Preparation of 1-amino-4-(benzyloxy)pyridin-1-ium 2,4,6- trimethylbenzenesulfonate Compound 251.1
A solution of 4-(benzyloxy) pyridine (2.0 g, 10.8 mmol) and amino 2,4,6- trimethylbenzenesulfonate (2.3 g, 10.8 mmol) in DCM (20 mL) was stirred at room temperature for 18 hours under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 201.10 [M+H]+. Step 2: Preparation of 5-(benzyloxy)-2-fluoropyrazolo[1,5-a] pyridine Compound 251.2
To a stirred mixture of 1-amino-4-(benzyloxy) pyridin-1-ium 2,4,6- trimethylbenzenesulfonate (4.3 g, 10.7 mmol and 2,2-difluoroethenyl 4- methylbenzenesulfonate (2.5 g, 10.7 mmol) in DMF (120 mL) was added K2CO3 (5.2 g, 37.6 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 90 ℃ for additional 1 hour. Desired product could be detected by LCMS. The reaction was quenched with H2O (200 mL) at room temperature. The resulting mixture was extracted with EA (200 mL x 3). The combined organic layers were washed with brine (150 mL x 2), dried over anhydrous Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 60% EtOAc in
petroleum ether) to afford (2.0 g, 76.9% yield) of 5-(benzyloxy)-2-fluoropyrazolo[1,5-a] pyridine as a yellow solid. LCMS observed m/z = 243.09 [M+H]+. Step 3: Preparation of 2-fluoropyrazolo[1,5-a] pyridin-5-ol Compound 251.3
A solution of 5-(benzyloxy)-2-fluoropyrazolo[1,5-a] pyridine (400 mg, 1.7 mmol) and Pd/C (10%, 404 mg, 0.4 mmol) in THF (5 mL) was stirred at room temperature for 1 hour under H2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was filtered, the filter cake was washed with THF (5 mL x 3). The filtrate was concentrated under reduced pressure to afford (220 mg, crude) of 2- fluoropyrazolo[1,5-a] pyridin-5-ol as a yellow oil. LCMS observed m/z = 151.04 [M-H]-. Step 4: Preparation of 2-fluoropyrazolo[1,5-a] pyridin-5-yl trifluoromethanesulfonate Compound 251.4
To a stirred solution of 2-fluoropyrazolo[1,5-a] pyridin-5-ol (210 mg, 1.4 mmol) and TEA (576 uL, 4.1 mmol) in DCM (5 mL) was added Tf2O (350 uL, 2.070 mmol) dropwise at 0 ℃ under N2 atmosphere. The resulting mixture was stirred at room temperature for additional 1 hour. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 60% EtOAc in petroleum ether) to afford (320 mg, 81.6% yield) of 2-fluoropyrazolo[1,5-a] pyridin-5-yl trifluoromethanesulfonate as a yellow solid. LCMS observed m/z = 284.99 [M+H]+. Step 5: Preparation of 2-fluoropyrazolo[1,5-a] pyridin-5-ylboronic acid Compound 251.5
To a stirred solution of 2-fluoropyrazolo[1,5-a] pyridin-5-yl trifluoromethanesulfonate (300 mg, 1.1 mmol) and bis(pinacolato)diboron (402 mg, 1.6
mmol) in 1,4-dioxane (6 mL) were added Pd(dppf)Cl2CH2Cl2 (86 mg, 0.1 mmol) and AcOK (311 mg, 3.2 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 100 ℃ for additional 2 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 181.05 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{2- fluoropyrazolo[1,5-a]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide Compound 251
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide, Intermediate AC (200 mg, 0.3 mmol) and 2- fluoropyrazolo[1,5-a] pyridin-5-ylboronic acid (68 mg, 0.4 mmol) in dioxane (5 mL) and H2O (1 mL) were added Pd(dppf)Cl2CH2Cl2 (21 mg, 0.1 mmol) and K2CO3 (105 mg, 0.8 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 100 ℃ for additional 3 hours. Desired product could be detected by LCMS. The reaction was quenched with H2O (8 mL). The aqueous layer was extracted with EA (10 mL x 3). The combined organic layers were washed with brine (8 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 33% B to 50%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.62) to afford (33.2 mg, 17.4% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{2- fluoropyrazolo[1,5-a]pyridin-5-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4-
carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ 10.26 (s, 1H), 8.70 (d, J = 7.3 Hz, 1H), 8.53 (s, 1H), 8.34 (t, J = 1.4 Hz, 1H), 8.07 (d, J = 8.4 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 7.55 (dd, J = 7.3, 2.0 Hz, 1H), 6.52 (d, J = 5.1 Hz, 1H), 5.41 (s, 2H), 4.54 (m, 1H), 3.52 (m, 3H), 3.25 (m, 1H), 3.10 – 2.96 (m, 3H), 2.85 (m, 1H), 2.67 (d, J = 9.0 Hz, 1H), 2.43 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 754.10 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 252 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-1'-methyl-4-oxo-7H,1'H-1,1',3,3a,7,7'- hexaaza-2,5'-biindenyl-7-yl)acetamide. Compound 252
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6- methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-2-bromo-6-ethyl-4-oxo-4,7-dihydro- 1,3,3a,7-tetraaza-7-indenyl]acetamide (60 mg, 76 µmol, Intermediate H), (1-methyl-1H- 1,7-diazainden-5-yl)boranediol 18.1 mg, 103 µmol) and K3PO4 (54.7 mg, 258 µmol) in a mixture of dioxane (340 µL) and H2O (110 µL) was added Pd(dppf)Cl2.CH2Cl2 (7.01 mg, 8.59 µmol). The mixture was stirred at 80 °C under N2 for 4 hours. The mixture was concentrated in vacuo. The residue was purified by reversed phase flash chromatography (Column: Xbridge C185 µm, 19 mm X 100 mm; mobile phase: 5-95% MeCN in H2O with 0.1% NH4OH modifier) to afford (30.1 mg, 47% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-1'-methyl-4-oxo-7H,1'H-1,1',3,3a,7,7'-hexaaza-2,5'-biindenyl-7-yl)acetamide as a light brown solid.1H NMR (400MHz, DMSO-d6) δ 10.44 (m, 2H), 9.00 (d, J = 2.0 Hz, 1H), 8.64 (d, J = 2.0 Hz, 1H), 8.55 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H),
7.71 (dd, J = 8.7, 2.1 Hz, 1H), 7.62 (d, J = 3.4 Hz, 1H), 6.60 (d, J = 3.5 Hz, 1H), 5.42 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.86 (s, 3H), 3.63 – 3.44 (m, 3H), 3.28 – 3.23 (m, 1H), 3.12 – 2.92 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 9.8 Hz, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.2 [M+H]+. EXAMPLE 253 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H-1,3,3',3a,3a',7-hexaaza-2,5'- biindenyl-7-yl)acetamide. Compound 253
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(1,7a-diaza-6- indenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.46 (s, 1H), 9.22 (d, J = 1.3 Hz, 1H), 8.54 (s, 1H), 8.13 (d, J = 2.2 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.90 – 7.78 (m, 2H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 6.74 (d, J = 2.2 Hz, 1H), 5.41 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.59 – 3.48 (m, 3H), 3.28 – 3.22 (m, 1H), 3.11 – 2.94 (m, 3H), 2.84 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 9.8 Hz, 1H), 2.43 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 736.2 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 254 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(1-methyl-1H-1,3-benzimidazol-5-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 254
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (1-methyl-1H- 1,3-benzimidazol-5-yl)boranediol.1H NMR (400MHz, DMSO-d6) δ 10.45 (s, 1H), 10.24 (s, 1H), 8.59 (s, 1H), 8.37 (d, J = 1.5 Hz, 1H), 8.28 (s, 1H), 8.11 – 8.01 (m, 2H), 7.98 (d, J = 2.1 Hz, 1H), 7.74 – 7.66 (m, 2H), 5.42 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 3.88 (s, 3H), 3.63 – 3.44 (m, 3H), 3.29 – 3.20 (m, 1H), 3.13 – 2.93 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.65 (d, J = 30.9 Hz, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.2 [M+H]+. EXAMPLE 255 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2'-methyl-4-oxo-7H-1,1',3,3a,3a',7- hexaaza-2,5'-biindenyl-7-yl)acetamide. Compound 255
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (2-methyl-1,3a- diaza-5-indenyl)boranediol.1H NMR (400MHz, DMSO-d6) δ 10.42 (s, 1H), 10.24 (s, 1H), 9.24 (t, J = 1.4 Hz, 1H), 8.59 (s, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.86
(s, 1H), 7.82 (dd, J = 9.4, 1.8 Hz, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.57 (d, J = 9.4 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.63 – 3.45 (m, 3H), 3.28 – 3.20 (m, 1H), 3.12 – 2.92 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 10.6 Hz, 1H), 2.45 (s, 3H), 2.35 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.3 [M+H]+. EXAMPLE 256 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [6-ethyl-2-(6-fluoro-2-methyl- 2H-indazol-5-yl)-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4- oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 256
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [6-ethyl-2-(6-fluoro-2-methyl-2H-indazol- 5-yl)-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.38 (s, 1H), 8.48 (s, 1H), 8.40 (d, J = 7.3 Hz, 1H), 8.35 (d, J = 5.4 Hz, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.89 (d, J = 2.1 Hz, 1H), 7.68 – 7.60 (m, 1H), 7.42 (d, J = 12.1 Hz, 1H), 5.33 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 4.11 (s, 3H), 3.45 - 3.41 (m, 3H), 3.20 - 3.15 m, 1H), 2.93 - 2.88 (m, 3H), 2.77 (d, J = 11.1 Hz, 1H), 2.63 – 2.56 (m, 1H), 2.33 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 768.4 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 257 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,3-dihydro-2- benzothiophen-5-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide. Compound 257
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,3-dihydro-2-benzothiophen-5-yl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl] acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.37 (s, 1H), 8.35 (d, J = 20.6 Hz, 1H), 7.99 (d, J = 8.5 Hz, 2H), 7.93 – 7.87 (m, 2H), 7.67 – 7.62 (m, 1H), 7.38 (d, J = 8.1 Hz, 1H), 5.32 (s, 2H), 4.46 (d, J = 12.5 Hz, 1H), 4.33 – 4.12 (m, 4H), 3.48 – 3.41 (m, 3H), 3.19 – 3.14 (m, 1H), 2.98 – 2.87 (m, 3H), 2.76 (d, J = 11.1 Hz, 1H), 2.62 – 2.54 (m, 1H), 2.34 (s, 3H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 754.2 [M+H]+. EXAMPLE 258 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2H-indazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 258
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[5-(benzyloxy)-6- methyl-4-pyrimidinyl]carbonyl}-1-piperazinyl)-2-bromo-6-ethyl-4-oxo-4,7-dihydro- 1,3,3a,7-tetraaza-7-indenyl]acetamide (70 mg, 100 µmol, Intermediate H), (2H-indazol-5- yl)boranediol (24.3 mg, 150 µmol) and K3PO4 (63.7 mg, 300 µmol) in a mixture of dioxane (667 µL) and H2O (100 µL) was added Pd(dppf)Cl2.CH2Cl2 (7.31 mg, 10 µmol). The mixture was stirred at 80 °C under N2 for 16 hours. The mixture was concentrated in vacuo.
The residue was purified by reversed phase flash chromatography (Column: Xbridge C185 µm, 19 mm X 100 mm; mobile phase: 5-95% MeCN in H2O with 0.1% NH4OH modifier) to afford (30.1 mg, 47% yield) of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2H-indazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a light brown solid. 1H NMR (400 MHz, DMSO) δ 13.29 (s, 1H), 10.44 (s, 1H), 10.25 (s, 1H), 8.57 (d, J = 16.3 Hz, 2H), 8.21 (s, 1H), 8.16 – 8.02 (m, 2H), 7.99 (d, J = 2.1 Hz, 1H), 7.82 – 7.61 (m, 2H), 5.43 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.54 (t, J = 10.8 Hz, 3H), 3.26 (d, J = 12.6 Hz, 1H), 3.03 (d, J = 10.1 Hz, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.68 (d, J = 10.7 Hz, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 736.2 [M+H]+. EXAMPLE 259 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 259
Step 1: Preparation of 3-methyl-6-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 4(3H)-quinazolinone. Compound 259.1
To a mixture of 6-bromo-3-methyl-3,4-dihydro-4-quinazolinone (478 mg, 2 mmol), KOAc (785 mg, 8 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3,2-dioxaborolane (762 mg, 3 mmol) in dioxane (4.5 mL) was added
Pd(dppf)Cl2.CH2Cl2 (163 mg, 200 µmol) at 25 °C under N2. The mixture was heated to 100 °C and continued to stir for 16 hours at the same temperature. The resulting mixture was cooled to 25 °C, filtered and the filter cake was washed with EtOAc (30 mL). The filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: with 0-100% EtOAc in hexanes) to afford (401 mg, 70% yield) of 3-methyl-6- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4(3H)-quinazolinone as a brown solid mixted with 30% pinacol. The mixture was used for nest step without further purification. LCMS observed m/z = 287.4 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 259
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 3-methyl-6- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4(3H)-quinazolinone. 1H NMR (400 MHz, DMSO) δ 10.47 (s, 1H), 10.28 (broad, 1H), 8.90 (d, J = 2.0 Hz, 1H), 8.59 (s, 1H), 8.50 (dd, J = 8.5, 2.1 Hz, 1H), 8.46 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.82 (d, J = 8.5 Hz, 1H), 7.71 (dd, J = 8.6, 2.1 Hz, 1H), 5.45 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.53 (s, 6H), 3.28 (s, 1H), 3.14 – 2.95 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.69 (d, J = 11.1 Hz, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 778.60 [M+H]+. EXAMPLE 260 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(7-fluoro-3,4- dihydro-1H-2-benzopyran-6-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 260
Step 1: Preparation of methyl 4-bromo-5-fluoro-2-iodobenzoate. Compound 260.1
To a stirred solution of methyl 2-amino-4-bromo-5-fluorobenzoate (2.5 g, 10.0 mmol) in ACN (20 mL) and HCl (50 mL, 300.0 mmol, 6 M in water) was added NaNO2 (1.0 g, 15.0 mmol) aqueous dropwise at 0 °C. The resulting mixture was stirred at 0 °C for 1 h. To the above mixture was added KI (3.3 g, 20.0 mmol) aqueous dropwise at 0 °C. The resulting mixture was stirred at room temperature for additional overnight. After completion of reaction, the resulting mixture was diluted with H2O (100 mL) at room temperature and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford methyl 4-bromo-5-fluoro-2-iodobenzoate (2.0 g, 52.3% yield) as an off-white solid.1H NMR (400 MHz, Chloroform-d) δ 8.23 (d, J = 6.6 Hz, 1H), 7.65 (d, J = 8.9 Hz, 1H), 3.96 (s, 3H). Step 2: Preparation of methyl 4-bromo-2-[(E)-2-ethoxyethenyl]-5-fluorobenzoate. Compound 260.2
To a stirred solution of methyl 4-bromo-5-fluoro-2-iodobenzoate (2.0 g, 5.5 mmol) and 2-[(E)-2-ethoxyethenyl]-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1.1 g, 5.5 mmol) in dioxane (40 mL) and H2O (10 mL) were added Pd(dppf)Cl2 (244 mg, 0.33 mmol) and NaHCO3 (1.4 g, 16.5 mmol) in portions at room temperature. The resulting mixture was
stirred at 80 °C for 1 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was quenched with H2O (30 mL) at room temperature and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 60% to 65% gradient in 5 min; detector, UV 254 nm) to afford methyl 4-bromo- 2-[(E)-2-ethoxyethenyl]-5-fluorobenzoate (620 mg, 37.2% yield) as a white solid. LCMS observed m/z = 303.00 [M+H] +. Step 3: Preparation of methyl 4-bromo-5-fluoro-2-(2-oxoethyl)benzoate. Compound 260.3
To a stirred solution of methyl 4-bromo-2-[(E)-2-ethoxyethenyl]-5-fluorobenzoate (580 mg, 1.9 mmol) in DCM (20 mL) was added TFA (0.2 mL) dropwise at 0 °C. The resulting mixture was stirred at 0 °C for 1 h. After completion of reaction, the reaction was quenched with saturated NaHCO3 aqueous at room temperature. The resulting mixture was extracted with DCM (3 x 30 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (589 mg) was used in the next step directly without further purification. LCMS observed m/z =
. [M+H] +. Step 4: Preparation of 2-[5-bromo-4-fluoro-2-(hydroxymethyl)phenyl]ethanol. Compound 260.4
To a stirred solution of methyl 4-bromo-5-fluoro-2-(2-oxoethyl)benzoate (580 mg, 2.1 mmol) in THF (10 mL) was added LiAlH4 (1.0 M in THF, 2 mL, 2.1 mmol) dropwise at -30 °C under nitrogen atmosphere. The resulting mixture was stirred at -30 °C for 30 min under nitrogen atmosphere. The reaction was quenched with saturated potassium sodium tartrate aqueous (50 mL) at -30 °C. After completion of reaction, the resulting mixture was extracted with Et2O (3 x 20 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under
reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% NH3.H2O), 18% to 20% gradient in 3 min; detector, UV 254 nm) to afford 2-[5-bromo-4- fluoro-2-(hydroxymethyl)phenyl]ethanol (204 mg, 38.4% yield) as a colorless oil. LCMS observed m/z = 248.98 [M+H] +. Step 5: Preparation of 6-bromo-7-fluoro-3,4-dihydro-1H-2-benzopyran. Compound 260.5
To a stirred solution of 2-[5-bromo-4-fluoro-2-(hydroxymethyl)phenyl]ethanol (204 mg, 0.8 mmol) in CPME (8 mL) was added NaH (64 mg, 1.6 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C for 10 min under nitrogen atmosphere. To the above mixture was added trimethyl phosphate (286 mg, 2.0 mmol) dropwise at 0 °C. The resulting mixture was stirred at room temperature for additional 4 h. The reaction was quenched with water (20 mL) at 0 °C. The resulting mixture was extracted with Et2O (3 x 20 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with n-pentane/EA (20/1) to afford 6-bromo-7-fluoro-3,4-dihydro-1H-2-benzopyran (94 mg, 50.3% yield) as a colorless oil.1H NMR (400 MHz, Chloroform-d) δ 7.33 (d, J = 6.7 Hz, 1H), 6.78 (d, J = 8.7 Hz, 1H), 4.71 (s, 2H), 3.96 (t, J = 5.7 Hz, 2H), 2.82 (t, J = 5.6 Hz, 2H). Step 6: Preparation of 2-(7-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane. Compound 260.6
To a stirred solution of 6-bromo-7-fluoro-3,4-dihydro-1H-2-benzopyran (50 mg, 0.2 mmol) in THF (10 mL) was added n-BuLi (2.5 M in n-hexane, 34 mg, 0.5 mmol) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was stirred at -78 °C for 1 h under nitrogen atmosphere. To the above mixture was added 4,4,5,5-tetramethyl-2-(propan-
2-yloxy)-1,3,2-dioxaborolane (48 mg, 0.3 mmol) dropwise at -78 °C. The resulting mixture was stirred at room temperature for additional 2 h. After completion of reaction, the reaction was quenched with water (20 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product (80 mg) was used in the next step directly without further purification. LCMS observed m/z = 279.15 [M+H] +. Step 7: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(7-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 260.7
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (40 mg, 0.05 mmol) and 2- (7-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (59 mg, 0.2 mmol) in dioxane (3 mL) and H2O (0.6 mL) were added Pd(dppf)Cl2 (4 mg, 0.005 mmol) and K2CO3 (14 mg, 0.1 mmol) in portions at room temperature. The resulting mixture was stirred at 90 °C for 1 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched with water (20 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 60% to 65% gradient in 5 min; detector, UV 254 nm) to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-(7-fluoro-3,4-dihydro-1H-2- benzopyran-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-
(trifluoromethyl)phenyl]acetamide (30 mg, 68.3% yield) as a brown solid. LCMS observed m/z = 860.26 [M+H] +. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(7- fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 260
Into a 4 mL sealed tube were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-(7-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (30 mg, 0.03 mmol) and TFA (0.5 mL) at room temperature. The resulting mixture was stirred at room temperature for 48 h. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: water (10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 45% B in 7 min; Wave Length: 254nm/220nm; RT1 (min): 6.57) to afford N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(7-fluoro-3,4-dihydro-1H-2-benzopyran-6-yl)-6- [4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (14.3 mg, 53.4% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.18 (s, 1H), 8.62 (s, 1H), 8.49 (d, J = 8.6 Hz, 1H), 8.00 (d, J = 7.1 Hz, 1H), 7.62 (d, J = 2.1 Hz, 1H), 7.59 – 7.50 (m, 1H), 6.86 (d, J = 10.8 Hz, 1H), 5.71 (d, J = 13.2 Hz, 1H), 5.20 (s, 2H), 4.90 – 4.75 (m, 3H), 4.02 (t, J = 5.7 Hz, 2H), 3.86 (t, J = 11.7 Hz, 2H), 3.58 – 3.49 (m, 1H), 3.35 – 3.22 (m, 2H), 3.19 – 3.02 (m, 1H), 2.95 – 2.75 (m, 4H), 2.59 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 770.15 [M+H] +. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 261
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,2-trimethylisoindolin-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 261
Step 1: Preparation of 6-bromo-2,3,3-trimethylisoindolin-1-one. Compound 261.1
To a stirred solution of 6-bromo-2-methyl-3H-isoindol-1-one (500 mg, 2.2 mmol) in THF (5 mL) was added NaH (265 mg, 11.1 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was warmed to 25 °C and stirred at 25 °C for 1 h under nitrogen atmosphere. Then the reaction mixture was cooled to 0 °C. To the above mixture was added iodomethane (413 uL, 6.6 mmol) dropwise over 1 min at 0 °C. The resulting mixture was warmed to 25 °C and stirred at 25 °C for 16 h under nitrogen atmosphere. The reaction was quenched with water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (20 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (3:1) to afford 6- bromo-2,3,3-trimethylisoindol-1-one (400 mg, 71% yield) as an off-white solid. LCMS observed m/z = 254.01 [M+H]+. Step 2: Preparation of 5-bromo-1,1,2-trimethylisoindoline. Compound 261.2
A solution of 6-bromo-2,3,3-trimethylisoindol-1-one (400 mg, 1.6 mmol) in borane- tetrahydrofuran complex (1.0 M in THF, 20 mL, 20.0 mmol) was stirred at 60 °C for 8 h under nitrogen atmosphere. The reaction was cooled to room temperature and quenched with MeOH. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (3:1) to afford 5-bromo-1,1,2-trimethylisoindoline (300 mg, 71% yield) as a dark blue oil. LCMS observed m/z = 240.03 [M+H]+. Step 3: Preparation of 1,1,2-trimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-3H-isoindole. Compound 261.3
To a stirred solution of 5-bromo-1,1,2-trimethylisoindoline (330 mg, 1.4 mmol) and bis(pinacolato)diboron (418 mg, 1.6 mmol) in dioxane (1 mL) were added Pd(dppf)Cl2.CH2Cl2 (112 mg, 0.1 mmol) and AcOK (674 mg, 6.9 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 12 h under nitrogen atmosphere. The mixture was cooled to room temperature and filtered. The filter cake was washed with CH2Cl2 (3 x 5 mL) and the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE / EA 5:1) to afford 1,1,2-trimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3H-isoindole (75 mg, 19% yield) as an off-white solid. LCMS observed m/z = 288.21 [M+H]+. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-(1,1,2-trimethyl-3H-isoindol-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 261.4
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (137 mg, 0.1 mmol) and 1,1,2-trimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3H-isoindole (75 mg, 0.2 mmol) in dioxane (2 mL) and H2O (0.4 mL) were added K2CO3 (87 mg, 0.6 mmol) and Pd(dppf)Cl2.CH2Cl2 (14 mg, 0.02 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2 h under nitrogen atmosphere. The mixture was cooled to room temperature and filtered. The filter cake was washed with CH2Cl2 (3x5 mL) and the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18, silica gel; mobile phase, MeCN in water (0.1% NH3.H2O), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(6-{4- [5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-(1,1,2- trimethyl-3H-isoindol-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (95 mg, 62% yield) as a light yellow solid. LCMS observed m/z = 869.32 [M+H]+. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,2- trimethylisoindolin-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 261
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-(1,1,2-trimethyl-3H-isoindol-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (90 mg, 0.1 mmol) in TFA (2 mL) was stirred at 50 °C for 3 h. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was redissolved in EtOAc (10 mL) and washed with sat. NaHCO3 (aq.3 x 5 mL). The organic layer was dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Mobile Phase A: 0.1% NH4HCO3 in water; Mobile Phase B: ACN; Flow rate: 60 mL/min) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(1,1,2-trimethylisoindolin-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (32.4 mg, 39% yield) as an off-white solid.1H NMR (400 MHz, Methanol-d4) δ 8.40 (s, 1H), 8.15 (m, 3H), 7.83 (d, J = 2.1 Hz, 1H), 7.65 – 7.58 (m, 1H), 7.35 (d, J = 7.9 Hz, 1H), 5.46 (s, 2H), 4.72 (m, 1H), 4.12 (s, 2H), 4.01 (s, 1H), 3.75 (m, 2H), 3.48 (t, J = 12.1 Hz, 1H), 3.14 (d, J = 8.0 Hz, 3H), 2.96 (d, J = 11.5 Hz, 1H), 2.81 (d, J = 11.5 Hz, 1H), 2.60 (s, 3H), 2.49 (s, 3H), 1.41 (s, 6H), 1.34 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 779.25 [M+H]+. Note: Two exchangeable proton were not observed in NMR spectra. EXAMPLE 262 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-isopropyl-2H-indazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 262
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(2-isopropyl- 2H-indazol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400 MHz, DMSO) δ 10.37 (s, 1H), 8.49 (d, J = 4.6 Hz, 2H), 8.44 (d, J = 1.4 Hz, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.94 – 7.87 (m, 2H), 7.65 (d, J = 9.0 Hz, 2H), 5.34 (s, 2H), 4.79 (p, J = 6.6 Hz, 1H), 4.47 (d, J = 12.4 Hz, 1H), 3.53 – 3.42 (m, 3H), 3.18 (d, J = 12.4 Hz, 1H), 2.95 (d, J = 9.9 Hz, 3H), 2.78 (d, J = 11.3 Hz, 1H), 2.60 (d, J = 10.3 Hz, 1H), 2.37 (s, 3H), 1.50 (d, J = 6.6 Hz, 6H), 1.15 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 778.8 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 263 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(6-quinazolinyl)-1,3,3a,7- tetraaza-7-indenyl) acetamide. Compound 263
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(6-quinazolinyl)-1,3,3a,7-tetraaza-7- indenyl) acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.52 (s, 1H), 9.80 (s, 1H), 9.36 (s, 1H), 8.93 (d, J = 1.9 Hz, 1H), 8.73 – 8.68 (m, 1H), 8.39 (s, 1H), 8.17 (d, J = 8.9 Hz, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.73 – 7.68 (m, 1H), 5.45 (s, 2H), 4.54
(d, J = 12.5 Hz, 1H), 3.58 – 3.49 (m, 3H), 3.31 - 3.23 (m, 1H), 3.09 – 2.93 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.71 – 2.65 (m, 1H), 2.40 (s, 3H), 1.23 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 748.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 264 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-2-(6-quinolyl)-1,3,3a,7-tetraaza-7- indenyl)acetamide. Compound 264
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(6-quinolyl)-1,3,2-dioxaborolane.1H NMR (400MHz, DMSO-d6) δ 10.45 (s, 1H), 10.25 (s, 1H), 8.97 (dd, J = 4.2, 1.7 Hz, 1H), 8.77 (d, J = 1.9 Hz, 1H), 8.58 (s, 1H), 8.55 (d, J = 8.2 Hz, 1H), 8.46 (dd, J = 8.9, 1.9 Hz, 1H), 8.15 (d, J = 8.8 Hz, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.61 (dd, J = 8.3, 4.1 Hz, 1H), 5.45 (s, 2H), 4.61 – 4.46 (m, 1H), 3.63 – 3.45 (m, 3H), 3.29 – 3.21 (m, 1H), 3.12 – 2.95 (m, 3H), 2.87 (d, J = 11.2 Hz, 1H), 2.77 – 2.63 (m, 1H), 2.45 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 747.3 [M+H]+. EXAMPLE 265 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(2,1-benzisoxazol-6-yl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide. Compound 265
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(2,1- benzisoxazol-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO- d6) δ 10.48 (s, 1H), 9.89 (s, 1H), 8.46 (s, 1H), 8.34 (d, J = 1.3 Hz, 1H), 8.05 (d, J = 8.5 Hz, 1H), 7.97 (d, J = 2.3 Hz, 1H), 7.92 (dd, J = 9.1, 1.1 Hz, 1H), 7.81 (dd, J = 9.0, 1.2 Hz, 1H), 7.71 (dd, J = 8.5, 2.1 Hz, 1H), 5.42 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 3.59 – 3.45 (m, 3H), 3.27 – 3.19 (m, 1H), 3.10 – 2.90 (m, 3H), 2.84 (d, J = 11.3 Hz, 1H), 2.72 – 2.60 (m, 1H), 2.41 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 737.2 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 266 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(2,1-benzisothiazol-5-yl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide. Compound 266
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(2,1- benzisothiazol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400MHz, DMSO-
d6) δ 10.46 (s, 1H), 9.97 (s, 1H), 8.71 (d, J = 1.6 Hz, 1H), 8.50 (s, 1H), 8.20 (dd, J = 9.2, 1.7 Hz, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.92 (d, J = 9.3 Hz, 1H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 5.42 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.61 – 3.44 (m, 3H), 3.28 – 3.21 (m, 1H), 3.11 – 2.92 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.67 (d, J = 10.7 Hz, 1H), 2.42 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 753.3 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 267 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-2-{p-[(1- pyrrolidinyl)carbonyl]phenyl}-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 267
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with [p- (dihydroxyboryl)phenyl](1-pyrrolidinyl)methanone. 1H NMR (400MHz, DMSO-d6) δ 10.41 (s, 1H), 10.25 (s, 1H), 8.57 (s, 1H), 8.15 (d, J = 8.4 Hz, 2H), 8.06 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.8, 2.2 Hz, 1H), 7.66 (d, J = 8.3 Hz, 2H), 5.41 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 3.59 – 3.37 (m, 7H), 3.29 – 3.22 (m, 1H), 3.09 – 2.94 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.73 – 2.62 (m, 1H), 2.45 (s, 3H), 1.85 (dq, J = 17.8, 6.8 Hz, 4H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 793.3 [M+H]+. EXAMPLE 268 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-2-(3,4,5-trimethoxyphenyl)- 1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 268
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (3,4,5- trimethoxyphenyl)boranediol.1H NMR (400MHz, DMSO-d6) δ 10.44 (s, 1H), 10.26 (s, 1H), 8.57 (s, 1H), 8.05 (d, J = 8.5 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.37 (s, 2H), 5.39 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 3.85 (s, 6H), 3.72 (s, 3H), 3.61 – 3.44 (m, 3H), 3.29 – 3.20 (m, 1H), 3.10 – 2.94 (m, 3H), 2.84 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 9.7 Hz, 1H), 2.44 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 786.3 [M+H]+. EXAMPLE 269 Synthesis of N-cyclopropylp-(7-{[N-2-chloro-4- (trifluoromethyl)phenylcarbamoyl]methyl}-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-2-indenyl)benzamide. Compound 269
The title compound was prepared using a similar procedure compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with N-cyclopropylp- (dihydroxyboryl)benzamide. 1H NMR (400MHz, DMSO-d6) δ 10.43 (s, 1H), 8.56 (d, J = 5.4 Hz, 2H), 8.17 (d, J = 8.3 Hz, 2H), 8.05 (d, J = 8.6 Hz, 1H), 8.01 – 7.89 (m, 3H), 7.71 (d, J = 8.1 Hz, 1H), 5.41 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 3.52 (d, J = 12.5 Hz, 3H), 3.25 (d,
J = 12.3 Hz, 1H), 3.03 (d, J = 10.1 Hz, 3H), 2.87 (td, J = 9.4, 5.8 Hz, 2H), 2.67 (s, 1H), 2.44 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H), 0.70 (dd, J = 7.1, 4.7 Hz, 2H), 0.64 – 0.53 (m, 2H). LCMS observed m/z = 779.3 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 270 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]{2-[p-(1- azetidinylsulfonyl)phenyl]-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl}acetamide. Compound 270
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with [p-(1- azetidinylsulfonyl)phenyl]boranediol.1H NMR (400MHz, DMSO-d6) δ 10.42 (s, 1H), 8.55 (s, 1H), 8.43 – 8.33 (m, 2H), 8.07 (d, J = 8.6 Hz, 1H), 8.02 – 7.90 (m, 3H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 5.43 (s, 2H), 4.54 (d, J = 12.3 Hz, 1H), 3.71 (t, J = 7.7 Hz, 5H), 3.62 – 3.46 (m, 3H), 3.29 – 3.21 (m, 1H), 3.12 – 2.94 (m, 3H), 2.86 (d, J = 11.3 Hz, 1H), 2.74 – 2.62 (m, 1H), 2.44 (s, 3H), 2.00 (p, J = 7.6 Hz, 2H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 815.3 [M+H]+. EXAMPLE 271 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-2-oxoindolin-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 271
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 1-methyl-5- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-indolinone. 1H NMR (400 MHz, DMSO) δ 10.46 (s, 2H), 8.37 (s, 1H), 8.06 (dd, J = 8.7, 2.8 Hz, 2H), 7.98 (dd, J = 9.9, 1.9 Hz, 2H), 7.70 (dd, J = 8.9, 2.2 Hz, 1H), 7.11 (d, J = 8.2 Hz, 1H), 5.38 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 3.65 (s, 2H), 3.55 – 3.45 (m, 3H), 3.23 (d, J = 12.2 Hz, 1H), 3.16 (s, 3H), 3.06 – 2.92 (m, 3H), 2.82 (d, J = 11.4 Hz, 1H), 2.66 (d, J = 10.8 Hz, 1H), 2.39 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 765.3 [M+H]+. EXAMPLE 272 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{3-methylpyrazolo[1,5-a]pyridin-5-yl}-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
Step 1: Preparation of {5-bromopyrazolo[1,5-a]pyridin-3-yl}methanol Compound 272.1
To a stirred solution of ethyl 5-bromopyrazolo[1,5-a] pyridine-3-carboxylate (2.0 g, 7.4 mmol) in THF (40 mL) was added Diisobutylaluminum hydride (1.0 M in THF) (29.7 mL, 29.7 mmol) dropwise at -60℃ under N2 atmosphere. The resulting mixture was stirred at room temperature for additional 16 hours. Desired product could be detected by LCMS. The reaction was quenched with 50 mL of potassium sodium tartrate (aq.) at room temperature. The resulting mixture was extracted with EA (50 mL x 3). The combined organic layers were washed with brine (40 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (1.0 g, 59.3% yield) of {5-bromopyrazolo[1,5-a] pyridin-3-yl} methanol as a yellow solid. LCMS observed m/z = 226.97 [M+H]+. Step 2: Preparation of 5-bromo-3-methylpyrazolo[1,5-a] pyridine Compound 272.2
To a stirred solution of {5-bromopyrazolo[1,5-a] pyridin-3-yl} methanol (1.0 g, 4.6 mmol) in TFA (30 mL) were added triethylsilane (5.4 g, 46.2 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at room temperature for additional 30 min. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (3.1 g, 84.6% yield) of 1-[4-bromo-2-(hydroxymethyl)phenyl]ethanol as a yellow oil. LCMS observed m/z = 210.98 [M+H]+. Step 3: Preparation of 3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl) pyrazolo[1,5-a] pyridine Compound 272.3
To a stirred solution of 5-bromo-3-methylpyrazolo[1,5-a] pyridine (405 mg, 1.9 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (731 mg, 2.9 mmol) in 1,4-dioxane (10 mL) were added Pd(dppf)Cl2CH2Cl2 (157 mg, 0.2 mmol) and AcOK (565 mg, 5.8 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 100 ℃ for additional 2 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 259.15 [M+H]+. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-5-ethyl-2-{3-methylpyrazolo[1,5-a] pyridin-5-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide: Compound 272.4
To a stirred solution of 3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine (147 mg, 0.6 mmol) in 1,4-dioxane (5 mL) and H2O (1 mL) were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (300 mg, 0.4 mmol), Pd(dppf)Cl2CH2Cl2 (31 mg, 0.1 mmol) and K2CO3 (158 mg, 1.2 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred at 100 ℃ for additional 2 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 2% B to 15%
B in 7min; Wave Length: 254/220 nm; RT1(min): 6.72) to afford (200 mg, 62.6% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-2-{3- methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 840.27 [M+H]+. Step 5: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{3-methylpyrazolo[1,5- a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 272
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-2-{3-methylpyrazolo[1,5-a]pyridin-5-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (240 mg, 0.3 mmol) in trifluoroacetic acid (5 mL) was stirred at 85 ℃ for 20 min under N2 atmosphere. Desired product could be detected by LCMS. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 2% B to 15% B in 7min; Wave Length: 254/220 nm; RT1(min): 6.72) to afford (37.8 mg, 17.6% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{3-methylpyrazolo[1,5-a]pyridin-5-yl}-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.24 (s, 1H), 8.70 (d, J = 7.3 Hz, 1H), 8.54 (s, 1H), 8.28 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.91 (s, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.44 (dd, J = 7.3, 1.9 Hz, 1H), 5.43 (s, 2H), 4.54 (m, 1H), 3.60 – 3.48 (m, 3H), 3.35 – 3.25 (m, 1H), 3.02 (m, 3H), 2.85 (m, 1H), 2.68 (m, 1H), 2.44 (s, 3H), 2.32 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 750.40 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra.
EXAMPLE 273 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{6'-fluoro-1- methyl-3'H-spiro[azetidine-3,1'-[2]benzofuran]-5'-yl}-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)acetamide.
Step 1: Preparation of (5-bromo-4-fluoro-2-iodophenyl) methanol. Compound 273.1
To a solution methyl 5-bromo-4-fluoro-2-iodobenzoate (2.0 g, 5.6 mmol) in diethyl ether (56 mL) were added LiBH4 (4.0M in THF 3.06 mL 12.2 mmol) and MeOH (0.56 mL, 13.9 mmol) at 0°C. The reaction mixture was stirred at 0°C for 1 hour. The resulting mixture was diluted with EA, and the mixture was washed sequentially with water and saturated brine, and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford (5-bromo-4-fluoro-2-iodophenyl) methanol (1.8 g, 97%yield) as a white solid. LCMS observed m/z = 333.05 [M+H] +. Step 2: Preparation of 1-bromo-5-(chloromethyl)-2-fluoro-4-iodobenzene. Compound 273.2
To a solution of (5-bromo-4-fluoro-2-iodophenyl) methanol (1.76 g, 5.3 mmol) in DCM (10 mL) were added SOCl2 (1.27 g, 10.7 mmol) and DMF (0.1 mL, 1.3 mmol) under nitrogen atmosphere. The mixture was stirred at 25°C for 2 h under nitrogen atmosphere.
The resulting mixture was concentrated under reduced pressure and purified by silica gel column chromatography, eluted with PE / EA (12/1) to afford 1-bromo-5-(chloromethyl)- 2-fluoro-4-iodobenzene (1.67 g, 89%yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 7.69 (d, J = 6.9 Hz, 1H), 7.64 (d, J = 7.6 Hz, 1H), 4.63 (s, 2H). Step 3: Preparation of tert-butyl 5'-bromo-6'-fluoro-3'H-spiro[azetidine-3,1'- isobenzofuran]-1-carboxylate. Compound 273.3
To a solution of 1-bromo-5-(chloromethyl)-2-fluoro-4-iodobenzene (1.68 g, 4.8 mmol) was dissolved in THF (3 mL) and cooled to -50° C. Isopropylmagnesium chloride lithium chloride complex (1.3 M in THF, 5.5 mL, 7.2 mmol) was added dropwise, keeping the temperature below -45° C under nitrogen atmosphere. The mixture was stirred at -50 °C for 1 h, followed by the addition of tert-butyl 3-oxoazetidine-1-carboxylate (988 mg, 5.8 mmol) dropwise at -78 °C. The reaction mixture was then allowed to warm to room temperature and stirred at room temperature for 2 h under nitrogen atmosphere and was then quenched with water. The aqueous phase was acidified to pH 5 with 6 M HCl. The reaction mixture was extracted with EA (2 x 5 mL). The combined organic phase was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford tert-butyl 5'-bromo-6'-fluoro-3'H-spiro[azetidine-3,1'- isobenzofuran]-1-carboxylate (870 mg, 50%yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 7.42 (d, J = 5.8, 1.1 Hz, 1H), 7.25 (d, J = 7.7 Hz, 1H), 5.11 – 5.06 (m, 2H), 4.33 (dd, J = 9.2, 1.2 Hz, 2H), 4.11 (dd, J = 9.3, 1.2 Hz, 2H), 1.50 (s, 9H). LCMS observed m/z = 358.10 [M+H] +. Step 4: Preparation of tert-butyl6'-fluoro-5'-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-3'H-spiro[azetidine-3,1'-isobenzofuran]-1-carboxylate. Compound 273.4
To a stirred solution of tert-butyl 5'-bromo-6'-fluoro-3'H-spiro[azetidine-3,1'- isobenzofuran]-1-carboxylate (300 mg, 0.84 mmol, 1.0 equiv.), B2Pin2 (255 mg, 1.0 mmol) and KOAc (165 mg, 1.7 mmol) in dioxane (3 mL) was added Pd(dppf)Cl2 (136 mg, 0.17 mmol) in portions at 25°C under nitrogen atmosphere. Then the resulting mixture was heated to 100 °C and stirred at 100 °C for 1 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (9/1) to afford tert-butyl 6'-fluoro-5'-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3'H- spiro [azetidine-3,1'- [2] benzofuran]-1-carboxylate (100 mg, crude) as a yellow oil. LCMS observed m/z = 305.10 [M-Boc] -. Step 5: Preparation of tert-butyl5'-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl) piperazin-1-yl)-4-(2-((2-chloro-4-(trifluoromethyl) phenyl) amino)-2-oxoethyl)- 5-ethyl-7-oxo-4,7-dihydro- [1,2,4] triazolo[1,5-a] pyrimidin-2-yl)-6'-fluoro-3'H- spiro[azetidine-3,1'-isobenzofuran]-1-carboxylate. Compound 273.5
To a solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate H (200 mg, 0.25 mmol), tert-butyl 6'- fluoro-5'-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-3'H-spiro[azetidine-3,1'-
isobenzofuran]-1-carboxylate(124 mg, 0.3 mmol) and K2CO3 (71 mg, 0.51 mmol) in dioxane (10 mL) and H2O (2 mL) was added Pd(dppf)Cl2 (41 mg, 0.05 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 1 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (9/1) to afford tert-butyl5'-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl) piperazin-1-yl)-4-(2-((2- chloro-4-(trifluoromethyl) phenyl) amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4] triazolo[1,5-a] pyrimidin-2-yl)-6'-fluoro-3'H-spiro[azetidine-3,1'-isobenzofuran]-1- carboxylate (100 mg, crude) as a brown solid. LCMS observed m/z = 987.10 [M+H] +. Step 6: Preparation of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(5-ethyl-2-(6'- fluoro-3'H-spiro[azetidine-3,1'-isobenzofuran]-5'-yl)-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl) piperazin-1-yl)-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin- 4(7H)-yl) acetamide.
A solution of tert-butyl 5'-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-4-({[2-chloro-4-(trifluoromethyl) phenyl] carbamoyl} methyl)-5-ethyl-7- oxo- [1,2,4] triazolo[1,5-a] pyrimidin-2-yl)-6'-fluoro-3'H-spiro[azetidine-3,1'- [2] benzofuran]-1-carboxylate (100 mg, 0.1 mmol) in TFA (3 mL) was heated to 80℃ and stirred at 80°C for 20 min under nitrogen atmosphere. After completion of reaction, the reaction mixture was cooled to 25 °C and concentrated under reduced pressure to afford N- [2-chloro-4-(trifluoromethyl) phenyl]-2-(5-ethyl-2- {6'-fluoro-3'H-spiro [azetidine-3,1'- [2] benzofuran]-5'-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7- oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl) acetamide (150 mg, crude) as a brown oil. LCMS observed m/z = 797.25 [M+H]+. Step 7: Preparation of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(5-ethyl-2-(6'- fluoro-1-methyl-3'H-spiro[azetidine-3,1'-isobenzofuran]-5'-yl)-6-(4-(5-hydroxy-6-
methylpyrimidine-4-carbonyl) piperazin-1-yl)-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin- 4(7H)-yl) acetamide.
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{6'-fluoro-3'H- spiro[azetidine-3,1'-[2]benzofuran]-5'-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide (150 mg, 0.19 mmol) and formaldehyde (153 mg, 1.9 mmol, 37%) in MeOH (3 mL) was stirred at 15 °C for 30 min under nitrogen atmosphere. To the above mixture was added NaBH3CN (24 mg, 0.38 mmol) in portions at 15°C. The resulting mixture was stirred at 15°C for additional 1 h. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (5/1) to afford N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-(5-ethyl-2-{6'-fluoro-1-methyl-3'H-spiro[azetidine-3,1'- [2]benzofuran]-5'-yl}-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide (27.3 mg, 18% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.81 (s, 1H), 9.14 (s, 1H), 8.62 (s, 1H), 8.46 (d, J = 8.7 Hz, 1H), 8.03 (d, J = 6.0 Hz, 1H), 7.87 (s, 1H), 7.63 (d, J = 2.0 Hz, 1H), 7.55 (dd, J = 6.8, 3.2 Hz, 1H), 5.78 – 5.66 (m, 1H), 5.23 (s, 2H), 5.13 (s, 2H), 4.90 – 4.76 (m, 1H), 4.43 – 3.81 (m, 2H), 3.53 (s, 5H), 3.25 (d, J = 8.1 Hz, 2H), 3.11 (s, 1H), 2.96 – 2.64 (m, 5H), 2.59 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H), 1.29 (s, 1H). LCMS observed m/z = 811.25 [M+H]+. EXAMPLE 274 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[(1R)-1-(trifluoromethyl)-1,3- dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 274
Step 1: Preparation of 6-bromo-3-hydroxy-3-(trifluoromethyl)-2-benzofuran-1-one Compound 274.1
To a stirred solution of 5-bromo-2-benzofuran-1,3-dione (10.0 g, 44.1 mmol) and CuI (0.8 g, 4.4 mmol) in THF (200 mL) was added KF (5.1 g, 88.1 mmol), 1,10- Phenanthroline (0.8 g, 4.4 mmol) and trimethyl(trifluoromethyl)silane (7.5 g, 52.9 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 50 ℃ for additional 8 hours. Desired product could be detected by LCMS. The mixture was allowed to cool down to room temperature and quenched with H2O (250 mL). The resulting mixture was extracted with EA (300 mL x 3). The combined organic layers were washed with brine (300 mL), dried over anhydrous Na2SO4. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 5% EtOAc in petroleum ether) to afford (4.7 g, 36.0% yield) of 6-bromo-3- hydroxy-3-(trifluoromethyl)-2-benzofuran-1-one as a yellow oil. LCMS observed m/z = 294.93 [M-H]-. Step 2: Preparation of 1-[4-bromo-2-(hydroxymethyl) phenyl]-2,2,2- trifluoroethano Compound 274.2 To a stirred solution of 6-bromo-3-hydroxy-3-(trifluoromethyl)-2-benzofuran-1-one (4.7 g, 15.8 mmol) in THF (80 mL) were added lithium aluminum hydride (1.5 g, 40.0 mmol) in portions at 0 ℃ under N2 atmosphere. The resulting mixture was stirred at room
temperature for additional 2.5 hours. Desired product could be detected by LCMS. The mixture was quenched with H2O (100 mL) at 0 ℃. The resulting mixture was extracted with EA (200 mL x 3). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 25% EtOAc in petroleum ether) to afford (2.0 g, 44.3% yield) of 1-[4-bromo-2-(hydroxymethyl) phenyl]-2,2,2-trifluoroethanol as a light-yellow oil. LCMS observed m/z = 282.97 [M-H]-. Step 3: Preparation of 5-bromo-1-(trifluoromethyl)-1,3-dihydro-2-benzofuran Compound 274.3
To a stirred solution of 1-[4-bromo-2-(hydroxymethyl) phenyl]-2,2,2- trifluoroethanol (2.0 g, 7.0 mmol) and tributylphosphane (2.8 g, 14.0 mmol) in toluene (5 mL) was added 1-[(dimethylcarbamoyl)imino]-3,3-dimethylurea (2.8 g, 16.1 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at room temperature for additional 2.5 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (1.7 g, 90.7% yield) of 5-bromo-1-(trifluoromethyl)-1,3-dihydro-2-benzofuran as a colorless oil. LCMS observed m/z = 266.96 [M+H]+. Step 4: Preparation of 4,4,5,5-tetramethyl-2-[1-(trifluoromethyl)-1,3-dihydro-2- benzofuran-5-yl]-1,3,2-dioxaborolane Compound 274.4
To a stirred mixture of 5-bromo-1-(trifluoromethyl)-1,3-dihydro-2-benzofuran (1.7 g, 6.3 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (2.4 g, 9.4 mmol) in 1,4-dioxane (20 mL) was added Pd(dppf)Cl2CH2Cl2 (0.5 g, 0.6 mmol) and AcOK (2.5 g, 25.2 mmol) in portions at room
temperature under N2 atmosphere. The resulting mixture was stirred at 100 ℃ for additional 2 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (1.7 g, 83.5% yield) of 4,4,5,5-tetramethyl-2-[1-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-1,3,2- dioxaborolane as a light-yellow solid. LCMS observed m/z = 315.13 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-[1-(trifluoromethyl)-1,3-dihydro-2-benzofuran- 5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide Compound 274.5
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide, Intermediate H (1.0 g, 1.3 mmol) and 4,4,5,5- tetramethyl-2-[1-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-1,3,2-dioxaborolane (597 mg, 1.9 mmol) in dioxane (20 mL) and H2O (4 mL) were added Pd(dppf)Cl2CH2Cl2 (104 mg, 0.1 mmol) and K2CO3 (525 mg, 3.8 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred at 100 ℃ for additional 16 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in DCM) to afford (850 mg, 74.8% yield) of 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-[1- (trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 896.24 [M+H]+.
Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[1-(trifluoromethyl)-1,3- dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1- yl}-5-ethyl-7-oxo-2-[1-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (800 mg, 0.9 mmol) in trifluoroacetic acid (10 mL) was stirred at 85 ℃ for 15 min under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeOH in H2O, 10% to 50% gradient in 10 min; detector, UV 254 nm to afford (705 mg, 97.8% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[1-(trifluoromethyl)-1,3-dihydro-2- benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. LCMS observed m/z = 806.24 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[(1R)-1- (trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide Compound 274
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[1-(trifluoromethyl)-1,3-dihydro-2- benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (200 mg, 0.2 mmol) was purified by SFC to afford (46.2 mg, 23.1% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-[(1R)-1-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5- yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 8.57 (s, 1H), 8.19 – 8.12 (m, 2H), 8.10 – 7.95 (m, 2H), 7.71 (m, 1H), 7.58 (d, J = 8.0 Hz, 1H), 5.91 (q, J = 7.1 Hz, 1H), 5.40 (s, 2H), 5.26 (s, 2H), 4.54 (m, 1H), 3.53 (m, 3H), 3.29 – 3.22 (m, 1H), 3.02 (m, 3H), 2.85 (m, 1H), 2.67 (m, 1H), 2.44 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 806.15 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 275 Synthesis of of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[(3S)-3- (trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide. Compound 275
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[3-(trifluoromethyl)-1,3-dihydro-2- benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (200 mg, 0.2 mmol) was purified by SFC to afford (58.8 mg, 29.4% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-[(3S)-3-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5- yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.45 (s, 1H), 8.61 (s, 1H), 8.19 – 8.12 (m, 2H), 8.06 – 7.96 (m, 2H), 7.73 (m, 1H), 7.55 (d, J = 8.0 Hz, 1H), 5.96 (q, J = 7.2 Hz, 1H), 5.42 (s, 2H), 5.29 (s, 2H), 4.57 (m, 1H), 3.51 (m, 3H), 3.29 – 3.17 (m, 1H), 3.05 (m, 3H), 2.87 (m, 1H), 2.64 (m, 1H), 2.44 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 806.15 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 276 Synthesis of of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[(1S)-1- (trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide.
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[1-(trifluoromethyl)-1,3-dihydro-2- benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (200 mg, 0.2 mmol) was purified by SFC to afford (9.0 mg, 4.5% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]- 2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2- [(1S)-1-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.55 (s, 1H), 8.18 – 8.12 (m, 2H), 8.10 – 7.97 (m, 2H), 7.72 (m, 1H), 7.53 (d, J = 8.0 Hz, 1H), 5.91
(q, J = 7.2 Hz, 1H), 5.41 (s, 2H), 5.26 (s, 2H), 4.56 (m, 1H), 3.53 (m, 3H), 3.29 – 3.20 (m, 1H), 3.06 (m, 3H), 2.83 (m, 1H), 2.65 (m, 1H), 2.41 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 806.10 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 277 Synthesis of of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[(3R)-3- (trifluoromethyl)-1,3-dihydro-2-benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide. Compound 277
The N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-[3-(trifluoromethyl)-1,3-dihydro-2- benzofuran-5-yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (200 mg, 0.2 mmol) was purified by SFC to afford (68.4 mg, 34.2% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-[(3R)-3-(trifluoromethyl)-1,3-dihydro-2-benzofuran-5- yl]-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.495 (s, 1H), 8.66 (s, 1H), 8.24 – 8.15 (m, 2H), 8.09 – 7.99 (m, 2H), 7.77 (m, 1H), 7.59 (d, J = 8.2 Hz, 1H), 5.99 (q, J = 7.3 Hz, 1H), 5.46 (s, 2H), 5.33 (s, 2H), 4.59 (m, 1H), 3.50 (m, 3H), 3.29 – 3.18 (m, 1H), 3.04 (m, 3H), 2.89 (m, 1H), 2.63 (m, 1H), 2.42 (s, 3H), 1.26 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 806.15 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 278 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{3H-spiro[2-benzofuran-1,3'- oxetan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
Compound 278
Step 1: Preparation of 5-bromospiro[2-benzofuran-1,3'-oxetan]-3-one. Compound 278.1
To a stirred solution of methyl 5-bromo-2-iodobenzoate (5.0 g, 14.7 mmol) in THF (50 mL) was added 3-oxetanone (1.0 g, 16.1 mmol) in portions at -78 °C under nitrogen atmosphere. To the above mixture was added chloro(propan-2-yl)magnesium; chlorolithium (2.0 M in THF) (8.1 mL, 16.1 mmol) dropwise over 5 min at -78 °C. The resulting mixture was stirred at -78 °C for additional 1.5 h. The reaction was quenched by the addition of sat. sodium hyposulfite (aq.) (50 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 40 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by reversed phase C18 silica gel column chromatography to afford (892 mg, 21% yield) of 5-bromospiro[2-benzofuran-1,3'-oxetan]-3-one as a yellow solid. LCMS observed m/z =254.96 [M+H]+. Step 2: Preparation of 3-[4-bromo-2-(hydroxymethyl)phenyl]oxetan-3-ol. Compound 278.2
To a stirred solution of 5-bromospiro[2-benzofuran-1,3'-oxetan]-3-one (893 mg, 3.5 mmol) in THF (10 mL) was added lithium borohydride (4.4 mL, 17.5 mmol, 4.0 M in THF) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 85 °C
for additional 3 h. The reaction was quenched with water (15 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (713 mg, 67% yield) of 3-[4-bromo-2- (hydroxymethyl)phenyl]oxetan-3-ol as a light yellow solid. LCMS observed m/z =258.99 [M+H]+. Step 3: Preparation of 3-[4-bromo-2-(hydroxymethyl)phenyl]oxetan-3-ol. Compound 278.3
To a stirred solution of 3-[4-bromo-2-(hydroxymethyl)phenyl]oxetan-3-ol (713 mg, 2.7 mmol) in DCM (10 mL) was added TsCl (629 mg, 3.3 mmol) and NaH (275 mg, 6.8 mmol, 60%) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 25 °C under nitrogen atmosphere. The reaction was quenched by the addition of water (15 mL) at 0 °C. The resulting mixture was extracted with CH2Cl2 (3 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (380 mg, 64% yield) of 5-bromo-3H-spiro[2-benzofuran-1,3'-oxetane] as a yellow oil. LCMS observed m/z =240.98 [M+H]+. Step 4: Preparation of 4,4,5,5-tetramethyl-2-{3H-spiro[2-benzofuran-1,3'-oxetan]- 5-yl}-1,3,2-dioxaborolane. Compound 278.4
To a stirred solution of5-bromo-3H-spiro[2-benzofuran-1,3'-oxetane] (300 mg, 1.2 mmol) in dioxane (1 mL) was added 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3,2-dioxaborolane (474 mg, 1.9 mmol), Pd(dppf)Cl2 (102 mg, 0.1 mmol)and AcOK (366 mg, 3.7 mmol) in portions at 25 °C under nitrogen atmosphere. The
resulting mixture was stirred for 1.5 h at 100 °C under nitrogen atmosphere. The crude product was used in the next step directly without further purification. LCMS observed m/z =289.15 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{3H-spiro[2-benzofuran-1,3'-oxetan]-5-yl}- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 278.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) in 1,4- dioxane (1 mL) /H2O (100 uL) was added 4,4,5,5-tetramethyl-2-{3H-spiro[2-benzofuran- 1,3'-oxetan]-5-yl}-1,3,2-dioxaborolane (55 mg, 0.2 mmol), Pd(dppf)Cl2 (10 mg, 0.01 mmol) and K2CO3 (52 mg, 0.4 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred at 100 °C for additional 4 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (42 mg, 35% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-{3H-spiro[2-benzofuran- 1,3'-oxetan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a brown solid. LCMS observed m/z =870.27 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{3H-spiro[2- benzofuran-1,3'-oxetan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 278
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-{3H-spiro[2-benzofuran-1,3'-oxetan]-5-yl}-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (42 mg, 0.01 mmol) in trifluoroacetic acid (1 mL) at 25 °C for 16 h under nitrogen atmosphere .The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: Water(0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 16% B to 39% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 4.93) to afford (11.4 mg, 31% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-{3H-spiro[2-benzofuran-1,3'- oxetan]-5-yl}-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.06 (s, 1H), 8.62 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.37 (d, J = 7.9 Hz, 1H), 8.18 (s, 1H), 7.82 (d, J = 8.0 Hz, 1H), 7.62 (d, J = 2.1 Hz, 1H), 7.59 – 7.54 (m, 1H), 5.72 (m, 1H), 5.20 (m, 4H), 5.12 (d, J = 7.0 Hz, 2H), 4.88 (d, J = 7.0 Hz, 3H), 3.86 (m, 2H), 3.55 (d, J = 13.0 Hz, 1H), 3.25 (d, J = 8.0 Hz, 2H), 3.11 (s, 1H), 2.85 (m, 2H), 2.59 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z =780.15 [M+H]+. EXAMPLE 279 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-2,2-dioxido-1,3- dihydrobenzo[c]isothiazol-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 279
Step 1: Preparation of 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1,3-dihydrobenzo[c]isothiazole 2,2-dioxide Compound 279.1
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 5-bromo-1- methyl-2,3-dihydro-1H-2λ⁶,1-benzisothiazole-2,2-dione. LCMS observed m/z = 310.443 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-2,2-dioxido-1,3- dihydrobenzo[c]isothiazol-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 279
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 1-methyl-5-
(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,3-dihydro-2λ⁶,1-benzisothiazole-2,2(1H)- dione. 1H NMR (400 MHz, DMSO) δ 10.43 (br, 2H), 8.58 (s, 1H), 8.16 – 8.01 (m, 3H), 8.01 – 7.92 (m, 1H), 7.72 (dd, J = 8.8, 2.2 Hz, 1H), 7.09 (d, J = 8.4 Hz, 1H), 5.40 (s, 2H), 4.80 (s, 2H), 4.54 (d, J = 12.3 Hz, 1H), 3.60 – 3.47 (m, 3H), 3.28 – 3.22 (m, 1H), 3.11 (s, 3H), 3.09 – 2.93 (m, 3H), 2.85 (d, J = 11.1 Hz, 1H), 2.67 (d, J = 10.8 Hz, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 801.120 [M+H]+. EXAMPLE 280 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(quinoxalin-6-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 280
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (6- quinoxalinyl)boranediol.1H NMR (400 MHz, DMSO) δ 10.54 (s, 1H), 10.30 (s, 1H), 9.19 – 8.99 (m, 2H), 8.84 (d, J = 1.9 Hz, 1H), 8.74 – 8.54 (m, 2H), 8.32 (d, J = 8.8 Hz, 1H), 8.13 (d, J = 8.6 Hz, 1H), 8.05 (d, J = 2.1 Hz, 1H), 7.78 (dd, J = 8.7, 2.1 Hz, 1H), 5.53 (s, 2H), 4.62 (d, J = 12.3 Hz, 1H), 3.61 (q, J = 11.4 Hz, 3H), 3.33 (d, J = 12.6 Hz, 1H), 3.11 (dd, J = 15.0, 5.9 Hz, 3H), 2.93 (d, J = 11.2 Hz, 1H), 2.76 (d, J = 11.2 Hz, 1H), 2.52 (s, 3H), 1.30 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 748.2 [M+H]+. EXAMPLE 281 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(3,4-dihydro-2H-1,4- benzoxazin-7-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide Compound 281
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(3,4-dihydro-2H-1,4-benzoxazin-7-yl)-6-ethyl- 5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl] acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.50 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 8.02 (s, 1H), 7.94 (s, 1H), 7.68 (d, J = 8.6 Hz, 1H), 7.49 – 7.42 (m, 1H), 7.35 (d, J = 1.9 Hz, 1H), 6.63 (d, J = 8.3 Hz, 1H), 6.34 (s, 1H), 5.32 (s, 2H), 4.51 (d, J = 12.3 Hz, 1H), 4.14 (t, J = 4.3 Hz, 2H), 3.48 – 3.43 (m, 4H), 3.24 – 3.15 (m, 2H), 3.00 (d, J = 7.6 Hz, 2H), 2.88 (t, J = 12.1 Hz, 1H), 2.77 (d, J = 11.1 Hz, 1H), 2.63 – 2.57 (m, 1H), 2.26 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 754.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 282 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2'-methyl-4-oxo-7H-3'-oxa-1,1',3,3a,7,7'- hexaaza-2,5'-biindenyl-7-yl)acetamide. Compound 282
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-1-oxa-3,4-diaza-6-indenyl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 9.21 – 9.13 (m, 1H), 8.69 – 8.62 (m, 1H), 8.23 (s, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.95 (s, 1H), 7.68 (d, J = 8.6 Hz, 1H), 5.40 (s, 2H), 4.52 (d, J = 12.2 Hz, 1H),
3.57 – 3.43 (m, 3H), 3.24 – 3.18 (m, 1H), 3.09 – 2.87 (m, 3H), 2.81 (d, J = 11.1 Hz, 1H), 2.72 (s, 3H), 2.65 (d, J = 10.8 Hz, 1H), 2.34 (s, 3H), 1.21 (t, J = 7.0 Hz, 3H). LCMS observed m/z = 752.3 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 283 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1-benzofuran-5-yl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl] acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1-benzofuran-5-yl)-6-ethyl-5-{4-[(5-hydroxy- 6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide as a brown solid. 1H NMR (400 MHz, DMSO) δ 10.43 (s, 1H), 10.24 (s, 1H), 8.59 (s, 1H), 8.44 (d, J = 1.7 Hz, 1H), 8.12 – 8.05 (m, 3H), 8.03 – 8.00 (m, 1H), 7.74 (d, J = 8.7 Hz, 1H), 7.63 – 7.58 (m, 1H), 7.09 (d, J = 2.2 Hz, 1H), 5.42 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 3.56 - 3.51 (m, 3H), 3.30 - 3.25 (m, 1H), 3.05 - 3.01 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.69 - 2.67 (m, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 736.3 [M+H]+. EXAMPLE 284 Synthesis of 2-(2-(2-aminobenzo[d]oxazol-6-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide Compound 284
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 6- (dihydroxyboryl)-1,3-benzoxazol-2-ylamine.1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 10.25 (s, 1H), 8.58 (d, J = 9.5 Hz, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.95 (ddd, J = 17.5, 8.5, 1.9 Hz, 3H), 7.79 – 7.63 (m, 3H), 7.30 (d, J = 8.1 Hz, 1H), 5.40 (s, 2H), 4.55 (d, J = 12.3 Hz, 1H), 3.54 (td, J = 12.4, 6.5 Hz, 3H), 3.29 – 3.22 (m, 1H), 3.03 (d, J = 9.5 Hz, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 10.5 Hz, 1H), 2.46 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.629 [M+H]+. EXAMPLE 285 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-1,3-benzothiazol-6-yl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 285
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-1,3-benzothiazol-6-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.43 (s, 1H), 8.78 (d, J = 1.6 Hz, 1H), 8.56 (s, 1H), 8.25 – 8.15 (m, 1H), 8.07
(d, J = 8.6 Hz, 1H), 8.02 (d, J = 8.5 Hz, 1H), 7.98 (d, J = 2.2 Hz, 1H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 5.42 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.62 – 3.43 (m, 3H), 3.28 – 3.21 (m, 1H), 3.09 – 2.95 (m, 3H), 2.83 (s, 3H), 2.82 – 2.78 (m, 1H), 2.71 – 2.64 (m, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 767.3 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 286 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(7-methoxy-2-methyl-2H-indazol-5-yl)- 4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 286
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(7-methoxy-2- methyl-2H-indazol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.43 (s, 1H), 10.24 (s, 1H), 8.59 (s, 1H), 8.43 (s, 1H), 8.11 – 8.04 (m, 2H), 7.98 (d, J = 2.1 Hz, 1H), 7.76 – 7.67 (m, 1H), 7.27 (d, J = 1.2 Hz, 1H), 5.40 (s, 2H), 4.60 – 4.43 (m, 1H), 4.16 (s, 3H), 3.98 (s, 3H), 3.61 – 3.44 (m, 3H), 3.28 – 3.18 (m, 1H), 3.11 – 2.94 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.72 – 2.61 (m, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 780.3 [M+H]+. EXAMPLE 287 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][6-ethyl-2-(7-fluoro-2-methyl- 2H-indazol-5-yl)-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4- oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 287
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with hydrogen chloride—2-(7-fluoro-2-methyl-2H-indazol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (1/1). 1H NMR (400MHz, DMSO-d6) δ 10.43 (s, 1H), 10.24 (s, 1H), 8.63 (d, J = 2.7 Hz, 1H), 8.58 (s, 1H), 8.36 (d, J = 1.2 Hz, 1H), 8.07 (d, J = 8.5 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.9, 2.1 Hz, 1H), 7.66 (dd, J = 12.5, 1.2 Hz, 1H), 5.41 (s, 2H), 4.59 – 4.47 (m, 1H), 4.23 (s, 3H), 3.61 – 3.46 (m, 3H), 3.30 – 3.21 (m, 1H), 3.12 – 2.94 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.67 (d, J = 9.5 Hz, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 768.3 [M+H]+. EXAMPLE 288 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,3-benzothiazol-5-yl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl] acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,3-benzothiazol-5-yl)-6-ethyl-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7- indenyl] acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.45 (s, 1H), 9.43 (s, 1H), 8.68 (d, J = 1.5 Hz, 1H), 8.35 (d, J = 10.5 Hz, 1H), 8.26 (d, J = 8.4 Hz, 1H), 8.19 – 8.12 (m, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.90 (d, J = 2.1 Hz, 1H), 7.67 – 7.58 (m, 1H), 5.37
(s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.51 – 3.49 (m, 3H), 3.23 – 3.16 (m, 1H), 3.00 – 2.86 (m, 3H), 2.77 (d, J = 11.1 Hz, 1H), 2.64 – 2.55 (m, 1H), 2.34 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 753.2 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 289 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2',3',5',6'-tetrahydro-3H- spiro[isobenzofuran-1,4'-pyran]-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
Step 1: Preparation of 5-bromo-2',3',5',6'-tetrahydro-3H-spiro[isobenzofuran-1,4'- pyran]-3-one. Compound 289.1
To a stirred mixture of methyl 5-bromo-2-iodobenzoate (2.0 g, 5.8 mmol) and tetrahydro-4H-pyran-4-one (0.6 g, 6.4 mmol) in THF (5 mL) was added Turbo Grignard (0.9 g, 6.4 mmol) dropwise at -78 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 12 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched by the addition of water (30 mL) at room temperature and extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (2 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 25% to 35% gradient in 10 min; detector, UV 220 nm) to afford 5-bromo-2',3',5',6'-
tetrahydro-3H-spiro[isobenzofuran-1,4'-pyran]-3-one (580 mg, 26% yield) as a yellow solid. LCMS observed m/z = 282.99 [M+H] +. Step 2: Preparation of 4-(4-bromo-2-(hydroxymethyl)phenyl)tetrahydro-2H-pyran- 4-ol. Compound 289.2
To a stirred mixture of 5-bromo-2',3',5',6'-tetrahydro-3H-spiro[isobenzofuran-1,4'- pyran]-3-one (410 mg, 1.5 mmol) in THF (6 mL) was added LiBH4 (0.7 mL, 3.0 mmol, 4.0 M in THF) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 12 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched by the addition of water (20 mL) at room temperature and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 20% gradient in 10 min; detector, UV 254 nm) to afford 4-(4-bromo-2- (hydroxymethyl)phenyl)tetrahydro-2H-pyran-4-ol (380 mg, 64% yield) as a yellow solid. LCMS observed m/z = 287.15 [M+H] +. Step 3: Preparation of 5-bromo-2',3',5',6'-tetrahydro-3H-spiro[isobenzofuran-1,4'- pyran]. Compound 289.3
Into a 25 mL 3-necked round-bottom flask were added 4-(4-bromo-2- (hydroxymethyl)phenyl)tetrahydro-2H-pyran-4-ol (380 mg, 1.3 mmol) and concentrated HCl (5 mL) dropwise at room temperature. The resulting mixture was stirred at 60 °C for 2 h. After completion of reaction, the reaction was quenched by the addition of water (20 mL) at room temperature and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash
chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 20% to 30% gradient in 10 min; detector, UV 254 nm) to afford 5- bromo-2',3',5',6'-tetrahydro-3H-spiro[isobenzofuran-1,4'-pyran] (220 mg, 50% yield) as a white solid. LCMS observed m/z = 269.14 [M+H] +. Step 4: Preparation of 4,4,5,5-tetramethyl-2-(2',3',5',6'-tetrahydro-3H- spiro[isobenzofuran-1,4'-pyran]-5-yl)-1,3,2-dioxaborolane. Compound 289.4
To a stirred mixture of 5-bromo-2',3',5',6'-tetrahydro-3H-spiro[isobenzofuran-1,4'- pyran] (220 mg, 0.8 mmol) and B2(Pin)2 (249 mg, 1.0 mmol) in dioxane (5 mL) were added Pd(dppf)Cl2 (60 mg, 0.1 mmol) and KOAc (240 mg, 2.4 mmol) in one portion at room temperature. The resulting mixture was stirred at 90 °C for 3 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched by the addition of water (20 mL) at room temperature and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford 4,4,5,5-tetramethyl-2-(2',3',5',6'- tetrahydro-3H-spiro[isobenzofuran-1,4'-pyran]-5-yl)-1,3,2-dioxaborolane (110 mg, 34% yield) as a yellow solid. LCMS observed m/z = 317.20 [M+H] +. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2',3',5',6'-tetrahydro- 3H-spiro[isobenzofuran-1,4'-pyran]-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide.
The title compound was prepared using similar procedure as compound 164 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-2-(2',3',5',6'-tetrahydro-3H-spiro[isobenzofuran-1,4'- pyran]-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. 1H NMR (400 MHz, Methanol -d4) δ 8.59 (s, 1H), 8.21 – 8.15 (m, 2H), 8.11 (s, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.64 – 7.61 (m, 1H), 7.34 (d, J = 7.9 Hz, 1H), 5.48 (s, 2H), 5.12 (s, 2H), 4.75 – 4.72 (m, 1H), 4.15 – 4.12 (m, 1H), 3.97 – 3.81 (m, 4H), 3.78 – 3.75 (m, 2H), 3.50 – 3.44 (m, 1H), 3.23 – 3.10 (m, 3H), 3.00 – 2.96 (m, 1H), 2.85 – 2.81 (m, 1H), 2.55 (s, 3H), 2.11 – 2.04 (m, 2H), 1.70 – 1.67 (m, 2H), 1.35 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 808.2 [M+H]+. Note: Two exchangeable protons were not visible in NMR spectra. EXAMPLE 290 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(3-fluoro-2- methylindazol-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Step 1: Preparation of 5-bromo-3-fluoro-2-methylindazole. Compound 290.1
To a stirred solution of 5-bromo-2-methylindazole (200 mg, 0.9 mmol) in ACN (5 mL) was added 4-(chloromethyl)-1-fluoro-1,4-diazabicyclo[2.2.2]octane-1,4-diium; bis(tetrafluoroboranuide) (370 mg, 1.0 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 3 h under nitrogen atmosphere. Then the reaction was quenched with water (10 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was
purified by silica gel column chromatography and eluted with PE/EA (5/1) to afford 5- bromo-3-fluoro-2-methylindazole (100 mg, 46% yield) as a white solid. LCMS observed m/z = 228.97 [M+H]+. Step 2: Preparation of 3-fluoro-2-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)-2H-indazole. Compound 290.2
To a stirred solution of 5-bromo-3-fluoro-2-methylindazole (200 mg, 0.9 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (443 mg, 1.7 mmol) in dioxane (5 mL) were added Pd(dppf)Cl2 (64 mg, 0.1 mmol) and KOAc (257 mg, 1.6mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 4 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to room temperature and concentrated under reduced pressure. This resulted in 3-fluoro-2-methyl-5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl) indazole (90 mg, crude) as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 277.14 [M+H]+. Step 3: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(3- fluoro-2-methylindazol-5-yl)-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide.
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(3-fluoro-2-methylindazol-5-yl)-6-[4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide.1H NMR (400 MHz, DMSO-d6) δ 10.29 (s, 1H), 8.54 (s, 1H),
8.36 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 8.9 Hz, 2H), 7.72 (d, J = 8.7 Hz, 1H), 7.61 (d, J = 9.2 Hz, 1H), 5.42 (s, 2H), 4.54 (m, 1H), 4.05 (s, 3H), 3.59 – 3.47 (m, 5H), 3.04 (m, 3H), 2.85 (m, 1H), 2.66 (s, 1H), 2.36 (m, 2H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 768.40 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 291 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-(difluoromethyl)-6- fluoro-2H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 291
Step 1: Preparation of 5-bromo-2-(difluoromethyl)-6-fluoro-2H-indazole. Compound 291.1
To a stirred mixture of 5-bromo-6-fluoro-2H-indazole (200 mg, 0.9 mmol) and K2CO3 (257 mg, 1.8 mmol) in EA (5 mL) was added 2,2-difluoro-2-(fluorosulfonyl)acetic acid (331 mg, 1.8 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h. After completion of reaction, the reaction was quenched with water (20 mL) at room temperature. The resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 5- bromo-2-(difluoromethyl)-6-fluoro-2H-indazole (170 mg, 52% yield) as a white solid. LCMS observed m/z = 264.95 [M+H] +. Step 2: Preparation of 2-(difluoromethyl)-6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2H-indazole. Compound 291.2
To a stirred mixture of 5-bromo-2-(difluoromethyl)-6-fluoro-2H-indazole (170 mg, 0.6 mmol) and 4,4,5,5-tetramethyl-2-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (305 mg, 1.2 mmol) in dioxane (5 mL) were added Pd(dppf)Cl2 (47 mg, 0.1 mmol) and KOAc (188 mg, 1.8 mmol) in one portion at room temperature. The resulting mixture was stirred at 80 °C for 3 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched with water (30 mL) at room temperature. The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 2-(difluoromethyl)-6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2H-indazole (130 mg, 45% yield) as a white solid. LCMS observed m/z = 313.10 [M+H] +. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2- (difluoromethyl)-6-fluoro-2H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
To a stirred mixture of 2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide, Intermediate AC (125 mg, 0.2 mmol, Intermediate) and 2-(difluoromethyl)-6-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H-indazole (64 mg, 0.4 mmol) in dioxane (3 mL) and H2O (0.3 mL) were added Pd(dppf)Cl2 (13 mg, 0.02 mmol,) and K2CO3 (74 mg, 0.6 mmol) in one portion at room temperature. The resulting mixture was stirred at 80 °C for 3 h under nitrogen atmosphere. After completion
of reaction, the reaction was quenched with water (30 mL) at room temperature. The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 26% B to 44% B in 9 min; Wave Length: 254 nm/220 nm; RT 1 (min): 8.37) to afford N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(2-(difluoromethyl)-6-fluoro-2H-indazol-5-yl)-5-ethyl-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (27.8 mg, 19% yield) as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.83 (s, 1H), 8.61 (s, 1H), 8.35 (s, 1H), 8.19 (d, J = 8.6 Hz, 1H), 8.02 – 7.59 (m, 3H), 7.51 – 7.47 (m, 1H), 5.49 (s, 2H), 4.75 – 4.72 (m, 1H), 4.14 – 3.92 (m, 1H), 3.79 – 3.73 (m, 2H), 3.57 – 3.44 (m, 1H), 3.26 – 3.08 (m, 3H), 3.01 – 2.98 (m, 1H), 2.87 – 2.81 (m, 1H), 2.50 (s, 3H), 1.37 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 804.10 [M+H] +. Note: Two exchangeable protons were not visible in NMR spectra. EXAMPLE 292 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-(2,2-difluoroethyl)- 2H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 292
Step 1: Preparation of 5-bromo-2-(2,2-difluoroethyl)-2H-indazole. Compound 292.1
To a stirred solution of 5-bromo-2H-indazole (500 mg, 2.5 mmol) and Cs2CO3 (2.5 g, 7.5 mmol) in DMF (6 mL) were added 2,2-difluoroethyl trifluoromethanesulfonate (652 mg, 3.0 mmol) dropwise at room temperature, after that, the resulting mixture was stirred at room temperature for 1 h. After completion of reaction, the reaction was quenched with water (30 mL) at room temperature, and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (3 x 60 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 30% to 40% gradient in 10 min; detector, UV 254 nm. This resulted in 5-bromo-2-(2,2-difluoroethyl)-2H-indazole (585 mg, 88% yield) as a light yellow solid. LCMS observed m/z = 260.90 [M+H]+. Step 2: Preparation of 2-(2,2-difluoroethyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-2H-indazole. Compound 292.2
To a stirred solution of 5-bromo-2-(2,2-difluoroethyl)-2H-indazole (35 mg, 0.13 mmol) and bis(pinacolato)diboron (68 mg, 0.26 mmol) in dioxane (2 mL) were added Pd(dppf)Cl2 (10 mg, 0.013 mmol) and AcOK (40 mg, 0.39 mmol) in portions at room temperature under nitrogen atmosphere, after that, the resulting mixture was heated to 100 ℃ and stirred at 100 ℃ for 1 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature and was filtered, the filter cake was washed with dioxane (1 x 2 mL). The filtrate was concentrated under reduced pressure. This resulted in 2-(2,2-difluoroethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H- indazole (50 mg, crude). LCMS observed m/z = 309.20 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2-(2,2-difluoroethyl)-2H-indazol-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 292.3
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (50 mg, 0.06 mmol) and 2- (2,2-difluoroethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H-indazole (23 mg, 0.07 mmol) in dioxane (2 mL) and H2O (0.4 mL) were added K2CO3 (26 mg, 0.18 mmol) and Pd(dppf)Cl2 (5 mg, 0.006 mmol) in portions at room temperature under nitrogen atmosphere, after that, the resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature, and was filtered, the filter cake was washed with dioxane (2 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 40% to 50% gradient in 5 min; detector, UV 254 nm. This resulted in 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(2-(2,2-difluoroethyl)-2H-indazol-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (48 mg, 72% yield) as a light yellow solid. LCMS observed m/z = 890.30 [M+H]+. Step 4: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-(2,2- difluoroethyl)-2H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 292
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2-(2,2-difluoroethyl)-2H-indazol-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (40 mg, 0.05 mmol) in DCM (2 mL) was added TFA (5 mL) dropwise at room temperature, after that, the resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 30 min. After completion of reaction, the mixture was allowed to cool down to room temperature and concentrated to dryness under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Phenyl 5 μm, 19*250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3+0.1% NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 30% B to 40% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 8.17) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)- 2-(2-(2-(2,2-difluoroethyl)-2H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (20.4 mg, 51% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 9.59 (s, 1H), 8.74 – 8.50 (m, 3H), 8.17 – 7.95 (m, 3H), 7.83 – 7.66 (m, 2H), 6.56 (tt, J = 54.8, 3.8 Hz, 1H), 5.42 (s, 2H), 5.02 (td, J = 14.8, 3.8 Hz, 2H), 4.55 (d, J = 12.3 Hz, 1H), 3.64 – 3.46 (m, 3H), 3.29 – 3.23 (m, 1H), 3.13 – 2.93 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.68 (d, J = 10.1 Hz, 1H), 2.48 – 2.39 (m, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 800.35 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 293 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1-(2,2-difluoroethyl)- 1H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 293
Step 1: Preparation of 1-(2,2-difluoroethyl)-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-indazole. Compound 293.1
To a stirred solution of 5-bromo-1-(2,2-difluoroethyl)indazole (30 mg, 0.12 mmol) and bis(pinacolato)diboron (58 mg, 0.24 mmol) in dioxane (2 mL) were added AcOK (34 mg, 0.36 mmol) and Pd(dppf)Cl2 (8 mg, 0.012 mmol) in portions at room temperature under nitrogen atmosphere, after that, the resulting mixture was heated to 100 ℃ and stirred at 100 ℃ for 2 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature and was filtered, the filter cake was washed with dioxane (2 mL). The filtrate was concentrated under reduced pressure. This resulted in 1- (2,2-difluoroethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole (50 mg, crude) as a brown solid. LCMS observed m/z = 309.25 [M+H]+. Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1-(2,2-difluoroethyl)-1H-indazol-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 293.2
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (50 mg, 0.06 mmol) and 1- (2,2-difluoroethyl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole (23 mg, 0.072 mmol) in dioxane (2 mL) and H2O (0.4 mL) were added K2CO3 (26 mg, 0.18 mmol) and Pd(dppf)Cl2 (5 mg, 0.006 mmol) in portions at room temperature under nitrogen atmosphere, after that, the resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 1 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature and was filtered, the filter cake was washed with dioxane (2 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 40% to 50% gradient in 20 min; detector, UV 254 nm. This resulted in 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (1-(2,2-difluoroethyl)-1H-indazol-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (45 mg, 80%yield) as a light yellow solid. LCMS observed m/z = 890.40 [M+H]+. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1-(2,2- difluoroethyl)-1H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1-(2,2-difluoroethyl)-1H-indazol-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (40 mg, 0.05 mmol) in DCM (2 mL) was added TFA (5 mL) dropwise at room temperature, after that, the resulting mixture was heated to 80 ℃ stirred at 80 ℃ for 30 min. After completion of reaction, the mixture was allowed to cool down to room temperature and concentrated to dryness under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Phenyl 5μm, 19*250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3+0.1% NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 34% B to 38% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 8.17) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)- 2-(2-(1-(2,2-difluoroethyl)-1H-indazol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (13.1 mg, 36% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 8.66 – 8.52 (m, 2H), 8.31 (s, 1H), 8.20 (dd, J = 8.8, 1.6 Hz, 1H), 8.08 (d, J = 8.6 Hz, 1H), 8.02 – 7.96 (m, 1H), 7.88 (d, J = 8.9 Hz, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 6.47 (tt, J = 54.7, 3.6 Hz, 1H), 5.43 (s, 2H), 4.99 (td, J = 15.3, 3.5 Hz, 2H), 4.55 (d, J = 12.3 Hz, 1H), 3.63 – 3.46 (m, 3H), 3.29 – 3.23 (m, 1H), 3.12 – 2.96 (m, 3H), 2.86 (d, J = 11.3 Hz, 1H), 2.73 – 2.64 (m, 1H), 2.48 – 2.39 (m, 3H), 1.32 – 1.14 (m, 3H). LCMS observed m/z = 800.40 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 294 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-1,2,3,4-tetrahydro-6- isoquinolyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 294
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-1,2,3,4-tetrahydro-6-isoquinolyl)-1,3,2-dioxaborolane. 1H NMR (400MHz, MeOD) δ 8.52 (s, 1H), 8.16 (d, J = 8.6 Hz, 1H), 8.07 – 7.98 (m, 2H), 7.82 (d, J = 2.1 Hz, 1H), 7.65 – 7.57 (m, 1H), 7.23 (d, J = 8.0 Hz, 1H), 5.45 (s, 2H), 4.71 (d, J = 12.7 Hz, 1H), 4.05 (d, J = 12.9 Hz, 1H), 3.93 (s, 2H), 3.84 – 3.66 (m, 2H), 3.52 – 3.39 (m, 1H), 3.23 – 3.01 (m, 7H), 2.95 (d, J = 11.6 Hz, 1H), 2.80 (d, J = 11.5 Hz, 1H), 2.67 (s, 3H), 2.52 (s, 3H), 1.32 (q, J = 8.2 Hz, 3H). LCMS observed m/z = 765.3 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra EXAMPLE 295 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-4-methyl-2H-indazol-5-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 295
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-4-methyl-2H-indazol-5-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.43 (s, 1H), 10.33 (s, 1H), 8.57 (d, J = 3.3 Hz, 2H), 8.06 (d, J = 8.4 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.84 (d, J = 9.1 Hz, 1H), 7.71 (dd, J = 8.7, 2.2 Hz, 1H), 7.49 (d, J = 9.0 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.2 Hz, 1H), 4.17 (s, 3H), 3.63 – 3.41 (m, 3H), 3.29 – 3.20 (m, 1H), 3.14 – 2.92 (m, 3H), 2.90 – 2.83 (m, 1H), 2.82 (s, 3H), 2.72 – 2.63 (m, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 764.3 [M+H]+. EXAMPLE 296
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1-acetyl-2,2-dimethyl-5- indolinyl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4- oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide.
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 1-[2,2-dimethyl- 5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-indolinyl]-1-ethanone. 1H NMR (400MHz, DMSO-d6) δ 10.42 (s, 1H), 8.55 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.92 (d, J = 9.2 Hz, 2H), 7.71 (dd, J = 8.9, 2.1 Hz, 1H), 5.38 (s, 2H), 4.53 (d, J = 12.3 Hz, 1H), 3.62 – 3.44 (m, 3H), 3.27 – 3.22 (m, 1H), 3.09 – 2.94 (m, 5H), 2.84 (d, J = 11.1 Hz, 1H), 2.71 – 2.61 (m, 1H), 2.44 (s, 3H), 2.35 (s, 3H), 1.54 (s, 6H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 807.3 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 297 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][6-ethyl-2-(4-fluoro-2-methyl- 1,3-benzoxazol-6-yl)-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}- 4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 297
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(4-fluoro-2- methyl-1,3-benzoxazol-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400MHz, DMSO-d6) δ 10.46 (s, 1H), 8.52 (s, 1H), 8.19 (d, J = 1.2 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.94 – 7.85 (m, 1H), 7.71 (dd, J = 8.7, 2.2 Hz, 1H), 5.41 (s, 2H), 4.53 (d, J = 12.4 Hz, 1H), 3.58 – 3.45 (m, 3H), 3.27 – 3.21 (m, 1H), 3.08 – 2.94 (m, 3H), 2.84 (d, J = 11.1 Hz, 1H), 2.68 (s, 3H), 2.67 – 2.62 (m, 1H), 2.43 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 769.2 [M+H]+. Note: One exchangeable proton not observed in NMR spectra. EXAMPLE 298 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-3-methyl-2H-indazol-5-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 298
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-3-methyl-2H-indazol-5-yl)-1,3,2-dioxaborolane. 1H NMR
(400MHz, DMSO-d6) δ 10.46 (s, 1H), 8.56 (s, 1H), 8.42 (d, J = 1.5 Hz, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.95 (dd, J = 9.1, 1.6 Hz, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.60 (d, J = 9.0 Hz, 1H), 5.42 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 4.07 (s, 3H), 3.61 – 3.44 (m, 3H), 3.28 – 3.21 (m, 1H), 3.10 – 2.93 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.71 – 2.67 (m, 1H), 2.65 (s, 3H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 764.3 [M+H]+. Note: One exchangeable proton not observed in NMR spectra EXAMPLE 299 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-7-methyl-2H-indazol-5-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 299
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-7-methyl-2H-indazol-5-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.44 (s, 1H), 10.30 (s, 1H), 8.57 (s, 1H), 8.46 (s, 1H), 8.34 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.77 (d, J = 1.5 Hz, 1H), 7.72 (dd, J = 8.6, 2.2 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 4.19 (s, 3H), 3.61 – 3.46 (m, 3H), 3.28 – 3.21 (m, 1H), 3.09 – 2.94 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.71 – 2.63 (m, 1H), 2.56 (s, 3H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 764.3 [M+H]+. EXAMPLE 300 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2'-methyl-4-oxo-7H,2'H-1,2',3,3',3a,7,7'- heptaaza-2,5'-biindenyl-7-yl)acetamide. Compound 300
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-2H-1,2,4-triazainden-6-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.48 (s, 1H), 9.25 (d, J = 1.8 Hz, 1H), 8.77 (t, J = 1.4 Hz, 1H), 8.53 (s, 1H), 8.36 (s, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 5.45 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 4.18 (s, 3H), 3.61 – 3.47 (m, 3H), 3.28 – 3.22 (m, 1H), 3.10 – 2.95 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.74 – 2.64 (m, 1H), 2.43 (s, 3H), 1.23 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. Note: One exchangeable proton not observed in NMR spectra EXAMPLE 301 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methyl-2H-benzo[d][1,2,3]triazol-5- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 301
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (2-methyl-2H- 1,2,3-benzotriazol-5-yl)boranediol.1H NMR (400 MHz, DMSO) δ 10.40 (br, 1H), 8.50 (d,
J = 23.8 Hz, 2H), 8.12 (dd, J = 9.0, 1.4 Hz, 1H), 7.99 (d, J = 9.2 Hz, 2H), 7.91 (d, J = 2.2 Hz, 1H), 7.69 – 7.61 (m, 1H), 5.36 (s, 2H), 4.47 (m, 4H), 3.45 (t, J = 10.9 Hz, 3H), 3.28 – 3.24 (m, 1H), 3.07 – 2.87 (m, 3H), 2.78 (d, J = 11.3 Hz, 1H), 2.63 – 2.58 (m, 1H), 2.37 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. Note: One exchangeable proton was not shown. EXAMPLE 302 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-propyl-2H-indazol-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 302
Step 1. Synthesis of 2-propyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H- indazole Compound 302.1
The title compound was prepared using a similar procedure as Compound 114.4, replacing 2-bromo-4H,6H,7H-pyrazolo[3,2-c][1,4]oxazine with 5-bromo-2-propyl-2H- indazole. LCMS observed m/z = 287.379 [M+H]+. Step 2. Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(2-propyl-2H-indazol-5- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 302
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-propyl-2H-indazol-5-yl)-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO) δ 10.38 (br, 2H), 8.53 – 8.36 (m, 3H), 8.01 (d, J = 8.6 Hz, 1H), 7.96 – 7.85 (m, 2H), 7.64 (d, J = 9.0 Hz, 2H), 5.34 (s, 2H), 4.47 (d, J = 12.5 Hz, 1H), 4.34 (t, J = 7.0 Hz, 2H), 3.52 – 3.41 (m, 3H), 3.19 (s, 1H), 2.95 (s, 3H), 2.77 (d, J = 11.3 Hz, 1H), 2.61 (s, 1H), 2.35 (s, 3H), 1.87 (p, J = 7.2 Hz, 2H), 1.15 (t, J = 7.4 Hz, 3H), 0.79 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 778.423 [M+H]+. EXAMPLE 303 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-((2,2- difluoroethyl)amino)benzo[d]oxazol-6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
Step 1: Preparation of 6-bromo-N-(2,2-difluoroethyl)-1,3-benzoxazol-2-amine. Compound 303.1
To a stirred solution of 6-bromo-2-chloro-1,3-benzoxazole (300 mg, 1.3 mmol) and DIEA (667 mg, 5.2 mmol) in DMF (10 mL) was added 2,2-difluoroethanamine (126 mg, 1.6 mmol) dropwise at room temperature. The resulting mixture was heated to 70 ℃ and stirred at 70 ℃ for 2 h. After that, the mixture was allowed to cool down to room temperature, quenched with water (20 mL), and extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 6-bromo-N- (2,2-difluoroethyl)-1,3-benzoxazol-2-amine (450 mg, crude) as a yellow solid. LCMS observed m/z = 276.95 [M+H]+. Step 2: Preparation of N-(2,2-difluoroethyl)-6-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1,3-benzoxazol-2-amine. Compound 303.2
To a stirred solution of 6-bromo-N-(2,2-difluoroethyl)-1,3-benzoxazol-2-amine (30 mg, 0.1 mmol) and bis(pinacolato)diboron (55 mg, 0.2 mmol) in dioxane (3 mL) were added KOAc (32 mg, 0.3 mmol) and Pd(dppf)Cl2 (8 mg, 0.01 mmol) in one portion at room temperature. The resulting mixture was heated to 100 ℃ and stirred at 100 ℃ for 2 h under nitrogen atmosphere. After completion of reaction, the mixture was allowed to cool down to room temperature and filtered, the filter cake was washed with dioxane (3 x 1 mL). The filtrate was concentrated under reduced pressure to afford N-(2,2-difluoroethyl)-6-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzoxazol-2-amine (50 mg, crude) as a brown solid. LCMS observed m/z = 325.20 [M+H]+. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-((2,2- difluoroethyl)amino)benzo[d]oxazol-6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide.
The title compound was prepared using similar procedure as compound 164 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-((2,2- difluoroethyl)amino)benzo[d]oxazol-6-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. 1H NMR (400 MHz, DMSO-d6) δ 10.45 (s, 1H), 8.75 – 8.66 (m, 1H), 8.55 (s, 1H), 8.11 – 7.92 (m, 4H), 7.72 (d, J = 8.7 Hz, 1H), 7.40 (d, J = 8.2 Hz, 1H), 6.20 (t, 1H), 5.40 (s, 2H), 4.61 – 4.46 (m, 1H), 3.84 – 3.70 (m, 2H), 3.54 – 3.48 (m, 2H), 3.26 – 3.15 (m, 4H), 3.12 – 2.94 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.71 – 2.63 (m, 1H), 2.44 (s, 2H), 1.29 – 1.14 (m, 3H). LCMS observed m/z = 816.2 [M+H]+. EXAMPLE 304 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-3'-methyl-4-oxo-7H,3'H-1,3,3',3a,7,7'- hexaaza-2,5'-biindenyl-7-yl)acetamide. Compound 304
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (1-methyl-1H- 1,4-diazainden-6-yl)boranediol. 1H NMR (400MHz, DMSO-d6) δ 10.47 (s, 1H), 10.25 (s, 1H), 9.11 (d, J = 1.8 Hz, 1H), 8.59 (s, 1H), 8.51 (s, 1H), 8.07 (d, J = 8.5 Hz, 1H), 7.99 (d, J
= 2.1 Hz, 1H), 7.81 (d, J = 3.2 Hz, 1H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 6.64 (d, J = 3.1 Hz, 1H), 5.44 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.92 (s, 3H), 3.62 – 3.46 (m, 3H), 3.29 – 3.21 (m, 1H), 3.11 – 2.94 (m, 3H), 2.86 (d, J = 11.3 Hz, 1H), 2.73 – 2.63 (m, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.3 [M+H]+. EXAMPLE 305 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3a,3a',7,7'-heptaaza-2,5'- biindenyl-7-yl)acetamide. Compound 305
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (1,3a,7-triaza-5- indenyl)boranediol.1H NMR (400MHz, DMSO-d6) δ 10.44 (s, 1H), 10.24 (s, 1H), 9.72 (d, J = 2.4 Hz, 1H), 9.12 (d, J = 2.4 Hz, 1H), 8.59 (s, 1H), 8.12 – 8.02 (m, 2H), 7.99 (d, J = 2.1 Hz, 1H), 7.81 (d, J = 1.4 Hz, 1H), 7.72 (dd, J = 8.6, 2.1 Hz, 1H), 5.42 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.61 – 3.46 (m, 3H), 3.28 – 3.21 (m, 1H), 3.11 – 2.95 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.73 – 2.63 (m, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 737.3 [M+H]+. EXAMPLE 306 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(1-methyl-5-methyl-1H-indazol-6-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 306
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (1-methyl-5- methyl-1H-indazol-6-yl)boranediol.1H NMR (400MHz, DMSO-d6) δ 10.43 (s, 1H), 10.24 (s, 1H), 8.58 (s, 1H), 8.11 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 8.02 (s, 1H), 7.97 (d, J = 1.9 Hz, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.69 (s, 1H), 5.42 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 4.06 (s, 3H), 3.61 – 3.47 (m, 3H), 3.29 – 3.23 (m, 1H), 3.13 – 2.97 (m, 3H), 2.87 (d, J = 11.2 Hz, 1H), 2.73 – 2.66 (m, 1H), 2.63 (s, 3H), 2.45 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 764.3 [M+H]+. EXAMPLE 307 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-(methylamino)benzo[d]oxazol-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 307
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with N-methyl[6- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3-benzoxazol-2-yl]amine. 1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 10.25 (s, 1H), 8.59 (s, 1H), 8.19 – 8.03 (m, 2H), 7.96 (ddd,
J = 18.3, 7.8, 1.8 Hz, 3H), 7.72 (dd, J = 8.7, 2.2 Hz, 1H), 7.34 (d, J = 8.1 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.3 Hz, 1H), 3.52 (t, J = 11.3 Hz, 3H), 3.26 (d, J = 11.6 Hz, 1H), 3.18 (d, J = 5.0 Hz, 1H), 3.10 – 2.97 (m, 3H), 2.93 (d, J = 4.8 Hz, 2H), 2.85 (d, J = 11.3 Hz, 1H), 2.68 (s, 1H), 2.46 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 766.3 [M+H]+. Note: One exchangeable proton was not shown. EXAMPLE 308 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(2-methyl-1,2,3,4-tetrahydro-7- isoquinolyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 308
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-1,2,3,4-tetrahydro-7-isoquinolyl)-1,3,2-dioxaborolane. 1H NMR (400MHz, MeOD) δ 8.53 (s, 1H), 8.17 (d, J = 8.6 Hz, 1H), 8.04 (d, J = 8.1 Hz, 1H), 7.97 (s, 1H), 7.83 (d, J = 2.0 Hz, 1H), 7.65 – 7.57 (m, 1H), 7.31 (d, J = 8.0 Hz, 1H), 5.46 (s, 2H), 4.72 (d, J = 12.7 Hz, 1H), 4.06 (d, J = 12.9 Hz, 1H), 4.00 (s, 2H), 3.76 (s, 2H), 3.45 (t, J = 12.1 Hz, 1H), 3.12 (d, J = 16.0 Hz, 7H), 2.96 (d, J = 11.7 Hz, 1H), 2.81 (d, J = 11.5 Hz, 1H), 2.71 (s, 3H), 2.52 (s, 3H), 1.33 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 765.3 [M+H]+. Note: 2 exchangeable protons were not visible in NMR spectra. EXAMPLE 309 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(2-oxo-6-indolinyl)-1,3,3a,7- tetraaza-7-indenyl) acetamide Compound 309
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(2-oxo-6-indolinyl)-1,3,3a,7-tetraaza-7- indenyl) acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H), 10.37 (s, 1H), 8.39 (s, 1H), 7.98 (d, J = 8.6 Hz, 1H), 7.90 (d, J = 2.1 Hz, 1H), 7.68 – 7.61 (m, 2H), 7.47 (d, J = 1.5 Hz, 1H), 7.27 (d, J = 7.8 Hz, 1H), 5.31 (s, 2H), 4.46 (d, J = 12.3 Hz, 1H), 3.48 (s, 2H), 3.46 – 3.39 (m, 3H), 3.20 – 3.10 (m, 1H), 2.98 – 2.86 (m, 3H), 2.76 (d, J = 11.2 Hz, 1H), 2.64 – 2.54 (m, 1H), 2.34 (s, 3H), 1.14 (q, J = 7.5, 6.0 Hz, 3H). LCMS observed m/z = 751.2 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 310 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-7H-1'-thia-1,3,3a,7,7'-pentaaza- 2,5'-biindenyl-7-yl) acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-7H-1'-thia-1,3,3a,7,7'-pentaaza-2,5'- biindenyl-7-yl) acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.46 (s, 1H), 10.25 (s, 1H), 9.26 (d, J = 2.1 Hz, 1H), 8.95 (d, J = 2.0 Hz, 1H), 8.60 (s, 1H), 8.08 (d, J = 8.6 Hz, 1H), 8.04 – 7.94 (m, 2H), 7.76 – 7.69 (m, 1H), 7.62 (d, J = 5.9 Hz, 1H), 5.44 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.63 – 3.47 (m, 3H), 3.33 – 3.22 (m, 1H), 3.10 – 2.97 (m,
3H), 2.87 (d, J = 11.2 Hz, 1H), 2.73 – 2.64 (m, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 753.2 [M+H]+. EXAMPLE 311 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(1-methyl-6-methyl-1H-indazol-5-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide.
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(1-methyl-6-methyl-1H-indazol-5-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, MeOD) δ 8.55 (s, 1H), 8.37 (s, 1H), 8.17 (d, J = 8.5 Hz, 1H), 8.01 (s, 1H), 7.81 (s, 1H), 7.61 (d, J = 8.6 Hz, 1H), 7.46 (s, 1H), 5.46 (s, 2H), 4.72 (d, J = 12.8 Hz, 1H), 4.14 – 4.07 (m, 1H), 4.05 (s, 3H), 3.84 – 3.70 (m, 2H), 3.52 – 3.41 (m, 1H), 3.21 – 3.08 (m, 3H), 2.97 (d, J = 11.7 Hz, 1H), 2.82 (d, J = 11.6 Hz, 1H), 2.76 (s, 3H), 2.53 (s, 3H), 1.34 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 764.3 [M+H]+. Note: 2 exchangeable protons were not visible in NMR spectra. EXAMPLE 312 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(1-methyl-3-methyl-1H-indazol-5-yl)-4- oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 312
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(1-methyl-3-methyl-1H-indazol-5-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, MeOD) δ 8.57 (d, J = 3.9 Hz, 2H), 8.27 (dd, J = 8.9, 1.5 Hz, 1H), 8.18 (d, J = 8.6 Hz, 1H), 7.83 (d, J = 2.0 Hz, 1H), 7.61 (dd, J = 8.2, 1.9 Hz, 1H), 7.56 (d, J = 8.9 Hz, 1H), 5.49 (s, 2H), 4.73 (d, J = 13.1 Hz, 1H), 4.14 – 4.05 (m, 1H), 4.01 (s, 3H), 3.85 – 3.71 (m, 2H), 3.52 – 3.40 (m, 1H), 3.21 – 3.10 (m, 3H), 2.97 (d, J = 11.7 Hz, 1H), 2.82 (d, J = 10.7 Hz, 1H), 2.57 (s, 3H), 2.53 (s, 3H), 1.35 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 764.3 [M+H]+. Note: 2 exchangeable protons were not visible in NMR spectra. EXAMPLE 313 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2'-methyl-4-oxo-7H,2'H-1,1',2',3,3a,7,7'- heptaaza-2,5'-biindenyl-7-yl)acetamide. Compound 313
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-methyl-2H-1,2,7-triazainden-5-yl)-1,3,2-dioxaborolane. 1H NMR
(400MHz, MeOD) δ 9.40 (d, J = 2.0 Hz, 1H), 8.99 (d, J = 2.1 Hz, 1H), 8.58 (s, 1H), 8.43 (s, 1H), 8.17 (d, J = 8.5 Hz, 1H), 7.82 (d, J = 2.0 Hz, 1H), 7.65 – 7.55 (m, 1H), 5.49 (s, 2H), 4.72 (d, J = 12.8 Hz, 1H), 4.28 (s, 3H), 4.12 (d, J = 13.1 Hz, 1H), 3.77 (t, J = 12.0 Hz, 2H), 3.46 (t, J = 11.9 Hz, 1H), 3.23 – 3.09 (m, 3H), 2.97 (d, J = 11.5 Hz, 1H), 2.82 (d, J = 11.4 Hz, 1H), 2.53 (s, 3H), 1.34 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. Note: 2 exchangeable protons were not visible in NMR spectra. EXAMPLE 314 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(1-methyl-1H-1,2,3-benzotriazol-5-yl)- 4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 314
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(1-methyl-1H-1,2,3-benzotriazol-5-yl)-1,3,2-dioxaborolane. 1H NMR (400MHz, MeOD) δ (s, 1H), 8.58 (s, 1H), 8.44 (d, J = 8.6 Hz, 1H), 8.17 (d, J = 8.4 Hz, 1H), 7.87 (d, J = 8.7 Hz, 1H), 7.83 (s, 1H), 7.61 (d, J = 8.9 Hz, 1H), 5.51 (s, 2H), 4.73 (d, J = 14.6 Hz, 1H), 4.37 (s, 3H), 4.11 (d, J = 12.8 Hz, 1H), 3.86 – 3.70 (m, 2H), 3.53 – 3.39 (m, 1H), 3.24 – 3.10 (m, 3H), 2.98 (d, J = 11.6 Hz, 1H), 2.83 (d, J = 11.5 Hz, 1H), 2.53 (s, 3H), 1.35 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. Note: 2 exchangeable protons were not visible in NMR spectra. EXAMPLE 315 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-morpholinobenzo[d]oxazol-6-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 315
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-(2-morpholino-1,3-benzoxazol-6-yl)-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO) δ 10.36 (br, 2H), 8.44 (s, 1H), 8.02 – 7.94 (m, 2H), 7.90 (d, J = 7.4 Hz, 2H), 7.64 (d, J = 8.7 Hz, 1H), 7.33 (d, J = 8.2 Hz, 1H), 5.32 (s, 2H), 4.46 (d, J = 12.4 Hz, 1H), 3.73 – 3.63 (m, 4H), 3.57 (d, J = 4.7 Hz, 4H), 3.49 – 3.37 (m, 3H), 3.20 – 3.14 (m, 1H), 2.94 (s, 3H), 2.76 (d, J = 11.0 Hz, 1H), 2.58 (s, 1H), 2.36 (s, 3H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 822.628 [M+H]+. EXAMPLE 316 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-(1,2,2-trimethyl-3H-indol-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 316
Step 1: Preparation of 1-acetyl-5-bromo-2H-indol-3-one. Compound 316.1
To a stirred solution of 1-acetyl-5-bromoindol-3-yl acetate (500 mg, 1.6 mmol) in EtOH (5 mL) and H2O (5 mL) was added Na2SO3 (532 mg, 4.2 mmol) in portions at room temperature. The resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 3 hours under nitrogen atmosphere. The mixture was cooled to room temperature, diluted with water (30 mL), and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 1-acetyl-5-bromo-2H-indol-3-one (420 mg, crude) as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 254.00 [M+H]+. Step 2: Preparation of 1-acetyl-5-bromo-2,2-dimethylindol-3-one. Compound 316.2
To a stirred solution of 1-acetyl-5-bromo-2H-indol-3-one (410 mg, 1.6 mmol) in THF (5 mL) was added sodium hydride (322 mg, 8.0 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C for 30 min under nitrogen atmosphere. To the above mixture was added iodomethane (458 mg, 3.2 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for additional 2 hours. Then the reaction was quenched with water (5 mL) at 0 °C and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 1-acetyl-5-bromo-2,2-dimethylindol-3-one (430 mg, crude) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 282.05 [M+H]+. Step 3: Preparation of 5-bromo-2,2-dimethyl-1H-indol-3-one. Compound 316.3
To a stirred solution of 1-acetyl-5-bromo-2,2-dimethylindol-3-one (420 mg, 1.4 mmol) in EtOH (2 mL) and H2O (2 mL) was added NaOH (178 mg, 4.4 mmol) in one portion at room temperature. The reaction mixture was heated to 50 °C and stirred at 50 °C for 2 hours under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (1:7) to afford 5-bromo-2,2-dimethyl-1H-indol-3- one (220 mg, 59.0% yield) as a yellow oil. LCMS observed m/z = 240.10 [M+H]+. Step 4: Preparation of 5-bromo-2,2-dimethyl-1,3-dihydroindole. Compound 316.4
A solution of 5-bromo-2,2-dimethyl-1H-indol-3-one (220 mg, 0.9 mmol, 1.0 equiv.) in triethylsilane (2 mL) and trifluoroacetic acid (2 mL) was stirred at room temperature for 3 hours under nitrogen atmosphere. Then the resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.1% NH3.H2O), 60% to 70% gradient in 10 min; detector, UV 254 nm, to afford 5-bromo-2,2-dimethyl-1,3- dihydroindole (120 mg, 57% yield) as a colorless oil. LCMS observed m/z = 226.10 [M+H]+. Step 5: Preparation of 5-bromo-1,2,2-trimethyl-3H-indole. Compound 316.5
To a stirred mixture of 5-bromo-2,2-dimethyl-1,3-dihydroindole (110 mg, 0.4 mmol) and formaldehyde (146 mg, 4.8 mmol) in methanol (3 mL) were added AcOH (29 mg, 0.4 mmol) and sodium cyanoboranuide (30 mg, 0.4 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 1 hour under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions:
column, C18; mobile phase, ACN in water (0.1% NH3.H2O), 80% to 95% gradient in 10 min; detector, UV 254 nm. This resulted in 5-bromo-1,2,2-trimethyl-3H-indole (65 mg, 51% yield) as a yellow oil. LCMS observed m/z = 239.95 [M+H]+. Step 6: Preparation of 1,2,2-trimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-3H-indole. Compound 316.6
To a stirred mixture of 5-bromo-1,2,2-trimethyl-3H-indole (60 mg, 0.2 mmol) and bis(pinacolato)diboron (76 mg, 0.3 mmol) in 1,4-dioxane (3 mL) were added AcOK (73 mg, 0.7 mmol) and Pd(dppf)Cl2CH2Cl2 (40 mg, 0.05 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 hours under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (1:1) to afford 1,2,2-trimethyl-5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-3H-indole (45 mg, 47% yield) as a colorless oil. LCMS observed m/z = 288.30 [M+H]+. Step 7: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-(1,2,2-trimethyl-3H-indol-5-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 316.7
To a stirred mixture of 1,2,2-trimethyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-3H-indole (36 mg, 0.1 mmol) and 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (100 mg, 0.1 mmol) in 1,4-
dioxane/H2O (5:1, 6 mL) were added Cs2CO3 (123 mg, 0.3 mmol) and Pd(dppf)Cl2CH2Cl2 (20 mg, 0.02 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 hours under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.1% NH3.H2O), 80% to 95% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(6-{4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-2-(1,2,2-trimethyl-3H- indol-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (70 mg, 40% yield) as a white solid. LCMS observed m/z = 967.30 [M+H]+. Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-2-(1,2,2-trimethyl-3H- indol-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. Compound 316
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-5-ethyl-7-oxo-2-(1,2,2-trimethyl-3H-indol-5-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)- N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (65 mg, 0.07 mmol) in trifluoroacetic acid (2 mL) was heated to 80 ℃ and stirred at 80 ℃ for 20 min under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18, 5 μm, 19*250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1% NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 42% B to 55%B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 7.47) to afford N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-2-(1,2,2-trimethyl-3H-indol-5-yl)-[1,2,4]triazolo[1,5-
a]pyrimidin-4-yl}acetamide (3.9 mg, 6% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.18 (s, 1H), 8.55 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.83 – 7.76 (m, 1H), 7.76 – 7.69 (m, 2H), 6.40 (d, J = 8.3 Hz, 1H), 5.36 (s, 2H), 4.54 (m, 1H), 3.61 – 3.45 (m, 3H), 3.29 -3.21 (m, 1H), 3.00 (s, 3H), 2.85 (s, 3H), 2.68 (s, 4H), 2.44 (s, 3H), 1.21 (s, 9H). LCMS observed m/z = 779.20 [M+H]+. Note: One exchangeable proton was not observed. EXAMPLE 317 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[7-(difluoromethoxy)-4- fluoropyrazolo[1,5-a]pyridin-5-yl]-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide
Step 1: Preparation of 1-amino-4-bromo-5-fluoro-2-methoxypyridin-1-ium. Compound 317.1
To a stirred solution of 4-bromo-5-fluoro-2-methoxypyridine (2.0 g, 9.7 mmol) in DCM (20 mL) was added amino 2,4,6-trimethylbenzenesulfonate (3.1 g, 14.6 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for additional 3 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure. The crude product was re-crystallized from EA (8 mL). The precipitated solids were collected by filtration and washed with EA (3 x 3 mL) to afford (1.0 g, 46% yield) of 1-amino-4-bromo-5-fluoro-2-methoxypyridin-1-ium as a white solid. LCMS observed m/z = 220.97 [M]+.
Step 2: Preparation of ethyl 5-bromo-4-fluoro-7-methoxypyrazolo[1,5-a]pyridine-3- carboxylate. Compound 317.2
To a stirred solution of 1-amino-4-bromo-5-fluoro-2-methoxypyridin-1-ium (130 mg, 0.6 mmol) and ethyl propiolate (69 mg, 0.7 mmol) in DMF (2 mL) was added K2CO3 (162 mg, 1.2 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for additional 1.5 h. After completed, poured the resulting mixture into water (5 mL) and extracted with EA (3 x 5 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford (30 mg, crude) of ethyl 5-bromo-4-fluoro-7-methoxypyrazolo[1,5-a]pyridine-3-carboxylate as a brown solid. LCMS observed m/z = 316.99 [M+H]+. Step 3: Preparation of 5-bromo-4-fluoropyrazolo[1,5-a]pyridin-7-ol. Compound 317.3
A solution of ethyl 5-bromo-4-fluoro-7-methoxypyrazolo[1,5-a]pyridine-3- carboxylate (270 mg, 0.9 mmol) in Hydrobromic acid(48%) (5 mL, 0.3 mmol) was stirred at 100°C for 2 h. After completed, the mixture was basified to pH 9 with NaOH(aq.). The resulting mixture was extracted with EtOEt (2 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford (150 mg, crude) of 5-bromo-4-fluoropyrazolo[1,5-a]pyridin-7-ol as a brown solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 230.95 [M+H]+. Step 4: Preparation of 5-bromo-7-(difluoromethoxy)-4-fluoropyrazolo[1,5- a]pyridine. Compound 317.4
To a stirred solution of 5-bromo-4-fluoropyrazolo[1,5-a]pyridin-7-ol (120 mg, 0.5 mmol) and sodium chlorodifluoroacetate (158 mg, 1.0 mmol) in DMF (3 mL) was added Cs2CO3 (254 mg, 0.8 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 100°C and stirred at 100 °C for 3 hours. After completed, poured the resulting mixture into water (5mL) and extracted with EA (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (30 mg, 20% yield) of 5-bromo-7-(difluoromethoxy)-4-fluoropyrazolo[1,5-a]pyridine as an off-white solid. LCMS observed m/z = 280.95 [M+H]+. Step 5: Preparation of 7-(difluoromethoxy)-4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine. Compound 317.5
To a stirred solution of 5-bromo-7-(difluoromethoxy)-4-fluoropyrazolo[1,5- a]pyridine (30 mg, 0.1 mmol) and bis(pinacolato)diboron (33 mg, 0.1 mmol) in dioxane (1 mL) were added AcOK (31 mg, 0.3 mmol) and Pd(dppf)Cl2 (16 mg, 0.1 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 80°C and stirred at 80°C for 2 hours. After completion of reaction, the reaction mixture was cooled to room temperature and used in the next step directly without further purification. LCMS observed m/z = 329.12 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[7- (difluoromethoxy)-4-fluoropyrazolo[1,5-a]pyridin-5-yl]-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide.
Compound 317
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[7-(difluoromethoxy)-4-fluoropyrazolo[1,5- a]pyridin-5-yl]-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide. 1H NMR (400 MHz, DMSO-d6) δ 10.46 (s, 1H), 8.27 (s, 1H), 8.15 – 8.01 (m, 1H), 7.97 (s, 1H), 7.80 – 7.66 (m, 1H), 7.63 – 7.39 (m, 1H), 7.34 – 7.20 (m, 1H), 7.15 (s, 1H), 5.42 (s, 2H), 4.81 – 4.61 (m, 1H), 4.60 – 4.42 (m, 1H), 3.52 (s, 2H), 3.07 (s, 2H), 2.92 – 2.67 (m, 2H), 2.36 (s, 2H), 2.22 – 1.84 (m, 2H), 1.76 (s, 1H), 1.24 (s, 5H). LCMS observed m/z = 820.19 [M+H]+. EXAMPLE 318 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(2-oxo-6-quinolyl)-1,3,3a,7- tetraaza-7-indenyl) acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(2-oxo-6-quinolyl)-1,3,3a,7-tetraaza-7- indenyl) acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 11.99 (s, 1H), 10.45 (s, 1H), 8.56 (s, 1H), 8.43 (d, J = 1.8 Hz, 1H), 8.25 – 8.18 (m, 1H), 8.11 – 8.02 (m, 2H), 7.99 (d, J = 2.1 Hz, 1H), 7.76 – 7.68 (m, 1H), 7.42 (d, J = 8.6 Hz, 1H), 6.59 – 6.54 (m, 1H), 5.41 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 3.60 – 3.46 (m, 3H), 3.24 (s, 1H), 3.07 – 2.95 (m,
3H), 2.85 (d, J = 11.1 Hz, 1H), 2.67 (d, J = 9.7 Hz, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 763.2 [M+H]+. Note: One exchangeable proton was not observed. EXAMPLE 319 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(2-ethyl-2H- indazol-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 319
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(2-ethyl-2H- indazol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane. 1H NMR (400 MHz, DMSO) δ 10.37 (br, 2H), 8.46 (d, J = 12.2 Hz, 3H), 8.00 (d, J = 8.7 Hz, 1H), 7.90 (d, J = 6.8 Hz, 2H), 7.64 (d, J = 9.0 Hz, 2H), 5.29 (d, J = 41.8 Hz, 2H), 4.59 – 4.37 (m, 3H), 3.52 – 3.40 (m, 3H), 3.17 (s, 1H), 2.94 (t, J = 11.8 Hz, 3H), 2.78 (d, J = 12.0 Hz, 1H), 2.58 (s, 1H), 2.37 (d, J = 4.1 Hz, 3H), 1.45 (t, J = 7.3 Hz, 3H), 1.19 – 1.09 (m, 3H). LCMS observed m/z = 764.697 [M+H]+. EXAMPLE 320 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(5,8-difluoroquinolin-6- yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 320
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (5,8-difluoro-6- quinolyl)boranediol.1H NMR (400 MHz, DMSO) δ 10.46 (s, 1H), 10.25 (s, 1H), 9.13 (dd, J = 4.3, 1.6 Hz, 1H), 8.71 (d, J = 8.5 Hz, 1H), 8.60 (s, 1H), 8.15 (dd, J = 11.2, 6.0 Hz, 1H), 8.07 (d, J = 8.5 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.85 (dd, J = 8.6, 4.2 Hz, 1H), 7.73 (d, J = 8.8 Hz, 1H), 5.45 (s, 2H), 4.56 (d, J = 12.4 Hz, 1H), 3.55 (q, J = 11.4 Hz, 3H), 3.29 (s, 1H), 3.05 (d, J = 11.9 Hz, 3H), 2.87 (d, J = 11.2 Hz, 1H), 2.68 (s, 1H), 2.46 (s, 3H), 1.24 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 783.487 [M+H]+. EXAMPLE 321 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(1-methyl-1H-indazol-5-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 321
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (1-methyl-1H- indazol-5-yl)boranediol.
1H NMR (400 MHz, DMSO) δ 10.45 (br, 1H), 10.26 (br, 1H), 8.56 (d, J = 16.9 Hz, 2H), 8.20 (s, 1H), 8.16 (d, J = 8.8 Hz, 1H), 8.08 (d, J = 8.6 Hz, 1H), 7.99 (s, 1H), 7.75 (dd, J = 24.2, 8.6 Hz, 2H), 5.43 (s, 2H), 4.55 (d, J = 12.3 Hz, 1H), 4.09 (s, 3H), 3.55 (q, J = 11.8 Hz, 3H), 3.26 (d, J = 12.1 Hz, 1H), 3.03 (d, J = 10.4 Hz, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.68 (d, J = 10.3 Hz, 1H), 2.46 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 750.3 [M+H]+. EXAMPLE 322 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-2-(8-(trifluoromethyl)quinolin-6-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 322
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with [8- (trifluoromethyl)-6-quinolyl]boranediol. 1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H), 10.25 (s, 1H), 9.16 – 9.06 (m, 2H), 8.79 – 8.75 (m, 1H), 8.72 – 8.65 (m, 1H), 8.60 (s, 1H), 8.08 (d, J = 8.5 Hz, 1H), 8.00 (d, J = 2.1 Hz, 1H), 7.82 – 7.77 (m, 1H), 7.73 (d, J = 8.2 Hz, 1H), 5.47 (s, 2H), 4.56 (d, J = 12.4 Hz, 1H), 3.55 (q, J = 11.5 Hz, 3H), 3.31 – 3.23 (m, 1H), 3.04 (dd, J = 19.4, 10.2 Hz, 3H), 2.88 (d, J = 11.2 Hz, 1H), 2.70 (d, J = 11.9 Hz, 1H), 2.46 (s, 3H), 1.27 – 1.21 (m, 3H). LCMS observed m/z = 815.3 [M+H]+. EXAMPLE 323 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2-methylquinolin-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 323
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (2-methyl-6- quinolyl)boranediol.1H NMR (400 MHz, DMSO) δ 10.46 (s, 1H), 10.25 (s, 1H), 8.70 (d, J = 1.9 Hz, 1H), 8.60 (s, 1H), 8.46 – 8.37 (m, 2H), 8.07 (dd, J = 20.1, 8.7 Hz, 2H), 8.00 (d, J = 2.1 Hz, 1H), 7.73 (dd, J = 8.8, 2.1 Hz, 1H), 7.50 (d, J = 8.5 Hz, 1H), 5.45 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.55 (q, J = 11.6 Hz, 3H), 3.27 (d, J = 12.0 Hz, 1H), 3.04 (d, J = 9.1 Hz, 3H), 2.87 (d, J = 11.4 Hz, 1H), 2.69 (s, 4H), 2.46 (s, 3H), 1.24 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 761.3 [M+H]+. EXAMPLE 324 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-2-(1-methyl-2-oxo-3,4-dihydro-6- quinolyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl) acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-2-(1-methyl-2-oxo-3,4-dihydro-6-quinolyl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl) acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 8.52 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 8.03 – 7.95 (m, 3H), 7.75 – 7.70 (m, 1H), 7.23 (d, J = 8.5 Hz, 1H), 5.40 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.58 – 3.47 (m, 3H), 3.29 (s, 3H), 3.25 (d, J = 12.3 Hz, 1H), 3.06 – 2.93 (m, 6H), 2.84 (d, J = 11.1 Hz, 1H), 2.70
– 2.67 (m, 1H), 2.61 – 2.59 (m, 1H), 2.43 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 779.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 325 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(2-oxo-5-indolinyl)-1,3,3a,7- tetraaza-7-indenyl) acetamide Compound 325
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-2-(2-oxo-5-indolinyl)-1,3,3a,7-tetraaza-7- indenyl) acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.56 (s, 1H), 10.48 (s, 1H), 10.24 (s, 1H), 8.59 (s, 1H), 8.04 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.71 (d, J = 8.1 Hz, 2H), 7.34 (t, J = 7.9 Hz, 1H), 6.95 (d, J = 7.7 Hz, 1H), 5.42 (s, 2H), 4.54 (d, J = 12.4 Hz, 1H), 3.82 (s, 2H), 3.52 - 3.50 (m, 3H), 3.31 - 3.23 (m, 1H), 3.10 – 2.97 (m, 3H), 2.85 (d, J = 11.1 Hz, 1H), 2.67 (d, J = 9.4 Hz, 1H), 2.46 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 751.3 [M+H]+. EXAMPLE 326 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-3'-fluoro-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3a,7,7a'- hexaaza-2,5'-biindenyl-7-yl) acetamide Compound 326
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-3'-fluoro-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-7H-1,1',3,3a,7,7a'-hexaaza-2,5'-biindenyl-7- yl) acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.46 (s, 1H), 8.72 (d, J = 7.4 Hz, 1H), 8.57 (s, 1H), 8.31 (d, J = 1.8 Hz, 1H), 8.22 (d, J = 3.6 Hz, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.75 – 7.69 (m, 1H), 7.53 – 7.48 (m, 1H), 5.43 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.60 – 3.45 (m, 3H), 3.29 – 3.23 (m, 1H), 3.09 – 2.96 (m, 3H), 2.86 (d, J = 11.3 Hz, 1H), 2.68 (d, J = 9.8 Hz, 1H), 2.45 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 754.2 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 327 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1-benzofuran-3-yl)-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl] acetamide Compound 327
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1-benzofuran-3-yl)-6-ethyl-5-{4-[(5-hydroxy- 6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.47 (s, 1H), 8.71 (s, 1H), 8.55 (s, 1H), 8.33 – 8.27 (m, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.72 (d, J = 8.2 Hz, 2H), 7.48 – 7.37 (m, 2H), 5.43 (s, 2H), 4.55 (d, J = 12.4 Hz, 1H), 3.62 – 3.47 (m, 3H), 3.30 – 3.23 (m, 1H), 3.09 – 2.97 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.71 – 2.65 (m, 1H), 2.44 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 736.3 [M+H]+. Note: the proton on -OH cannot be seen. EXAMPLE 328 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](3'-chloro-6-ethyl-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-7H,1'H-1,1',3,3a,7,7'- hexaaza-2,5'-biindenyl-7-yl)acetamide.
Compound 328
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 2-(3-chloro-1H- 1,7-diazainden-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane.1H NMR (400MHz, DMSO- d6) δ 12.31 (s, 1H), 10.47 (s, 1H), 9.05 (d, J = 2.0 Hz, 1H), 8.60 – 8.53 (m, 2H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.82 (d, J = 1.6 Hz, 1H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 5.43 (s, 2H), 4.54 (d, J = 12.3 Hz, 1H), 3.62 – 3.45 (m, 3H), 3.28 – 3.23 (m, 1H), 3.13 – 2.97 (m, 3H), 2.85 (d, J = 11.3 Hz, 1H), 2.68 (d, J = 10.7 Hz, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 770.2 [M+H]+. Note: One exchangeable proton was not observed. EXAMPLE 329 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]{2-[p- (aminosulfonylamino)phenyl]-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl}acetamide. Compound 329
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with N-
aminosulfonyl[p-(dihydroxyboryl)phenyl]amine. 1H NMR (400MHz, DMSO-d6) δ 10.41 (d, J = 9.3 Hz, 1H), 10.25 (s, 1H), 9.91 (s, 1H), 8.58 (s, 1H), 8.05 (dd, J = 8.6, 3.7 Hz, 1H), 8.02 – 7.94 (m, 2H), 7.77 (d, J = 8.4 Hz, 1H), 7.71 (dd, J = 8.7, 2.1 Hz, 1H), 7.36 – 7.12 (m, 2H), 6.62 (d, J = 8.6 Hz, 1H), 5.62 (s, 1H), 5.36 (d, J = 11.5 Hz, 2H), 4.60 – 4.44 (m, 1H), 3.60 – 3.43 (m, 3H), 3.28 – 3.19 (m, 1H), 3.12 – 2.90 (m, 3H), 2.83 (d, J = 11.4 Hz, 1H), 2.62 (d, J = 14.3 Hz, 1H), 2.44 (s, 3H), 1.26 – 1.15 (m, 3H). LCMS observed m/z = 790.2 [M+H]+. EXAMPLE 330 Synthesis of [p-(7-{[N-2-chloro-4-(trifluoromethyl)phenylcarbamoyl]methyl}-6- ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-2-indenyl)phenoxy]acetic acid. Compound 330
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with [p- (dihydroxyboryl)phenoxy]acetic acid.1H NMR (400MHz, DMSO-d6) δ 13.16 (s, 1H), 10.42 (s, 1H), 10.25 (s, 1H), 8.58 (s, 1H), 8.08 – 8.02 (m, 2H), 8.02 (d, J = 2.2 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.7, 2.1 Hz, 1H), 7.09 – 7.00 (m, 2H), 5.38 (s, 2H), 4.74 (s, 2H), 4.59 – 4.44 (m, 1H), 3.62 – 3.45 (m, 3H), 3.28 – 3.20 (m, 1H), 3.09 – 2.92 (m, 3H), 2.84 (d, J = 11.0 Hz, 1H), 2.71 – 2.61 (m, 1H), 2.45 (s, 3H), 1.20 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 770.3 [M+H]+. EXAMPLE 331 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl](6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-[p-(3-methylureido)phenyl]-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide. Compound 331
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with 4,4,5,5- tetramethyl-2-[p-(3-methylureido)phenyl]-1,3,2-dioxaborolane. 1H NMR (400MHz, DMSO-d6) δ 10.42 (s, 1H), 8.82 (s, 1H), 8.54 (s, 1H), 8.06 (d, J = 8.6 Hz, 1H), 7.99 – 7.92 (m, 3H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 7.53 – 7.51 (m, 1H), 6.13 (d, J = 4.8 Hz, 1H), 5.37 (s, 2H), 4.53 (d, J = 12.2 Hz, 1H), 4.49 (q, J = 1.8 Hz, 1fH), 4.21 (q, J = 1.9 Hz, 1H), 3.52 (q, J = 11.5 Hz, 3H), 3.27 – 3.19 (m, 1H), 3.06 – 2.94 (m, 3H), 2.83 (d, J = 11.2 Hz, 1H), 2.69 – 2.61 (m, 4H), 2.43 (s, 3H), 1.20 (t, J = 7.5 Hz, 3H). LCMS observed m/z =
. [M+H]+. EXAMPLE 332 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-2-[2-(3-oxetanyl)-2H-indazol-5-yl]-4- oxo-1,3,3a,7-tetraaza-7-indenyl) acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-2-[2-(3-oxetanyl)-2H-indazol-5-yl]-4-oxo-1,3,3a,7- tetraaza-7-indenyl) acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.45 (s, 1H), 10.25 (s, 1H), 8.61 – 8.55 (m, 2H), 8.39 (s, 1H), 8.19 – 8.14 (m, 1H), 8.10 – 8.06 (m, 1H), 7.99 (t, J = 2.6 Hz, 1H), 7.84 (d, J = 8.9 Hz, 1H), 7.75 – 7.69 (m, 1H), 6.14 – 6.07 (m,
1H), 5.42 (s, 2H), 5.09 – 4.97 (m, 4H), 4.55 (d, J = 12.1 Hz, 1H), 3.62 – 3.41 (m, 3H), 3.31 - 3.25 (m, 1H), 3.03 - 3.00 (m, 3H), 2.86 (d, J = 11.5 Hz, 1H), 2.72 – 2.64 (m, 1H), 2.46 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 792.3 [M+H]+. EXAMPLE 333 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-2-[p-(methylsulfinyl) phenyl]-4-oxo- 1,3,3a,7-tetraaza-7-indenyl) acetamide Compound 333
The title compound was prepared using similar procedure as compound 164 to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl) carbonyl]-1-piperazinyl}-2-[p-(methylsulfinyl) phenyl]-4-oxo-1,3,3a,7- tetraaza-7-indenyl) acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.44 (s, 1H), 10.25 (s, 1H), 8.59 (s, 1H), 8.32 – 8.26 (m, 2H), 8.06 (d, J = 8.6 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.85 (d, J = 8.5 Hz, 2H), 7.75 – 7.69 (m, 1H), 5.42 (s, 2H), 4.55 (d, J = 12.5 Hz, 1H), 3.58 – 3.50 (m, 3H), 3.30 - 3.22 (m, 1H), 3.09 – 2.96 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.81 (s, 3H), 2.68 (d, J = 9.7 Hz, 1H), 2.46 (s, 3H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 758.2 [M+H]+. EXAMPLE 334 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2-(difluoromethoxy)-4- fluoropyrazolo[1,5-a]pyridin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 334
Step 1: Preparation of 1-tert-butyl 3-methyl 2-(4-bromo-3-fluoropyridin-2- yl)propanedioate. Compound 334.1
To a solution of 1-tert-butyl 3-methyl propanedioate (2.7 g, 15.5 mmol) in DMF (10 mL) was added NaH (495 mg, 20.6 mmol) at 0 °C. The mixture was stirred at 0 °C for 0.5 hour. Then a solution of 4-bromo-2,3-difluoropyridine (2.0 g, 10.3 mmol) in DMF (3 mL) was added dropwise at 0 °C. The resulting mixture was allowed to warm to room temperature and stirred for 16 hours. After completion of reaction, the reaction mixture was quenched by addition of water (30 mL). The aqueous layer was extracted with ethyl acetate (3x20 mL). The combined organic phases were washed with brine (3 x 5 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (3 g, 83.6% yield) of 1-tert-butyl 3-methyl 2-(4-bromo-3- fluoropyridin-2-yl)propanedioate as a colorless oil. LCMS observed m/z = 348.05 [M+H]+. Step 2: Preparation of methyl 2-(4-bromo-3-fluoropyridin-2-yl)acetate. Compound 334.2
To a stirred solution of 1-tert-butyl 3-methyl 2-(4-bromo-3-fluoropyridin-2- yl)propanedioate (3 g, 8.6 mmol) in DCM (20 mL) was added TFA (5 mL) dropwise at room temperature. The resulting mixture was heated to 45 °C and stirred at 45 °C for 1 h.
After completion of reaction, the reaction mixture was concentrated under reduced pressure. The residue was dissolved in H2O (5 mL). The mixture basified to pH 8 with saturated NaHCO3 (aq.). The aqueous layer was extracted with EA (3 x 4 mL). The combined organic phases were washed with brine (2x3 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford (1.8 g, 84.2% yield) of methyl 2-(4-bromo- 3-fluoropyridin-2-yl)acetate as a light yellow oil. LCMS observed m/z = 248.00 [M+H]+. Step 3: Preparation of 1-amino-4-bromo-3-fluoro-2-(2-methoxy-2- oxoethyl)pyridin-1-ium 2,4,6-trimethylbenzenesulfonate Compound 334.3
To a stirred solution of methyl 2-(4-bromo-3-fluoropyridin-2-yl)acetate (1.8 g, 7.3 mmol) in DCM (20 mL) was added amino 2,4,6-trimethylbenzenesulfonate (2.3 g, 10.9 mmol) at room temperature. The resulting mixture was stirred at room temperature for 4 h. After completion of reaction, the reaction mixture was concentrated under reduced pressure. The crude product was re-crystallized from EA (5 mL) to afford (2 g, 59.3% yield) of 1- amino-4-bromo-3-fluoro-2-(2-methoxy-2-oxoethyl)pyridin-1-ium 2,4,6- trimethylbenzenesulfonate as a white solid. LCMS observed m/z = 262.85 [M] +. Step 4: 5-bromo-4-fluoropyrazolo[1,5-a]pyridin-2-ol.
To a stirred solution of 1-amino-4-bromo-3-fluoro-2-(2-methoxy-2- oxoethyl)pyridin-1-ium 2,4,6-trimethylbenzenesulfonate (1 g, 2.2 mmol) in DMF (10 mL) was added K2CO3 (0.6 g, 4.3 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 16 h. After completion of reaction, the reaction mixture was quenched by addition of water (20 mL). The aqueous layer was extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with brine (3 x 10mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by silica gel column chromatography (eluted: with 20%
EA in PE) to afford (220 mg, 44.1%yield) of 5-bromo-4-fluoropyrazolo[1,5-a]pyridin-2-ol as a white solid. LCMS observed m/z = 231.00 [M+H]+. Step 5: Preparation of 5-bromo-2-(difluoromethoxy)-4-fluoropyrazolo[1,5- a]pyridine. Compound 334.5
To a stirred solution of 1-amino-4-bromo-3-fluoro-2-(2-methoxy-2- oxoethyl)pyridin-1-ium 2,4,6-trimethylbenzenesulfonate (1 g, 2.2 mmol) in DMF (10 mL) was added K2CO3 (0.6 g, 4.3 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for16 h. After completion of reaction, the reaction mixture was quenched by addition of water (20 mL). The aqueous layer was extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with brine (3x10mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by silica gel column chromatography (eluted: with 20% EA in PE) to afford (72 mg, 21.1%yield) of 5-bromo-2-(difluoromethoxy)-4- fluoropyrazolo[1,5-a]pyridine as a yellow solid. LCMS observed m/z = 280.95 [M+H]+. Step 6: Preparation of 2-(difluoromethoxy)-4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine. Compound 334.6
To a stirred solution of 5-bromo-2-(difluoromethoxy)-4-fluoropyrazolo[1,5- a]pyridine (72 mg, 0.3 mmol) and bis(pinacolato)diboron (97 mg, 0.4 mmol) in dioxane (5 mL) was added AcOK (50mg, 0.5 mmol) and Pd(dppf)Cl2 (375 mg, 0.5 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 2 h. After completion of reaction, the reaction mixture was cooled to room temperature and used in the next step directly without further purification. LCMS observed m/z = 329.20 [M+H] +.
Step 7: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2- (difluoromethoxy)-4-fluoropyrazolo[1,5-a]pyridin-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide Compound 334
To a stirred solution of 2-(difluoromethoxy)-4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (45 mg, 0.1 mmol) in dioxane (4 mL) was added 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate AC (80 mg, 0.1 mmol), K2CO3 (31 mg, 0.2 mmol), H2O (1 mL) and Pd(dppf)Cl2 (8 mg, 0.01 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 3 h. After completion of reaction, the reaction mixture was quenched by addition of water (3 mL). The aqueous layer was extracted with EA (3 x 3 mL). The combined organic phases were washed with brine (3 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by Prep-HPLC (Column: XBridge BEH C185μm, 19*250mm; mobile phase: 30-37% ACN in water(10 mmol/L NH4HCO3+0.1%NH3.H2O)) to afford (3.5 mg, 3.7% yield) N-[2-chloro-4- (trifluoromethyl)phenyl]-2-{2-[2-(difluoromethoxy)-4-fluoropyrazolo[1,5-a]pyridin-5-yl]- 5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 10.06 (s, 1H), 8.54 (s, 1H), 8.40 (d, J = 7.3 Hz, 1H), 8.12 – 7.85 (m, 3H), 7.76 – 7.69 (m, 1H), 7.20 (t, J = 7.0 Hz, 1H), 5.56 – 5.37 (m, 3H), 4.55 (m, 1H), 3.61 – 3.44 (m, 3H), 3.28 – 3.23 (m, 1H), 3.15 – 2.79 (m, 5H), 2.75 – 2.63 (m, 1H), 2.44 (s, 3H), 1.22 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 820.20 [M+H] +. EXAMPLE 335
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[1-(difluoromethyl)-4- fluoro-2-oxopyrazolo[1,5-a]pyridin-5-yl]-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide Compound 335
Step 1: Preparation of 5-bromo-1-(difluoromethyl)-4-fluoropyrazolo[1,5-a]pyridin- 2-one. Compound 335.1
To a stirred solution of 1-amino-4-bromo-3-fluoro-2-(2-methoxy-2- oxoethyl)pyridin-1-ium 2,4,6-trimethylbenzenesulfonate (1.0 g, 2.2 mmol) in DMF (10 mL) was added K2CO3 (0.6 g, 4.3 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for16 h. After completion of reaction, the reaction mixture was quenched by addition of water (20 mL). The aqueous layer was extracted with ethyl acetate (3 x 20 mL). The combined organic phases were washed with brine (3x10mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by silica gel column chromatography (eluted: with 20% EA in PE) to afford (187 mg, 54.9% yield) of 5-bromo-1-(difluoromethyl)-4- fluoropyrazolo[1,5-a]pyridin-2-one as a white solid as a yellow solid. LCMS observed m/z = 280.95 [M+H] +. Step 2: Preparation of 2-(difluoromethoxy)-4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine. Compound 335.2
To a stirred solution of 5-bromo-1-(difluoromethyl)-4-fluoropyrazolo[1,5-a]pyridin- 2-one (187 mg, 0.7 mmol) and bis(pinacolato)diboron (253 mg, 1.0 mmol) in dioxane (5 mL) was added AcOK (131 mg, 1.3 mmol) and Pd(dppf)Cl2 (97 mg, 0.13 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h. After completion of reaction, the reaction mixture was cooled to room temperature and used in the next step directly without further purification. LCMS observed m/z = 329.20 [M+H] +. Step 3: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[1- (difluoromethyl)-4-fluoro-2-oxopyrazolo[1,5-a]pyridin-5-yl]-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide. Compound 335
To a stirred solution of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide, Intermediate AC (70 mg, 0.1 mmol) in dioxane (4 mL) was added 1-(difluoromethyl)-4-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridin-2-one (39 mg, 0.1 mmol), K2CO3 (28 mg, 0.2 mmol), H2O (0.5 mL) and Pd(dppf)Cl2 (7 mg, 0.01 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 80 °C for additional 3 h. After completion of reaction, the reaction mixture was quenched by addition of water 3 mL. The aqueous layer was extracted with EA (3 x 3 mL). The combined organic phases were washed with brine (3 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by Prep-HPLC (Column: XBridge BEH C185μm,
19*250mm; mobile phas: 30-37% ACN in Water(10 mmol/L NH4HCO3+0.1%NH3.H2O)) to afford (10.2 mg, 12.4%yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{2-[1- (difluoromethyl)-4-fluoro-2-oxopyrazolo[1,5-a]pyridin-5-yl]-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}acetamide as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 10.33 (s, 2H), 8.65 (d, J = 7.2 Hz, 1H), 8.54 (s, 1H), 8.07 (d, J = 8.7 Hz, 1H), 7.98 (d, J = 2.1 Hz, 1H), 7.75 – 7.35 (m, 3H), 6.76 (s, 1H), 5.42 (s, 2H), 4.55 (m, 1H), 3.29 – 3.22 (m, 1H), 3.61 – 3.48 (m, 3H), 3.13 – 2.97 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.74 – 2.64 (m, 1H), 2.44 (s, 3H), 1.23 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 820.15 [M+H] +. EXAMPLE 336 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(8-methylquinolin-6-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 336
The title compound was prepared using a similar procedure as compound 164, replacing (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)boronic acid with (8-methyl-6- quinolyl)boranediol. 1H NMR (400 MHz, DMSO) δ 10.47 (s, 1H), 8.98 (dd, J = 4.2, 1.8 Hz, 1H), 8.60 (d, J = 1.9 Hz, 1H), 8.56 – 8.47 (m, 2H), 8.34 (d, J = 2.0 Hz, 1H), 8.09 (d, J = 8.6 Hz, 1H), 7.99 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.61 (dd, J = 8.3, 4.2 Hz, 1H), 5.44 (s, 2H), 4.54 (d, J = 12.5 Hz, 1H), 3.54 (q, J = 12.0 Hz, 3H), 3.24 (s, 1H), 3.12 – 2.94 (m, 3H), 2.86 (d, J = 11.2 Hz, 1H), 2.80 (s, 3H), 2.70 (d, J = 5.9 Hz, 1H), 2.43 (s, 3H), 1.23 (t, J = 7.5 Hz, 3H). One exchangeable proton was not shown. Note: δ 2.54 is DMSO. LCMS observed m/z = 761.3 [M+H]+. EXAMPLE 337
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(4,4-dimethyl-1,4- azasilinan-1-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
The title compound was prepared using similar procedure as compound 164 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(4,4-dimethyl-1,4-azasilinan-1-yl)-5-ethyl-6- (4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.28 (s, 1H), 10.17 (s, 1H), 8.51 (s, 1H), 7.95 (d, J = 8.6 Hz, 1H), 7.90 (d, J = 2.1 Hz, 1H), 7.68 – 7.63 (m, 1H), 5.14 (s, 2H), 4.44 (d, J = 12.4 Hz, 1H), 3.70 – 3.57 (m, 4H), 3.49 – 3.36 (m, 3H), 3.16 (t, J = 12.4 Hz, 1H), 2.96 – 2.82 (m, 3H), 2.71 (d, J = 11.3 Hz, 1H), 2.53 (d, J = 10.7 Hz, 1H), 2.38 (s, 3H), 1.09 (t, J = 7.4 Hz, 3H), 0.70 – 0.56 (m, 4H), 0.00 (s, 6H). LCMS observed m/z = 747.3 [M+H]+. EXAMPLE 338 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-{5-methyl-6-oxo-4H,7H-pyrazolo[1,5-a]pyrazin-2-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 338
Step 1: Preparation of (5-bromo-2H-pyrazol-3-yl)methanol. Compound 338.1
To a stirred solution of methyl 5-bromo-2H-pyrazole-3-carboxylate (5.0 g, 24.4 mmol) in THF (70 mL) was added BH3.THF (73.1 mL, 73.1 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature. After completion of reaction, the reaction was cooled to 0 °C, HCl (2 N, 20 mL) aqueous solution was added and the reaction was heated to 650 °C and stirred at 65 °C for 1 h. The reaction was cooled to 25 °C and concentrated under reduced pressure. The resulting mixture was poured into water and extracted with EA (3 x 10 mL). The combined organic phases were washed with brine (3 x 5 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford (3.5 g, crude) of (5-bromo-2H-pyrazol-3-yl)methanol as a white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 177.01 [M+H]+. Step 2: Preparation of 3-(azidomethyl)-5-bromo-2H-pyrazole. Compound 338.2
To a stirred solution of (5-bromo-2H-pyrazol-3-yl)methanol (3.5 g, 19.8 mmol) in THF (50 mL) was added DPPA (6.0 g, 21.8 mmol) and DBU (3.6 g, 23.7 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at room temperature. After completion of reaction, the reaction mixture was quenched by addition of saturated NaHCO3 aqueous solution (20 mL). The resulting mixture was poured into water and extracted with EA (3 x 20 mL). The combined organic phases were washed with brine (2 x 10 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by silica gel column chromatography (eluted: with 22% EtOAc in petroleum ether) to afford (1.8 g, 45% yield) 3-(azidomethyl)-5-bromo-2H-pyrazole as a colorless oil. LCMS observed m/z = 202.02 [M+H]+. Step 3: Preparation of ethyl 2-[5-(azidomethyl)-3-bromopyrazol-1-yl]acetate.
Compound 338.3
To a stirred solution of 3-(azidomethyl)-5-bromo-2H-pyrazole (1.7 g, 8.4 mmol) and ethyl bromoacetate (1.4 g, 8.4 mmol) in DMF (15 mL) was added K2CO3 (2.3 g, 16.8 mmol) in portions at room temperature. The resulting mixture was stirred for additional 3 h at room temperature. After completed, poured the resulting mixture into water and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluted: with 20% EtOAc in petroleum ether) to afford (1.7 g, 70% yield) of ethyl 2-[5-(azidomethyl)-3- bromopyrazol-1-yl]acetate as a colorless oil. LCMS observed m/z = 288.11 [M+H]+. Step 4: Preparation of 2-bromo-4H,5H,7H-pyrazolo[1,5-a]pyrazin-6-one. Compound 338.4
To a stirred solution of ethyl 2-[5-(azidomethyl)-3-bromopyrazol-1-yl]acetate (1.7 g, 5.9 mmol) in THF (15 mL) and H2O (1.5 mL) was added PPh3 (4.6 g, 17.7 mmol) in portions at room temperature. The resulting mixture was stirred for additional 4 h at room temperature. After completed, the resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography (eluted: with 5% MeOH in DCM) to afford (650 mg, 51% yield) of 2-bromo-4H,5H,7H-pyrazolo[1,5-a]pyrazin-6-one as a white solid. LCMS observed m/z = 216.02 [M+H]+. Step 5: Preparation of 2-bromo-5-methyl-4H,7H-pyrazolo[1,5-a]pyrazin-6-one. Compound 338.5
To a stirred solution of 2-bromo-4H,5H,7H-pyrazolo[1,5-a]pyrazin-6-one (320 mg, 1.5 mmol) in DMF (10 mL) were added Cs2CO3 (965 mg, 3.0 mmol) and MeI (210 mg, 1.5 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. After completed, poured the resulting mixture into water and extracted with EA (3 x 8 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford (200 mg, crude) of 2-bromo-5-methyl-4H,7H-pyrazolo[1,5-a]pyrazin-6-one as a white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 230.11 [M+H]+. Step 6: Preparation of 5-methyl-2-(tributylstannyl)-4H,7H-pyrazolo[1,5-a]pyrazin- 6-one. Compound 338.6
To a stirred solution of 2-bromo-5-methyl-4H,7H-pyrazolo[1,5-a]pyrazin-6-one (150 mg, 0.7 mmol) in dioxane (10 mL) were added 1,1,1,2,2,2-hexabutyldistannane (567 mg, 1.0 mmol) and Pd(PPh3)4 (75 mg, 0.1 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 100 °C and stirred at 100 °C for 48 hours. After completed, poured the resulting mixture into water and extracted with EA (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography(eluted: with 45% EtOAc in petroleum ether) to afford (80 mg, 27% yield) of 5-methyl-2-(tributylstannyl)-4H,7H-pyrazolo[1,5-a]pyrazin-6-one as a colorless oil. LCMS observed m/z = 442.11 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-{5-methyl-6-oxo-4H,7H-pyrazolo[1,5-a]pyrazin-2-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 338
To a stirred solution of 5-methyl-2-(tributylstannyl)-4H,7H-pyrazolo[1,5-a]pyrazin- 6-one (35 mg, 0.1 mmol) and 2-[6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (45 mg, 0.1 mmol) in DMF (3 mL) was added Pd(PPh3)4 (9 mg, 0.1 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 120 °C and stirred at 120 °C for 2 hours. After completed, the residue was purified by reversed-phase flash chromatography (Column: Xselect CSH C185 m, 19 mm X 250 mm; mobile phase: 60-72% MeOH in Water (0.1% FA)) to afford (7.2 mg, 14% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-{5-methyl-6-oxo- 4H,7H-pyrazolo[1,5-a]pyrazin-2-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, Chloroform-d) δ 9.08 (s, 1H), 8.54 – 8.44 (m, 1H), 7.70 – 7.63 (m, 1H), 7.60 – 7.52 (m, 1H), 7.05 (s, 1H), 6.44 (s, 1H), 5.22 (s, 2H), 4.91 (s, 2H), 4.69 (s, 2H), 4.49 – 4.34 (m, 2H), 4.02 – 3.90 (m, 2H), 3.21 (s, 3H), 3.05 – 2.94 (m, 2H), 2.77 (s, 2H), 1.43 – 1.22 (m, 3H).LCMS observed m/z = 631.30 [M+H]+. EXAMPLE 339 Synthesis of N-(1-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)- 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin- 6-yl)pyrrolidin-3-yl)-5-hydroxy-N,6-dimethylpyrimidine-4-carboxamide Compound 339
Step 1: Preparation of methyl 2-(3-((tert-butoxycarbonyl)(methyl)amino)pyrrolidin- 1-yl)-3-oxopentanoate. Compound 339.1
To a solution of tert-butyl methyl(pyrrolidin-3-yl)carbamate (1.0 g, 5 mmol) and methyl 2-chloro-3-oxopentanoate (1.23 g, 7.5 mmol) in ACN (20 mL) was added TEA (1.01 g, 10 mmol) in one portion at 25°C. The reaction mixture was heated to 60 ℃ and stirred at 60 ℃ for 2 hours. When the start material was consumed up, the resulting mixture was quenched with water (10 mL), and extracted with EA (2 x 10 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with mobile phase, EA/PE (10/1) to afford methyl 2-(3-((tert- butoxycarbonyl)(methyl)amino)pyrrolidin-1-yl)-3-oxopentanoate (740 mg, 38%) as a yellow oil. LCMS observed m/z = 329.15 [M+H]+. Step 2: Preparation of tert-butyl (1-(3-amino-6-ethyl-2-imino-4-oxo-1,2,3,4- tetrahydropyrimidin-5-yl)pyrrolidin-3-yl)(methyl)carbamate. Compound 339.2
To a solution of 1-aminoguanidine hydrochloride (430 mg, 3.9 mmol) in EtOH (10 mL) was added tetrabutylammonium hydroxide (40% in water, 3.1 g, 4.9 mmol) at 25°C. The reaction mixture was heated to 55 ℃ and stirred at 55 ℃ for 30 minutes. Methyl 2-(3- ((tert-butoxycarbonyl)(methyl)amino)pyrrolidin-1-yl)-3-oxopentanoate (640 mg, 2.0 mmol) was added and the resulting mixture was heated to 90 ℃ and stirred at 90 ℃ for 16 hours. The EtOH was removed under vacuum. The residue was dissolved in water, extracted with DCM (2 x 10 mL) and the combined organic extracts were dried over Na2SO4, and concentrated. The residue was purified by silica gel column chromatography, eluted with mobile phase, DCM/MeOH (5/1) to afford tert-butyl (1-(3-amino-6-ethyl-2-imino-4-oxo- 1,2,3,4-tetrahydropyrimidin-5-yl)pyrrolidin-3-yl)(methyl)carbamate (395 mg, 49% yield) as a yellow oil. LCMS observed m/z = 353.10 [M+H]+. Step 3: Preparation of tert-butyl (1-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)(methyl)carbamate. Compound 339.3
To a solution of tert-butyl (1-(3-amino-6-ethyl-2-imino-4-oxo-1,2,3,4- tetrahydropyrimidin-5-yl)pyrrolidin-3-yl)(methyl)carbamate (395 mg, 1.1 mmol) and 3,6- dihydro-2H-pyran-4-carbaldehyde (150 mg, 1.3 mmol) in NMP (7 mL) was added anhydrous FeCl3 (363 mg, 2.2 mmol) in one portion at 25°C. The reaction mixture was heated to 55 ℃ and stirred at 55 ℃ for 16 hours. The reaction mixture was diluted with DCM and water, filtered through celite, the filtrate was extracted with DCM (2 x 10mL) and the combined organic extracts were dried over Na2SO4. This resulted in tert-butyl (1- (2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)pyrrolidin-3-yl)(methyl)carbamate(440mg, 53% yield)as a brown oil. LCMS observed m/z = 445.25 [M+H]+.
Step 4: Preparation of tert-butyl (1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)(methyl)carbamate. Compound 339.4
To a solution of tert-butyl (1-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)(methyl)carbamate (435 mg, 1.0 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (355 mg, 1.0 mmol, Intermediate) in DMF (7 mL) was added DIPEA (164 mg, 1.3 mmol) in one portion at 25°C. The resulting mixture was stirred at 25°C for 3 hours. The resulting mixture was quenched with water (10 mL), and extracted with EA (2 x 10 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with mobile phase, EA / PE (1 / 4). to afford tert-butyl (1- (4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3- yl)(methyl)carbamate (269 mg, 32%yield) as a yellow oil. LCMS observed m/z = 680.40 [M+H]+. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(3-(methylamino)pyrrolidin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 339.5
tert-butyl tert-butyl (1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)(methyl)carbamate (164 mg, 0.24 mmol) in TFA (1 mL) at 25 ℃ was heated to 45 ℃ and stirred at 45 ℃ for 1 hours. The reaction mixture was poured into sat. NaHCO3 (5 mL) and extracted with DCM (2 x 5 mL). The combined the organic layers were dried over anhydrous Na2SO4. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (10 / 1) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-6-(3-(methylamino)pyrrolidin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (55 mg, 35% yield) as a white solid. LCMS observed m/z = 580.15 [M+H]+. Step 6: Preparation of 5-(benzyloxy)-N-(1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)-N,6- dimethylpyrimidine-4-carboxamide. Compound 339.6
To a solution of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (25 mg, 0.1 mmol, Intermediate) in ACN (2 mL) were added NMI (27 mg, 0.3 mmol), TCFH (26 mg,
0.1 mmol) and N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-6-(3-(methylamino)pyrrolidin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (55 mg, 1.0 mmol) at 25 °C. The resulting mixture was stirred at 25 °C for 3 hours. The resulting mixture was quenched with water (5 mL), extracted with EtOAc (2 x 5 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The cruder resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 806.40 [M+H]+. Step 7: Preparation of N-(1-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)pyrrolidin-3-yl)-5-hydroxy-N,6-dimethylpyrimidine-4-carboxamide. Compound 339
A mixture of 5-(benzyloxy)-N-(1-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)-N,6- dimethylpyrimidine-4-carboxamide (54 mg, 0.07 mmol) in TFA (1.5 mL) at 25 ℃ was heated to 80 ℃ and stirred at 80 ℃ for 1 hours. The resulting mixture was quenched by addition of a solution of sat. NaHCO3 (aq, 5 mL), extracted with DCM (2 x 5 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 19 mm * 250 mm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 57% B to 70% B in 10min; Wave Length: 254 nm/220 nm; RT1(min): 5.88) to afford N-(1-(4-(2- ((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pyrrolidin-3-yl)-5- hydroxy-N,6-dimethylpyrimidine-4-carboxamide (12.6 mg, 26% yield) as a light yellow
solid.1H NMR (400 MHz, Acetonitrile-d3) δ 11.44 – 10.79 (m, 1H), 8.82 (s, 1H), 8.62 (s, 1H), 8.34 (d, J = 8.7 Hz, 1H), 7.84 (d, J = 2.1 Hz, 1H), 7.63 (dd, J = 8.7, 2.1 Hz, 1H), 6.92 (td, J = 2.9, 1.4 Hz, 1H), 5.82 – 5.63 (m, 1H), 5.22 (d, J = 2.2 Hz, 2H), 4.31 (t, J = 2.8 Hz, 2H), 3.88 (t, J = 5.5 Hz, 2H), 3.71 – 3.57 (m, 1H), 3.57 – 3.48 (m, 1H), 3.47 – 3.34 (m, 1H), 3.28 – 3.16 (m, 3H), 3.11 – 2.91 (m, 2H), 2.66 – 2.57 (m, 2H), 2.50 (s, 3H), 1.36 – 1.24 (m, 5H). LCMS observed m/z = 716.30 [M+H]+. Note: One exchangeable proton was not observed. EXAMPLE 340 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-1,4-diazepan-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Step 1: Preparation of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-1,4-diazepane-1-carboxylate.
To a solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-iodo-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (100 mg, 0.2 mmol) and tert-butyl 1,4-diazepane-1-carboxylate (330 mg, 1.7 mmol) in DMSO (10 mL) was added AgBF4 (64 mg, 0.3 mmol) in portions at room temperature. The reaction mixture was irradiated with microwave radiation for 5 h at 120 °C. After completion of reaction, the reaction mixture was cooled to room temperature and quenched by addition of water (10 mL). The aqueous layer was extracted with ethyl acetate (3 x 10 mL). The combined organic phases were washed with brine (3 x 4 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified by reversed-phase flash chromatography (Column, C18 silica gel; mobile phase: 10-100% MeCN in Water (0.1% FA)) to afford (24 mg, 21.4% yield) of tert-butyl 4-[4-({[2- chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-1,4-diazepane-1-carboxylate as a yellow solid. LCMS observed m/z = 680.25 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[6-(1,4-diazepan- 1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide. Compound 340.2
To a stirred solution of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-1,4-diazepane-1-carboxylate (24 mg, 0.03 mmol) in DCM (3 mL) was added a solution of HCl (gas) in 1,4-dioxane (1.0 mL) dropwise at room temperature. The resulting mixture was stirred for 2 h at room temperature. After completion of reaction, the reaction mixture was concentrated under reduced pressure to afford (20 mg, 97.7% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[6-(1,4-diazepan-
1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide as a yellow solid. LCMS observed m/z = 580.20. [M+H]+. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-1,4- diazepan-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 340.3
To a mixture of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (9 mg, 0.04 mmol), NMI (8.5 mg, 0.1 mmol) and TCFH (19 mg, 0.07 mmol) in ACN (2 mL) was added N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[6-(1,4-diazepan-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (20 mg, 0.03 mmol) in portions at room temperature. The resulting mixture was stirred for additional 3 h at room temperature. After completion of reaction, the reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluted: with 50% EA in PE) to afford (25 mg, 89.9% yield) of 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]-1,4-diazepan-1-yl}-2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 806.27. [M+H]+ Step 4: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-1,4-diazepan-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 340
Into a 25 mL round-bottom flask were added 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]-1,4-diazepan-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (23 mg, 0.03 mmol) and TFA (2 mL) at room temperature under nitrogen atmosphere. The mixture was heated to 80 °C and stirred for 1 h at 80 °C. After completion of reaction, the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep-HPLC (Column: Xbridge BEH Phenyl 5 m, 19 mm * 250 mm; mobile phase: 50-70% ACN in Water (0.1% FA)) to afford (3.7 mg, 17.5% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl]acetamide. 1H NMR (400 MHz, Chloroform-d) δ 8.95 (s, 1H), 8.78 – 8.59 (m, 1H), 8.49 (d, J = 8.7 Hz, 1H), 7.66 (d, J = 2.0 Hz, 1H), 7.56 (d, J = 7.7 Hz, 1H), 6.99 (s, 1H), 5.08 (s, 2H), 4.57 (s, 1H), 4.40 (d, J = 3.2 Hz, 2H), 4.32 – 4.13 (m, 1H), 3.95 (t, J = 4.6 Hz, 2H), 3.83 – 3.68 (m, 3H), 3.66 – 3.57 (m, 1H), 3.21 – 3.15 (m, 1H), 3.12 – 2.99 (m, 3H), 2.74 (s, 2H), 2.65 (d, J = 5.1 Hz, 3H), 2.07 (m, 2H), 1.22 (t, J = 8.5 Hz, 3H). LCMS observed m/z = 716.25 [M+H] +. Note: One exchangeable proton was not observed in NMR spectra. EXAMPLE 341 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-[(5R)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5-methyl-1,4- diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 341
Step 1: Preparation of tert-butyl (7R)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7- methyl-1,4-diazepane-1-carboxylate. Compound 341.1
A solution of tert-butyl (7R)-7-methyl-1,4-diazepane-1-carboxylate (500 mg, 2.3 mmol) and methyl 2-chloro-3-oxopentanoate (422 mg, 2.6 mmol) in ACN (5 mL) was treated with K2CO3 (967 mg, 7.0 mmol) in one portion at 25 ℃. The reaction mixture was stirred at 25 ℃ for 16 hours under nitrogen atmosphere. The resulting mixture was diluted with H2O (30 mL) and extracted with EA (3 x 30 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 7% EtOAc in petroleum ether) to afford (620 mg, 78% yield) of tert-butyl (7R)-4-(1-methoxy- 1,3-dioxopentan-2-yl)-7-methyl-1,4-diazepane-1-carboxylate as a light-yellow oil. LCMS observed m/z = 343.20 [M+H]+. Step 2: Preparation of tert-butyl (7R)-4-{2-bromo-5-ethyl-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl}-7-methyl-1,4-diazepane-1-carboxylate. Compound 341.2
A solution of tert-butyl (7R)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7-methyl-1,4- diazepane-1-carboxylate (600 mg, 1.8 mmol) and 5-bromo-2H-1,2,4-triazol-3-amine, Intermediate B (286 mg, 1.8 mmol) in EtOH (4 mL) was treated with H3PO4 (172 mg, 1.8 mmol) in one portion at 25 ℃. The reaction mixture was heated to 80 ℃ and stirred at 80 ℃ for 16 hours under N2 atmosphere. The mixture was allowed to cool down to room temperature. Then the mixture was acidified to pH 8~9 with TEA and added di-tert-butyl dicarbonate (184 mg, 0.8 mmol) in one portion at 25 ℃. The reaction mixture was stirred at 25 ℃ for 16 hours. The mixture was purified by reversed phase C18 silica gel column chromatography to afford (160 mg, 20% yield) of tert-butyl (7R)-4-{2-bromo-5-ethyl-7- oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}-7-methyl-1,4-diazepane-1-carboxylate as a brown yellow solid. LCMS observed m/z =453.25 [M+H]+. Step 3: Preparation of tert-butyl (7R)-4-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate. Compound 341.3
A solution of tert-butyl (7R)-4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl}-7-methyl-1,4-diazepane-1-carboxylate (160 mg, 0.4 mmol) and 2-(3,6- dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (111 mg, 0.5 mmol) in dioxane (3 mL) and H2O (1 mL) was treated with Pd(dppf)Cl2CH2Cl2 (28 mg, 0.04 mmol) and Na2CO3 (112 mg, 1.1 mmol) in one portion at 25 ℃. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 16 hours under N2 atmosphere. The mixture was allowed to cool down to room temperature. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: with 8% MeOH in dichloromethane) to afford (135 mg, 84% yield) of tert-butyl (7R)-4-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-
1,4-diazepane-1-carboxylate as a brown yellow solid. LCMS observed m/z = 459.30 [M+H]+. Step 4: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[(5R)-5-methyl-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 341.4
of tert-butyl (7R)-4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate (90 mg, 0.1 mmol) in DCM (4 mL) was treated with TFA (1 mL) in one portion at 25 ℃. The reaction mixture was stirred at 25 ℃ for 30 min. The mixture was concentrated in vacuo to afford (100 mg, crude) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-[(5R)-5-methyl-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide as a brown yellow solid. The crude product was used directly in the next step without further purification. LCMS observed m/z = 594.30 [M+H]+. Step 5: Preparation of 2-{6-[(5R)-4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-5-methyl-1,4-diazepan-1-yl]-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 341.5
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-[(5R)-5-methyl-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl]acetamide (90 mg, 0.2 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (41 mg, 0.2 mmol) in ACN (3 mL) was treated with TCFH (51 mg, 0.2 mmol) and NMI (62 mg, 0.8 mmol) in one portion at 25 ℃. The reaction mixture was stirred at 25 ℃ for 1 hour. The resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography (eluent: with 5% MeOH in dichloromethane) to afford (95 mg, 76% yield) of 2-{6-[(5R)-4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-5-methyl-1,4-diazepan-1-yl]-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a brown yellow oil. LCMS observed m/z = 820.35 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[(5R)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5-methyl- 1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide.
A solution of 2-{6-[(5R)-4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-5- methyl-1,4-diazepan-1-yl]-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (90 mg, 0.1 mmol) in TFA (3 mL) at 25 ℃ for 16 hours. The resulting mixture was concentrated under reduced pressure and dissolved in DMF (2 mL). The mixture was purified by preparatory HPLC (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 30-48% MeCN in H2O) to afford (21.4 mg, 26% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[(5R)- 4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5-methyl-1,4-diazepan-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.32 (s, 2H), 8.59 (s, 1H), 8.11 – 8.02 (m, 1H), 7.97 (t, J = 3.0 Hz, 1H), 7.78 – 7.69 (m, 1H), 6.83 (s, 1H), 5.37 – 5.23 (m, 2H), 4.31 – 4.24 (m, 2H), 3.81 (t, J = 5.4 Hz, 2H), 3.55 (m, 3H), 3.25 – 3.06 (m, 2H), 3.01 – 2.88 (m, 2H), 2.82 – 2.70 (m, 2H), 2.68 (s, 1H), 2.45 (m, 4H), 2.17 – 2.08 (m, 1H), 1.76 (m, 1H), 1.32 – 1.03 (m, 6H). LCMS observed m/z = 730.55 [M+H]+. EXAMPLE 342 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-[(5S)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5-methyl-1,4- diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Step 1: Preparation of tert-butyl (7S)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7- methyl-1,4-diazepane-1-carboxylate. Compound 342.1
To a stirred mixture of tert-butyl (7S)-7-methyl-1,4-diazepane-1-carboxylate (800 mg, 3.7 mmol) and methyl 2-chloro-3-oxopentanoate (922 mg, 5.6 mmol) in ACN (10 mL) was added TEA (755 mg, 7.5 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 60 °C and stirred at 60 °C for 2 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3/1) to afford tert-butyl (7S)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7-methyl-1,4- diazepane-1-carboxylate (1.0 g, 78% yield) as a yellow oil. LCMS observed m/z = 343.22 [M+H] +. Step 2: Preparation of 2-bromo-5-ethyl-6-[(5S)-5-methyl-1,4-diazepan-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyri midin-7-one. Compound 342.2
To a stirred mixture of tert-butyl (7S)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7- methyl-1,4-diazepane-1-carboxylate (800 mg, 2.3 mmol) and 5-bromo-2H-1,2,4-triazol-3- amine, Intermediate B (381 mg, 2.3 mmol) in EtOH (5 mL) was added H3PO4 (229 mg, 2.3 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C overnight under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure to give 2-bromo- 5-ethyl-6-[(5S)-5-methyl-1,4-diazepan-1-yl]-4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one (800 mg, 96% yield) as a brown oil. LCMS observed m/z = 355.08 [M+H]+. Step 3: Preparation of tert-butyl (7S)-4-{2-bromo-5-ethyl-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl}-7-methyl-1,4-diazepane-1-carboxylate. Compound 342.3
To a stirred mixture of 2-bromo-5-ethyl-6-[(5S)-5-methyl-1,4-diazepan-1-yl]-4H- [1,2,4]triazolo[1,5-a]pyrimidin-7-one (800 mg, 2.3 mmol) and TEA (1.14 g, 11.3 mmol) in MeOH (10 mL) was added Boc2O (983 mg, 4.5 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford tert-butyl (7S)- 4-{2-bromo-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}-7-methyl-1,4- diazepane-1-carboxylate (300 mg, 29% yield) as a light yellow solid. LCMS observed m/z = 455.13 [M+H]+. Step 4: Preparation of tert-butyl (7S)-4-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4H-[1,2,4] triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate. Compound 342.4
To a stirred mixture of tert-butyl (7S)-4-{2-bromo-5-ethyl-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl}-7-methyl-1,4-diazepane-1-carboxylate (250 mg, 0.5 mmol) and 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (173 mg, 0.8 mmol) in dioxane (5 mL) and H2O (1 mL) were added Pd(dppf)Cl2.CH2Cl2 (45 mg, 0.05 mmol) and Na2CO3 (175 mg, 1.6 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C overnight under nitrogen atmosphere. After the reaction was completed, the resulting mixture was diluted with water (20 mL). The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/4) to afford tert-
butyl (7S)-4-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate (120 mg, 48% yield) as a white solid. LCMS observed m/z = 459.26 [M+H] +. Step 5: Preparation of tert-butyl (7S)-4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate. Compound 342.5
To a stirred mixture of tert-butyl (7S)-4-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4H-[1,2,4] triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate (120 mg, 0.3 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (95 mg, 0.3 mmol) in DMF (2 mL) was added DIEA (101 mg, 0.8 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 3 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was diluted with water (20 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/4) to afford tert-butyl (7S)-4-[4-({[2-chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane- 1-carboxylate (90 mg, 50% yield) as a light yellow solid. LCMS observed m/z = 694.27 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[(5S)-5-methyl-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 342.6
To a stirred solution of tert-butyl (7S)-4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-7-methyl-1,4-diazepane-1-carboxylate (80 mg, 0.1 mmol) in DCM (1 mL) was added TFA (1 mL) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure to give N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[(5S)-5-methyl-1,4-diazepan-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (80 mg, crude) as a green oil. LCMS observed m/z = 594.21 [M+H] +. Step 7: Preparation of 2-{6-[(5S)-4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-5-methyl-1,4-diazepan-1-yl]-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide.
To a stirred mixture of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[(5S)-5-methyl-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (90 mg, 0.2 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-
carboxylic acid, Intermediate A (44 mg, 0.2 mmol) in ACN (2 mL) were added TCFH (64 mg, 0.2 mmol) and NMI (62 mg, 0.8 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was diluted with water (10 mL). The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2 / MeOH (10/1) to afford 2-{6-[(5S)-4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]-5-methyl-1,4-diazepan-1-yl]-2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (80 mg, 64% yield) as a brown oil. LCMS observed m/z = 820.29 [M+H] +. Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[(5S)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5-methyl- 1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide.
A solution of 2-{6-[(5S)-4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-5- methyl-1,4-diazepan-1-yl]-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (75 mg, 0.09 mmol) in TFA (3 mL) was stirred at room temperature overnight under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product (80 mg) was purified by Prep- HPLC with the following conditions (Column: X-Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B:
ACN; Flow rate: 60 mL/min; Gradient: 33% B to 56% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 5.27) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-[(5S)-4-(5-hydroxy-6-methylpyrimi dine-4-carbonyl)-5- methyl-1,4-diazepan-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide(34.3 mg, 51% yield) as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.58 (d, J = 26.4 Hz, 1H), 8.22 – 8.13 (m, 1H), 7.86 – 7.80 (m, 1H), 7.68 – 7.60 (m, 1H), 6.99 – 6.93 (m, 1H), 5.47 – 5.30 (m, 2H), 4.44 – 4.06 (m, 4H), 3.97 – 3.87 (m, 2H), 3.87 – 3.48 (m, 2H), 3.36 (s, 1H), 3.27 – 3.13 (m, 1H), 3.13 – 2.72 (m, 3H), 2.65 (s, 2H), 2.58 – 2.49 (m, 3H), 2.37 – 2.17 (m, 1H), 2.15 – 1.87 (m, 1H), 1.38 – 1.17 (m, 6H). LCMS observed m/z = 730.40 [M+H] +. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 343 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(3-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,7- diazabicyclo[4.2.0]octan-7-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 343
Step 1: Preparation of 3,6-dihydro-2H-pyran-4-carbaldehyde. Compound 343.1
In a 50-mL round bottom flask, to a solution of 3,6-dihydro-2H-pyran-4-carbonitrile (2.2 g, 20.1 mmol) in toluene (25 mL) was added dropwise DIBAL-H (1.0 M solution in hexanes, 21 mL, 21.2 mmol) at -10 °C under N2 atmosphere. The reaction mixture was stirred at -10 °C for another 1 hour. The mixture was acidified to pH 5 with HCl (aq.). The two layers were separated, and the aqueous layer was extracted twice with DCM. The combined organic layers were washed with brine and dried over Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 3,6-dihydro-2H-pyran-
4-carbaldehyde (800 mg, 35%yield) as a colorless liquid.1H NMR (400 MHz, Chloroform- d) δ 9.44 (s, 1H), 6.78 (tt, J = 2.9, 1.5 Hz, 1H), 4.37 (q, J = 2.7 Hz, 2H), 3.77 (t, J = 5.5 Hz, 2H), 2.35 – 2.24 (m, 2H). Step 2: Preparation of tert-butyl 7-(1-methoxy-1,3-dioxopentan-2-yl)-3,7- diazabicyclo[4.2.0]octane-3-carboxylate. Compound 343.2
To a solution of tert-butyl 3,7-diazabicyclo[4.2.0]octane-3-carboxylate (450 mg, 2.1 mmol) and methyl 2-chloro-3-oxopentanoate (523 mg, 3.2 mmol, Intermediate) in ACN (10 mL) was added TEA (429 mg, 4.2 mmol) in one portion at 25°C. The resulting mixture was heated to 60 ℃ and stirred at 60 ℃ for 2 hours. The resulting mixture was quenched with water (5 mL), and extracted with EA (2 x 10 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with mobile phase, EA / PE (5 / 1). This resulted in tert-butyl 7-(1-methoxy-1,3-dioxopentan-2-yl)-3,7- diazabicyclo[4.2.0]octane-3-carboxylate (407 mg, 40%yield) as a yellow solid. LCMS observed m/z = 341.25 [M+H]+. Step 3: Preparation of tert-butyl 7-(3-amino-6-ethyl-2-imino-4-oxo-1,2,3,4- tetrahydropyrimidin-5-yl)-3,7-diazabicyclo[4.2.0]octane-3-carboxylate. Compound 343.3
To a solution of 1-aminoguanidine hydrochloride (260 mg, 2.4 mmol) in EtOH (4 mL) was added tetrabutylammonium hydroxide (40% in water, 1.5 g, 2.4 mmol) at 25℃. The reaction mixture was heated to 55 ℃ and stirred at 55 ℃ for 30 minutes. Tert-butyl 7- (1-methoxy-1,3-dioxopentan-2-yl)-3,7-diazabicyclo[4.2.0]octane-3-carboxylate (400 mg, 1.2 mmol) was added and the resulting mixture was heated to 90 ℃ and stirred at 90 ℃ for 16 hours. The EtOH was removed under vacuum. The residue was dissolved in water, extracted with DCM (2 x 10 mL) and the combined organic extracts were dried over Na2SO4, and concentrated. The residue was purified by silica gel column chromatography,
eluted with mobile phase, DCM / MeOH (5 / 1) to afford tert-butyl 7-(3-amino-6-ethyl-2- imino-4-oxo-1,2,3,4-tetrahydropyrimidin-5-yl)-3,7-diazabicyclo[4.2.0]octane-3- carboxylate (121 mg, 20% yield) as a yellow oil. LCMS observed m/z = 365.20 [M+H]+. Step 4: Preparation of tert-butyl 7-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-3,7-diazabicyclo[4.2.0]octane-3- carboxylate. Compound 343.4
To a solution of tert-butyl 7-(3-amino-6-ethyl-2-imino-4-oxo-1,2,3,4- tetrahydropyrimidin-5-yl)-3,7-diazabicyclo[4.2.0]octane-3-carboxylate (90 mg, 0.25 mmol) and 3,6-dihydro-2H-pyran-4-carbaldehyde (83 mg, 0.5 mmol, Intermediate) in EtOH/AcOH (1 mL/2 mL) was added Cu(AcO)2 (22 mg, 0.12 mmol) in one portion at 25℃. The reaction mixture was heated to 70 ℃ and stirred at 70 ℃ for 16 hours. The resulting mixture was quenched with water (5 mL), extracted with EtOAc (2 x 3 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (10/1) to afford tert-butyl 7-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-3,7- diazabicyclo[4.2.0]octane-3-carboxylate (80 mg, 21%yield) as a yellow oil. LCMS observed m/z = 457.20 [M+H]+. Step 5: Preparation of tert-butyl 7-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-3,7-diazabicyclo[4.2.0]octane-3- carboxylate. Compound 343.5
To a solution of tert-butyl 7-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-3,7-diazabicyclo[4.2.0]octane-3-carboxylate (35 mg, 0.08 mmol) and N-(2-chloro-4-(trifluoromethyl)phenyl)-2-iodoacetamide (33.44 mg, 0.1 mmol) in DMF (2 mL) was added DIPEA (20 mg, 0.15 mmol) in one portion at 25°C. The resulting mixture was stirred at 25°C for 3 hours. The resulting mixture was quenched with water (2 mL), and extracted with EA (2 x 3 mL). The combined organic layers were washed with brine (2 x 3 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 692.35 [M+H]+. Step 6: Preparation of 2-(6-(3,7-diazabicyclo[4.2.0]octan-7-yl)-2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide.
A mixture of tert-butyl 7-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)-3,7-diazabicyclo[4.2.0]octane-3-carboxylate (29 mg, 0.042 mmol) in TFA (1 mL) at 25 ℃ was stirred at 25 ℃ for 1 hours. The reaction mixture was poured into sat. NaHCO3 (aq, 2 mL) and extracted with DCM (2 x 3 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated
under reduced pressure. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 592.30 [M+H]+. Step 7: Preparation of 2-(6-(3-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)-3,7- diazabicyclo[4.2.0]octan-7-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 343.7
To a solution of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (11 mg, 0.04 mmol, Intermediate) in ACN (1 mL) were added NMI (11 mg, 0.13 mmol), TCFH (12 mg, 0.04 mmol) and 2-(6-(3,7-diazabicyclo[4.2.0]octan-7-yl)-2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (25 mg, 0.042 mmol) at 25 °C. The resulting mixture was stirred at 25 °C for 3 hours. The resulting mixture was quenched with water (2 mL), extracted with EtOAc (2 x 2 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The cruder resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 818.40 [M+H]+. Step 8: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(3-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,7- diazabicyclo[4.2.0]octan-7-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 343
A mixture of 2-(6-{3-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-3,7- diazabicyclo[4.2.0]octan-7-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (24 mg, 0.03 mmol) in TFA (1 mL) at 25 ℃ was stirred at 25 ℃ for 8 hours. The resulting mixture was quenched by addition of a solution of sat. NaHCO3 (aq, 2 mL), extracted with DCM (2 x 3 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 19 mm * 250 mm; Mobile Phase A: water(0.1% FA), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 57% B to 70% B in 10min; Wave Length: 254 nm/220 nm; RT1(min): 5.88) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-6-(3-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,7-diazabicyclo[4.2.0]octan-7- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (1.2 mg, 6% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.95 (s, 1H), 8.97 (d, J = 10.0 Hz, 1H), 8.75 (d, J = 12.9 Hz, 1H), 8.50 (d, J = 8.7 Hz, 1H), 7.66 (s, 1H), 7.56 (d, J = 9.5 Hz, 1H), 7.02 (s, 1H), 5.41 – 5.34 (m, 1H), 5.20 – 5.05 (m, 3H), 4.76 – 4.69 (m, 1H), 4.63 – 4.56 (m, 1H), 4.45 – 4.35 (m, 3H), 4.33 – 4.25 (m, 1H), 4.07 – 3.99 (m, 2H), 3.99 – 3.91 (m, 2H), 3.48 – 3.44 (m, 1H), 3.21 – 3.10 (m, 3H), 2.85 – 2.80 (m, 2H), 2.79 – 2.74 (m, 2H), 2.27 (d, J = 7.5 Hz, 1H), 1.30 – 1.28 (m, 3H). LCMS observed m/z = 728.30 [M+H]+. EXAMPLE 344 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl](5-{4-[(E)-4-(p- methoxyphenyl)-4-oxo-2-butenoyl]-1-piperazinyl}-2-(3,6-dihydro-2H-pyran-4-yl)-6- ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide Compound 344
The title compound was prepared using similar procedure as compound 32 to afford N-[2-chloro-4-(trifluoromethyl) phenyl](5-{4-[(E)-4-(p-methoxyphenyl)-4-oxo-2- butenoyl]-1-piperazinyl}-2-(3,6-dihydro-2H-pyran-4-yl)-6-ethyl-4-oxo-1,3,3a,7-tetraaza- 7-indenyl)acetamide as a faint yellow solid.1H NMR (400 MHz, DMSO) δ 10.33 (s, 1H), 8.00 (d, J = 9.1 Hz, 3H), 7.90 (s, 1H), 7.77 (d, J = 15.1 Hz, 1H), 7.65 (d, J = 8.8 Hz, 1H), 7.42 (d, J = 15.1 Hz, 1H), 7.04 (d, J = 8.6 Hz, 2H), 6.77 (s, 1H), 5.26 (s, 2H), 4.45 (d, J = 12.5 Hz, 1H), 4.22 – 4.15 (m, 2H), 4.03 (d, J = 12.5 Hz, 1H), 3.81 (s, 3H), 3.73 (t, J = 5.4 Hz, 2H), 3.47 – 3.31 (m, 3H), 3.26 – 3.13 (m, 2H), 3.00 – 2.90 (m, 2H), 2.88 – 2.79 (m, 1H), 2.70 (t, J = 12.0 Hz, 2H), 1.13 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 754.3 [M+H]+. EXAMPLE 345 Synthesis of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(1-(5-hydroxy-6-methylpyrimidine-4-carbonyl)octahydro-4H-pyrrolo[3,2- b]pyridin-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 345
Step 1: Preparation of tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)octahydro-1H- pyrrolo[3,2-b]pyridine-1-carboxylate. Compound 345.1
A solution of tert-butyl octahydro-1H-pyrrolo[3,2-b]pyridine-1-carboxylate (300 mg, 1.3 mmol), TEA (268 mg, 2.7 mmol) and methyl 2-chloro-3-oxopentanoate (327 mg, 2.0 mmol, Intermediate) in ACN (5 mL) was heated to 60 ℃ and stirred at 60 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (9/1) to afford tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)octahydro-1H-pyrrolo[3,2-b]pyridine- 1-carboxylate (200 mg, 42% yield) as a yellow oil. LCMS observed m/z = 355.30 [M+H]+. Step 2: Preparation of 2-bromo-5-ethyl-6-(octahydro-4H-pyrrolo[3,2-b]pyridin-4- yl)-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one. Compound 345.2
A solution of tert-butyl 4-(1-methoxy-1,3-dioxopentan-2-yl)octahydro-1H- pyrrolo[3,2-b]pyridine-1-carboxylate (200 mg, 0.6 mmol) and 3-bromo-1H-1,2,4-triazol-5- amine, Intermediate A (184 mg, 1.1 mmol, Intermediate) in para-toluene sulfonate (97 mg, 0.6 mmol) was heated to 140 ℃ and stirred at 140 ℃ for 1.5 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 367.15 [M+H]+. Step 3: Preparation of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1-carboxylate. Compound 345.3
A solution of 2-bromo-5-ethyl-6-(octahydro-4H-pyrrolo[3,2-b]pyridin-4-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one (200 mg, 0.5 mmol) in MeOH (2 mL) were added Boc2O (131 mg, 0.6 mmol) and TEA (110 mg, 1.1 mmol) dropwise at 25 °C under nitrogen atmosphere. The resulting mixture was stirred for 1 h at 25 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 45% to 50% gradient in 10 min; detector, UV 254 nm to afford tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1-carboxylate (60 mg, 23% yield) as a yellow solid. LCMS observed m/z = 467.15 [M+H]+. Step 4: Preparation of tert-butyl 4-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1- carboxylate. Compound 345.4
A solution of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1-carboxylate (60 mg, 0.1 mmol) in Dioxane (3 mL) and H2O (1 mL) were added 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (40 mg, 0.2 mmol), Na2CO3 (41 mg, 0.4 mmol) and Pd(dppf)Cl2 (10 mg, 0.01 mmol) in one portion at 25 °C. The resulting mixture was heated to 100 ℃ and stirred at 100 ℃ for 1.5 hours under nitrogen atmosphere. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure, collected the residue. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (9/1) to afford tert-butyl 4-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-
oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2- b]pyridine-1-carboxylate (100 mg, crude) as a brown solid. LCMS observed m/z = 471.30 [M+H]+. Step 5: Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2- b]pyridine-1-carboxylate. Compound 345.5
A solution of tert-butyl 4-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7- dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1- carboxylate (100 mg, 0.2 mmol) in DMF (1 mL) were added N-(2-chloro-4- (trifluoromethyl)phenyl)-2-iodoacetamide (93 mg, 0.3 mmol) and DIPEA (55 mg, 0.4 mmol) in portions at 25 °C. The resulting mixture was stirred for 1 h at 25 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (9/1) to afford tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1-carboxylate (50 mg, 33%yield) as a yellow oil. LCMS observed m/z = 706.45 [M+H]+. Step 6: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(octahydro-4H-pyrrolo[3,2-b]pyridin-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 345.6
A solution of tert-butyl 4-(4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)octahydro-1H-pyrrolo[3,2-b]pyridine-1-carboxylate (50 mg, 0.1 mmol) in TFA (2 mL) was stirred for 30 min at 0 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 606.25 [M+H]+. Step 7: Preparation of 2-(6-(1-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)octahydro-4H-pyrrolo[3,2-b]pyridin-4-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 345.7
A solution of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(octahydro-4H-pyrrolo[3,2-b]pyridin-4-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (50 mg, 0.2 mmol) in ACN (2 mL) were added 5- (benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (24 mg, 0.2 mmol), NMI (24 mg, 0.5 mmol) and TCFH (23 mg, 0.2 mmol) dropwise at 25 °C. The reaction mixture was stirred at 25°C for 1 hour. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in water, extracted with EA (3 x 1 mL) and the combined organic extracts were dried over Na2SO4, and concentrated to afford 2-(6-(1-(5-
(benzyloxy)-6-methylpyrimidine-4-carbonyl)octahydro-4H-pyrrolo[3,2-b]pyridin-4-yl)-2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (2-chloro-4-(trifluoromethyl)phenyl)acetamide (10 mg crude) as a white solid. LCMS observed m/z = 832.30 [M+H]+. Step 9: Preparation of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-(1-(5-hydroxy-6-methylpyrimidine-4-carbonyl)octahydro-4H- pyrrolo[3,2-b]pyridin-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 345
A solution of 2-(6-(1-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)octahydro- 4H-pyrrolo[3,2-b]pyridin-4-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (10 mg, 0.01 mmol) in TFA (0.5 mL) was heated to 80 ℃ and stirred at 80 ℃ for 20 minute. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure, collected the residue. The residue was purified by reversed-phase flash chromatography with the following column: X bridge BEH Phenyl5μm, 19*250 mm; Mobile Phase A: Water (0.1%FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 62% B to 75%B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 6.9 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(1-(5-hydroxy-6-methylpyrimidine-4-carbonyl)octahydro-4H- pyrrolo[3,2-b]pyridin-4-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (3.1 mg, 33% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 8.96 (m, 1H), 8.89 – 8.65 (m, 1H), 8.51 (d, J = 8.6 Hz, 1H), 7.72 – 7.65 (m, 1H), 7.56 (d, J = 8.8 Hz, 1H), 7.01 (d, J = 7.7 Hz, 1H), 5.28 – 5.01 (m, 2H), 4.59 – 4.35 (m, 3H), 4.16 (s, 1H), 3.98 – 3.81 (m, 3H), 3.71 – 3.43 (m, 2H), 3.35 – 2.94 (m, 2H), 2.94 – 2.54 (m, 7H), 2.24 (s, 1H), 2.00 – 1.65 (m, 4H), 1.57 – 1.30 (m, 4H).LCMS observed m/z = 742.40 [M+H]+. EXAMPLE 346
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-[3-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,8- diazabicyclo[4.2.0]octan-8-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Step 1: Preparation of tert-butyl 8-(1-methoxy-1,3-dioxopentan-2-yl)-3,8- diazabicyclo[4.2.0]octane-3-carboxylate. Compound 346.1
To a stirred solution of tert-butyl 3,8-diazabicyclo[4.2.0]octane-3-carboxylate (510 mg, 2.4 mmol) and methyl 2-chloro-3-oxopentanoate (435 mg, 2.6 mmol) in ACN (20 mL) was added Et3N (729 mg, 7.2 mmol) in portions at room temperature. The reaction mixture was heated to 60 °C and stirred at 60 °C for 2 hours. After completed, poured the resulting mixture into water (20 mL) and extracted with EA (3 x 15 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 18% EtOAc in petroleum ether) to afford (510 mg, 62% yield) of tert-butyl 8- (1-methoxy-1,3-dioxopentan-2-yl)-3,8-diazabicyclo[4.2.0]octane-3-carboxylate as a light yellow oil. LCMS observed m/z = 341.20 [M+H]+. Step 2: Preparation of tert-butyl 8-(4-ethyl-2-imino-6-oxo-1,3-dihydropyrimidin-5- yl)-3,8-diazabicyclo[4.2.0]octane-3-carboxylate).
Compound 346.2
To a solution of guanidine (149 mg, 2.5 mmol) in EtOH (15 mL) was added tetrabutylazanium oxidanide (819 mg, 3.1 mmol) at room temperature. The reaction was heated to 55 °C and stirred at 55 °C for 0.5 hours. Tert-butyl 8-(1-methoxy-1,3-dioxopentan- 2-yl)-3,8-diazabicyclo[4.2.0]octane-3-carboxylate (430 mg, 1.2 mmol) was added to the mixture. The mixture was heated to 90 °C and stirred at 90 °C for 2 hours. After completed, the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was dissolved in water (10 mL). The aqueous phase was extracted with EA (3 x 15 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography (Column: C18 silica gel; mobile phase: 20%-40% MeCN in H2O) to afford (380 mg, 86% yield) of tert-butyl 8-(4-ethyl-2-imino-6- oxo-1,3-dihydropyrimidin-5-yl)-3,8-diazabicyclo[4.2.0]octane-3-carboxylate as a light yellow solid. LCMS observed m/z = 350.21 [M+H]+. Step 3: Preparation of tert-butyl 8-(1-amino-4-ethyl-2-imino-6-oxo-3H-pyrimidin- 5-yl)-3,8-diazabicyclo[4.2.0]octane-3-carboxylate. Compound 346.3
To a solution of tert-butyl 8-(4-ethyl-2-imino-6-oxo-1,3-dihydropyrimidin-5-yl)- 3,8-diazabicyclo[4.2.0]octane-3-carboxylate (380 mg, 1.1 mmol) in DCM (10 mL) was added amino 2,4,6-trimethylbenzenesulfonate (234 mg, 1.1 mmol) at 0 °C. The resulting mixture was stirred for 2 h at room temperature. After completed, poured the resulting mixture into water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash
chromatography(Column:C18 silica gel; mobile phase: 30%-40% ACN in Water) to afford (105 mg, 26% yield) tert-butyl 8-(1-amino-4-ethyl-2-imino-6-oxo-3H-pyrimidin-5-yl)-3,8- diazabicyclo[4.2.0]octane-3-carboxylate as a light yellow solid. LCMS observed m/z = 365.23 [M+H]+. Step 4: Preparation of tert-butyl 8-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-3,8-diazabicyclo[4.2.0]octane-3-carboxylate. Compound 346.4
To a stirred solution of tert-butyl 8-(1-amino-4-ethyl-2-imino-6-oxo-3H-pyrimidin- 5-yl)-3,8-diazabicyclo[4.2.0]octane-3-carboxylate (120 mg, 0.3 mmol) and 3,6-dihydro- 2H-pyran-4-carbaldehyde (111 mg, 1.0 mmol) in NMP (5 mL) was added FeCl3 (107 mg, 0.7 mmol) in portions at room temperature. The reaction mixture was heated to 55 °C and stirred at 55 °C for 4 hours. After completed, the resulting mixture was filtered, the filter cake was washed with DCM (3 x 3 mL). The filtrate was poured into water (5 mL) and extracted with DCM (3 x 5 mL). The combined organic phase was washed with brine (2 x 5 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford (30 mg, crude) of tert-butyl 8-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-3,8-diazabicyclo[4.2.0]octane-3-carboxylate as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 457.25 [M+H]+. Step 5: Preparation of tert-butyl 8-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-3,8-diazabicyclo[4.2.0]octane-3-carboxylate. Compound 346.5
To a stirred solution of tert-butyl 8-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-3,8-diazabicyclo[4.2.0]octane-3-carboxylate (25 mg, 0.1 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (24 mg, 0.1 mmol) in DMF (1 mL) was added DIEA (21 mg, 0.2 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h. After completed, poured the resulting mixture into water (2 mL) and extracted with EA (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 45% EtOAc in petroleum ether) to afford (25 mg, 65% yield) of tert-butyl 8-[4-({[2-chloro-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-3,8- diazabicyclo[4.2.0]octane-3-carboxylate as a brown oil. LCMS observed m/z = 692.25 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(6-{3,8- diazabicyclo[4.2.0]octan-8-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide. Compound 346.6
To a stirred solution of tert-butyl 8-[4-({[2-chloro-4- (fluoromethyl)phenyl]carbamoyl}methyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]-3,8-diazabicyclo[4.2.0]octane-3-carboxylate (50 mg, 0.1 mmol) in DCM (2 mL) was added TFA (17 mg, 0.2 mmol) dropwise at room temperature. The resulting mixture was stirred for additional 30 min at room temperature. After completed, the resulting mixture was concentrated under vacuum to afford (35 mg, crude) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(6-{3,8-diazabicyclo[4.2.0]octan-8- yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)acetamide as a brown oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 592.25 [M+H]+. Step 7: Preparation of 2-(6-{3-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-3,8- diazabicyclo[4.2.0]octan-8-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 346.7
To a stirred solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(6-{3,8- diazabicyclo[4.2.0]octan-8-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide (35 mg, 0.1 mmol) and 5-(benzyloxy)-6- methylpyrimidine-4-carboxylic acid, Intermediate A (14 mg, 0.1 mmol) in ACN (3 mL) were added NMI (15 mg, 0.2 mmol) and TCFH (17 mg, 0.1 mmol) in portions at room temperature. The resulting mixture was stirred for additional 2 h at room temperature. After completed, poured the resulting mixture into water (3 mL) and extracted with EA (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 45% MeOH in DCM) to afford (20 mg, 41% yield) of 2-(6-{3-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-3,8-diazabicyclo[4.2.0]octan-
8-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)- N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a brown oil. LCMS observed m/z = 818.27 [M+H]+. Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[3-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3,8- diazabicyclo[4.2.0]octan-8-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide.
A solution of 2-(6-{3-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-3,8- diazabicyclo[4.2.0]octan-8-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (10 mg, 0.1 mmol) in TFA (1 mL) was stirred for 3 h at room temperature. After completed, the mixture was basified to pH 8 with Na2CO3 (aq.). The aqueous phase was extracted with EA (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography(Column: XBridge BEH C18 5 μm, 30*150mm; mobile phase 31%-41% ACN in Water) to afford (0.5 mg, 6% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[3-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-3,8-diazabicyclo[4.2.0]octan-8-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 12.01 – 11.72 (m, 1H), 8.98 – 8.76 (m, 1H), 8.56 – 8.39 (m, 1H), 7.73 – 7.62 (m, 1H), 7.61 – 7.48 (m, 1H), 7.05 – 6.94 (m, 1H), 5.27 – 4.88 (m, 3H), 4.64 – 4.15 (m, 4H), 4.07 – 3.73 (m, 3H), 3.49 – 3.19 (m, 2H), 3.14 – 2.92 (m, 2H), 2.75 (s, 2H), 2.67 – 2.33 (m, 3H), 2.22 (s, 1H), 2.10 – 1.95 (m, 1H), 1.72 (s, 1H), 1.29 (s, 1H), 1.26 – 0.99 (m, 3H). LCMS observed m/z = 728.30 [M+H]+. Note: One exchangeable proton was not observed in NMR spectra.
EXAMPLE 347 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro-2H- pyrrolo[3,2-b]pyridin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Step 1: Preparation of tert-butyl 1-(1-methoxy-1,3-dioxopentan-2-yl)-hexahydro- 2H-pyrrolo[3,2-b]pyridine-4-carboxylate. Compound 347.1
A solution of tert-butyl octahydropyrrolo[3,2-b]pyridine-4-carboxylate (2.0 g, 8.837 mmol) in ACN (20 mL) was treated with methyl 2-chloro-3-oxopentanoate (4.4 g, 26.511 mmol) at room temperature under nitrogen atmosphere followed by the addition of K2CO3 (3.7 g, 26.511 mmol) in portions at room temperature. The resulting mixture was stirred for 16 hours at room temperature under nitrogen atmosphere. The solids were removed by filtration and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% EtOAc in petroleum ether) to afford (1.5 g, 48% yield) of tert-butyl 1-(1-methoxy-1,3-dioxopentan-2-yl)-hexahydro-2H- pyrrolo[3,2-b]pyridine-4-carboxylate (1.5 g, 47.89%) as a yellow oil. LCMS observed m/z = 355.22 [M+H]+. Step 2: Preparation of 2-bromo-5-ethyl-6-{octahydropyrrolo[3,2-b]pyridin-1-yl}- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one.
Compound 347.2
A solution of tert-butyl 1-(1-methoxy-1,3-dioxopentan-2-yl)-hexahydro-2H- pyrrolo[3,2-b]pyridine-4-carboxylate (1.5 g, 4.232 mmol) and 5-bromo-2H-1,2,4-triazol-3- amine, Intermediate B (690 mg, 4.232 mmol) in EtOH (7 mL) was treated with H3PO4 (410 mg, 4.232 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 80 °C under nitrogen atmosphere. To the above was added H3PO4 (820 mg, 8.464 mmol) at room temperature again. The resulting mixture was stirred for 24 hours at 80 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The mixture was neutralized to pH 7 with 10% NaOH aqueous solution. The resulting mixture was concentrated under reduced pressure. The residue was purified by reverse phase column (eluent: with 9% MeOH in H2O) to afford (568 mg, 36% yield) of 2-bromo-5-ethyl-6-{octahydropyrrolo[3,2-b]pyridin-1-yl}-4H-[1,2,4]triazolo[1,5- a]pyrimidin-7-one as a yellow oil. LCMS observed m/z = 367.08 [M+H]+. Step 3: Preparation of 6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-4H-[1,2,4]triazolo[1,5- a]pyrimidin-7-one. Compound 347.3
To a stirred solution of 2-bromo-5-ethyl-6-{octahydropyrrolo[3,2-b]pyridin-1-yl}- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one (250 mg, 0.681 mmol) and 5-(benzyloxy)-6- methylpyrimidine-4-carboxylic acid, Intermediate A (250 mg, 1.024 mmol) in DMF (5 mL) were added HATU (388 mg, 1.022 mmol) and DIEA (280 mg, 2.166 mmol) at 25 °C. The resulting mixture was stirred for 3 hours at this temperature. The reaction mixture was purified by reversed-phase flash chromatography (eluent: with 9% MeOH in H2O) to afford (90 mg, 22% yield) of 6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-4H-
[1,2,4]triazolo[1,5-a]pyrimidin-7-one as a light-yellow solid. LCMS observed m/z = 593.15 [M+H]+. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 347.4
To a stirred mixture of 6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-4H-[1,2,4]triazolo[1,5- a]pyrimidin-7-one (86 mg, 0.145 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2- iodoacetamide (63 mg, 0.174 mmol) in DMF (1 mL) was added DIEA (56.19 mg, 0.435 mmol) in portions at 25 °C. Then the resulting mixture was stirred at 45 °C for 4 hours under nitrogen atmosphere. The reaction was quenched with water (5 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (3 x 5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (57 mg, 47% yield) of 2-(6- {4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-hexahydro-2H-pyrrolo[3,2-b]pyridin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 828.16 [M+H]+ Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 347.5
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (15 mg, 0.018 mmol) and 2-(3,6-dihydro-2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (5.7 mg, 0.027 mmol) in dioxane (5 mL) were added a solution of K2CO3 (7.5 mg, 0.054 mmol) in H2O (1 mL) in one portion under N2 atmosphere. To the above mixture was added Pd(dppf)Cl2CH2Cl2 (1.5 mg, 0.002 mmol) under N2 atmosphere. The resulting mixture was stirred for additional 16 hours at 100 °C. The residue was purified by reversed phase C18 silica gel column chromatography to afford (14 mg, 92% yield) of 2-(6-{4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]-hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide as a light-yellow solid. LCMS observed m/z = 832.29 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro-2H- pyrrolo[3,2-b]pyridin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 347
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-hexahydro- 2H-pyrrolo[3,2-b]pyridin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (14 mg, 0.017 mmol) in TFA (5 mL) was stirred at 80 °C under nitrogen atmosphere for 15 min. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparatory HPLC (Column: XBridge BEH C185 μm, 30 * 150mm; mobile phase: 30-42% MeCN in H2O) to afford (2.7 mg, 21% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.35 (s, 1H), 8.48 (m, 1H), 8.11 – 7.91 (m, 2H), 7.72 (d, J = 8.6 Hz, 1H), 7.06 (m, 1H), 6.84 (s, 1H), 5.31 (s, 2H), 4.43 (s, 1H), 4.26 (s, 2H), 3.81 (s, 2H), 3.23 – 2.73 (m, 6H), 2.47 – 1.90 (m, 7H), 1.89 – 1.31 (m, 5H), 1.29 – 1.15 (m, 3H). LCMS observed m/z = 742.35 [M+H]+. EXAMPLE 348 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 348
Step 1: Preparation of 2-(3,4-dihydro-1H-2-benzopyran-5-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane. Compound 348.1
To a stirred solution of 4-bromo-2,3-dihydro-1-benzofuran (50 mg, 0.2 mmol) and bis(pinacolato)diboron (77 mg, 0.3 mmol) in dioxane (10 mL) were added KOAc (61 mg, 0.6 mmol) and Pd(dppf)Cl2 (18 mg, 0.02 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 3 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 2-(2,3-dihydro-1-benzofuran-4- yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (30 mg, 48.2% yield) as a colorless oil. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. Compound 348.2
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide, Intermediate H (50 mg, 0.06 mmol) and 2- (2,3-dihydro-1-benzofuran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (18 mg, 0.07 mmol) in dioxane (3 mL) and H2O (0.5 mL) were added K2CO3 (22 mg, 0.1 mmol) and Pd(dppf)Cl2 (4 mg, 0.01 mmol) in one portion at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 8 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1-benzofuran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (20 mg, 38.6% yield) as a white solid. LCMS observed m/z = 828.26 [M+H] +. Step 3: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1- benzofuran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 348
Into a 8 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1-benzofuran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide (20 mg, 0.02 mmol) and TFA (3 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 1 h. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions (Column: X Bridge BEH C18 5 μm, 30*150 mm; mobile phase A: water (10 mmol/L NH4HCO3), mobile phase B: ACN; flow rate: 60 mL/min; gradient: 34% B to 50% B in 7 min; wave length: 254 nm / 220 nm; RT1 (min): 6.55) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(2,3-dihydro-1-benzofuran-4- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (2.4 mg, 12.5% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.81 (s, 1H), 9.08 (s, 1H), 8.62 (s, 1H), 8.52 (d, J = 8.7 Hz, 1H), 7.77 (d, J = 7.8 Hz, 1H), 7.62 (s, 1H), 7.57 – 7.52 (m, 1H), 7.24 (t, J = 8.0 Hz, 1H), 6.93 (d, J = 7.9 Hz, 1H), 5.69 (d, J = 12.9 Hz, 1H), 5.20 (s, 2H), 4.89 – 4.79 (m, 1H), 4.66 (t, J = 8.8 Hz, 2H), 3.86 (t, J = 11.7 Hz, 2H), 3.73 (t, J = 8.8 Hz, 2H), 3.58 – 3.48 (m, 1H), 3.24 (d, J = 7.8 Hz, 2H), 3.15 – 3.05 (m, 1H), 2.92 – 2.78 (m, 2H), 2.59 (s, 3H), 1.39 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 738.15 [M+H] +. EXAMPLE 349 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[4-(trifluoromethyl)bicyclo[2.2.2] octan-1-yl]acetamide Compound 349
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[4-(trifluoromethyl)bicyclo [2.2.2]octan-1-yl]acetamide. Compound 349.1
To a stirred mixture of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (50 mg, 0.08 mmol) and 4- (trifluoromethyl)bicyclo[2.2.2]octan-1-amine (18 mg, 0.09 mmol) in DCM (10 mL) were added DIEA (31 mg, 0.2 mmol) and 2,4,6-trichloro-1,3,5-triazine (30 mg, 0.2 mmol) in portions at 25°C. The resulting mixture was heated to 40 °C and stirred at 40 °C for 2 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 2-(6-{4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[4-(trifluoromethyl)bicyclo[2.2.2]octan-1-
yl]acetamide (20 mg, 31.3% yield) as a white solid. LCMS observed m/z = 790.36 [M+H] +. Step 2: Preparation of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[4-(trifluoromethyl)bicyclo[2.2.2] octan-1-yl]acetamide. Compound 349
Into a 20 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[4-(trifluoromethyl) bicyclo[2.2.2]octan-1-yl]acetamide (30 mg, 0.04 mmol) and TFA (5 mL) dropwise at room temperature. The resulting mixture was stirred at 90 °C for 30 min. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X bridge BEH C185μm, 19*250 mm; mobile phase A: water (10 mmol/L NH4HCO3), mobile phase B: MeOH; flow rate: 25 mL/min; gradient: 50% B to 65% B in 10 min; wave length: 254 nm / 220 nm; RT1 (min): 8.03) to afford 2- [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[4- (trifluoromethyl)bicyclo[2.2.2]octan-1-yl]acetamide (4.9 mg, 17.3% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 12.06 (s, 1H), 8.65 (s, 1H), 6.94 – 6.86 (m, 1H), 6.24 (s, 1H), 5.64 (d, J = 13.0 Hz, 1H), 4.80 (s, 3H), 4.39 (q, J = 2.8 Hz, 2H), 3.94 (t, J = 5.4 Hz, 2H), 3.83 (t, J = 11.5 Hz, 2H), 3.53 – 3.51 (m, 1H), 3.12 (d, J = 10.1 Hz, 3H), 2.90 – 2.82 (m, 2H), 2.76 – 2.70 (m, 2H), 2.66 (s, 3H), 1.95 – 1.91 (m, 6H), 1.84 – 1.76 (m, 6H), 1.30 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 700.30 [M+H] +. EXAMPLE 350
Synthesis of N-(6,6-difluorobicyclo[3.1.0]hexan-3-yl)-2-(2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 350
To a stirred solution of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy- 6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetic acid, Intermediate S (100 mg, 0.2 mmol) and DIEA (74 mg, 0.6 mmol) in DCM (5 mL) were added bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (97 mg, 0.4 mmol) and 6,6-difluorobicyclo[3.1.0]hexan-3-amine (30 mg, 0.23 mmol) in portions at 25°C. The resulting mixture was stirred at 25° for 2 h and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 ml/min; Gradient: 30% B to 50% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 5.73) to afford N-(6,6-difluorobicyclo[3.1.0]hexan-3- yl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (20.8 mg, 17% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.56 (s, 1H), 8.41 (d, J = 7.7 Hz, 1H), 6.81 (d, J = 2.8 Hz, 1H), 4.88 (s, 2H), 4.52 (d, J = 12.4 Hz, 1H), 4.27 (q, J = 2.9 Hz, 2H), 4.11 – 3.96 (m, 1H), 3.82 (t, J = 5.4 Hz, 2H), 3.53 – 3.44 (m, 5H), 3.02 – 2.93 (m, 1H), 2.87 (d, J = 7.8 Hz, 2H), 2.79 (d, J = 11.3 Hz, 1H), 2.61 (d, J = 11.3 Hz, 1H), 2.44 (s, 3H), 2.24 – 2.10 (m, 4H), 1.90 – 1.79 (m, 2H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 640.30 [M+H]+. Note: Two exchangeable protons were not observed in NMR spectra. EXAMPLE 351
Synthesis of N-(4,4-difluorocyclohexyl)-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 351
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-(4,4-difluorocyclohexyl)aceta mide. Compound 351.1
To a stirred mixture of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (50 mg, 0.08 mmol) and 4,4- difluorocyclohexan-1-amine (13 mg, 0.1 mmol) in DMF (1 mL) were added bis(2-oxo-1,3- oxazolidin-3-yl)phosphinoyl chloride (41 mg, 0.2 mmol) and DIEA (32 mg, 0.2 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The resulting mixture was diluted with water (20 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined
organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-4-yl)-N-(4,4-difluorocyclohexyl) acetamide (60 mg, crude) as a brown oil. LCMS observed m/z = 732.34 [M+H]+. Step 2: Preparation of N-(4,4-difluorocyclohexyl)-2-[2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]aceta mide. Compound 351
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- (4,4-difluorocyclohexyl)acetamide (55 mg, 0.07 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product (50 mg) was purified by Prep-HPLC with the following conditions (Column: X-bridge BEH Shield RP18, 5 μm, 19*250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 18% B to 27% B in 10 min; Wave Length: 254 nm/220 nm; RT1 (min): 9.28) to afford N-(4,4-difluorocyclohexyl)-2-[2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyri midine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (29.5 mg, 58% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.18 (s, 1H), 8.58 (s, 1H), 8.35 (d, J = 7.6 Hz, 1H), 6.81 (s, 1H), 4.92 (s, 2H), 4.52 (d, J = 12.4 Hz, 1H), 4.30 – 4.23 (m, 2H), 3.93 – 3.69 (m, 3H), 3.56 – 3.42 (m, 3H), 3.32 – 3.16 (m, 3H), 3.03 – 2.93 (m, 1H), 2.93 – 2.83 (m, 2H), 2.79 (d, J = 11.2 Hz, 1H), 2.62 (d, J = 10.8 Hz, 1H), 2.45 (s, 3H), 2.22 – 1.84 (m, 4H), 1.85 – 1.74 (m, 2H), 1.58 – 1.44 (m, 2H), 1.15 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 642.30 [M+H]+.
EXAMPLE 352 Synthesis of N-{3-cyclobutylbicyclo[1.1.1]pentan-1-yl}-2-[2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl] acetamide Compound 352
To a stirred solution of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid, Intermediate S (30 mg, 0.05 mmol, 1.0 equiv.) and 3- cyclobutylbicyclo[1.1.1]pentan-1-amine (7 mg, 0.05 mmol, 1.0 equiv.) in DCM (0.5 mL) were added HATU (19 mg, 0.05 mmol, 1.0 equiv.) and DIEA (29 mg, 0.2 mmol, 4.0 equiv.) in portions at 25 ℃. The resulting mixture was stirred at 25 ℃ for 4 hours under nitrogen atmosphere. Then the resulting mixture was quenched with H2O (10 mL), extracted with DCM (3 x 15 mL). The combined organic layers were washed with H2O (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure, collected the residue. The residue was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 19 mm * 250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 45% B to 75% B in 9 min; Wave Length: 254 nm/220 nm; RT1 (min): 7.17) to afford N-{3-cyclobutylbicyclo[1.1.1]pentan- 1-yl}-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpy rimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (11 mg, 29% yield) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ 10.25 (s, 1H), 8.87 (s, 1H), 8.59 (s, 1H), 6.83 – 6.82 (m, 1H), 4.88 (s, 2H), 4.55 – 4.49 (m, 1H), 4.27 (q, J = 2.9 Hz, 2H), 3.82 (t, J = 5.4 Hz, 2H), 3.50 (s, 4H), 3.02 - 2.95 (m, 1H), 2.91 - 2.74 (m, 1H), 2.91 – 2.73 (m, 3H), 2.64 – 2.61 (m, 1H), 2.54 – 2.53 (m, 1H), 2.45 (s, 3H), 2.41 (t, J = 7.6 Hz, 1H), 2.00 – 1.88 (m, 2H), 1.78 -1.76 (m, 7H), 1.72 – 1.62 (m, 3H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 644.45[M+H]+.
EXAMPLE 353 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-(4-fluorophenyl)bicyclo[1.1.1]pentan-1-yl]acetamide Compound 353
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[3-(4-fluorophenyl)bicyclo[1.1.1]pentan-1-yl]acetamide. Compound 353.1
To a stirred solution of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (60 mg, 0.1 mmol), DIEA (37 mg, 0.3 mmol) and HATU (56 mg, 0.1 mmol) in DCM (5 mL) was added 3-(4- fluorophenyl)bicyclo[1.1.1]pentan-1-amine (19 mg, 0.1 mmol) in portions at room
temperature. The resulting mixture was stirred for 3 h at room temperature. After completion of reaction, the reaction mixture was quenched by addition of water (3 mL). The aqueous layer was extracted with DCM (3x 5 mL). The combined organic phases were washed with brine (3 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford (50 mg, crude) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[3-(4-fluorophenyl)bicyclo[1.1.1]pentan-1-yl]acetamide as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 774.31 [M+H]+. Step 2: Preparation of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-(4-fluorophenyl)bicyclo[1.1.1]pentan-1-yl]acetamide. Compound 353
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[3-(4-fluorophenyl)bicyclo[1.1.1]pentan-1-yl]acetamide (30 mg, 0.04 mmol) in DCM (2 mL) was added boron trichloride (0.4 mL, 1M in DCM, 0.4 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for additional 2 h at room temperature. After completion of reaction, the reaction mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 μm, 30 mm * 150 mm; mobile phase: 43-66% ACN in water (0.1% FA)) to afford (6.1 mg, 21.8% yield) of 2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[3-(4- fluorophenyl)bicyclo[1.1.1]pentan-1-yl]acetamide as a white solid.1H NMR (400 MHz,
Chloroform-d) δ 12.27 (s, 1H), 8.69 (s, 1H), 7.26 – 7.11 (m, 2H), 7.10 – 6.88 (m, 3H), 6.81 (s, 1H), 5.62 (s, 1H), 4.90 (s, 2H), 4.82 (d, J = 12.4 Hz, 1H), 4.40 (s, 2H), 3.94 (t, J = 5.1 Hz, 2H), 3.91 – 3.73 (m, 2H), 3.55 (s, 1H), 3.27 – 3.03 (m, 3H), 2.99 – 2.80 (m, 2H), 2.80 – 2.59 (m, 5H), 2.38 (s, 6H), 1.35 – 1.29 (m, 3H). LCMS observed m/z = 684.30 [M+H]+. EXAMPLE 354 Synthesis of N-{3-[3-(1,1-difluoroethyl)bicyclo[1.1.1]pentan-1- yl]bicyclo[1.1.1]pentan-1-yl}-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy- 6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide. Compound 354
To a mixture of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid, Intermediate S (30 mg, 0.06 mmol) in DCM (5 mL) were added 3-[3-(1,1- difluoroethyl)bicyclo[1.1.1]pentan-1-yl]bicyclo[1.1.1]pentan-1-amine (13 mg, 0.06 mmol), HATU (32 mg, 0.09 mmol) and DIPEA (22. mg, 0.2 mmol) in portions at room temperature. The reaction mixture was stirred at room temperature for 5 hours. The resulting mixture was quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (PE/ EA = 5/1) to afford N-{3-[3-(1,1-difluoroethyl)bicyclo[1.1.1]pentan-1- yl]bicyclo[1.1.1]pentan-1-yl}-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy- 6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (13 mg, 32% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.24 (s, 1H), 8.89 (s, 1H), 8.59 (s, 1H), 6.82 (s, 1H), 4.87 (s, 2H), 4.52 (d, J = 12.7 Hz, 1H), 4.27 (d, J = 3.2 Hz, 2H), 3.82 (t, J = 5.5 Hz, 2H), 3.60 (s, 5H), 3.26 (d, J = 12.1 Hz, 1H), 2.98 (t,
J = 12.1 Hz, 1H), 2.92 – 2.72 (m, 3H), 2.62 (d, J = 10.6 Hz, 1H), 2.45 (s, 3H), 1.84 (s, 6H), 1.65 (s, 6H), 1.52 (t, J = 18.7 Hz, 3H), 1.13 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 720.30 [M+H]+. EXAMPLE 355 Synthesis of N-(2-chloro-4-(chlorodifluoromethoxy)phenyl)-2-(2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
The title compound was prepared using similar procedure as compound 68 to afford N-(2-chloro-4-(chlorodifluoromethoxy)phenyl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl- 6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.26 (s, 1H), 8.53 (s, 1H), 7.83 (d, J = 9.0 Hz, 1H), 7.64 (d, J = 2.7 Hz, 1H), 7.38 (dd, J = 8.9, 2.8 Hz, 1H), 6.90 – 6.80 (m, 1H), 5.26 (s, 2H), 4.52 (d, J = 12.5 Hz, 1H), 4.26 (q, J = 2.8 Hz, 2H), 3.80 (t, J = 5.4 Hz, 2H), 3.59 – 3.42 (m, 4H), 3.28 – 3.15 (m, 3H), 2.98 (q, J = 8.2 Hz, 3H), 2.80 (d, J = 11.2 Hz, 1H), 2.63 (d, J = 10.8 Hz, 1H), 2.43 (s, 3H), 1.19 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 734.2 [M+H]+. EXAMPLE 356 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{4-[(trifluoromethyl)sulfanyl]phenyl} acetamide. Compound 356
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- {4-[(trifluoromethyl)sulfanyl] phenyl}acetamide (40 mg, 0.05 mmol) and 2-(3,6-dihydro- 2H-pyran-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (12 mg, 0.1 mmol) in dioxane (3 mL) and H2O (0.5 mL) were added Na2CO3 (13 mg, 0.1 mmol) and Pd(dppf)Cl2 (3 mg, 0.005 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 8 h under nitrogen atmosphere. After the reaction was completed, the mixture was cooled down to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X bridge BEH Shield RP18, 5μm, 19 * 250 mm; mobile phase A: water (10 mmol/L NH4HCO3), mobile phase B: ACN; flow rate: 25 mL/min; gradient: 31% B to 40% B in 10 min; wave length: 254 nm / 220 nm; RT1 (min): 8.22) to afford 2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (3.7 mg, 10.3% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.84 (s, 1H), 9.04 (s, 1H), 8.61 (s, 1H), 7.65 (d, J = 8.4 Hz, 2H), 7.59 – 7.54 (m, 2H), 6.97 (s, 1H), 5.68 (d, J = 12.6 Hz, 1H), 5.06 (s, 2H), 4.88 – 4.78 (m, 2H), 4.44 – 4.37 (m, 2H), 3.95 (t, J = 5.4 Hz, 2H), 3.82 (t, J = 11.6 Hz, 2H), 3.53 (d, J = 12.9 Hz, 1H), 3.19 (d, J = 7.6 Hz, 2H), 3.10 (d, J = 5.4 Hz, 1H), 2.75 (s, 3H), 2.59 (s, 3H), 1.35 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 700.30 [M+H] +. EXAMPLE 357 Synthesis of N-{6,6-difluorospiro[3.3]heptan-2-yl}-2-[2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 357
To a stirred solution of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid, Intermediate S (30 mg, 0.1 mmol) and 6,6-difluorospiro[3.3]heptan-2-amine (10 mg, 0.1 mmol) in DCM (3 mL) were added DIEA (15 mg, 0.1 mmol) and HATU (33 mg, 0.1 mmol) in portions at room temperature. The resulting mixture was stirred for additional 2 h at room temperature. After completed, poured the resulting mixture into water (3 mL) and extracted with DCM (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography (Column: Xselect CSH C185 m, 19 mm * 250 mm; mobile phase: 33%-56% ACN in H2O) to afford (13.5 mg, 34% yield) of N-{6,6-difluorospiro[3.3]heptan-2-yl}-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 12.09 (s, 1H), 8.65 (s, 1H), 6.93 (s, 1H), 6.64 – 6.51 (m, 1H), 5.71 – 5.55 (m, 1H), 4.97 – 4.70 (m, 3H), 4.48 – 4.36 (m, 2H), 4.36 – 4.24 (m, 1H), 4.01 – 3.90 (m, 2H), 3.89 – 3.74 (m, 2H), 3.52 (s, 1H), 3.24 – 3.03 (m, 3H), 2.92 – 2.75 (m, 2H), 2.73 (s, 2H), 2.68 – 2.58 (m, 5H), 2.61 – 2.46 (m, 4H), 2.09 – 2.04 (m, 2H), 1.40 – 1.21 (m, 3H). LCMS observed m/z = 654.35 [M+H]+. EXAMPLE 358 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[1-(trifluoromethyl)-2-oxabicyclo[2.2.2]octan-4-yl]acetamide. Compound 358
To a stirred solution of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid (28 mg, 0.05 mmol) and 1-(trifluoromethyl)-2-oxabicyclo[2.2.2]octan-4- amine (15.6 mg, 0.08 mmol) in ACN (0.5 mL) were added TCFH (15.0 mg, 0.05 mmol) and NMI (10.9 mg, 0.1 mmol) in portions at 25 °C. Then the resulting mixture was stirred at 25 °C for 4 h. After completion of reaction, the resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC (Column: Sunfire C18 5 m, 30 mm * 150 mm; Mobile Phase: ACN in water(0.1% FA)) to afford (8.7 mg, 23% yield) of 2-[2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[1- (trifluoromethyl)-2-oxabicyclo[2.2.2]octan-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.27 (s, 1H), 8.54 (s, 1H), 8.28 (s, 1H), 6.87 – 6.79 (m, 1H), 4.89 (s, 2H), 4.52 (d, J = 12.5 Hz, 1H), 4.28 (q, J = 2.8 Hz, 2H), 3.96 (s, 2H), 3.82 (t, J = 5.4 Hz, 2H), 3.57 – 3.41 (m, 4H), 3.24 (s, 2H), 2.98 (m, 1H), 2.87 (q, J = 7.7 Hz, 2H), 2.79 (d, J = 11.3 Hz, 1H), 2.61 (d, J = 11.2 Hz, 1H), 2.44 (s, 3H), 2.14 – 1.83 (m, 8H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 702.25[M+H]+. EXAMPLE 359 Synthesis of N-{3-cyclopropylbicyclo[1.1.1]pentan-1-yl}-2-[2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 359
A solution of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid (30 mg, 0.1 mmol) in DMF (1 mL) was treated with HATU (33 mg, 0.1 mmol) and DIEA (37 mg, 0.3 mmol) at room temperature under nitrogen atmosphere followed by the addition of 3-cyclopropylbicyclo[1.1.1]pentan-1-amine (9 mg, 0.1 mmol) in portions at room temperature. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min mL/min; Gradient: 25% B to 35%B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 7.65) to afford (12.2 mg, 32% yield) of N-{3-cyclopropylbicyclo[1.1.1]pentan- 1-yl}-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.23 (s, 1H), 8.85 (s, 1H), 8.58 (s, 1H), 6.82 (s, 1H), 4.86 (s, 1H), 4.52 (m, 1H), 4.27 (d, J = 3.1 Hz, 1H), 3.82 (t, J = 5.4 Hz, 1H), 3.48 (m, 2H), 3.30 (s, 11H), 2.98 (m, 1H), 2.92 (s, 1H), 2.82 (m, 2H), 2.62 (m, 1H), 2.45 (s, 2H), 1.83 (s, 1H), 1.75 (s, 4H), 1.13 (t, J = 7.5 Hz, 2H), 0.45 – 0.33 (m, 2H), 0.16 – 0.04 (m, 2H). LCMS observed m/z = 630.30 [M+H]+. EXAMPLE 360 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-(trifluoromethyl)quinolin-8-yl)acetamide Compound 360
The title compound was prepared using similar procedure as compound 68 to afford 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5- (trifluoromethyl)quinolin-8-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 11.18 (s, 1H), 9.14 (d, J = 4.2 Hz, 1H), 8.66 (d, J = 8.3 Hz, 1H), 8.54 (d, J = 11.5 Hz, 2H), 8.06 (d, J = 8.4 Hz, 1H), 7.89 (dd, J = 8.7, 4.2 Hz, 1H), 6.78 (d, J = 3.4 Hz, 1H), 5.56 (s, 2H), 4.52 (d, J = 12.6 Hz, 1H), 4.20 (q, J = 2.9 Hz, 2H), 3.78 (t, J = 5.4 Hz, 2H), 3.51 (td, J = 12.3, 7.0 Hz, 4H), 3.28 – 3.16 (m, 3H), 2.98 (tt, J = 12.8, 5.0 Hz, 3H), 2.83 (d, J = 11.3 Hz, 1H), 2.66 (d, J = 10.8 Hz, 1H), 2.43 (s, 3H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 719.3 [M+H]+. EXAMPLE 361 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-(3,3,3-trifluoropropyl)bicyclo [1.1.1]pentan-1-yl]acetamide. Compound 361
To a stirred mixture of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid (30 mg, 0.06 mmol) and 3-(3,3,3-trifluoropropyl)bicyclo[1.1.1]pentan-1- amine (12 mg, 0.07 mmol) in DCM (1 mL) were added bis(2-oxo-1,3-oxazolidin-3-
yl)phosphinoyl chloride (22 mg, 0.09 mmol) and DIEA (22 mg, 0.2 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product (30 mg) was purified by Prep-HPLC with the following conditions (Column: X-select CSH C185 m, 19 mm * 250 mm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 46% B to 69% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 6.19) to afford 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-(3,3,3-trifluoropropyl)bicyclo[1.1.1]pentan-1-yl]acetamide (7.3 mg, 18% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.22 (s, 1H), 8.87 (s, 1H), 8.58 (s, 1H), 6.82 (s, 1H), 4.87 (s, 2H), 4.52 (d, J = 12.4 Hz, 1H), 4.27 (s, 2H), 3.85 – 3.78 (m, 2H), 3.53 – 3.43 (m, 3H), 3.33 – 3.31 (m, 1H), 3.31 – 3.19 (m, 2H), 2.98 (t, J = 11.2 Hz, 1H), 2.88 – 2.76 (m, 3H), 2.62 (d, J = 11.2 Hz, 1H), 2.45 (s, 3H), 2.25 – 2.11 (m, 2H), 1.87 (s, 6H), 1.76 – 1.67 (m, 2H), 1.14 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 686.45 [M+H]+. EXAMPLE 362 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[4-(trifluoromethyl)bicyclo[2.2.1] heptan-1-yl]acetamide. Compound 362
Step 1: Preparation of 4-(trifluoromethyl)bicyclo[2.2.1]heptan-1-amine. Compound 362.1
To a stirred solution of 4-(trifluoromethyl)bicyclo[2.2.1]heptane-1-carboxylic acid (100 mg, 0.5 mmol) and TEA (53 mg, 0.5 mmol) in ACN (4 mL) was added DPPA (145 mg, 0.5 mmol) dropwise at 25 °C. The resulting mixture was stirred at 50 °C for 3 h. Then the mixture was allowed to cool down to 25°C. The reaction mixture was added conc. HCl (1.5 mL) dropwise. The resulting mixture was stirred at 90 °C for 3.5 h and stirred at 85 °C for additional 15 h. After completion of reaction, the resulting mixture was cooled down to 25 °C and basified to pH 7 with saturated NaHCO3 (aq.). The resulting mixture was extracted with CH2Cl2 (3 x 25 mL). The combined organic layers were washed with water (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give 4-(trifluoromethyl)bicyclo[2.2.1]heptan-1-amine (80 mg, crude) as a white solid. LCMS observed m/z = 180.09 [M+H] +. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[4-(trifluoromethyl)bicyclo [2.2.1]heptan-1-yl]acetamide. Compound 362.2
To a stirred mixture of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (39 mg, 0.06 mmol) and 4- (trifluoromethyl)bicyclo[2.2.1]heptan-1-amine (13 mg, 0.07 mmol) in ACN (4 mL) were added TCFH (44 mg, 0.2 mmol) and NMI (15 mg, 0.2 mmol) in portions at 25 °C. The resulting mixture was stirred at 25 °C for 4 h under nitrogen atmosphere. After completion of reaction, the resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford 2-(6-{4-
[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-2-(3,6-dihydro-2H-pyran- 4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[4- (trifluoromethyl)bicyclo[2.2.1]heptan-1-yl]acetamide (20 mg, 40.5% yield) as a white solid. LCMS observed m/z = 828.26 [M+H] +. Step 3: Preparation of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[4-(trifluoromethyl)bicyclo[2.2.1] heptan-1-yl]acetamide. Compound 362
Into a 8 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[4-(trifluoromethyl) bicyclo[2.2.1]heptan-1-yl]acetamide (20 mg, 0.02 mmol) and TFA (3 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 1 h. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C185 μm, 30*150 mm; mobile phase A: water (10 mmol/L NH4HCO3), mobile phase B: ACN; flow rate: 60 mL/min; gradient: 34% B to 50% B in 7 min; wave length: 254 nm / 220 nm; RT1 (min): 6.55) to afford 2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[4-(tri fluoromethyl)bicyclo[2.2.1]heptan-1-yl]acetamide (2.2 mg, 12.5% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 8.62 (s, 1H), 6.94 – 6.85 (m, 1H), 6.69 (s, 1H), 5.67 (d, J = 13.1 Hz, 1H), 4.89 – 4.77 (m, 3H), 4.44 – 4.34 (m, 2H), 3.94 (t, J = 7.4 Hz, 2H), 3.87 – 3.77 (m, 2H), 3.56 – 3.46 (m, 1H), 3.17 – 3.07 (m, 3H), 2.86 (d, J = 11.3 Hz, 2H), 2.80 – 2.69 (m, 2H), 2.59 (s, 3H), 2.06 – 2.01 (m, 4H), 1.98 (s, 2H), 1.81 (d, J =
10.9 Hz, 2H), 1.68 (s, 2H), 1.30 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 686.30 [M+H] +. EXAMPLE 363 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-(trifluoromethyl)pyridin-2-yl)acetamide. Compound 363
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(5-(trifluoromethyl)pyridin-2-yl)acetamide. Compound 363.1
To a stirred solution of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (100 mg, 0.2 mmol) in DCM (5 mL) were added 5-(trifluoromethyl)pyridin-2-amine (40 mg, 0.3 mmol) and DIEA (63 mg, 0.6 mmol) dropwise at 25 °C, followed by the addition of bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl
chloride (166 mg, 0.8 mmol) dropwise at 25 °C. The resulting mixture was heated to 40 °C and stirred at 40 °C for 5 hours. After completion of reaction, the resulting mixture was cooled to 25 °C and quenched with H2O (10 mL). The resulting mixture was extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 2-(6-(4-(5- (benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-(trifluoromethyl)pyridin- 2-yl)acetamide (60mg, crude). The crude product was used in the next step directly without further purification. LCMS observed m/z = 759.29 [M+H]+. Step 2: Preparation of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-(trifluoromethyl)pyridin-2-yl)acetamide. Compound 363
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)pyridin-2-yl]acetamide (60 mg, 0.1 mmol) in DCM (2 mL) was added boron trichloride (2 mL) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for additional 2 hours. After completion of reaction, the reaction was quenched with water at 25 °C and extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by prep- HPLC with the following conditions (Mobile Phase A: water (10 mmol/L NH4HCO3); Mobile Phase B: ACN; Flow Rate: 25; Wave Length: 254 nm / 220 nm) to give 2-(2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5- (trifluoromethyl)pyridin-2-yl)acetamide (6.4 mg, 11.3% yield) as an off-white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.89 (s, 1H), 9.22 (s, 1H), 8.61 (d, J = 9.7 Hz, 2H), 8.25
(s, 1H), 8.01 – 7.94 (m, 1H), 7.00 (s, 1H), 5.68 (d, J = 13.1 Hz, 1H), 5.13 (s, 2H), 4.82 (d, J = 12.7 Hz, 1H), 4.39 (d, J = 2.9 Hz, 2H), 3.94 (m, 2H), 3.95 – 3.75 (m, 2H), 3.52 (s, 1H), 3.17 – 3.10 (m, 3H), 2.92 – 2.70 (m, 4H), 2.61 (s, 3H), 1.38 – 1.32 (m, 3H). LCMS observed m/z = 669.25 [M+H]+. EXAMPLE 364 Synthesis of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 364
Step 1: Preparation of tert-butyl 4-{4-[({2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}carbamoyl)methyl]-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate Compound 364.1
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (60 mg, 0.1 mmol) and 2-chloro-4-[(trifluoromethyl)sulfanyl] aniline (36 mg, 0.2 mmol) in DCM (3 mL) were added DIEA (48 mg, 0.4 mmol) and bis(2-oxo-1,3-oxazolidin-3-yl) phosphinoyl chloride (63 mg, 0.2 mmol) at room temperature under N2 atmosphere. The resulting
mixture was stirred for additional 16 hours at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL). The resulting mixture was extracted with DCM (5 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (55 mg, 64.1% yield) of tert-butyl 4-{4-[({2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}carbamoyl)methyl]-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate as a yellow solid. LCMS observed m/z = 698.16 [M+H]+. Step 2: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl]acetamide Compound 364.2
of tert-butyl 4-{4-[({2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}carbamoyl)methyl]-2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate (50 mg, 0.072 mmol) and TFA (500 uL) in DCM (2 mL) was stirred for 30 min at room temperature under N2 atmosphere. Desired product could be detected by LCMS. Desired product could be detected by LCMS. The crude product was used in the next step directly without further purification. LCMS observed m/z = 598.16 [M+H]+. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-acetamide Compound 364.3
To a stirred solution of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl]acetamide (48 mg, 0.1 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (20 mg, 0.1 mmol) in DMF (1 mL) were added HATU (37 mg, 0.1 mmol) and DIEA (52 mg, 0.4 mmol) at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 1 hour at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (3 mL) and extracted with EA (3 mL x 3). The combined organic layers were washed with brine (2 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 5% MeOH in DCM) to afford (60 mg, 90.7 % yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}acetamide as a light yellow solid. LCMS observed m/z = 824.23 [M+H]+. Step 4: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 364
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- {2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (60 mg, 0.1 mmol) in TFA (3 mL) was stirred for 18 hours at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 30% B to 50%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.87) to afford (17.3 mg, 31.7% yield) of N-{2- chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6- [4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 12.09 (s, 1H), 8.98 (s, 1H), 8.63 (s, 1H), 8.43 (d, J = 8.7 Hz, 1H), 7.68 (d, J = 2.1 Hz, 1H), 7.57 (dd, J = 8.7, 2.1 Hz, 1H), 7.01 – 6.95 (m, 1H), 5.62 (m, 1H), 5.10 (s, 2H), 4.80 (m, 1H), 4.37 (q, J = 2.8 Hz, 2H), 3.92 (t, J = 5.4 Hz, 2H), 3.81 (m, 2H), 3.53 (m, 1H), 3.18 (d, J = 8.0 Hz, 2H), 3.10 (m, 1H), 2.89 – 2.69 (m, 4H), 2.65 (s, 3H), 1.34 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 734.30 [M+H]+. EXAMPLE 365 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-(trifluoromethyl)quinolin-8-yl)acetamide Compound 365
The title compound was prepared using similar procedure as compound 68 to afford 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5- (trifluoromethyl)quinolin-8-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 11.18 (s, 1H), 9.14 (d, J = 4.2 Hz, 1H), 8.66 (d, J = 8.3 Hz, 1H), 8.54 (d, J = 11.5 Hz, 2H), 8.06 (d, J = 8.4 Hz, 1H), 7.89 (dd, J = 8.7, 4.2 Hz, 1H), 6.78 (d, J = 3.4 Hz, 1H), 5.56 (s, 2H), 4.52 (d, J = 12.6 Hz, 1H), 4.20 (q, J = 2.9 Hz, 2H), 3.78 (t, J = 5.4 Hz, 2H), 3.51 (td, J = 12.3, 7.0 Hz, 4H), 3.28 – 3.16 (m, 3H), 2.98 (tt, J = 12.8, 5.0 Hz, 3H), 2.83 (d, J = 11.3 Hz, 1H), 2.66 (d, J = 10.8 Hz, 1H), 2.43 (s, 3H), 1.18 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 719.3 [M+H]+. EXAMPLE 366 Synthesis of N-(1,1-difluorospiro[2.3]hexan-5-yl)-2-(2-(3,6-dihydro-2H-pyran-4- yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 366
To a solution of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid (50 mg, 0.1 mmol) and DIEA (37 mg, 0.3 mmol). in DCM (3 mL) were added bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (49 mg, 0.2 mmol) and 1,1-
difluorospiro[2.3]hexan-5-amine (15 mg, 0.12 mmol) in portions at 25°C. After that, the resulting mixture was stirred for 30 min at 25°C. After completion of reaction, the resulting mixture was concentrated to dryness under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: MeOH; Flow rate: 60 mL/min; Gradient: 31% B to 54% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 7.3) to afford N-(1,1-difluorospiro[2.3]hexan-5-yl)-2- (2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (36.2 mg, 57.4%yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.25 (s, 1H), 8.80 (m, 1H), 8.56 (s, 1H), 6.82 (s, 1H), 4.92 (s, 2H), 4.52 (d, J = 12.5 Hz, 1H), 4.44 – 4.34 (m, 1H), 4.31 – 4.24 (m, 2H), 3.81 (t, J = 5.5 Hz, 2H), 3.57 – 3.44 (m, 3H), 3.29 – 3.19 (m, 2H), 3.01 – 2.93 (m, 1H), 2.94 – 2.85 (m, 2H), 2.84 – 2.76 (m, 1H), 2.66 – 2.59 (m, 1H), 2.49 – 2.42 (m, 4H), 2.42 – 2.19 (m, 4H), 1.43 (m, 2H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 640.45 [M+H]+. EXAMPLE 367 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[1-meth yl-2-oxo-6-(trifluoromethyl)pyridin-3-yl]acetamide Compound 367
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(1-methyl-2-oxo-6-(trifluoromethyl)-1,2-dihydropyridin-3- yl)acetamide. Compound 367.1
To a stirred solution of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (50 mg, 0.1 mmol), DIEA (21 mg, 0.2 mmol) and 3-amino-1-methyl-6-(trifluoromethyl)pyridin-2-one (16 mg, 0.1 mmol) in DCM (2 mL) was added bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (41 mg, 0.2 mmol) in portions at 25 °C. The mixture was stirred at 40 °C for 2 h. The residue was purified prep- TLC PE / EA (0/1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[1-methyl-2-oxo-6-(trifluoromethyl)pyridin-3-yl]acetamide (26 mg, 40% yield) as a white solid. LCMS observed m/z:789.30[M+H] + Step 2: Preparation of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimi dine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(1-methyl-2-oxo-6-(trifluoromethyl)-1,2-dihydropyridin-3-yl)acetamide. Compound 367
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[1-methyl-2-oxo-6-(trifluoromethyl)pyridin-3-yl]acetamide (25 mg, 0.03 mmol) in DCM (1 mL) was added boron trichloride (0.5 mL) dropwise at 0 °C under nitrogen atmosphere. The mixture was stirred at 0 °C for 30 min. The residue was purified by reversed-phase flash chromatography with the following conditions: column, Column: Sunfire C185 m, 30 mm * 150 mm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 37% B to 60% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 5.47 to give 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[1-meth yl-2-oxo-6-(trifluoromethyl)pyridin-3-yl]acetamide (1.9 mg, 8% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.22 (s, 1H), 9.17 (s, 1H), 8.66 (s, 1H), 8.25 (d, J = 7.2 Hz, 1H), 7.00 (s, 1H), 6.76 (d, J = 8.0 Hz, 1H), 5.59 (s, 1H), 5.11 (s, 2H), 4.83 (s, 1H), 4.36 (s, 2H), 3.93 (t, J = 5.1 Hz, 2H), 3.86 (s, 2H), 3.75 (s, 3H), 3.56 (s, 1H), 3.11 (s, 4H), 2.89 (s, 2H), 2.72 (s, 4H), 1.32 (t, J = 5.9 Hz, 3H). LCMS observed m/z = 699.35[M+H] +. EXAMPLE 368 Synthesis of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide Compound 368
Step 1: Preparation of N-((2-bromo-5- (trifluoromethyl)phenyl)carbamothioyl)benzamide. Compound 368.1
To a stirred solution of 2-bromo-5-(trifluoromethyl)aniline (2.0 g, 8.3 mmol) in acetone (20 mL) was added benzoyl isothiocyanate (2.0 g, 10.0 mmol) in portions at 25 °C. The resulting mixture was stirred at 25 °C for 12 hours. After completion of reaction, the resulting mixture was concentrated under reduced pressure to afford N-((2-bromo-5- (trifluoromethyl)phenyl)carbamothioyl)benzamide (2.0 g, crude) as an off-white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 402.96 [M+H]+. Step 2: Preparation of 1-(2-bromo-5-(trifluoromethyl)phenyl)thiourea. Compound 368.2
Into a 50 mL round bottom were added N-((2-bromo-5- (trifluoromethyl)phenyl)carbamothioyl)benzamide (2.0 g, 5.0 mmol and NaOH (20 mL, 5% aq) dropwise at 25 °C. The resulting mixture was heated to 90 ℃ and stirred at 90 ℃ for 20 minutes. After completion of reaction, the resulting mixture was cooled to 25 °C and diluted with H2O (10 mL). The resulting mixture was extracted with EA (3 x 15 mL). The combined organic layers were washed brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 1-(2-bromo-5- (trifluoromethyl)phenyl)thiourea (2.3 g, crude) as an off-white solid. LCMS observed m/z = 298.94 [M+H]+. Step 3: Preparation of 4-bromo-7-(trifluoromethyl)benzo[d]thiazol-2-amine. Compound 368.3 To a stirred solution of 1-(2-bromo-5-(trifluoromethyl)phenyl)thiourea (2.0 g, 6.7 mmol) in AcOH (10 mL) was added Br2 (4.0 g, 26.8 mmol) dropwise at 25 °C. The resulting
mixture was heated to 110 ℃ and stirred at 110 ℃ for 12 hours. After completion of reaction, the resulting mixture was cooled to 25 °C and quenched with H2O (20 mL). The mixture was neutralized to pH = 8 with saturated NaHCO3 (aq.). The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure, collected the residue. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford 4-bromo-7- (trifluoromethyl)benzo[d]thiazol-2-amine (1.2 g, 60.3% yield) as a yellow solid. LCMS observed m/z = 296.92 [M+H]+. Step 4: Preparation of 4-bromo-7-(trifluoromethyl)benzo[d]thiazole. Compound 368.4
To a stirred solution of 4-bromo-7-(trifluoromethyl)benzo[d]thiazol-2-amine (1.0 g, 3.4 mmol) in THF (10 mL) was added isopentyl nitrite (789 mg, 6.8 mmol) dropwise at 25 ℃. The reaction mixture was heated to 70 ℃ and stirred at 70 ℃ for 4 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 ℃ and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 4-bromo-7-(trifluoromethyl)benzo[d]thiazole (488 mg, 51.4% yield) as a yellow solid. LCMS observed m/z = 281.91 [M+H]+. Step 5: Preparation of tert-butyl(7-(trifluoromethyl)benzo[d]thiazol-4-yl)carbamate. Compound 368.5
To a stirred solution of 4-bromo-7-(trifluoromethyl)benzo[d]thiazole (200 mg, 0.7 mmol), tert-butyl carbamate (100 mg, 0.8 mmol), XPhos (68 mg, 0.1 mmol) and Cs2CO3 (693 mg, 2.1 mmol) in dioxane (10 mL) was added Pd2(dba)3 (65 mg, 0.1 mmol) in one portion at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 4 hours under nitrogen atmosphere. After completion of reaction, the
resulting mixture was cooled to 25 °C and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford tert- butyl(7-(trifluoromethyl)benzo[d]thiazol-4-yl)carbamate (200 mg, 89.3% yield) as a yellow solid. LCMS observed m/z = 319.06 [M+H]+. Step 6: Preparation of 7-(trifluoromethyl)benzo[d]thiazol-4-amine. Compound 368.6
To a stirred solution of tert-butyl (7-(trifluoromethyl)benzo[d]thiazol-4- yl)carbamate (200 mg, 0.6 mmol) in dioxane (10 mL) was added HCl (2 mL, 4 M in dioxane) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 4 hours. After completion of reaction, the resulting mixture was concentrated under reduced pressure to afford 7-(trifluoromethyl)benzo[d]thiazol-4-amine (100 mg, crude). LCMS observed m/z = 219.01 [M+H]+. Step 7: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide. Compound 368.7
To a stirred solution of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (100 mg, 0.2 mmol), DIEA (63 mg, 0.6 mmol) and 7-(trifluoromethyl)benzo[d]thiazol-4-amine (53 mg, 0.3 mmol) in DCM (1 mL) was added bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (166 mg, 0.8 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 40 °C and stirred at 40 °C for 2 hours under nitrogen atmosphere. After completion of reaction, the resulting
mixture was cooled to 25 °C and quenched with H2O (10 mL). The aqueous layer was extracted with EA (3 x 15 mL). After filtration, the filtrate was concentrated under reduced pressure to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)- 2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide (150 mg, crude) . LCMS observed m/z = 815.26 [M+H]+. Step 8: Preparation of 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(7-(trifluoromethyl)benzo[d] thiazol-4-yl)acetamide. Compound 368
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(7-(trifluoromethyl) benzo[d]thiazol-4-yl)acetamide (100 mg, 0.1 mmol) in DCM (1 mL) was added boron trichloride (1 mL) dropwise 0 °C. The resulting mixture was stirred at 25 °C for 2 hours. After completion of reaction, the reaction was quenched with water at 25 °C and extracted with CH2Cl2 (3 x 10 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by prep-HPLC with the following conditions (Mobile Phase A: water (10 mmol/L NH4HCO3); Mobile Phase B: ACN; Flow Rate: 60 min; Wave Length: 254 nm / 220 nm) to afford 2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(7- (trifluoromethyl)benzo[d]thiazol-4-yl)acetamide (9.9 mg, 11.1% yield) as an off-white solid.1H NMR (400 MHz, Chloroform-d) δ 12.03 (s, 1H), 9.97 (s, 1H), 8.99 (s, 1H), 8.64 (s, 1H), 8.57 (d, J = 8.3 Hz, 1H), 7.77 (d, J = 8.4 Hz, 1H), 7.11 (s, 1H), 5.66 (d, J = 12.7 Hz, 1H), 5.22 (s, 2H), 4.82 (d, J = 12.7 Hz, 1H), 4.42 – 4.37 (m, 2H), 3.98 – 3.91 (m, 2H), 3.90
– 3.80 (m, 2H), 3.53 (s, 1H), 3.25 – 3.16 (m, 2H), 3.16 – 3.06 (m, 1H), 2.89 (d, J = 11.8 Hz, 2H), 2.78 (s, 2H), 2.65 (s, 3H), 1.42 – 1.33 (m, 3H). LCMS observed m/z = 725.30 [M+H]+. EXAMPLE 369 Synthesis of N-(4-difluoromethoxy-2-fluorophenyl) [2-(3,6-dihydro-2H-pyran-4- yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo- 1,3,3a,7-tetraaza-7-indenyl]acetamide Compound 369
The title compound was prepared using similar procedure as compound 68 to afford N-(4-difluoromethoxy-2-fluorophenyl) [2-(3,6-dihydro-2H-pyran-4-yl)-6-ethyl-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7- indenyl]acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.33 (s, 1H), 8.41 (s, 1H), 7.78 (t, J = 8.9 Hz, 1H), 7.18 (s, 1H), 6.96 – 6.93 (m, 1H), 6.75 (s, 1H), 5.16 (s, 2H), 4.44 (d, J = 12.4 Hz, 1H), 4.19 (d, J = 3.1 Hz, 2H), 3.73 (t, J = 5.5 Hz, 2H), 3.43 - 3.39 (m, 5H), 3.17 – 3.14 (m, 2H), 2.93 - 2.86 (m, 3H), 2.73 (d, J = 11.3 Hz, 1H), 2.55 (d, J = 10.8 Hz, 1H), 2.34 (s, 3H), 1.10 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 684.4 [M+H]+. EXAMPLE 370 Synthesis of N-(2-chloro-4-difluoromethoxyphenyl) [2-(3,6-dihydro-2H-pyran-4- yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo- 1,3,3a,7-tetraaza-7-indenyl] acetamide Compound 370
The title compound was prepared using similar procedure as compound 68 to afford N-(2-chloro-4-difluoromethoxyphenyl) [2-(3,6-dihydro-2H-pyran-4-yl)-6-ethyl-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7- indenyl] acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.13 (s, 1H), 8.38 (s, 1H), 7.62 (d, J = 8.9 Hz, 1H), 7.36 (d, J = 2.8 Hz, 1H), 7.13 – 7.10 (m, 1H), 6.78 (s, 1H), 5.16 (s, 2H), 4.44 (d, J = 12.6 Hz, 1H), 4.19 (s, 2H), 3.74 (t, J = 5.5 Hz, 2H), 3.43 – 3.36 (m, 5H), 3.18 – 3.13 (m, 2H), 2.96 – 2.84 (m, 3H), 2.73 (d, J = 11.5 Hz, 1H), 2.55 (d, J = 10.8 Hz, 1H), 2.34 (s, 3H), 1.12 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 700.37 [M+H]+. EXAMPLE 371 Synthesis of N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}-2-[2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 371
Step 1: Preparation of benzyl N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1- yl}carbamate. Compound 371.1
To a stirred solution of 2,2-difluoro-3-methylbicyclo[1.1.1]pentane-1-carboxylic acid (63 mg, 0.4 mmol, 1.0 equiv.) and TEA (236 mg, 2.4 mmol) in toluene (1 mL) was added DPPA (128 mg, 0.5 mmol) dropwise at room temperature. The resulting mixture was stirred at 110 °C for 3 h under nitrogen atmosphere. The mixture was allowed to cool down to room temperature. To the above mixture was added BnOH (126 mg, 1.2 mmol) dropwise at room temperature. The resulting mixture was stirred at 90 °C for additional 16 h. The mixture was allowed to cool down to room temperature. To the above mixture was added TEA (236 mg, 2.4 mmol) and Ac2O (159 mg, 1.6 mmol) dropwise at room temperature. The resulting mixture was stirred at room temperature for additional 1 h. The reaction was quenched with sat. NaHCO3 (aq.) at room temperature. The resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford benzyl N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}carbamate (70 mg, crude) as a light brown oil. The compound lacks UV, staining, or LCMS signals. Due to the difficulty of purification, the crude product was used in the next step directly without further purification. Step 2: Preparation of 2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-amine. Compound 371.2
To a stirred solution of benzyl N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1- yl}carbamate (70 mg, 0.3 mmol) and concentrated HCl (14 mg, 0.6 mmol) in MeOH (2 mL) was added Pd/C (10%, 6 mg, 0.1 mmol) in one portion at room temperature. The resulting mixture was stirred at room temperature for 3 h under hydrogen atmosphere (50 atm) using a pressured reactor. After completion of reaction, the resulting mixture was filtered, the filter cake was washed with MeOH (3 x 1 mL). The filtrate was concentrated under reduced pressure to afford 2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-amine (5 mg, crude) as a colorless oil. The compound lacks UV, staining, or LCMS signals. Due to the difficulty of purification, the crude product was used in the next step directly without further purification. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}acetamide. Compound 371.3
To a stirred solution of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid, Intermediate S (20 mg, 0.03 mmol) and bis(2-oxo-1,3- oxazolidin-3-yl)phosphinoyl chloride (16 mg, 0.06 mmol) in DCM (1 mL) were added DIEA (42 mg, 0.3 mmol) and 2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-amine (5 mg, 0.04 mmol) dropwise at room temperature. The resulting mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 30% to 32% gradient in 3 min; detector, UV 254 nm) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}acetamide (3.7 mg, 15.2% yield) as a brown solid. LCMS observed m/z = 730.32 [M+H] +. Step 4: Preparation of N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}-2-[2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 371
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}acetamide (3.7 mg, 0.005 mmol) in DCM (0.5 mL) was added TFA (2.5 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for overnight. After completion of reaction, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm X 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 42% B in 7 min; Wave Length: 254 nm / 220 nm; RT1 (min): 6.52) to afford N-{2,2-difluoro-3-methylbicyclo[1.1.1]pentan-1-yl}-2-[2-(3,6-dihydro-2H- pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (1.4 mg, 40.3% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.22 (s, 1H), 9.30 (s, 1H), 8.58 (s, 1H), 6.82 (s, 1H), 4.94 (s, 2H), 4.52 (d, J = 12.3 Hz, 1H), 4.27 (d, J = 3.3 Hz, 2H), 3.82 (t, J = 5.6 Hz, 2H), 3.57 – 3.40 (m, 4H), 3.28 – 3.18 (m, 1H), 3.09 – 2.84 (m, 4H), 2.80 (d, J = 11.1 Hz, 1H), 2.62 (d, J = 11.0 Hz, 2H), 2.45 (s, 3H), 2.04 (s, 2H), 1.76 (t, J = 10.7 Hz, 2H), 1.21 (s, 2H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 640.40 [M+H] +. EXAMPLE 372 Synthesis of 2-[2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{6-fluorospiro[3.3]heptan-2-yl}acetamide Compound 372
To a stirred solution of [2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyri midine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetic acid (40 mg, 0.07 mmol, 1.0 equiv.), bis(2-oxo-1,3-oxazolidin-3- yl)phosphinoylchloride (29 mg, 0.1 mmol, 1.5 equiv.) and DIEA (20 mg, 0.1 mmol, 2.
Equiv.) in DCM (3 mL) was added 6-fluorospiro[3.3]heptan-2-amine (10 mg, 0.08 mmol, 1.0 equiv.) in portions at 25 °C. The mixture was heated to 45 °C and stirred at 45 °C for 16 h. After the reaction was completed, the reaction mixture was poured into water (5 mL) and extracted with DCM (3 x 10 mL). The combined organic phase was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: Column: X-select CSH C185 m, 19 mm X 250 mm; Mobile Phase A: Water (0.1%FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 33% B to 42%B in 10 min; Wave Length: 254 nm/220 nm RT1(min): 8.63 to give 2-[2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-{6-fluorospiro[3.3]heptan-2-yl}acetamide (3.5 mg, 90 % purity) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.22 (s, 1H), 8.68 (s, 1H), 6.95 (s, 1H), 6.54 (s, 1H), 5.62 (s, 1H), 4.86 – 4.79 (m, 4H), 4.40 (s, 2H), 4.29 (s, 1H), 3.94 (s, 2H), 3.83 (s, 2H), 3.54 (s, 1H), 3.12 (s, 3H), 2.83 (m, 2H), 2.70 (s, 2H), 2.56 – 2.39 (m, 3H), 2.28 – 2.12 (m, 3H), 2.10 – 2.08 (m, 3H), 2.01 – 1.95(m, 2H), 1.39 – 1.21 (m, 3H). LCMS observed m/z = 636.40 [M+H] +. EXAMPLE 373 Synthesis of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
Step 1: Preparation of tert-butyl 4-(4-(2-((5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)- 5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 373.1
To a stirred solution of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(3,6- dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid (100 mg, 0.2 mmol) and 5-chloro-7-(trifluoromethyl)-1,3-benzothiazol-4-amine (52 mg, 0.2 mmol) in THF (2 mL) were added pyridine (165 uL, 2.0 mmol) and 2,4,6-trichloro- 1,3,5-triazine (100 mg, 0.5 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 60 °C and stirred at 60 °C for 3 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C, quenched with H2O (2 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 4-(4- (2-((5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)-2-(3,6-dihydro- 2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (40 mg, 27% yield) as an off-white solid. LCMS observed m/z = 723.35 [M+H]+. Step 2: Preparation of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2- (2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 373.2
A solution of tert-butyl 4-(4-(2-((5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4- yl)amino)-2-oxoethyl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (40 mg, 0.04 mmol) in TFA (0.5 mL) was stirred for 30 min at 25 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to afford N-(5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (40 mg, crude) as a brown yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 623.15 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide. Compound 373.3
To a stirred solution of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (24 mg, 0.1 mmol) and pyridine (15 mg, 0.2 mmol) in DMF (1 mL) were added EDCI (18 mg, 0.1 mmol) and HOBT (13 mg, 0.1 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting mixture was stirred for 30 min at 25 °C under nitrogen atmosphere. To the above mixture was added N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(3,6-
dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (40 mg, 0.06 mmol) in one portion at 25 °C. The resulting mixture was stirred for additional 2 hours at 25 °C under nitrogen atmosphere. Then the reaction was quenched with H2O (5 mL) at 25 °C and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (5 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide (4 mg, 7% yield) as an off-white solid. LCMS observed m/z = 849.40 [M+H]+. Step 4: Preparation of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2- (3,6-dihydro-2H-pyran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 373
A stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(3,6-dihydro-2H-pyran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide (20 mg, 0.02 mmol) in TFA (1 mL) was heated to 80 °C and stirred at 80 °C for 30 min under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Mobile Phase A: 0.1% FA in water; Mobile Phase B: ACN; Flow rate: 60 mL/min) to afford N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(3,6-dihydro-2H-pyran-4-yl)-5- ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (2.5 mg, 12% yield) as an off-white solid.1H NMR (400 MHz, Methanol-d4) δ 9.47 (s, 1H), 8.59 (s, 1H), 8.04 (s, 1H), 7.07 – 7.02 (m, 1H), 5.46 (s, 2H), 4.72 (m, 1H), 4.37 (q, J = 2.8 Hz, 2H), 4.11 (m, 1H), 3.93 (t, J = 5.5 Hz, 2H), 3.75 (s, 2H), 3.44 (m, 1H), 3.21 (d, J = 7.8 Hz, 3H), 2.95 (m, 1H), 2.80 (m, 1H), 2.66 (s, 2H), 2.54 (s, 3H), 1.39 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 759.20 [M+H]+. EXAMPLE 374 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-7-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- [1,2,4]triazolo[1,5-a]pyridin-8-yl]acetamide Compound 374
Step 1: Preparation of 5-bromo-4-ethylpyridin-2-amine. Compound 374.1
A solution of 4-ethylpyridin-2-amine (25.0 g, 204.6 mmol), NBS (40.0 g, 225.0 mmol) and ammonium acetate (1577 mg, 20.4 mmol) in ACN (250 mL) was stirred at 25 °C for 3 hours under nitrogen atmosphere. The reaction was quenched by H2O (500 mL). The resulting mixture was extracted with EtOAc (3 x 500 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the mixture was concentrated in vacuo to afford (35.0 g, 85% yield) of 5-bromo-4-ethylpyridin-2-amine as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 201.01 [M+H]+.
Step 2: Preparation of 5-bromo-4-ethyl-3-iodopyridin-2-amine. Compound 374.2
A solution of 5-bromo-4-ethylpyridin-2-amine (25.0 g, 124.3 mmol), TFA (28.3 g, 248.6 mmol) and NIS (41.9 g, 186.5 mmol) in DMF (250 mL) was stirred at 55 °C for 4 hours under nitrogen atmosphere. The reaction was quenched by saturated Na2S2O4 solution (250 mL). The resulting mixture was extracted with EtOAc (3 x 300 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 25% EtOAc in petroleum ether) to afford (27.0 g, 66% yield) of 5-bromo-4-ethyl-3-iodopyridin-2-amine as a yellow solid. LCMS observed m/z = 328.91 [M+H]+. Step 3: Preparation of 1,2-diamino-5-bromo-4-ethyl-3-iodopyridin-1-ium 2,4,6- trimethylbenzenesulfonate. Compound 374.3
A solution of 5-bromo-4-ethyl-3-iodopyridin-2-amine (7.7 g, 23.5 mmol) and amino 2,4,6-trimethylbenzenesulfonate (10.1 g, 47.1 mmol) in DCM (50 mL) was stirred for 1 hour at 0 °C under nitrogen atmosphere. The reaction mixture was then transferred to 25 ℃ and stirred for another 16 hours under N2 atmosphere. After filtration, the filter cake was dried under reduced pressure to afford (8.0 g, 62% yield) of 1,2-diamino-5-bromo-4-ethyl- 3-iodopyridin-1-ium 2,4,6-trimethylbenzenesulfonate as an off-white solid. Step 4: Preparation of 3,6-dihydro-2H-pyran-4-carbaldehyde. Compound 374.4
To a solution of 3,6-dihydro-2H-pyran-4-carbonitrile (10.0 g, 91.6 mmol) in toluene (80 mL) at 0 °C under nitrogen atmosphere was added DIBAL-H (96.2 mL, 96.2 mmol, 1.0
M in toluene). After stirred for 1 hour, the reaction was quenched by 3M HCl (100 mL) and extracted by EtOAc (3 x 100 ml) and dried over anhydrous magnesium sulfate. The mixture was concentrated in vacuo to afford (8.0, 77% yield) of 3,6-dihydro-2H-pyran-4- carbaldehyde as a colorless oil. LCMS observed m/z = 113.05 [M+H]+. Step 5: Preparation of 6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-8-iodo- [1,2,4] triazolo[1,5-a] pyridine. Compound 374.5
To a mixture of 1,2-diamino-5-bromo-4-ethyl-3-iodopyridin-1-ium 2,4,6- trimethylbenzenesulfonate (8.0 g, 14.7 mmol) in DMF (75 mL) was added 3,6-dihydro-2H- pyran-4-carbaldehyde (4.9 g, 44.2 mmol) and Na2S2O5 (5.6 g, 29.5 mmol) at 25 ℃. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours under N2 atmosphere. The reaction was quenched by H2O (100 mL) and extracted by EtOAc (3 x 100 ml). The resulting mixture was washed with brine (3 x 100 ml) and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (2.5 g, 39% yield) of 6-bromo-2-(3,6-dihydro-2H- pyran-4-yl)-7-ethyl-8-iodo-[1,2,4]riazolo 1,5-a]pyridine as an off-white solid. LCMS observed m/z = 433.99 [M+H]+. Step 6: Preparation of 6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-8-[(E)-2- ethoxyethenyl]-7-ethyl-[1,2,4]triazolo[1,5-a]pyridine. Compound 374.6
A solution of 6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-8-iodo- [1,2,4]triazolo[1,5-a]pyridine (2.0 g, 4.6 mmol), 2-[(E)-2-ethoxyethenyl]-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (1.1 g, 5.5 mmol), Pd(dppf)Cl2CH2Cl2 (0.3 g, 0.4 mmol) and Na2CO3 (2.4 g, 23.0 mmol) in dioxane (30 mL) and H2O (15 mL) was stirred for 16 hours at 90 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent:
with 10% EtOAc in petroleum ether) to afford (1.2 g, 68% yield) of 6-bromo-2-(3,6- dihydro-2H-pyran-4-yl)-8-[(E)-2-ethoxyethenyl]-7-ethyl-[1,2,4]triazolo[1,5-a]pyridine as a white solid. LCMS observed m/z = 378.07 [M+H]+. Step 7: Preparation of 2-[6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl- [1,2,4]triazolo[1,5-a]pyridin-8-yl]acetaldehyde. Compound 374.7
A solution of 6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-8-[(E)-2-ethoxyethenyl]-7- ethyl-[1,2,4]triazolo[1,5-a]pyridine (1.0 g, 2.644 mmol) and HCl (6M) (10 mL, 329.1 mmol) in THF (10 mL) was stirred at 25 °C for 16 hours under nitrogen atmosphere. The mixture was concentrated in vacuo to afford (1.0 g, crude) 2-[6-bromo-2-(3,6-dihydro-2H- pyran-4-yl)-7-ethyl-[1,2,4]triazolo[1,5-a]pyridin-8-yl]acetaldehyde as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 350.07 [M+H]+. Step 8: Preparation of [6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl- [1,2,4]triazolo[1,5-a]pyridin-8-yl]acetic acid. Compound 374.8
A solution of 2-[6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl- [1,2,4]triazolo[1,5-a]pyridin-8-yl]acetaldehyde (1.0 g, 2.8 mmol), NaClO2 (3.8 g, 42.8 mmol), NaH2PO4 (5.1 g, 42.8 mmol) and 2-methyl-2-butene (4.0 g, 57.1 mmol) in 2-methyl- 2-propanol (10 mL) was stirred for 30 min at 25 °C under nitrogen atmosphere. The reaction was quenched by H2O (20 mL). The resulting mixture was extracted with EtOAc (3 x 20 ml). The combined organic layers were washed with brine (3 x 20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (500 mg, 47% yield) of [6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-
[1,2,4]triazolo[1,5-a]pyridin-8-yl]acetic acid as a white solid. LCMS observed m/z = 366.07 [M+H]+. Step 9: Preparation of 2-[6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl- [1,2,4]triazolo[1,5-a]pyridin-8-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 374.9
A solution of [6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-[1,2,4]triazolo[1,5- a]pyridin-8-yl]acetic acid (500 mg, 1.3 mmol), T3P (2.5 g, 3.9 mmol, 50% in EtOAc), TEA (5 mL, 35.9 mmol) and 2-chloro-4-(trifluoromethyl)aniline (0.53 g, 2.7 mmol) in EA (6 mL) was stirred at 25 °C for 2 hours under nitrogen atmosphere. The reaction was quenched by H2O (10 mL). The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (120 mg, 16% yield) of 2-[6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl- [1,2,4]triazolo[1,5-a]pyridin-8-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a white solid. LCMS observed m/z = 543.05 [M+H]+. Step 10: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-7-ethyl-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyridin-8-yl]acetamide. Compound 374.10
A solution of 2-[6-bromo-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl- [1,2,4]triazolo[1,5-a]pyridin-8-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (120 mg, 0.2 mmol), {1,3-bis[2,6-bis(pentan-3-yl)phenyl]-4,5-dichloro-2,3-dihydro-1H- imidazol-2-yl}dichloro(2-methyl-1lambda4-pyridin-1-yl)palladium (18 mg, 0.02 mmol), Cs2CO3 (143 mg, 0.4 mmol) and piperazine (57 mg, 0.6 mmol) in toluene (10 mL) was stirred at 100 °C for 16 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 60% EtOAc in petroleum ether) to afford (7 mg, 5% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyridin-8-yl]acetamide as a white solid. LCMS observed m/z = 549.20 [M+H]+. Step 11: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-[1,2,4]triazolo[1,5- a]pyridin-8-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 374.11
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran- 4-yl)-7-ethyl-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyridin-8-yl]acetamide (5 mg, 0.009 mmol), 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (2 mg, 0.01 mmol), HATU (3.4 mg, 0.009 mmol) and DIEA (5 mg, 0.04 mmol) in DMF (1 mL) was stirred at 25 °C for 16 hours under nitrogen atmosphere. The reaction was quenched by H2O (5 mL). The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (2 x 2 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo to afford (5 mg, crude) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-
[1,2,4]triazolo[1,5-a]pyridin-8-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 775.25 [M+H]+. Step 12: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro- 2H-pyran-4-yl)-7-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- [1,2,4]triazolo[1,5-a]pyridin-8-yl]acetamide. Compound 374
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,6-dihydro-2H-pyran-4-yl)-7-ethyl-[1,2,4]triazolo[1,5-a]pyridin-8-yl)-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide (5 mg, 0.006 mmol) in TFA (3 mL) was stirred at 25 °C for 6 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparatory HPLC (Column: XBridge Prep OBD C18 Column, 30*150 mm, 5μm; mobile phase: 22-52% MeCN in H2O) to afford (0.9 mg, 18% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,6-dihydro-2H-pyran-4- yl)-7-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- [1,2,4]triazolo[1,5-a]pyridin-8-yl]acetamide as a brown yellow solid.1H NMR (400 MHz, Methanol-d4) δ 8.58 (s, 1H), 8.29 (d, J = 8.5 Hz, 1H), 8.04 (s, 1H), 7.76 (s, 1H), 7.59 (d, J = 8.8 Hz, 1H), 7.00 (s, 1H), 4.35 (s, 4H), 3.95 (t, J = 5.4 Hz, 2H), 3.56 (s, 2H), 3.50 (s, 2H), 3.15 (d, 3H), 3.02 (d, 3H), 2.72 (s, 2H), 2.43 (s, 3H), 0.92 (s, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 685.15 [M+H]+. EXAMPLE 375 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-(difluoromethoxy)-2- (1,3-dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 375
Step 1: Preparation of diethyl 2-(4-(tert-butoxycarbonyl)piperazin-1-yl)malonate. Compound 375.1
To a stirred solution of tert-butyl piperazine-1-carboxylate (5.0 g, 26.8 mmol) and TEA (8.2 g, 80.4 mmol) in ACN (40 mL) was added diethyl 2-chloromalonate (5.2 g, 26.8 mmol) dropwise at room temperature. The resulting mixture was heated to 60 °C and stirred at 60 °C for 16 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent with 20% EtOAc in petroleum ether) to afford diethyl 2-(4-(tert-butoxycarbonyl)piperazin-1-yl)malonate (5.1 g, 55.3% yield) as a colorless oil. LCMS observed m/z = 354.19 [M+H] +. Step 2: Preparation of tert-butyl 4-(2-bromo-5-hydroxy-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 375.2
To a stirred solution of diethyl 2-(4-(tert-butoxycarbonyl)piperazin-1-yl)malonate (5.1 g, 14.8 mmol) and 3-bromo-1H-1,2,4-triazol-5-amine (2.9 g, 17.8 mmol) in DMF (40 mL) was added DBU (6.7 g, 44.4 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was heated to 120 °C and stirred at 120 °C for 16 h under nitrogen atmosphere. After completion of the reaction, the mixture was allowed to cool
down to room temperature and dissolved in water (50 mL). The resulting mixture was acidified to pH = 2 with HCl (1 M, 20 ml). And the solid was collected by filtration and washed with water (3 x 30 mL), then dried to afford tert-butyl 4-(2-bromo-5-hydroxy-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (2.5 g, 41.2% yield) as a grey white solid. LCMS observed m/z = 415.07 [M+H] +. Step 3: Preparation of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-hydroxy-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate.
To a stirred solution of tert-butyl 4-(2-bromo-5-hydroxy-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (2.5 g, 6.0 mmol) and N-(2- chloro-4-(trifluoromethyl)phenyl)-2-iodoacetamide (2.4 g, 6.6 mmol) in DMF (40 mL) was added DIEA (1.6 g, 12.0 mmol) dropwise at room temperature. The resulting mixture was heated to 45 °C and stirred at 45 °C for 2 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 50% gradient in 20 min; detector, UV 254 nm) to afford tert-butyl 4-(2-bromo-4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2- oxoethyl)-5-hydroxy-7-oxo-4,7-dihydro-[1,2,4]triazolo [1,5-a]pyrimidin-6-yl)piperazine- 1-carboxylate (1.8 g, 46.7% yield) as a white solid. LCMS observed m/z = 650.07 [M+H] +. Step 4: Preparation of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-(difluoromethoxy)-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 375.4
To a stirred solution of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-hydroxy-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (1.8 g, 2.8 mmol) in ACN (20 mL) was added Na2CO3 (440 mg, 4.2 mmol) in portions at room temperature. The resulting mixture was heated to 60 °C and stirred at 60 °C for 10 min under nitrogen atmosphere. To the above mixture was added TMSCF2Br (1.1 g, 5.6 mmol) dropwise at room temperature, and then heated to 60 °C and stirred at 60 °C for additional 2 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was dissolved in water (50 mL), extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent with 67% EtOAc in petroleum ether) to afford tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-(difluoromethoxy)-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (900 mg, 46.4% yield) as a colorless oil. LCMS observed m/z = 700.06 [M+H] +. Step 5: Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-(difluoromethoxy)-2-(1,3- dihydroisobenzofuran-5-yl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate. Compound 375.5
To a stirred solution of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-(difluoromethoxy)-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (900 mg, 1.3 mmol) and 2- (1,3-dihydroisobenzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (474 mg, 1.9 mmol) in dioxane (10 mL) and H2O (1 mL) were added Na2CO3 (272 mg, 2.6 mmol), P(t- Bu)3 Pd G3 (74 mg, 0.13 mmol) and P(t-Bu)3HBF4 (75 mg, 0.26 mmol) in one portion at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 16 h under nitrogen atmosphere. After completion of the reaction, the mixture was allowed to cool down to room temperature, dissolved in water (30 mL), and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent with 33% EtOAc in petroleum ether) to afford tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-(difluoromethoxy)-2-(1,3- dihydroisobenzofuran-5-yl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (200 mg, 21.3% yield) as a white solid. LCMS observed m/z = 740.19 [M+H] +. Step 6: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5- (difluoromethoxy)-2-(1,3-dihydroisobenzofuran-5-yl)-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 375.6
To a stirred solution of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-(difluoromethoxy)-2-(1,3- dihydroisobenzofuran-5-yl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (200 mg, 0.3 mmol) in DCM (5 mL) was added TFA (2.5 mL) dropwise at room temperature. The resulting mixture was stirred for 30 min at room temperature. After completion of the reaction, the resulting mixture was concentrated under reduced pressure to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-(difluoromethoxy)- 2-(1,3-dihydroisobenzofuran-5-yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (170 mg, crude) as a white solid. LCMS observed m/z = 640.14 [M+H] +. Step 7: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5- (difluoromethoxy)-2-(1,3-dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 375
To a stirred solution of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5- (difluoromethoxy)-2-(1,3-dihydroisobenzofuran-5-yl)-7-oxo-6-(piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (90 mg, 0.14 mmol) and 5-hydroxy-6- methylpyrimidine-4-carboxylic acid, Intermediate AB (24 mg, 0.16 mmol) in ACN (3 mL)
were added TCFH (43 mg, 0.16 mmol) and NMI (35 mg, 0.42 mmol) in one portion at room temperature. The resulting mixture was stirred for 30 min at room temperature. After completion of the reaction, the mixture was quenched with water (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 ExRs 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 36% B to 57% B in 7min; Wave Length: 254 nm / 220 nm; RT1 (min): 6.72) to afford N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(5-(difluoromethoxy)-2-(1,3-dihydroisobenzofuran-5-yl)-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (25.2 mg, 22.4% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.34 (s, 1H), 10.26 – 10.07 (m, 1H), 8.54 (s, 1H), 8.09 – 7.99 (m, 3H), 7.98 – 7.96 (m, 1H), 7.81 – 7.60 (m, 2H), 7.46 (d, J = 7.8 Hz, 1H), 5.10 (s, 2H), 5.06 (d, J = 4.3 Hz, 4H), 3.76 (s, 2H), 3.39 – 3.34 (m, 2H), 3.23 – 3.17 (m, 2H), 3.12 – 3.06 (m, 2H), 2.43 (s, 3H). LCMS observed m/z = 776.25 [M+H] +. EXAMPLE 376 Synthesis of 2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide Compound 376
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-(trifluoromethyl)bicyclo[4.2.0]octa- 1(6),2,4-trien-2-yl)acetamide.
Compound 376.1
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin- 1 -yl)-2-bromo-5-ethyl-7 -oxo-[ 1 , 2, 4]tri azolof 1 ,5-a]pyrimidin-4(7H)- yl)-N-(5-(trifluoromethyl)bicyclo[4.2.0]octa-l(6),2,4-trien-2-yl)acetamide (60 mg, 0.08 mmol) and 2-(l,3-dihydro-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (21 mg, 0.09 mmol) in dioxane (5 mL) and H2O (0.5 mL) were added K2CO3 (32 mg, 0.24 mmol) and Pd(dppf)C12 (6 mg, 0.01 mmol) in one portion at room temperature under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 8 h under nitrogen atmosphere. After completion of the reaction, the mixture was allowed to cool down to room temperature, dissolved in water (10 mL), and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with water (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure, purified by silica gel column chromatography (eluent with 8% MeOH in DCM) to afford 2- (6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-l-yl)-2-(l,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[l, 2, 4]tri azolof l,5-a]pyrimidin-4(7H)-yl)-N-(5- (trifluoromethyl)bicyclo[4.2.0]octa-l(6),2,4-trien-2-yl)acetamide (30 mg, 47.3% yield) as a yellow solid. LCMS observed m/z = 820.31 [M+H] +.
Step 2: Preparation of 2-(2-(l,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-l-yl)-7-oxo-[l, 2, 4]tri azolof 1,5- a]pyrimidin-4(7H)-yl)-N-(5-(trifluoromethyl)bicyclo [4.2.0]octa-l(6),2,4-trien-2- yl)acetamide.
Compound 376
Into a 40 mL vial were added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-(trifluoromethyl) bicyclo[4.2.0]octa- 1(6),2,4-trien-2-yl)acetamide (40 mg, 0.05 mmol) and TFA (5 mL) dropwise at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The crude product (40 mg) was purified by Prep- HPLC with the following conditions (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 45% B in 11.5 min; Wave Length: 254 nm / 220 nm; RT1 (min): 11.17) to afford 2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (11.4 mg, 30.2% yield) as a light yellow solid.1H NMR (400 MHz, Chloroform-d) δ 9.00 (s, 1H), 8.61 (s, 1H), 8.23 – 8.11 (m, 2H), 7.54 – 7.45 (m, 1H), 7.41 – 7.32 (m, 2H), 5.76 – 5.63 (m, 1H), 5.24 – 5.07 (m, 6H), 4.89 – 4.75 (m, 1H), 3.92 – 3.78 (m, 2H), 3.59 – 3.45 (m, 1H), 3.30 – 3.18 (m, 4H), 3.18 – 3.04 (m, 3H), 2.94 – 2.75 (m, 2H), 2.59 (s, 3H), 1.36 (t, J = 7.6 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 730.25 [M+H] +. EXAMPLE 377 Synthesis of 2-[2-(azetidin-1-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide Compound 377
Step 1: Preparation of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 377.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (100 mg, 0.1 mmol) in DCM (5 mL) was added boron trichloride (5 mL) in portions at room temperature. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. Then the resulting mixture was concentrated under reduced pressure. The residue was purified by reverse phase flash to afford 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy- 6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl}-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (55 mg, 62% yield) as a white solid. LCMS observed m/z = 689.12 [M+H]+. Step 2: Preparation of 2-[2-(azetidin-1-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compoun
To a stirred solution of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (50 mg, 0.1 mmol) and azetidine (5 mg, 0.1 mmol) in IPA (2 mL) was added TEA (22 mg, 0.2 mmol) in portions at room temperature. The reaction mixture was heated to 60 °C and stirred at 60 °C for 5 hours under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep- HPLC with the following conditions (Column: Xselect CSH C18, 5 m, 19 mm * 250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 51% B to 74% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 7.27) to afford 2-[2- (azetidin-1-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa- 1(6),2,4-trien-2-yl]acetamide (1.2 mg, 2% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.69 (s, 1H), 8.51 (s, 1H), 7.46 (m, 2H), 5.09 (s, 2H), 4.51 (m, 1H), 3.95 (m, 4H), 3.50 (m, 3H), 3.26 (s, 3H), 3.17 (s, 2H), 2.91 – 2.94 (m, 3H), 2.77 (m, 2H), 2.60 (m, 1H), 2.42 (s, 3H), 2.36 – 2.26 (m, 2H), 1.14 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 667.35 [M+H]+. EXAMPLE 378 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(2,3- dihydrobenzo[b][1,4]dioxin-5-yl)-5-ethyl-6-(4-(2-(methylsulfonamido)benzoyl)piperazin- 1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 378
The title compound was prepared using a similar procedure as Example 97, using Intermediate G.1H NMR (400 MHz, DMSO) δ 10.39 (s, 1H), 9.26 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.9 Hz, 1H), 7.46 (d, J = 3.8 Hz, 2H), 7.34 (qt, J = 7.6, 4.0 Hz, 3H), 7.01 (d, J = 8.0 Hz, 1H), 6.92 (t, J = 7.9 Hz, 1H), 5.37 (s, 2H), 4.53 (d, J = 12.9 Hz, 1H), 4.30 (s, 3H), 3.51 (s, 2H), 3.06 (s, 6H), 2.84 (s, 1H), 1.26 – 1.15 (m, 6H), 0.78 (s, 1H). LCMS observed m/z = 815.2 [M+H]+. EXAMPLE 379 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(2- (cyclopropanesulfonamido)benzoyl)piperazin-1-yl)-2-(2,3-dihydrobenzo[b][1,4]dioxin-5- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
The title compound was prepared using a similar procedure as Example 97, using Intermediate G.1H NMR (400 MHz, DMSO) δ 10.39 (s, 1H), 9.26 (s, 1H), 8.07 (d, J = 8.6 Hz, 1H), 7.97 (s, 1H), 7.72 (d, J = 8.9 Hz, 1H), 7.46 (d, J = 3.8 Hz, 2H), 7.34 (qt, J = 7.6, 4.0 Hz, 3H), 7.01 (d, J = 8.0 Hz, 1H), 6.92 (t, J = 7.9 Hz, 1H), 5.37 (s, 2H), 4.53 (d, J = 12.9 Hz, 1H), 4.30 (s, 3H), 3.51 (s, 2H), 3.06 (s, 6H), 2.84 (s, 1H), 1.26 – 1.15 (m, 6H), 0.78 (s,
1H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 841.2 [M+H]+. EXAMPLE 380 Synthesis of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide Compound 380
Step 1: Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2- yl)acetamide. Compound 380.1
To a solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide, Intermediate R (45 mg, 0.1 mmol) and 2-(benzo[d][1,3]dioxol-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (16 mg, 0.1 mmol) in 1,4-dioxane/H2O (1 mL/0.2 mL) were added KOAc (14 mg, 0.1 mmol) and Pd(dppf)Cl2 (4 mg, 0.01 mmol) at 25°C under N2 atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 1 hours. The resulting mixture was cooled down to
room temperature and concentrated under reduced pressure, collected the residue. The residue was purified by silica gel column chromatography, eluted with PE/EA (0/100) to afford 2-(2-(benzo[d][1,3]dioxol-5-yl)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3- chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide (40 mg, 84% yield) as a yellow solid. LCMS observed m/z = 856.45 [M+H]+. Step 2: Preparation of 2-(2-(benzo[d][1,3]dioxol-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 380
A mixture of 2-(2-(benzo[d] [1,3] dioxol-5-yl)-6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl) piperazin-1-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2- yl)acetamide (45 mg, 0.1 mmol) in TFA (2 mL) was heated to 80 ℃ and stirred at 80 ℃ for 20 min. The resulting mixture was quenched by addition of a solution of NaHCO3 in H2O (3 mL), extracted with DCM (2 x 2 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions Column: Xbridge BEH Shield RP185 μm, 30*150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min ; Gradient: 35% B to 50% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.85 to afford 2-(2-(benzo[d][1,3]dioxol- 5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide (8.3 mg, 20% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 10.21 (s, 1H), 8.59 (s, 1H), 7.72 (s, 1H), 7.68 (d, J = 8.2 Hz, 1H), 7.55 (d, J = 1.6 Hz, 1H), 7.06 (d, J = 8.2 Hz, 1H), 6.12 (s, 2H), 5.34 (m, 3H), 3.27 (s, 1H), 3.17 –
3.10 (m, 4H), 3.03 (d, J = 8.6 Hz, 3H), 2.89 – 2.81 (m, 1H), 2.71 – 2.63 (m, 1H), 2.46 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H).LCMS observed
EXAMPLE 381 Synthesis of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]- 2-[2-(2,3-dihydro-1-benzofuran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 381
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzofuran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 381.1
To a solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide, Intermediate R (45 mg, 0.1 mmol) and 2-(2,3-dihydrobenzofuran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (15 mg, 0.1 mmol) in 1,4-dioxane/H2O (1 mL/0.2 mL) were added K2CO3 (23 mg, 0.2 mmol) and Pd(dppf)Cl2 (4 mg, 0.01 mmol) at 25 °C under N2 atmosphere. The reaction
mixture was heated to 90 ℃ and stirred at 90 ℃ for 1 hours. The resulting mixture was cooled down to room temperature and concentrated under reduced pressure, collected the residue. The residue was purified by silica gel column chromatography, eluted with PE/EA (1/9) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (2,3-dihydrobenzofuran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide (30 mg, 63% yield) as a yellow solid. LCMS observed m/z = 854.25 [M+H]+. Step 2: Preparation of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl)-2-(2-(2,3-dihydrobenzofuran-6-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 381
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(2,3-dihydrobenzofuran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide (30 mg, 0.1 mmol) in TFA (2 mL) was heated to 80 ℃ and stirred at 80 ℃ for 20 min. The resulting mixture was quenched by addition of a solution of saturated NaHCO3 (3 mL) aqueous solution, extracted with DCM (2 x 2 mL). The combined the organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions Column: Xbridge BEH Shield RP185 μm, 30*150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 50% B in 8 min; Wave Length: 254 nm/220 nm; RT1 (min): 7.3 to afford N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]-2-[2-(2,3-dihydro-1-benzofuran-6- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (5.3 mg, 13% yield) as a white solid.1H NMR (400 MHz, 7 (s, 1H), 7.85 – 7.58 (m, 2H),
7.54 – 7.25 (m, 2H), 5.35 (s, 2H), 4.73 – 4.35 (m, 3H), 3.64 – 3.43 (m, 3H), 3.31 – 3.22 (m, 3H), 3.21 – 3.07 (m, 4H), 3.03 (q, J = 6.2, 5.3 Hz, 3H), 2.88 – 2.81 (m, 1H), 2.71 – 2.62 (m, 1H), 2.45 (s, 3H), 1.22 (q, J = 7.5, 7.0 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 764.20 [M+H]+. EXAMPLE 382 Synthesis of 2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl]-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N- [5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide Compound 382
Step 1: Preparation of tert-butyl 4-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7- oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl) piperazine-1-carboxylate. Compound 382.1
To a stirred solution of tert-butyl 4-(1-amino-4-ethyl-2-imino-6-oxo-3H-pyrimidin- 5-yl)piperazine-1-carboxylate (215 mg, 0.6 mmol) in EtOH (10 mL) was added EtONa (108 mg, 1.5 mmol) in portions at room temperature. The resulting mixture was stirred at 80 °C for 30 min under nitrogen atmosphere. The mixture was allowed to cool down to 25 °C and followed by the addition of ethyl 3-oxabicyclo[3.1.0]hexane-6-carboxylate (119 mg, 0.7 mmol) in portions at 25 °C. The resulting mixture was heated to 85 °C and stirred at 85 °C for 8 h. After the reaction was completed, the mixture was allowed to cool down to 25 °C, the resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromato h l t d ith DCM/M OH (12/1) t afford tert-butyl 4-(5-ethyl-2-
{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (100 mg, 36.3% yield) as a white solid. LCMS observed m/z = 431.23 [M+H] +. Step 2: Preparation of tert-butyl 4-[4-(2-ethoxy-2-oxoethyl)-5-ethyl-2-{3- oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate. Compound 382.2
To a stirred solution of tert-butyl 4-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7- oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (100 mg, 0.2 mmol) and ethyl bromoacetate (40 mg, 0.2 mmol) in DMF (5 mL) was added DIEA (75 mg, 0.5 mmol) dropwise at 25 °C. The resulting mixture was heated to 90 °C and stirred at 90 °C for 3 h. After the reaction was completed, the mixture was allowed to cool down to 25 °C. The resulting mixture was quenched with water (5 mL) at 25 °C and extracted with EtOAc (3 x 20 mL). The combined layers were washed with brine (20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford tert-butyl 4-[4-(2-ethoxy-2-oxoethyl)-5-ethyl-2-{3- oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate (80 mg, 66.4% yield) as a white solid. LCMS observed m/z = 517.27 [M+H] +. Step 3: Preparation of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl-2-{3- oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-4-yl}acetic acid. Compound 382.3
To a stirred solution of tert-butyl 4-[4-(2-ethoxy-2-oxoethyl)-5-ethyl-2-{3- oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate (61 mg, 0.1 mmol) in ACN (5 mL) was added LiOH.H2O aqueous solution (49 mg in 0.5 mL H2O, 1.1 mmol,). The resulting mixture was stirred at 25 °C for 2 h. After the reaction was completed, the resulting mixture was concentrated under vacuum. The mixture was acidified to pH 5 with HCl (aq.). The aqueous layer was extracted with EtOAc (3 x 20 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl-2- {3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (50 mg, 86.3% yield) as a white solid. LCMS observed m/z = 427.21 [M+H] +. Step 4: Preparation of tert-butyl 4-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7- oxo-4-({[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 382.4
To a stirred solution of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-5-ethyl-2-{3- oxabicyclo[3.1.0] hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (80 mg, 0.1 mmol) and 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-amine (36 mg, 0.2 mmol) in DCM (5 mL) were added DIEA (63 mg, 0.5 mmol) and 2,4,6-trichloro-1,3,5- triazine (60 mg, 0.3 mmol) in portions at 25 °C under nitrogen atmosphere. The resulting 812 of 1111
mixture was stirred at 25 °C for 8 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM / MeOH (10/1) to afford tert-butyl 4-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-4-({[5-(trifluoromethyl) bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)-[1,2,4]triazolo[1,5-a]pyrimidin- 6-yl)piperazine-1-carboxylate (60 mg, 55.3% yield) as a white solid. LCMS observed m/z = 658.29 [M+H] +. Step 5: Preparation of 2-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 382.5
To a stirred solution of tert-butyl 4-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7- oxo-4-({[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]carbamoyl}methyl)- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (60 mg, 0.09 mmol) in DCM (1 mL) was added TFA (1 mL) dropwise at room temperature. The resulting mixture was stirred at 25 °C for 2 h. After the reaction was completed, the resulting mixture was concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 2-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7- oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (45 mg, 88.4% yield) as a colorless oil. LCMS observed m/z = 558.24 [M+H] +. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4- trien-2-yl]acetamide. Compound 382.6
To a stirred mixture of 2-(5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (45 mg, 0.08 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid, Intermediate A (23 mg, 0.1 mmol) in DMF (5 mL) were added HATU (61 mg, 0.2 mmol) and DIEA (26 mg, 0.2 mmol) dropwise at 25°C. The resulting mixture was stirred at 25°C for 3 h. After the reaction was completed, the mixture was quenched with water (5 mL) at 25 °C and extracted with EtOAc (3 x 20 mL). The combined layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH (10/1) to afford 2- (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-5-ethyl-2-{3- oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5- (trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (20 mg, 31.2% yield) as a colorless oil. LCMS observed m/z = 415.21 [M+H] +. Step 7: Preparation of 2-{5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}-N-[5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 382
Into a 4 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide (20 mg, 0.02 mmol) and TFA (1 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 1 h. After the reaction was completed, the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH C18 5 μm, 30*150 mm; mobile phase A: water (10 mmol/L NH4HCO3), mobile phase B: ACN; flow rate: 60 mL/min; gradient: 34% B to 50% B in 7 min; wave length: 254 nm / 220 nm; RT1 (min): 6.55) to afford 2-{5-ethyl-6-[4-(5-hydroxy- 6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-2-{3-oxabicyclo[3.1.0]hexan-6-yl}-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]acetamide (2.3 mg, 12%.5 yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.85 (s, 1H), 8.64 (s, 1H), 8.60 (s, 1H), 7.41 – 7.34 (m, 2H), 5.60 (s, 1H), 4.91 (s, 2H), 4.77 (s, 1H), 4.05 (d, J = 9.0 Hz, 2H), 3.82 (d, J = 8.8 Hz, 4H), 3.53 (s, 1H), 3.30 (s, 4H), 3.11 (s, 2H), 2.92 (q, J = 7.5 Hz, 2H), 2.81 – 2.73 (m, 2H), 2.58 (s, 3H), 2.53 – 2.48 (m, 2H), 1.28 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 694.20 [M+H] +. EXAMPLE 383 Synthesis of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]- 2-[2-(2,3-dihydro-1-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 383
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-
[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 383.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (40 mg, 0.05 mmol) and 2-(2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (14 mg, 0.06 mmol) in dioxane (1 mL) and H2O (200 uL) were added K2CO3 (20 mg, 0.1 mmol) and Pd(dppf)Cl2 (4 mg, 0.005 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature, diluted with H2O (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 20% to 70% gradient in 15 min; detector, UV 254 nm. This resulted in 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1-benzofuran-5-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (20 mg, 47.2% yield) as a brown solid. LCMS observed m/z = 854.27 [M+H]+. Step 2: Preparation of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl)-2-(2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-
methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 383
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(2,3-dihydro-1-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (20 mg, 0.02 mmol) in TFA (2 mL) was heated to 80 °C and stirred for 20 min at 80 °C under air atmosphere. The resulting mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions:(Column: XBridge BEH C18, 5 μm, 30 * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 35% B to 70% B in 8 min; Wave Length: 254 nm/220 nm; RT1(min): 7.2). This resulted in N-[3- chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]-2-[2-(2,3-dihydro-1- benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (9.0 mg, 50.2% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 8.56 (s, 1H), 7.98 (d, J = 1.7 Hz, 1H), 7.88 (m, 1H), 7.71 (s, 1H), 6.88 (d, J = 8.4 Hz, 1H), 5.33 (s, 2H), 4.61 (t, J = 8.7 Hz, 2H), 4.53 (m, 1H), 4.29 (s, 1H),3.26 (m, J = 9.2 Hz,4H), 3.13 (m, J = 3.9 Hz, 5H), 3.00 (t, J = 8.5 Hz, 4H), 2.84 (m, 1H), 2.69 – 2.64 (m, 1H), 2.44 (s, 3H), 1.20 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 764.15 [M+H]+. EXAMPLE 384 Synthesis of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]- 2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide) Compound 384
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 384.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (40 mg, 0.05 mmol) and 2-(2,3-dihydro-1-benzofuran-5-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (14 mg, 0.06 mmol) in dioxane (2 mL) and H2O (400 uL) were added K2CO3 (20 mg, 0.1 mmol,) and Pd(dppf)Cl2 (4 mg, 0.005 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature, diluted with H2O (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column C18 silica gel; mobile phase MeCN in water (0.1% FA), 30% to 70%
gradient in 15 min; detector, UV 254 nm. This resulted in 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1-benzofuran-5-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (30 mg, 71.2% yield) as a brown solid. LCMS observed m/z = 854.27 [M+H]+. Step 2: Preparation of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 384
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (20 mg, 0.02 mmol) in TFA (2 mL) was heated to 80 °C and stirred for 20 min at 80 °C under air atmosphere. The resulting mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: (Column: XBridge BEH C18, 5 μm, 30 *150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 50% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.62). This resulted in N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]-2-[2-(1,3-dihydro-2-benzofuran-5- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide) (3.4 mg, 18.2% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.54 (s, 1H), 8.56 (s, 1H), 8.04 (d, J = 7.6 Hz, 2H), 7.71 (s, 1H), 7.46 (d, J = 8.0 Hz, 1H), 5.35 (s, 2H), 5.06 (d, J = 5.7 Hz, 4H), 4.54 (m, 1H), 3.27 (m, 4H), 3.17 – 3.09 (m, 5H), 3.02 (q, J = 7.5, 7.0 Hz, 3H), 2.85 (m, 1H), 2.67 (m, 1H), 2.44 (s, 3H), 1.21 (t, J = 7 M+H]+.
EXAMPLE 385 Synthesis of 2-[2-(2H-1,3-benzodioxol-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide) Compound 385
Step 1: Preparation of 2-(2-(benzo[d][1,3]dioxol-4-yl)-6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2- yl)acetamide. Compound 385.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (50 mg, 0.06 mmol) and 2H-1,3-benzodioxol-4-ylboronic acid (20 mg, 0.1 mmol) in dioxane (2 mL) and H2O (400 uL) were added K2CO3 (25 mg, 0.2 mmol) and Pd(dppf)Cl2 (4 mg, 0.006 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours under
nitrogen atmosphere. Then the resulting mixture was cooled to room temperature, diluted with H2O (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 20% to 70% gradient in 15 min; detector, UV 254 nm. This resulted in 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[3-chloro- 5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (26 mg, 49.2% yield) as a white solid. LCMS observed m/z = 856.25 [M+H]+. Step 2: Preparation of 2-(2-(benzo[d][1,3]dioxol-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 385
A solution of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2- yl]acetamide (20 mg, 0.02 mmol) in TFA (2 mL) was heated to 80 °C and stirred for 30 min at 80 °C under air atmosphere. The resulting mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: (Column: Xselect CSH C18, 5 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 ml/min; Gradient: 59% B to 82% B in 9 min; Wave Length: 254 nm/220 nm; RT1 (min): 5.3). This resulted in 2-[2-(2H-1,3- benzodioxol-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[3-chloro-5- (trifluoromethyl)b mide) (5.2 mg, 28.1% yield) as
a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.52 (s, 1H), 8.55 (s, 1H), 7.71 (s, 1H), 7.52 (dd, J = 8.1, 1.3 Hz, 1H), 7.06 (dd, J = 7.7, 1.3 Hz, 1H), 6.98 (d, J = 7.9 Hz, 1H), 6.17 (s, 2H), 5.35 (s, 2H), 4.56 (s, 1H), 3.53 (m, 4H), 3.14 (d, J = 6.2 Hz, 4H), 3.03 (d, J = 8.5 Hz, 4H), 2.85 (m, 1H), 2.67 (t, J = 2.0 Hz, 1H), 2.44 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 766.20 [M+H]+. EXAMPLE 386 Synthesis of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]- 2-[2-(1,3-dihydro-2-benzofuran-4-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 386
Step 1: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-4-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide. Compound 386.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-
[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (50 mg, 0.06 mmol) and 2-(1,3-dihydro-2-benzofuran-4-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (30 mg, 0.1 mmol) in dioxane (2 mL) and H2O (400 uL) were added K2CO3 (25 mg, 0.2 mmol) and Pd(dppf)Cl2 (4 mg, 0.006 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, diluted with H2O (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 20% to 70% gradient in 15 min; detector, UV 254 nm. This resulted in 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-4-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (20 mg, 38.1% yield) as a white solid. LCMS observed m/z = 854.27 [M+H]+. Step 2: Preparation of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl)-2-(2-(1,3-dihydroisobenzofuran-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 386
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-4-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (19 mg, 0.02 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 20 min under
air atmosphere. The resulting mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: (Column: Xselect CSH C18, 5 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 ml/min; Gradient: 40% B to 80% B in 10 min; Wave Length: 254 nm/220 nm; RT1 (min): 3.25; 7.28). This resulted in N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]-2-[2-(1,3-dihydro-2-benzofuran-4- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (1.8 mg, 10.1% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.58 (s, 1H), 8.42 (m, 1H), 8.05 (t, J = 4.6 Hz, 1H), 7.70 (s, 1H), 7.46 (d, J = 4.4 Hz, 2H), 5.36 (d, J = 5.2 Hz, 4H), 5.07 (s, 2H), 4.54 (m, 1H), 3.18 – 3.09 (m, 5H), 3.26 (d, J = 8.0 Hz, 4H), 3.07 – 2.98 (m, 3H), 2.85 (m, 1H), 2.67 (m, 1H), 2.41 (d, J = 3.0 Hz, 3H), 1.21 (t, J = 7.8 Hz, 3H). LCMS observed m/z = 764.25 [M+H]+. EXAMPLE 387 Synthesis of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]- 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 387
Step 1: 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 387.1
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (100 mg, 0.1 mmol) and potassium acetate (36 mg, 0.4 mmol) in DMF (500 uL) /DMSO (500 uL) were added dimethylamine hydrochloride (16 mg, 0.2 mmol) in portions at 25 °C. Then the resulting mixture was heated to 120 °C and stirred at 120 °C for 16 h under nitrogen atmosphere. The reaction was quenched with water (5 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with brine (2 x 5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was purified by reversed phase C18 silica gel column chromatography to afford (61 mg, 63% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2- (dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a brown solid. LCMS observed m/z = 779.27 [M+H]+. Step 2: N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 387
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (60 mg, 0.08 mmol) in TFA (1 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: SunFire C185 m, 19 mm x 250 mm; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 25 mL/min mL/min; Gradient: 55% B to 64%B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 7.88) to afford (5.8 mg, 10% yield) of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.45 (s, 1H), 10.23 (s, 1H), 8.58 (s, 1H), 7.70 (s, 1H), 5.18 (s, 2H), 4.51 (m, 1H), 3.48 (m, 3H), 3.25 – 3.11 (m, 5H), 2.96 (s, 9H), 2.78 (m, 1H), 2.60 (m, 1H), 2.45 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 689.20 [M+H]+. EXAMPLE 388 Synthesis of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]- 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 388
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa- 1(6),2,4-trien-2-yl]acetamide. Compound 388.1
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (50 mg, 0.1 mmol) in dioxane (3 mL) and H2O (0.3 mL) was treated with 2,3-dihydro-1,4- benzodioxin-5-ylboronic acid (13 mg, 0.1 mmol) and K2CO3 (25 mg, 0.2 mmol) at room temperature under nitrogen atmosphere, followed by the addition of Pd(PPh3)4 (7 mg, 0.01 mmol) in portions at room temperature. The resulting mixture was stirred for 16 h at 100 °C under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 10 mL). The organic layers were concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with (eluent: with 15% MeOH in DCM) to afford (17 mg, 32% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3- dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo-[124]triazolo[1,5-a]pyrimidin-4-yl)-N-[3-
chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide as a yellow solid. LCMS observed m/z = 870.29 [M+H]+. Step 2: Preparation of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]-2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide. Compound 388
2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3- dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[3- chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (14 mg, 0.1 mmol) was treated with TFA (3 mL) at room temperature. The resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 36% B to 50%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.7) to afford (1.9 mg, 15% yield) of N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl]-2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.50 (s, 1H), 7.70 (s, 1H), 7.36 (dd, J = 7.7, 1.7 Hz, 1H), 7.01 (dd, J = 8.1, 1.7 Hz, 1H), 6.92 (t, J = 7.8 Hz, 1H), 5.34 (s, 2H), 4.54 (m, 1H), 4.30 (s, 4H), 3.53 (m, 3H), 3.14 (d, J = 5.7 Hz, 5H), 3.01 (d, J = 8.6 Hz, 3H), 2.84 (m, 1H), 2.65 (s, 2H), 2.42 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 780.35 [M+H]+.
EXAMPLE 389 Synthesis of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)- 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (isochroman-5-yl)-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 389
Step 1: Preparation of 2-(isochroman-5-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane. Compound 389.1 To a stirred solution of 5-bromoisochromane (50 mg, 0.2 mmol) and 4,4,5,5- tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (72 mg, 0.3 mmol) in dioxane (5 mL) were added KOAc (46 mg, 0.5 mmol) and Pd(dppf)Cl2 (17 mg, 0.02 mmol) in one portion at room temperature under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. After completion of the reaction, the crude product was used in the next step directly without further purification. LCMS observed m/z = 261.16 [M+H] +. Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(isochroman-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4-trien-2- yl)acetamide. Compound 389.2
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide, Intermediate R (40 mg, 0.05 mmol) and 2-(3,4-dihydro-1H-2-benzopyran-8-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (19 mg, 0.08 mmol) in dioxane (5 mL) and H2O (0.5 mL) were added K2CO3 (20 mg, 0.15 mmol) and Pd(dppf)Cl2 (4 mg, 0.005 mmol) in one portion at room temperature. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. When the start materials were consumed, the mixture was allowed to cool down to room temperature, dissolved in water (10 mL), and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with water (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (eluent with 3% MeOH in DCM) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-2- (isochroman-5-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifleoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide (15 mg, 35.4% yield) as a brown oil. LCMS observed m/z = 868.29 [M+H] +. Step 3: Preparation of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (isochroman-5-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 389
Into a 4 mL vial were added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2- yl)acetamide)piperazin-1-yl)-5-ethyl-2-(isochroman-5-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4-trien-2- yl)acetamide (15 mg, 0.02 mmol) and TFA (1 mL) dropwise at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X-select CSH C185 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 60% B to 80% B in 9 min; Wave Length: 254 nm / 220 nm; RT1 (min): 5.23) to afford N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(isochroman-5-yl)-7-oxo-[1,2,4]triazolo [1,5-a]pyrimidin-4(7H)-yl)acetamide (2.7 mg, 19.3% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.80 (s, 1H), 8.92 (s, 1H), 8.62 (s, 1H), 8.04 (d, J = 7.7 Hz, 1H), 7.40 (s, 1H), 7.32 – 7.29 (m, 1H), 7.13 (d, J = 7.6 Hz, 1H), 5.69 (s, 1H), 5.17 (s, 2H), 4.87 (s, 3H), 4.00 (t, J = 5.8 Hz, 2H), 3.91 – 3.82 (m, 2H), 3.54 (s, 1H), 3.43 (t, J = 4.5 Hz, 2H), 3.37 (t, J = 5.6 Hz, 2H), 3.26 – 3.16 (m, 4H), 3.12 (s, 1H), 2.91 – 2.79 (m, 2H), 2.59 (s, 3H), 1.43 – 1.33 (m, 3H). LCMS observed m/z = 778.15 [M+H] +. EXAMPLE 390 Synthesis of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)- 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (isochroman-8-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
Compound 390
Step 1: Preparation of 2-(isochroman-8-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane. Compound 390.1
To a stirred solution of 8-bromo-3,4-dihydro-1H-2-benzopyran (50 mg, 0.2 mmol) and 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2- dioxaborolane (72 mg, 0.3 mmol) in dioxane (5 mL) were added KOAc (46 mg, 0.5 mmol) and Pd(dppf)Cl2 (17 mg, 0.02 mmol) in one portion at room temperature under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. After completion of the reaction, the crude product was used in the next step directly without further purification. LCMS observed m/z = 261.16 [M+H] +. Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(isochroman-8-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4-trien-2- yl)acetamide. Compound 390.2
To a stirred solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide, Intermediate R (40 mg, 0.05 mmol) and 2-(3,4-dihydro-1H-2-benzopyran-8-yl)-4,4,5,5- tetramethyl-1,3,2-dioxaborolane (19 mg, 0.1 mmol) in dioxane (5 mL) and H2O (0.5 mL) were added K2CO3 (20 mg, 0.1 mmol) and Pd(dppf)Cl2 (4 mg, 0.005 mmol) in one portion at room temperature. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. When the start materials were consumed, the mixture was allowed to cool down to room temperature, dissolved in water (10 mL), and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with water (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (eluent with 3% MeOH in DCM) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-2- (isochroman-8-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(3-chloro-5- (trifluoro methyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)acetamide (15 mg, 35.3% yield) as a brown oil. LCMS observed m/z = 868.29 [M+H] +. Step 3: Preparation of N-(3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4- trien-2-yl)-2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2- (isochroman-8-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 390
Into a 4 mL vial were added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(isochroman-8-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(3-chloro-5-(trifluoromethyl)bicyclo [4.2.0]octa-1(6),2,4-trien-2- yl)acetamide (15 mg, 0.02 mmol) in TFA (1 mL) dropwise at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The crude product (30 mg) was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 60% B to 80% B in 9 min; Wave Length: 254 nm / 220 nm; RT1 (min): 5.68) to afford N-(3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl)-2-(5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(isochroman-8-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (2.8 mg, 20.2% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.82 (s, 1H), 8.87 (s, 1H), 8.62 (s, 1H), 8.08 (d, J = 7.2 Hz, 1H), 7.39 (s, 1H), 7.33 – 7.29 (m, 1H), 7.27 – 7.25 (m, 1H), 5.69 (m, 1H), 5.27 (s, 2H), 5.16 (s, 2H), 4.84 (m, 1H), 4.01 (t, J = 5.7 Hz, 2H), 3.91 – 3.82 (m, 2H), 3.54 (s, 1H), 3.43 (t, J = 4.4 Hz, 2H), 3.20 (s, 4H), 3.11 (s, 1H), 2.99 (t, J = 5.7 Hz, 2H), 2.90 – 2.80 (m, 2H), 2.60 (s, 3H), 1.41 – 1.35 (m, 3H). LCMS observed m/z = 778.20 [M+H] +. EXAMPLE 391 Synthesis of 2-[2-(azetidin-1-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide Compound 391
Step 1: Preparation of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide. Compound 391.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide, Intermediate R (100 mg, 0.1 mmol) in DCM (2 mL) was added boron trichloride (1 mL) dropwise at 25 °C under nitrogen atmosphere. The mixture was stirred at 25 °C for 1 h. Then the resulting mixture was concentrated under reduced pressure to give 2-{2-bromo-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa- 1(6),2,4-trien-2-yl]acetamide (50 mg, 53% yield) as a yellow solid. LCMS observed m/z = 723.09 [M+H] +. Step 2: Preparation of 2-[2-(azetidin-1-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-chloro-5-(trifl h l)bi l [420] 1(6)24 rien-2-yl]acetamide.
Compound 391
To a mixture of 2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[3-chloro-5- (trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (40 mg, 0.05 mmol) and azetidine (12.6 mg, 0.2 mmol) in ACN (5 mL) was added Cs2CO3 (36 mg, 0.1 mmol) in one portion at 25°C under nitrogen atmosphere. The resulting mixture was heated to 50°C and stirred at 50°C for 4d under N2 atmosphere. Then the mixture was allowed to cool down to 25°C. The resulting mixture was filtered, the filter cake was washed with ACN (3 x 5 mL). The filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X-Bridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 27% B to 50% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 7.8) to afford 2-[2-(azetidin-1-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-[3-chloro-5-(trifluoromethyl)bicyclo[4.2.0]octa-1(6),2,4-trien-2-yl]acetamide (3.7 mg, 96.8% purity) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.04 (s, 1H), 9.02 (s, 1H), 8.63 (s, 1H), 7.46 (s, 1H), 5.63 (d, J = 13.0 Hz, 1H), 4.99 (s, 2H), 4.80 (d, J = 12.7 Hz, 1H), 4.17 (t, J = 7.5 Hz, 4H), 3.88 – 3.78 (m, 2H), 3.52 (s, 1H), 3.45 – 3.39 (m, 2H), 3.24 – 3.19 (m, 2H), 3.18 – 3.03 (m, 3H), 2.79 (m, 2H), 2.64 (d, J = 1.7 Hz, 3H), 2.44 (m, 2H), 1.32 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 701.20 [M+H] +. EXAMPLE 392 Synthesis of 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide Compound 392
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dime thylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{4-(trifluoromethyl)sulfanyl]phenyl}aceta mide. Compound 392.1
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (100 mg, 0.1 mmol) and dimethylamine (2 M in THF) (12 uL, 0.2 mmol) in DMSO (2.5 mL) and DMF (2.5 mL) was added KOAc (37 mg, 0.4 mmol) at 25 °C. The mixture was heated to 120 °C and stirred at 120 °C for 2 h under nitrogen atmosphere. Then the mixture was cooled down to 25 °C. The reaction was quenched by the addition of H2O (10 mL) at 25°C. The resulting mixture was extracted with EA (3 x 15 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with DCM/MeOH(10/1) to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{4-[(trifluoromethyl)sulfanyl]phenyl} acetamide (15 mg, 15% yield) as a white solid. LCMS observed m/z = 751.27 [M+H] +.
Step 2: Preparation of 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{4-(trifluoromethyl)sulfanyl]phenyl}aceta mide. Compound 392
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (11 mg, 0.02 mmol) in TFA (2 mL) was heated to 80°C and stirred at 80 °C for 30 min under nitrogen atmosphere. After the reaction was completed, the mixture was cooled down to 25 °C. The resulting mixture was concentrated under reduced pressure. The crude product (10 mg) was purified by Prep- HPLC with the following conditions (Column: X-Bridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 ml/min; Gradient: 27% B to 50% B in 10 min; Wave Length: 254 nm/220 nm; RT1(min): 7.8) to afford 2-[2-(dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (1.4 mg, 96.5 % purity) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.84 (s, 1H), 9.22 (s, 1H), 8.61 (s, 1H), 7.65 (d, J = 8.5 Hz, 2H), 7.60 – 7.52 (m, 2H), 5.65 (d, J = 13.1 Hz, 1H), 4.94 (s, 2H), 4.79 (d, J = 12.9 Hz, 1H), 3.88 – 3.75 (m, 2H), 3.50 (s, 1H), 3.17 (s, 9H), 2.86 – 2.70 (m, 2H), 2.59 (s, 3H), 1.32 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 661.25 [M+H] +. EXAMPLE 393 Synthesis of 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl)-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N- (4-((trifluoromethyl)thio) phenyl)acetamide Compound 393
Step 1: Preparation of 2-(isochroman-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane Compound 393.1
To a stirred mixture of 6-bromo-3,4-dihydro-1H-2-benzopyran (50 mg, 0.2 mmol), KOAc (70 mg, 0.7 mmol) and 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1,3,2-dioxaborolane) (71 mg, 0.3 mmol) in dioxane (5 mL) was added Pd(dppf)Cl2 (17 mg, 0.02 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 90 °C and stirred at 90 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 2-(3,4-dihydro-1H-2-benzopyran-6-yl)- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (50 mg, 81.2% yield) as a white solid. LCMS observed m/z = 216.16[M+H] + Step 2: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(4-((trifluoromethyl) thio)phenyl)acetamide Compound 393.2
To a stirred mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-
yl)-N-(4-((trifluoromethyl)thio)phenyl) acetamide (100 mg, 0.1 mmol), K2CO3 (53 mg, 0.4 mmol) and 2-(isochroman-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (33 mg, 0.1 mmol) in dioxane (10 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (18 mg, 0.03 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture heated to 100 °C and stirred at 100 °C for 3 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Cl2/MeOH 10/1) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-5-ethyl-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(4-((trifluoro methyl)thio)phenyl)acetamide (40 mg, 37% yield) as a yellow solid. LCMS observed m/z = 840.29[M+H] + Step 3: Preparation of 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-((trifluoromethyl)thio)phenyl)acetamide. Compound 393
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-5-ethyl-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(4- ((trifluoromethyl)thio) phenyl) acetamide (40 mg, 0.05 mmol) in TFA (3 mL) was heated to 90 °C and stirred at 80 °C for 20 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (40 mg) was purified by Prep- HPLC with the following conditions (Column: X-Bridge BEH C18 5 μm, 30*150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 33% B to 50% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.27) to afford 2-(5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1- yl)-2-(isochroman-6-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(4- ((trifluoromethyl)thio) phenyl)acetamide (1.6 mg,4%yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.91 (s, 1H), 8.51 (s, 1H), 7.88 (d, J = 7.0 Hz, 2H), 7.75 (d, J = 8.8 Hz, 2H), 7.70 (d, J = 8.7 Hz, 2H), 7.17 (d, J = 8.3 Hz, 1H), 5.27 (s, 2H), 4.73 (s, 2H), 4.54 (d, J
= 12.4 Hz, 1H), 3.90 (t, J = 5.7 Hz, 2H), 3.53 (d, J = 11.7 Hz, 3H), 3.31 – 3.27 (m, 1H), 3.01 (d, J = 11.2 Hz, 3H), 2.86 (d, J = 5.6 Hz, 3H), 2.51 – 2.50 (m, 1H), 2.43 (s, 3H), 1.20 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 750.20[M+H]+. EXAMPLE 394 Synthesis of 2-[2-(2H-1,3-benzodioxol-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide Compound 394
Step 1: Preparation of 2-(2-(benzo[d][1,3]dioxol-4-yl)-6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(4-((trifluoromethyl)thio)phenyl)acetamide. Compound 394.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- {4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (50 mg, 0.06 mmol), 2H-1,3-benzodioxol- 4-ylboronic acid (21 mg, 0.1 mmol), and K2CO3 (26 mg, 0.2 mmol) in dioxane (2 mL) and H2O (400 uL) was added Pd(dppf)Cl2 (5 mg, 0.006 mmol) in one portion at room temperature unde ure was heated to 80 ℃ and
stirred at 80 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (50 mL) and extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18, silica gel; mobile phase, MeCN in water (0.1% FA), 20% to 70% gradient in 15 min; detector, UV 254 nm. This resulted in 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (34 mg, 64.2% yield) as a brown solid. LCMS observed m/z = 828.25 [M+H]+. Step 2: Preparation of 2-(2-(benzo[d][1,3]dioxol-4-yl)-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(4-((trifluoromethyl)thio)phenyl)acetamide. Compound 394
A solution of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (30 mg, 0.04 mmol) in TFA (2 mL) was heated to 80 °C and stirred for 20 min at 80 °C under air atmosphere. Then the resulting mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: (Column: XBridge BEH C18, 5 μm, 30 * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 34% B to 50% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 5.93). This resulted in 2-[2-(2H-1,3-benzodioxol-4-yl)-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (3.3 mg, 11.2% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.92 – 10.87 (m, 1H), 8.55 (d, J = 1.9 Hz, 1H), 7.77 – 7.66 (m, 4H), 7.47 (dd, J = 8.1, 1.3 Hz, 1H), 7.04 (dd,
J = 7.8, 1.3 Hz, 1H), 6.94 (t, J = 7.9 Hz, 1H), 6.14 (s, 2H), 5.25 (s, 2H), 4.54 (m, 1H), 3.54 (m, 5H), 2.99 (d, J = 8.9 Hz, 3H), 2.85 (m, 1H), 2.67 (m, 1H), 2.44 (s, 3H), 1.23 (m, 1H), 1.19 (d, J = 7.5 Hz, 2H). LCMS observed m/z = 738.25 [M+H]+. EXAMPLE 395 Synthesis of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,4-dihydro- 1H-2-benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 395
Step 1: Preparation of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetic acid. Compound 395.1
A solution of ethyl 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetate (280 mg, 0.4 mmol) in EtOH (6 mL) and H2O (3 mL) was treated with NaOH (81 mg, 2.0 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The residue was acidified to pH 6 with HCl (aq.). The aqueous layer was extracted with CH2Cl2 (3 x 10 mL). The organic layers were concentrated under reduced pressure. The crude
product was purified by reversed phase C18 silica gel column chromatography to afford (60 mg, 23% yield) of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2- (3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)acetic acid as a white solid. LCMS observed m/z = 665.72 [M+H]+. Step 2: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 395.2
A solution of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl)acetic acid (55 mg, 0.1 mmol) in DCM (5 mL) was treated with DIEA (32 mg, 0.2 mmol) and bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (42 mg, 0.2 mmol) at room temperature under nitrogen atmosphere followed by the addition of 2-chloro-4- [(trifluoromethyl)sulfanyl]aniline (24 mg, 0.1 mmol) in portions at room temperature. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 10 mL). The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 60% EtOAc in petroleum ether) to afford (60 mg, 83% yield) 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}acetamide as a white solid. LCMS observed m/z = 874.34 [M+H]+.
Step 3: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,4- dihydro-1H-2-benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 395
2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(3,4- dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2- chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (60 mg, 0.1 mmol) was treated with TFA (4 mL) at room temperature. The resulting mixture was stirred for 16 at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The crude product was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 36% B to 55%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 7.05) to afford (9.3 mg, 17% yield) of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(3,4-dihydro-1H-2-benzopyran-6- yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.42 (s, 1H), 8.00 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 2.1 Hz, 1H), 7.90 (d, J = 4.6 Hz, 2H), 7.69 (dd, J = 8.7, 1H), 7.19 (d, J = 8.4 Hz, 1H), 5.38 (s, 2H), 4.74 (s, 2H), 4.54 (m, 1H), 3.91 (t, J = 5.7 Hz, 2H), 3.52 (m, 3H), 3.00 (m, 3H), 2.91 – 2.84 (m, 2H), 2.82 (s, 1H), 2.64 (s, 2H), 2.40 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 784.20 [M+H]+. EXAMPLE 396 Synthesis of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 396
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- {2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 396.1
To a stirred mixture of (6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)acetic acid (100 mg, 0.2 mmol) in DCM (1 mL) were added 2-chloro-4- [(trifluoromethyl) sulfanyl] aniline (59 mg, 0.3 mmol), bis(2-oxo-1,3-oxazolidin-3- yl)phosphinoyl chloride (88 mg, 0.3 mmol) and DIEA (67 mg, 0.5 mmol) in portions at 25 °C. Then the resulting mixture was stirred at rt for 16 h under nitrogen atmosphere. The reaction was quenched by the addition of water (3 mL) at room temperature. The resulting mixture was extracted with CH2Cl2 (3 x 3 mL). The resulting mixture was concentrated under reduced pressure. The crude product was purified by reversed phase C18 silica gel column chromatography to afford (61 mg, 42% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide as a white solid. LCMS observed m/z = 785.23 [M+H]+. Step 2: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triaz l [15 ] i idi 4 l] id
Compound 396
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(dimethylamino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (50 mg, 0.06 mmol) in TFA (1 mL) was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Shield RP18, 5μm, 19x250mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 33% B to 42%B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 9.07) to afford (14.2 mg, 31% yield) of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.56 (s, 1H), 8.00 – 7.92 (m, 2H), 7.70 (dd, J = 8.6, 2.1 Hz, 1H), 5.21 (s, 2H), 4.51 (m, 1H), 3.49 (m, 4H), 2.96 (s, 10H), 2.78 (m, 1H), 2.60 (m, 1H), 2.44 (s, 3H), 1.16 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 695.20 [M+H]+. EXAMPLE 397 Synthesis of N-p-chlorodifluoromethoxyphenyl[2-(2,3-dihydro-1-benzofuran-5-yl)- 6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo- 1,3,3a,7-tetraaza-7-indenyl]acetamide
The title compound was prepared using similar procedure as compound 11 afford N- p-chlorodifluoromethoxyphenyl[2-(2,3-dihydro-1-benzofuran-5-yl)-6-ethyl-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7- indenyl]acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.71 (s, 1H), 8.45 (s, 1H), 7.88 (s, 1H), 7.78 (d, J = 8.4 Hz, 1H), 7.63 (d, J = 8.7 Hz, 2H), 7.27 (d, J = 8.6 Hz, 2H), 6.80 (d, J = 8.3 Hz, 1H), 5.16 (s, 2H), 4.53 (t, J = 8.7 Hz, 2H), 4.46 (d, J = 12.7 Hz, 1H), 3.49 – 3.40 (m, 3H), 3.21 – 3.12 (m, 4H), 2.99 – 2.85 (m, 2H), 2.76 (d, J = 11.2 Hz, 1H), 2.59 (d, J = 11.0 Hz, 1H), 2.36 (s, 3H), 1.12 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 736.3 [M+H]+. EXAMPLE 398 Synthesis of N-p-chlorodifluoromethoxyphenyl(6-ethyl-5-{4-[(5-hydroxy-6- methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-2-(6-isochromanyl)-4-oxo-1,3,3a,7- tetraaza-7-indenyl)acetamide Compound 398
The title compound was prepared using similar procedure as compound 3 to afford N-p-chlorodifluoromethoxyphenyl(6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-2-(6-isochromanyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.75 (s, 1H), 8.43 (s, 1H), 7.81 (m, 2H), 7.63 (d, J = 8.8 Hz, 2H), 7.27 (d, J = 8.6 Hz, 2H), 7.10 (d, J = 8.3 Hz, 1H), 5.18 (s, 2H), 4.66 (s, 2H), 4.47 (d, J = 12.4 Hz, 1H), 3.83 (t, J = 5.7 Hz, 2H), 3.52 – 3.39 (m, 3H), 3.20 – 3.16 (m, 1H), 2.95 – 2.87 (m, 3H), 2.79 – 2.75 (m, 3H), 2.59 (d, J = 10.7 Hz, 1H), 2.36 (s, 3H), 1.12 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 750.3 [M+H]+. EXAMPLE 399 Synthesis of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(2,3-dihydro- 1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compoun
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 399.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- {2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (44 mg, 0.054 mmol) and 2,3- dihydro-1,4-benzodioxin-5-ylboronic acid (29 mg, 0.162 mmol) in dioxane (5 mL) were added K2CO3 (22 mg, 0.162 mmol) and H2O (1 mL). To the above mixture was added Pd(dppf)Cl2CH2Cl2 (8.75 mg, 0.011 mmol) in one portion under N2 atmosphere. The resulting mixture was stirred for additional 3 hours at 100 °C. The reaction was quenched with H2O (10 mL) at room temperature. The resulting mixture was extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 9% MeOH in DCM) to afford (41 mg, 87% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro- 1,4-benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4-
[(trifluoromethyl)sulfanyl]phenyl}acetamide as a light-brown solid. LCMS observed m/z = 876.22 [M+H]+. Step 2: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(2,3- dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 399
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (41 mg, 0.047 mmol) in TFA (3 mL) was stirred for 16 hours at 25 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparatory HPLC (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; mobile phase: 38-47% MeCN in H2O) to afford (3.9 mg, 10% yield) of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(2,3-dihydro-1,4- benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.51 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.93 (d, J = 2.2 Hz, 1H), 7.72 – 7.66 (m, 1H), 7.35 (dd, J = 7.5, 1.8 Hz, 1H), 7.00 (dd, J = 8.0, 1.7 Hz, 1H), 6.92 (t, J = 7.9 Hz, 1H), 5.36 (s, 2H), 4.54 (m, 1H), 4.29 (s, 4H), 3.53 (m, 3H), 3.32 (m, 1H) 3.02 (d, J = 8.9 Hz, 3H), 2.84 (m, 1H), 2.65 (s, 1H), 2.43 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 786.30 [M+H]+. EXAMPLE 400 Synthesis of 2-[2-(2H-1,3-benzodioxol-4-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide Compound 400
Step 1: Preparation of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 400.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- {2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (35 mg, 0.04 mmol) and 2H-1,3- benzodioxol-4-ylboronic acid (10 mg, 0.06 mmol) in dioxane (1 mL) were added Pd(dppf)Cl2CH2Cl2 (3 mg, 0.004 mmol), H2O (0.2 mL) and Cs2CO3 (41 mg, 0.1 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 4 h at 100 oC under nitrogen atmosphere. The reaction was quenched with water (50 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in CH2Cl2) to afford (60 mg, crude) of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)- 6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide as a white solid. LCMS observed m/z = 862.20 [M+H]+.
Step 2: Preparation of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 400
A solution of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide (30 mg, 0.03 mmol) in TFA (12 mL) was treated overnight at room temperature .The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions (Column: YMC Triart C18 ExRs5 m, 20 mm X 250 mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 40% B to 50%B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 7.92) to afford (3 mg, 12% yield) of 2-[2-(2H-1,3-benzodioxol-4-yl)-5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}acetamide as a white solid.1H NMR (400 MHz, DMSO- d6) δ 9.61 (s, 1H), 8.54 (s, 1H), 7.99 (d, J = 8.6 Hz, 1H), 7.93 (d, J = 2.1 Hz, 1H), 7.69 (dd, J = 8.6, 2.2 Hz, 1H), 7.51 (d, J = 8.1 Hz, 1H), 7.05 (d, J = 7.5 Hz, 1H), 6.96 (t, J = 7.9 Hz, 1H), 6.16 (s, 2H), 5.37 (s, 2H), 4.53 (m, 1H), 3.50 (m, 3H), 3.25 (s, 1H), 3.01 (d, J = 9.4 Hz, 3H), 2.84 (m, 1H), 2.66 (m, 1H), 2.43 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 772.35 [M+H]+. EXAMPLE 401 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(dimethylamino)-5-ethyl- 6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin -4-yl]-2,2-difluoroacetamide Compound 401
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 401.1
To a solution of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide (60 mg, 0.06 mmol) and dimethylamine (4 mg, 0.09 mmol) in DMSO (2.5 mL) and DMF (2.5 mL) was added KOAc (18 mg, 0.2 mmol) in portions. The reaction mixture was heated to 120 °C and stirred at 120 °C for 4 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, FA (0.1%) in ACN, 50% to 70% gradient in 10 min; detector, UV 254 nm, to afford 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyri midin-4-yl)-N- [2-chloro-4-(tri luoromethyl)phenyl]acetamide (10 mg, 19% yield) as a brown solid. LCMS observed m/z = 789.24 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydro xy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-2,2-difluoro acetamide.
Compound 401
A mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(dimethyl amino)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]-2,2-difluoroace tamide (8 mg, 0.01 mmol) in TFA (0.5 mL) was stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure. The crude product (13 mg) was purified by Prep-HPLC with the following conditions (Column: X select CSH C185 m, 30 mm X 150 mm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 50% B to 60%B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.17) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (dimethylamino)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin -4-yl]-2,2-difluoroacetamide (6.0 mg, 81% yield) as an off-white solid.1H NMR (400 MHz, Chloroform-d) δ 12.40 (s, 1H), 9.05 (s, 1H), 8.86 (s, 1H), 8.50 (d, J = 8.7 Hz, 1H), 7.67 (d, J = 2.0 Hz, 1H), 7.57 - 7.54 (m, 1H), 6.98 (t, 1H), 5.65 (d, J = 13.2 Hz, 1H), 5.04 (s, 2H), 4.81 (d, J = 12.7 Hz, 1H), 3.85 (t, J = 11.5 Hz, 2H), 3.53 (t, J = 12.4 Hz, 1H), 3.14 (s, 9H), 2.86 - 2.76 (m, 2H), 1.31 (q, J = 11.3, 9.3 Hz, 3H). LCMS observed m/z = 699.19 [M+H] +. EXAMPLE 402 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-1H-2- benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-] pyri midin-4-yl]-2,2-difluoroacetamide Compound 402
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl) phenyl]acetamide. Compound 402.1
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide (70 mg, 0.1 mmol) and 2-(3,4-dihydro-1H- 2-benzopyran-6-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (22 mg, 0.1 mmol), Na2CO3 (15 mg, 0.1 mmol) in dioxane (4 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (5 mg, 0.007 mmol) under nitrogen atmosphere. The mixture was heated to 80 °C and stirred at 80 °C for 2 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by Prep-TLC (PE/EA 0/1) to afford 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimi din-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (34 mg, 53% yield) as a light yellow solid. LCMS observed m/z = 878.25 [M+H] +. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro- 1H-2-benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-
carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimi din-4-yl]-2,2- difluoroacetamide. Compound 402
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(3,4-dihydro-1H-2-benzopyran-6-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl] -2,2-difluoroacetamide (32 mg, 0.04 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C. The resulting mixture was concentrated under reduced pressure. The crude product (35 mg) was purified by Prep- HPLC with the following conditions (Column: X Bridge BEH Shield RP185 m, 30 mm *150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 47% B in 10min; Wave Length: 254 nm/220 nm; RT1 (min): 8.43) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(3,4-dihydro-1H-2- benzopyran-6-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-] pyri midin-4-yl]-2,2-difluoroacetamide (5.3 mg, 19% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 12.40 (s, 1H), 9.17 (s, 1H), 8.87 (s, 1H), 8.53 (d, J = 8.7 Hz, 1H), 8.14 – 8.04 (m, 2H), 7.62 (d, J = 2.0 Hz, 1H), 7.59 – 7.52 (m, 1H), 7.13 (s, 1H), 7.02 – 6.96 (m, 1H), 5.71 (d, J = 13.2 Hz, 1H), 5.20 (s, 2H), 4.85 (s, 3H), 4.04 (t, J = 5.7 Hz, 2H), 3.89 (q, J = 12.4 Hz, 2H), 3.56 (t, J = 12.4 Hz, 1H), 3.25 (q, J = 7.5 Hz, 2H), 3.17 – 3.11 (m, 1H), 2.96 (t, J = 5.7 Hz, 2H), 2.92 – 2.79 (m, 2H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 788.15 [M+H] +. EXAMPLE 403 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(6-{4-[6-(difluoromethyl)- 5-hydroxypyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide Compound 403
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 403.1
To a mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]-2,2-difluoroacetamide (20 mg, 0.02 mmol), 2,3-dihydro-1,4- benzodioxin-5-ylboronic acid (4 mg, 0.02 mmol) and Cs2CO3 (24 mg, 0.07 mmol) in dioxane (3 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (2 mg, 0.002 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (20 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (PE/ EA = 5/1) to afford 2-(6-{4-[5-(benzyloxy)-6- (difluoromethyl)pyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-
(trifluoromethyl)phenyl]acetamide (5 mg, 22%) as a white solid. LCMS observed m/z = 880.23 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(6-{4-[6- (difluoromethyl)-5-hydroxypyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4- 5 benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide. Compound 403
A mixture of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (5 mg, 0.01 mmol) in TFA (2 mL) was stirred at room temperature for 2 hours. The resulting mixture was quenched with H2O (5 mL) and extracted with DCM (3 x 5 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by prep-TLC (PE/ EA = 5/1) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-(6-{4-[6- (difluoromethyl)-5-hydroxypyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4- benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)acetamide (1.8 mg, 40% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 12.40 (s, 1H), 9.27 (s, 1H), 8.87 (s, 1H), 8.49 (d, J = 8.7 Hz, 1H), 7.65 (dd, J = 7.7, 1.7 Hz, 1H), 7.61 (s, 1H), 7.58 – 7.51 (m, 1H), 7.14 – 6.84 (m, 3H), 5.71 (d, J = 13.2 Hz, 1H), 5.23 (s, 2H), 4.84 (d, J = 12.8 Hz, 1H), 4.37 (dd, J = 20.2, 5.0 Hz, 4H), 3.88 (t, J = 12.7 Hz, 2H), 3.55 (t, J = 12.5 Hz, 1H), 3.25 (d, J = 7.6 Hz, 2H), 3.13 (t, J = 12.2 Hz, 1H), 2.86 (dd, J = 31.2, 11.8 Hz, 2H), 1.39 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 790.15 [M+H]+. EXAMPLE 404 Synthesis of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[6-(difluoromethyl)-5- hydroxypyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide
Compound 404
Step 1: Preparation of 2-(2-(benzo[d][1,3]dioxol-4-yl)-6-(4-(5-(benzyloxy)-6- (difluoromethyl)pyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 404.1
To a stirred solution of 2-(6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N- [2-chloro-4-(trifluoromethyl)phenyl]acetamide (50 mg, 0.06 mmol) and 2-(2H-1,3- benzodioxol-4-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (30 mg, 0.1 mmol) in dioxane (2 mL) and H2O (0.4 mL) were added Pd(dppf)Cl2 (4 mg, 0.006 mmol) and K2CO3 (25 mg, 0.2 mmol) in one portion at room temperature under nitrogen atmosphere. The reaction mixture was heated to 90 ℃ and stirred at 90 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was cooled to room temperature, quenched with H2O (50 mL), and extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA),
20% to 70% gradient in 15 min; detector, UV 254 nm. This resulted in 2-[2-(2H-1,3- benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6-(difluoromethyl)pyrimidine-4- carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-[2-chloro- 4-(trifluoromethyl)phenyl]acetamide (20 mg, 38.2% yield) as a white solid. LCMS 5 observed m/z = 866.22 [M+H]+. Step 2: Preparation of 2-(2-(benzo[d][1,3]dioxol-4-yl)-6-(4-(6-(difluoromethyl)-5- hydroxypyrimidine-4-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 404
A solution of 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[5-(benzyloxy)-6- (difluoromethyl)pyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (20 mg, 0.02 mmol) in TFA (2 mL) was stirred for 30 min at 80 °C under air atmosphere. The resulting mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by Prep-HPLC with the following conditions: (Column: Xselect CSH C18, 5 m, 30 mm * 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 ml/min mL/min; Gradient: 59% B to 82% B in 9 min; Wave Length: 254 nm/220 nm; RT1 (min): 5.3). This resulted in 2-[2-(2H-1,3-benzodioxol-4-yl)-6-{4-[6-(difluoromethyl)-5- hydroxypyrimidine-4-carbonyl]piperazin-1-yl}-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (1.7 mg, 9.1% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 12.40 (s, 1H), 9.20 (s, 1H), 8.91(s, 1H), 8.53 (dd, J = 8.1, 1.3 Hz, 1H), 7.75 (dd, J = 7.7, 1.3 Hz, 1H), 7.65 (d, J = 7.9 Hz, 1H), 7.55 (d, 1H), 7.32 (s, 3H), 6.22 (s, 2H), 5.74 (m, 1H), 5.24 (s, 2H), 4.85 (d, J = 8.5 Hz, 1H), 3.89 (dd, J = 8.1, 1.3 Hz, 2H), 3.55 (m, 1H),3.30 (J = 7.9 Hz, 2H), 3.25 (m, 1H), 2.85 (m, 2H), 2.74 (s, 3H). LCMS observed m/z = 776.10 [M+H]+. EXAMPLE 405
Synthesis of N-(2-chloro-4-chlorodifluoromethoxyphenyl) [2-(2,3-dihydro-1- benzofuran-5-yl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide
The title compound was prepared using similar procedure as compound 11 to afford N-(2-chloro-4-chlorodifluoromethoxyphenyl) [2-(2,3-dihydro-1-benzofuran-5-yl)-6-ethyl- 5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl]acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.26 (s, 1H), 8.46 (s, 1H), 7.91 (s, 1H), 7.82 - 7.76 (m, 2H), 7.58 (s, 1H), 7.32 – 7.30 (m, 1H), 6.82 (d, J = 8.3 Hz, 1H), 5.25 (s, 2H), 4.54 (t, J = 8.7 Hz, 2H), 4.46 (d, J = 12.5 Hz, 1H), 3.47 - 3.40 (m, 4H), 3.21 - 3.16 (m, 2H), 2.97 - 2.89 (m, 3H), 2.76 (d, J = 11.3 Hz, 1H), 2.58 (d, J = 11.3 Hz, 1H), 2.36 (s, 3H), 1.14 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 770.3 [M+H]+. EXAMPLE 406 Synthesis of N-(2-chloro-4-chlorodifluoromethoxyphenyl) (6-ethyl-5-{4-[(5- hydroxy-6-methyl-4-pyrimidinyl)carbonyl]-1-piperazinyl}-2-(6-isochromanyl)-4-oxo- 1,3,3a,7-tetraaza-7-indenyl)acetamide
The title compound was prepared using similar procedure as compound 3 to afford N-(2-chloro-4-chlorodifluoromethoxyphenyl) (6-ethyl-5-{4-[(5-hydroxy-6-methyl-4- pyrimidinyl)carbonyl]-1-piperazinyl}-2-(6-isochromanyl)-4-oxo-1,3,3a,7-tetraaza-7- indenyl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.28 (s, 1H), 8.44 (s, 1H), 7.83 (d, J = 5.0 Hz, 2H), 7.77 (d, J = 9.0 Hz, 1H), 7.58 (s, 1H), 7.33-7.30 (m, 1H), 7.12
(d, J = 8.5 Hz, 1H), 5.27 (s, 2H), 4.67 (s, 2H), 4.46 (d, J = 12.4 Hz, 1H), 3.84 (t, J = 5.7 Hz, 2H), 3.49 – 3.39 (m, 2H), 3.20 - 3.15 (m, 2H), 3.0 - 2.88 (m, 3H), 2.84 – 2.73 (m, 3H), 2.58 (d, J = 11.3 Hz, 1H), 2.36 (s, 3H), 1.14 (t, J = 7.5 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 786.2 [M+H]+. 5 EXAMPLE 407 Synthesis of N-(2-chloro-4-chlorodifluoromethoxyphenyl) [2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1- piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide Compound 407
The title compound was prepared using similar procedure as compound 13 to afford N-(2-chloro-4-chlorodifluoromethoxyphenyl) [2-(1,3-dihydro-5-isobenzofuranyl)-6-ethyl- 5-{4-[(5-hydroxy-6-methyl-4-pyrimidinyl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7- tetraaza-7-indenyl] acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.27 (s, 1H), 8.45 (s, 1H), 7.98 (d, J = 7.8 Hz, 2H), 7.77 (d, J = 9.0 Hz, 1H), 7.59 (d, J = 2.7 Hz, 1H), 7.39 (d, J = 7.9 Hz, 1H), 7.33 - 7.30 (m, 1H), 5.27 (s, 2H), 4.99 (d, J = 6.2 Hz, 4H), 4.46 (d, J = 12.4 Hz, 1H), 3.49 – 3.40 (m, 2H), 3.19 - 3.16 (m, 2H), 2.95 - 2.88 (m, 3H), 2.76 (d, J = 11.2 Hz, 1H), 2.59 (d, J = 11.2 Hz, 1H), 2.36 (s, 3H), 1.15 (t, J = 7.5 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 770.2 [M+H]+. EXAMPLE 408 Synthesis
N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-6-(4-(2,4-dihydroxy-5-isopropylbenzoyl) piperazin-1-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 408
To a stirred solution of 2,4-dihydroxy-5-isopropylbenzoic acid (19 mg, 0.1 mmol), TCFH (35 mg, 0.1 mmol) and NMI (34 mg, 0.4 mmol) in ACN (1 mL) was added N-[2- chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (50 mg, 0.1 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 40 °C and stirred at 40 °C for 4 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, CAN in water (10 mmol/L NH4HCO3), 40% to 60% gradient in 20 minutes; detector, UV 254 nm) to give N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2- (1,3-dihydroisobenzofuran-5-yl)-6-(4-(2,4-dihydroxy-5-isopropylbenzoyl) piperazin-1-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (10.1 mg, 15.0% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.39 (s, 1H), 9.51 (d, J = 10.2 Hz, 2H), 8.10 – 8.01 (m, 3H), 7.98 (s, 1H), 7.72 (d, J = 8.9 Hz, 1H), 7.46 (d, J = 8.0 Hz, 1H), 6.90 (s, 1H), 6.37 (s, 1H), 5.40 (s, 2H), 5.06 (d, J = 5.1 Hz, 4H), 4.04 (s, 2H), 3.53 – 3.41 (m, 2H), 3.15 – 2.95 (m, 5H), 2.72 (d, J = 10.8 Hz, 2H), 1.26 – 1.18 (m, 3H), 1.14 (d, J = 6.9 Hz, 6H). LCMS observed m/z = 780.40 [M+H]+. EXAMPLE 409 Synthesis of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 409
Step 1: Preparation of tert-butyl 4-{4-[({2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}carbamoyl)methyl]-2-(1,3-dihydro-2-benzofuran-5-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxy late. Compound 409.1
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (100 mg, 0.2 mmol) and 2-chloro-4-[(trifluoromethyl)sulfanyl]aniline (43 mg, 0.2 mmol) in DMA (2 mL) were added bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (97 mg, 0.4 mmol) and DIEA (74 mg, 0.6 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was heated 40 °C and stirred at 40 °C overnight under nitrogen atmosphere. After the reaction was completed, the residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 10% to 80% gradient in 30 min; detector, UV 254 nm to afford tert-butyl 4-{4-[({2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}carbamoyl)methyl]-2-(1,3-dihydro-2-benzofuran-5-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxylate (70 mg, 50% yield) as a light yellow solid. LCMS observed m/z = 734.21 [M+H]+.
Step 2: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide.
of tert-butyl 4-{4-[({2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl} carbamoyl)methyl]-2-(1,3-dihydro-2-benzofuran-5-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl}piperazine-1-carboxy late (60 mg, 0.08 mmol) in DCM (1 mL) was added TFA (1 mL) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 30 min under nitrogen atmosphere. After the reaction was completed, the resulting mixture was concentrated under reduced pressure to give N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (60 mg, crude) as a yellow oil. LCMS observed m/z = 634.15 [M+H] +. Step 3: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4-[(trifluoro methyl)sulfanyl]phenyl} acetamide. Compound 409.3
To a stirred mixture of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(1,3- dihydro-2-benzo furan-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (70 mg, 0.1 mmol) and NMI (45 mg, 0.5 mmol) in ACN (2 mL) were added 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (32 mg, 0.1 mmol) and 5 TCFH (46 mg, 0.2 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 1 h under nitrogen atmosphere. After the reaction was completed, the resulting mixture was diluted with water (10 mL). The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (0-100%) to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl- 7-oxo-[1,2,4]tria zolo[1,5-a]pyrimidin-4-yl)-N-{2-chloro-4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (60 mg, 63% yield) as a brown oil. LCMS observed m/z = 860.23 [M+H]+. Step 4: Preparation of N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 409
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-{2-chloro-4-[(trifluoromethyl) sulfanyl]phenyl}acetamide (55 mg, 0.06 mmol) in TFA (2 mL) was stirred at 80 °C for 1 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (60 mg) was purified by Prep- HPLC with the following conditions (Column: X-Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B:
ACN; Flow rate: 60 mL/min; Gradient: 32% B to 55% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 7.05) to afford N-{2-chloro-4-[(trifluoromethyl)sulfanyl]phenyl}- 2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (26.7 mg, 5 53% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 9.91 (br, 2H), 8.57 (s, 1H), 8.10 – 7.98 (m, 3H), 7.94 (d, J = 2.0 Hz, 1H), 7.69 (dd, J = 8.4, 2.0 Hz, 1H), 7.46 (d, J = 8.0 Hz, 1H), 5.39 (s, 2H), 5.06 (d, J = 4.8 Hz, 4H), 4.54 (d, J = 12.4 Hz, 1H), 3.63 – 3.48 (m, 3H), 3.25 (s, 1H), 3.14 – 2.95 (m, 3H), 2.85 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 10.4 Hz, 1H), 2.45 (s, 3H), 1.22 (t, J = 7.6 Hz, 3H). LCMS observed m/z = 770.30 [M+H]+. EXAMPLE 410 Synthesis of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methyl pyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 410
Step 1: Preparation of tert-butyl 4-(4-(2-((2-chloro-4- ((trifluoromethyl)thio)phenyl)amino)-2-oxoethyl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl- 7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 410.1
To a stirred mixture of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic
acid (90 mg, 0.2 mmol), DIEA (66 mg, 0.5 mmol) and 2-chloro-4- ((trifluoromethyl)thio)aniline (39 mg, 0.2 mmol) in DCM (5 mL) was added Bop-Cl(87 mg, 0.3 mmol) in portions at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 3 h under nitrogen atmosphere. The resulting mixture was 5 concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (10 mmol/L NH4HCO3), 50% to 100% gradient in 10 min; detector, UV 254 nm. to give tert-butyl 4-(4-(2-((2-chloro-4-((trifluoromethyl)thio)phenyl)amino)-2- oxoethyl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]tria zolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (80 mg, 63% yield) as an off-white solid. LCMS observed m/z = 734.21 [M+H] +. Step 2: Preparation of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide. Compound 410.2
A mixture of tert-butyl 4-(4-(2-((2-chloro-4-((trifluoromethyl)thio)phenyl)amino)- 2-oxoethyl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (70 mg, 0.1 mmol) in TFA (1 mL) and DCM (3 mL) was stirred at room temperature for 1h. The resulting mixture was concentrated under vacuum to give N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (55 mg, 90% yield) as a yellow solid. LCMS observed m/z = 634.15 [M+H] + Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- ((trifluoromethyl)thio)phenyl)acetamide. Compound 410.3
To a stirred mixture of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(2,3- dihydroben zofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (50 mg, 0.08 mmol), TCFH (33 mg, 0.1 mmol) and 5- (benzyloxy)-6-methylpyrimidine-4-carboxylic acid (21 mg, 0.09 mmol) in ACN (3 mL) was added NMI (20 mg, 0.2 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under vacuum. The residue was purified by Prep- TLC (CH2Cl2 / MeOH 10/1) to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl) piperazin-1-yl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a] pyrimi din-4(7H) -yl)-N-(2-chloro-4-((trifluoromethyl)thio) phenyl)acetamide (60 mg, 88% yield) as a yellow solid. LCMS observed m/z = 860.23 [M+H] + Step 4: Preparation of N-(2-chloro-4-((trifluoromethyl)thio)phenyl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 410
A mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-2-(2,3-dihy drobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)-N-(2-chloro-4-((trifluo romethyl)thio) phenyl)acetamide (50 mg, 0.05 mmol) in TFA (2 mL) was stirred at 80 °C for 20 min. The resulting mixture was concentrated under vacuum.
The crude product (50 mg) was purified by Prep-HPLC with the following conditions (Column: X-select CSH C185 m, 30 mm X 150 mm; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 60% B to 70%B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 5.9) to afford N-(2-chloro-4-5 ((trifluoromethyl)thio)phenyl)-2-(2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5- hydroxy-6-methyl pyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (25.2 mg, 55% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.36 (s, 1H), 10.24 (s, 1H), 8.59 (s, 1H), 8.04 – 7.97 (m, 2H), 7.94 (d, J = 2.1 Hz, 1H), 7.88 (dd, J = 8.3, 1.9 Hz, 1H), 7.69 (dd, J = 8.6, 2.1 Hz, 1H), 6.88 (d, J = 8.4 Hz, 1H), 5.37 (s, 2H), 4.68 – 4.50 (m, 3H), 3.62 – 3.46 (m, 2H), 3.26 (t, J = 8.6 Hz, 3H), 3.03 – 2.98 (m, 3H), 2.84 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 12.9 Hz, 1H), 2.45 (s, 3H), 1.21 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 768.30 [M-H]-. EXAMPLE 411 Synthesis of 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]- N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide Compound 411
Step 1: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 411.1
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (100 mg, 0.1 mmol) in dioxane (2 mL) and H2O (0.2 mL) was treated with K2CO3 (53 mg, 0.4 mmol) and Pd(PPh3)4 (15 mg, 0.01 mmol) at room temperature under nitrogen atmosphere followed by the addition of 2,3-dihydro- 1,4-benzodioxin-5-ylboronic acid (28 mg, 0.2 mmol) in portions at room temperature. The resulting mixture was stirred for 16 h at 100 °C under nitrogen atmosphere. The reaction was quenched with water (15 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 20 mL). The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 70% EtOAc in petroleum ether) to afford (35 mg, 33% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide as a yellow solid. LCMS observed m/z = 842.87 [M+H]+. Step 2: Preparation of 2-[2-(2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]-N-{4-[(trifluoromethyl)sulfanyl]phenyl}acetamide. Compound 411
2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(2,3- dihydro-1,4-benzodioxin-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide (5 mg, 0.1 mmol) was treated with TFA (2 mL) at room temperature. The resulting mixture was stirred for 16 h at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min mL/min; Gradient: 35% B to 45%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.53) to afford (2.3 mg, 49% yield) of 2-[2- (2,3-dihydro-1,4-benzodioxin-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]-N-{4- [(trifluoromethyl)sulfanyl]phenyl}acetamide as a dark yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H), 8.55 (s, 1H), 7.77 – 7.66 (m, 4H), 7.30 (d, J = 7.8 Hz, 1H), 6.99 (dd, J = 8.3, 1.7 Hz, 1H), 6.90 (t, J = 7.9 Hz, 1H), 5.24 (s, 2H), 4.55 (m, 1H), 4.31 – 4.24 (m, 4H), 3.54 (m, 3H), 3.01 (s, 2H), 2.85 (m, 2H), 2.44 (s, 3H), 2.35 (s, 2H), 2.30 (s, 1H), 1.21 (m, 3H). LCMS observed m/z = 752.40 [M+H]+. EXAMPLE 412 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(6-(3,3- difluoroazetidin-1-yl)-5-hydroxypyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 412
Step 1: Preparation of 5-(benzyloxy)-4,6-dichloropyrimidine. Compound 412.1
To a stirred mixture of 4,6-dichloropyrimidin-5-ol (2.0 g, 12.1 mmol) and benzyl bromide (2.3 g, 13.3 mmol) in DMF (25 mL) was added K2CO3 (2.2 g, 15.7 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 3 h. The reaction was quenched with water at room temperature. The resulting mixture was extracted with EA (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to afford 5-(benzyloxy)-4,6-dichloropyrimidine (2.6 g, 84% yield) as a white solid. LCMS observed m/z = 255.00 [M+H]+. Step 2: Preparation of 5-(benzyloxy)-4-chloro-6-(3,3-difluoroazetidin-1- yl)pyrimidine. Compound 412.2
To a stirred mixture of 5-(benzyloxy)-4,6-dichloropyrimidine (500 mg, 1.9 mmol) and 3,3-difluoroazetidine (191 mg, 2.0 mmol) in i-PrOH (8 mL) was added TEA (595 mg, 5.8 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 50 °C for 3 h. The resulting mixture was concentrated under reduced pressure. 5 The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 5-(benzyloxy)-4-chloro-6-(3,3-difluoroazetidin-1-yl)pyrimidine (530 mg, 86% yield) as a yellow solid. LCMS observed m/z = 312.06 [M+H]+. Step 3: Preparation of methyl 5-(benzyloxy)-6-(3,3-difluoroazetidin-1- yl)pyrimidine-4-carboxylate. Compound 412.3
To a stirred solution of 5-(benzyloxy)-4-chloro-6-(3,3-difluoroazetidin-1- yl)pyrimidine (200 mg, 0.6 mmol) in MeOH (5 mL) was added Pd(dppf)Cl2 (47 mg, 0.06 mmol) and TEA (259 mg, 2.4 mmol) dropwise at room temperature. The resulting mixture was stirred at 100 °C for 24 h under CO (g) atmosphere (50 atm) in a pressure tank. After completion of the reaction, the mixture was cooled to room temperature and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford methyl 5-(benzyloxy)-6-(3,3- difluoroazetidin-1-yl)pyrimidine-4-carboxylate (62 mg, 28% yield) as a yellow solid. LCMS observed m/z = 336.11 [M+H]+. Step 4: Preparation of 5-(benzyloxy)-6-(3,3-difluoroazetidin-1-yl)pyrimidine-4- carboxylic acid. Compound 412.4
To a stirred mixture of methyl 5-(benzyloxy)-6-(3,3-difluoroazetidin-1- yl)pyrimidine-4-carboxylate (57 mg, 0.2 mmol) in MeOH (3 mL) was added a solution of NaOH (8 mg, 0.2 mmol) in H2O (1 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 1 h. The reaction was diluted with water (5 mL) 5 and acidified with HCl (2 M) to PH~6 at 0 °C. The resulting mixture was extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 5-(benzyloxy)-6-(3,3-difluoroazetidin-1-yl)pyrimidine-4-carboxylic acid (60 mg, crude) as a yellow solid. LCMS observed m/z =
. [M+H]+. Step 5: Preparation of 2-(6-(4-(5-(benzyloxy)-6-(3,3-difluoroazetidin-1- yl)pyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 412.5
To a stirred mixture of 5-(benzyloxy)-6-(3,3-difluoroazetidin-1-yl)pyrimidine-4- carboxylic acid (60 mg, 0.2 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (169 mg, 0.3 mmol, Intermediate) in ACN (3 mL) were added TCFH (57 mg, 0.2 mmol) and NMI (46 mg, 0.5 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. After completion of the reaction, the mixture was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with EA (3 x 15 mL). The combined organic layers were washed with brine (15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-(6-{4-[5-
(benzyloxy)-6-(3,3-difluoroazetidin-1-yl)pyrimidine-4-carbonyl]piperazin-1-yl}-2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-4-(trifluoromethyl)phenyl] acetamide (140 mg, crude) as a yellow solid. LCMS observed m/z = 905.26 [M+H]+. 5 Step 6: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(6-(3,3- difluoroazetidin-1-yl)-5-hydroxypyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 412
A solution of 2-(6-(4-(5-(benzyloxy)-6-(3,3-difluoroazetidin-1-yl)pyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (135 mg, 0.1 mmol) in TFA (3 mL) was stirred at room temperature for 1 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column: Xselect CSH C185 m, 30 mm X 150 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 48% B to 85% B in 8 min; Wave Length: 254 nm/220 nm; RT 1 (min): 7.4) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)- 2-(6-(4-(6-(3,3-difluoroazetidin-1-yl)-5-hydroxypyrimidine-4-carbonyl)piperazin-1-yl)-2- (1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (17.4 mg, 13% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 2H), 8.23 – 8.14 (m, 1H), 8.12 – 8.02 (m, 3H), 7.98 (d, J = 2.1 Hz, 1H), 7.75 – 7.73 (m, 1H), 7.46 (d, J = 7.9 Hz, 1H), 5.41 (s, 2H), 5.06 (d, J = 5.1 Hz, 4H), 4.65 – 4.51 (m, 5H), 3.94 – 3.81 (m, 1H), 3.55 – 3.52 (m, 2H), 3.25 (s, 1H), 3.08 – 2.93 (m, 3H), 2.85 – 2.82 (m, 1H), 2.73 – 2.62 (m, 1H), 1.24 – 1.21 (m, 3H). LCMS observed m/z = 815.25 [M+H]+. EXAMPLE 413
Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-6-[4-(2,4-dihydroxy-5-methylbenzoyl)piperazin-1-yl]-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 413
Step 1: Preparation of 2,4-dihydroxy-5-methylbenzoic acid. Compound 413.1
To a stirred solution of 1,3-benzenediol, 4-methyl- (50 mg, 0.4 mmol) in H2O (5 mL) were added KHCO3 (8 mg, 0.08 mmol) in one portion at room temperature under CO2 atmosphere. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 3 hours under CO2 atmosphere. Then the resulting mixture was cooled to room temperature, acidified to pH 6 with HCl (1 N), and then extracted with EtOAc (3 x30 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2,4- dihydroxy-5-methylbenzoic acid (60 mg, 88.2% yield) as a pink solid. LCMS observed m/z = 168.03 [M+H]+. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-6-(4-(2,4-dihydroxy-5-methylbenzoyl)piperazin-1-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 413
To a stirred solution N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (20 mg, 0.03 mmol) and 2,4-dihydroxy-5-methylbenzoic acid (6 mg, 0.03 mmol) in ACN (1 mL) were added TCFH (11 mg, 0.04 mmol) and NMI (5 mg, 0.07 mmol) in one portion at room temperature under air atmosphere. The reaction mixture was heated to 50 ℃ and stirred at 50 ℃ for 12 h under nitrogen atmosphere. Then the resulting mixture was cooled to room temperature, quenched with H2O (50 mL) and extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (100 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: YMC-Actus Triart C18, ExRS, 5 m, 30 mm x 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 58% B to 65% B in 8 min; Wave Length: 254 nm/220 nm; RT1 (min): 7.6) to afford N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-6-[4-(2,4-dihydroxy-5- methylbenzoyl)piperazin-1-yl]-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (3.2 mg, 13.1% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.10 – 8.01 (m, 3H), 7.92 (s, 1H), 7.67 (d, J = 8.6 Hz, 1H), 7.46 (d, J = 8.2 Hz, 1H), 6.88 (s, 1H), 6.37 (s, 1H), 5.35 (s, 2H), 5.06 (d, J = 5.3 Hz, 4H), 4.04 (s, 2H), 3.45(m, 4H)3.02 (d, J = 7.6 Hz, 5H), 2.71 (m, 2H), 2.02 (s, 3H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 752.20 [M+H]+. EXAMPLE 414 Synthesis of N-[2-chloro-6-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 414
Step 1: Preparation of 1-chloro-3-cyclopropoxy-2-nitro-5-(trifluoromethyl)benzene Compound 414.1
To a stirred solution of cyclopropanol (48 mg, 0.8 mmol) in THF (10 mL) was added NaH (60%, 35 mg, 0.9 mmol) in portions at 0 ℃ under N2 atmosphere, the resulting mixture was stirred for additional 0.5 hours at 0 ℃. Then the 1-chloro-3-fluoro-2-nitro-5- (trifluoromethyl) benzene (200 mg, 0.8 mmol) in THF (10 mL) was added to the mixture at 0 ℃, the resulting mixture was stirred for additional 1 hour at 60 ℃. Desired product could be detected by LCMS. The reaction was quenched with H2O (10 mL) at room temperature. The resulting mixture was extracted with EA (10 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 2% EtOAc in petroleum ether) to afford (140 mg, 60.5% yield) of 1-chloro-3- cyclopropoxy-2-nitro-5-(trifluoromethyl)benzene as a white solid. LCMS observed m/z = 282.01 [M+H]+. Step 2: Preparation of 2-chloro-6-cyclopropoxy-4-(trifluoromethyl)aniline Compound 414.2
To a stirred mixture of 1-chloro-3-cyclopropoxy-2-nitro-5-(trifluoromethyl)benzene (125 mg, 0.5 mmol) and tetrahydroxydiborane (120 mg, 1.3 mmol) in dimethylformamide (5 mL) was added bipyridine (1 mg, 0.1 mmol) in portions at 0 ℃ under air atmosphere. The resulting mixture was stirred for additional 5 min at 0 ℃.
Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (85 mg, 76.1% yield) of 2-chloro-6- cyclopropoxy-4-(trifluoromethyl)aniline as a yellow oil. LCMS observed m/z = 252.03 5 [M+H]+. Step 3: Preparation of tert-butyl 4-[4-({[2-chloro-6-cyclopropoxy-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate Compound 414.3
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (100 mg, 0.2 mmol) and DIEA (74 mg, 0.6 mmol) in DCM (3 mL) was added cyanuric chloride (42 mg, 0.2 mmol) and 2-chloro-6-cyclopropoxy-4-(trifluoromethyl)aniline (53 mg, 0.2 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 3 hours at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL). The resulting mixture was extracted with DCM (5 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 80% EtOAc in petroleum ether) to afford (62 mg, 42.9% yield) of tert-butyl 4-[4-({[2-chloro-6- cyclopropoxy-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate as a white solid. LCMS observed m/z = 758.26 [M+H]+. Step 4: Preparation of N-[2-chloro-6-cyclopropoxy-4-(trifluoromethyl)phenyl]-2- [2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide
A mixture of tert-butyl 4-[4-({[2-chloro-6-cyclopropoxy-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (60 mg, 0.1 mmol) and TFA (1 mL) in DCM (4 mL) was stirred for 15 min at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The crude product was used in the next step directly without further purification. LCMS observed m/z = 658.21 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-6-cyclopropoxy-4- (trifluoromethyl)phenyl]acetamide Compound 414.5
To a stirred mixture of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (20 mg, 0.1 mmol) and N-[2-chloro-6-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (55 mg, 0.1 mmol) in DMF (3 mL) were added HATU (38 mg, 0.1 mmol) and DIEA (54 mg, 0.4 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 1 hour at room temperature. Desired product could be detected by LCMS. The reaction was
quenched with H2O (10 mL), the resulting mixture was extracted with EA (10 mL x 4). The combined organic layers were washed with brine (10 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 20% EtOAc in5 petroleum ether) to afford (35 mg, 47.4% yield) of 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-6-cyclopropoxy-4- (trifluoromethyl)phenyl]acetamid as a yellow solid. LCMS observed m/z = 884.28 [M+H]+. Step 6: Preparation of N-[2-chloro-6-cyclopropoxy-4-(trifluoromethyl)phenyl]-2- [2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 414
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-6-cyclopropoxy-4-(trifluoromethyl)phenyl]acetamide (30 mg, 0.1 mmol) in TFA (3 mL) was stirred for 16 hours at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C185 m, 30 mm X 150 mm; Mobile Phase A: Water(10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 23% B to 35%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 5.1) to afford (12.4 mg, 44.6% yield) of N-[2-chloro-6-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.17 (s, 1H), 8.55 (s, 1H), 8.07 (d, J = 6.6 Hz, 2H), 7.57 (m, 2H), 7.48 (d, J = 8.2 Hz, 1H), 5.22 (s, 2H), 5.08 (d, J = 8.2 Hz, 3H), 4.53 (m, 1H), 4.12 (s, 1H), 3.52
(m, 3H), 3.01 (s, 3H), 2.83 (m, 1H), 2.70 – 2.61 (m, 2H), 2.43 (s, 2H), 2.35 – 2.31 (m, 1H), 1.24 (t, J = 7.4 Hz, 3H), 0.76 (d, J = 6.3 Hz, 2H), 0.59 (s, 2H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 794.35 [M+H]+. EXAMPLE 415 5 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-(3-methoxyazetidin-1- yl)pyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 415
Step 1: Preparation of 5-(benzyloxy)-4-chloro-6-(3-methoxyazetidin-1- yl)pyrimidine. Compound 415.1
To a stirred solution of 5-(benzyloxy)-4,6-dichloropyrimidine (500 mg, 2.0 mmol) and TEA (0.8 mL, 6.0 mmol) in 2-propanol (10 mL) was added 3-methoxyazetidine hydrochloride (291 mg, 2.4 mmol) in portions at 25°C, after that, the resulting mixture was stirred for 1 h at room temperature. After completion of reaction, the reaction was quenched by the addition of water (30 mL) at room temperature and extracted with EA (3 x 30 mL). The combined organic layers were washed with water (80 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10 / 1) to afford 5-
(benzyloxy)-4-chloro-6-(3-methoxyazetidin-1-yl)pyrimidine (540 mg, 90% yield) as a white solid. LCMS observed m/z = 306.15 [M+H]+. Step 2: Preparation of methyl methyl 5-(benzyloxy)-6-(3-methoxyazetidin-1- yl)pyrimidine-4-carboxylate. 5 Compound 415.2
To a stirred solution of 5-(benzyloxy)-4-chloro-6-(3-methoxyazetidin-1- yl)pyrimidine (200 mg, 0.7 mmol) and TEA (132 mg, 1.4 mmol) in MeOH (8 mL) and DMSO (4 mL) were added Pd(dppf)Cl2CH2Cl2 (53 mg, 0.07 mmol). The flask was evacuated and refilled with nitrogen three times and then evacuated and refilled with carbon monoxide two times. The resulting mixture was heated to 100 ℃ under an atmosphere of carbon monoxide for 84 h. The mixture was allowed to cool down to room temperature and was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 20% to 25% gradient in 5 min; detector, UV 254 nm. This resulted in methyl methyl 5-(benzyloxy)-6-(3-methoxyazetidin-1-yl)pyrimidine-4-carboxylate (26 mg, 12% yield) as a white solid. LCMS observed m/z = 330.15 [M+H]+. Step 3: Preparation of 5-(benzyloxy)-6-(3-methoxyazetidin-1-yl)pyrimidine-4- carboxylic acid. Compound 415.3
To a stirred solution of methyl 5-(benzyloxy)-6-(3-methoxyazetidin-1- yl)pyrimidine-4-carboxylate (20 mg, 0.06 mmol) in MeOH (5 mL) was added NaOH (5 mg, 0.12 mmol) in H2O (3 mL) dropwise at room temperature, after that, the resulting mixture was stirred for 30 min at room temperature. After completion of reaction, the mixture was
neutralized to pH 7 with HCl (aq.), and was extracted with EA (3 x 2 mL). The combined organic layers were washed with H2O (5 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 5-(benzyloxy)-6-(3- methoxyazetidin-1-yl)pyrimidine-4-carboxylic acid (19 mg, crude) as a white solid. LCMS 5 observed m/z = 316.05 [M+H]+. Step 4: Preparation of 2-(6-(4-(5-(benzyloxy)-6-(3-methoxyazetidin-1- yl)pyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 415.4
To a stirred solution of 5-(benzyloxy)-6-(3-methoxyazetidin-1-yl)pyrimidine-4- carboxylic acid (19 mg, 0.06 mmol) and NMI (15 mg, 0.18 mmol) in ACN (3 mL) were added TCFH (17 mg, 0.06 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (44 mg, 0.072 mmol) in portions at room temperature, after that, the resulting mixture was heated to 40 ℃ and stirred for overnight at 40 ℃. After completion of reaction, the resulting mixture was quenched with water (10 mL), and was extracted with EA (3 x 10 mL). The combined organic layers were washed with water (30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-(6-(4-(5-(benzyloxy)-6-(3-methoxyazetidin-1-yl)pyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (50 mg, crude) as a yellow oil. LCMS observed m/z = 899.50 [M+H]+.
Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-(3-methoxyazetidin-1- yl)pyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. 5 Compound 415
A solution of 2-(6-(4-(5-(benzyloxy)-6-(3-methoxyazetidin-1-yl)pyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (30 mg, 0.03 mmol) in trifluoroacetaldehyde (2 mL) was heated to 80 ℃ and stirred for overnight at 80 ℃. After completion of reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Shield RP18, 5μm, 19*250mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 41% B to 61%B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 6.5) to afford N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6- (3-methoxyazetidin-1-yl)pyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (5.1 mg, 19% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 10.01 (s, 1H), 8.16 – 8.00 (m, 4H), 7.98 (s, 1H), 7.72 (dd, J = 8.6, 2.2 Hz, 1H), 7.46 (d, J = 8.1 Hz, 1H), 5.40 (s, 2H), 5.12 – 4.99 (m, 4H), 4.58 – 4.48 (m, 1H), 4.46 – 4.38 (m, 2H), 4.33 – 4.24 (m, 2H), 4.03 – 3.99 (m, 2H), 3.55 – 3.51 (m, 3H), 3.26 – 3.24 (m, 3H), 3.08 – 2.97 (m, 3H), 2.83 (d, J = 10.8 Hz, 1H), 1.25 – 1.19 (m, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 809.45 [M+H]+. EXAMPLE 416
Synthesis of N-[2-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 416
Step 1: Preparation of 2-cyclopropoxy-1-nitro-4-(trifluoromethyl)benzene. Compound 416.1
A solution of cyclopropanol (138 mg, 2.4 mmol) in THF (5 mL) was treated with NaH (192 mg, 4.8 mmol, 60%) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 0.5 h at 0 °C under nitrogen atmosphere. To the above mixture was added 2- fluoro-1-nitro-4-(trifluoromethyl)benzene (500 mg, 2.4 mmol) dropwise at room temperature. The resulting mixture was stirred for additional 1 h at room temperature. The reaction was quenched with water (20 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 30 mL). The organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 8% EtOAc in petroleum ether) to afford (480 mg, 81% yield) of 2-cyclopropoxy-1-nitro-4- (trifluoromethyl)benzene as a yellow oil. LCMS observed m/z = 247.17 [M+H]+. Step 2: Preparation of 2-cyclopropoxy-4-(trifluoromethyl)aniline. Compound 416.2
A solution of 2-cyclopropoxy-1-nitro-4-(trifluoromethyl)benzene (400 mg, 1.6 mmol) in EA (5 mL) was treated with Pd/C (200 mg, 0.2 mmol, 10%) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at room temperature
under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with EtOAc (3 x 10 mL). The filtrate was concentrated under reduced pressure. This resulted in (290 mg, 82% yield) of 2-cyclopropoxy-4-(trifluoromethyl)aniline as a yellow oil. LCMS observed m/z = 217.19 [M+H]+. 5 Step 3: Preparation of tert-butyl 4-[4-({[2-cyclopropoxy-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate. Compound 416.3
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (100 mg, 0.2 mmol) and DIEA (74 mg, 0.6 mmol) in DCM (3 mL) was added cyanuric chloride (42 mg, 0.2 mmol) and 2-chloro-6-cyclopropoxy-4-(trifluoromethyl)aniline (53 mg, 0.2 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 3 hours at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL). The resulting mixture was extracted with DCM (5 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 80% EtOAc in petroleum ether) to afford (62 mg, 42.9% yield) of tert-butyl 4-[4-({[2-chloro-6- cyclopropoxy-4-(trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1- carboxylate as a white solid. LCMS observed m/z = 758.26 [M+H]+. Step 4: Preparation of N-[2-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 416.4
tert-butyl 4-[4-({[2-cyclopropoxy-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (100 mg, 0.138 mmol) in DCM (0.5 mL) was treated with TFA (2 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 30 min at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 624.64 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-cyclopropoxy-4- (trifluoromethyl)phenyl]acetamide. Compound 416.5
A solution of N-[2-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (130 mg, 0.2 mmol) in DMF (2 mL) was treated with HATU (118 mg, 0.3 mmol) and DIEA (135 mg, 1.0 mmol) at room temperature under nitrogen atmosphere followed by the addition of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (61 mg, 0.3 mmol) in portions at room temperature. The resulting mixture was stirred for 1 h at room temperature under nitrogen atmosphere. The reaction was quenched with water (10 mL) at
room temperature. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 850.32 5 [M+H]+. Step 6: Preparation of N-[2-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 416
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-cyclopropoxy-4-(trifluoromethyl)phenyl]acetamide (130 mg, 0.32mmol) in DCM (1 mL) was treated with TFA (10 mL) at room temperature. The resulting mixture was stirred for 16 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 33% B to 47%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.53) to afford (27.8 mg, 24% yield) of N-[2-cyclopropoxy-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5- ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 9.98 (s, 2H), 8.58 (d, J = 1.4 Hz, 1H), 8.16 (d, J = 8.5 Hz, 1H), 8.02 (d, J = 6.1 Hz, 2H), 7.60 (s, 1H), 7.45 (d, J = 8.2 Hz, 1H), 7.30 (d, J = 8.2 Hz, 1H), 5.38 (s, 2H), 5.05 (d, J = 5.0 Hz, 3H), 4.54 (m, 1H), 4.17 – 4.11 (m, 1H), 3.53 (m, 3H), 3.26 (m, 2H), 3.06 – 2.95 (m, 3H), 2.84 (m, 1H), 2.67 (m, 1H), 2.45 (d, J = 1.4 Hz, 3H), 1.19 (t, J = 7.3 Hz, 3H), 0.91 (m, 2H), 0.83 (d, J = 3.2 Hz, 2H). m/z = 760.30 [M+H]+.
EXAMPLE 417 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide 5 Compound 417
Step 1: Preparation of 5H,6H,7H-1lambda5-cyclopenta[b]pyridin-1-one Compound 417.1
To a stirred mixture of 5H,6H,7H-cyclopenta[b]pyridine (1.5 g, 12.6 mmol) in DCM (20 mL) was added m-CPBA (2.3 g, 13.5 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 2 hours at room temperature. The reaction was quenched with 1M aq. NaOH (30 mL). The resulting mixture was extracted with DCM (30 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 136.07 [M+H]+. Step 2: Preparation of 5H,6H,7H-cyclopenta[b]pyridine-2-carbonitrile Compound 417.2
To a stirred mixture of 5H,6H,7H-1lambda5-cyclopenta[b]pyridin-1-one (1.0 g, 7.0 mmol) and trimethylsilyl cyanide (2.1 g, 21.1 mmol) in ethylene dichloride (12 mL) was added diethylcarbamoyl-chloride (1.9 g, 14.0 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 16
hours at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (20 mL) at room temperature. The resulting mixture was extracted with DCM (20 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was 5 purified by silica gel column chromatography (eluent: with 80% EtOAc in petroleum ether) to afford (350 mg, 34.5% yield) of 5H,6H,7H-cyclopenta[b]pyridine-2-carbonitrile as a white solid. LCMS observed m/z = 145.07 [M+H]+. Step 3: Preparation of 5H,6H,7H-cyclopenta[b]pyridine-2-carboxylic acid Compound 417.3
To a stirred mixture of 5H,6H,7H-cyclopenta[b]pyridine-2-carbonitrile (200 mg, 1.4 mmol) in EtOH (2 mL) were added NaOH (277 mg, 6.9 mmol) and H2O (1 mL) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 2 hours at 85 ℃. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was acidize to pH 6 with HCl and purified by reversed phase C18 silica gel column chromatography to afford (180 mg, 79.5% yield) of 5H,6H,7H-cyclopenta[b]pyridine-2-carboxylic acid as a yellow oil. LCMS observed m/z = 164.06 [M+H]+. Step 4: Preparation of N, N-diethyl-5H,6H,7H-cyclopenta[b]pyridine-2- carboxamide Compound 417.4
To a stirred solution of 5H,7H-furo[3,4-b]pyridine-2-carboxylic acid (430 mg, 2.6 mmol) and diethylamine (190 mg, 2.6 mmol) in DCM (1 mL) were added NMI (641 mg, 7.8 mmol) and TCFH (1.5 g, 5.2 mmol) in portions at 25 ℃. Then the resulting mixture was stirred at 25 ℃ for 3 hours. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (150 mg,
69.7% yield) of N, N-diethyl-5H,7H-furo[3,4-b]pyridine-2-carboxamide as a white solid. LCMS observed m/z = 219.14 [M+H]+. Step 5: Preparation of N, N-diethyl-3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2- carboxamide 5 Compound 417.5
A mixture of N, N-diethyl-5H,6H,7H-cyclopenta[b]pyridine-2-carboxamide (60 mg, 0.3 mmol) and triisopropyl borate (155 mg, 0.8 mmol) in THF (1 mL) was added LDA (207 uL, 0.4 mmol, 2.0 M in THF) at 0 ℃, then the mixture was stirred for 10 min at 0 ℃ under N2 atmosphere. The H2O2 (128 uL, 1.7 mmol, 30%) was added to the mixture. The resulting mixture was stirred for additional 1 hour at room temperature. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure, purified by silica gel column chromatography (eluent: with 40% EtOAc in petroleum ether) to afford (16 mg, 24.9% yield) of N, N-diethyl-3-hydroxy-5H,6H,7H- cyclopenta[b]pyridine-2-carboxamide as a yellow solid. LCMS observed m/z = 235.14 [M+H]+. Step 6: Preparation of 3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2-carboxylic acid Compound 417.6
A solution of N, N-diethyl-3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2- carboxamide (10 mg, 0.1 mmol) in con. HCl (1 mL) was stirred for 16 hours at 110 ℃ under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 180.06 [M+H]+. Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Compound 417
To a stirred solution of 3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2-carboxylic acid (7 mg, 0.1 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (15 mg, 0.1 mmol) in DMF (0.5 mL) were added HATU (11 mg, 0.1 mmol) and DIEA (16 mg, 0.1 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 1 hour at room temperature. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C185 μm, 30*150mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 50% B to 80% B in 8 min; Wave Length: 254nm/ 220nm nm; RT1(min): 6.75) to afford (2.3 mg, 11.9% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5- yl)-5-ethyl-6-(4-{3-hydroxy-5H,6H,7H-cyclopenta[b]pyridine-2-carbonyl}piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.20 – 10.05 (s, 1H), 8.09 – 8.01 (m, 3H), 7.95 (s, 1H), 7.69 (d, J = 8.7 Hz, 1H), 7.45 (d, J = 7.9 Hz, 1H), 7.13 (s, 1H), 5.37 (s, 2H), 5.06 (d, J = 5.0 Hz, 4H), 4.55 (m, 1H), 3.59 (m, 1H), 3.49 (s, 2H), 3.24 (s, 1H), 3.07 – 2.95 (m, 3H), 2.83 (m, 5H), 2.70 – 2.65 (s, 1H), 2.05 (d, J = 7.5 Hz, 2H), 1.21 (t, J = 7.6 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 763.45 [M+H]+. EXAMPLE 418 Synthesis of N-[2-chloro-5-cyclopropyl-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 418
Step 1: Preparation of 1-bromo-4-chloro-5-nitro-2-(trifluoromethyl)benzene. Compound 418.1
A solution of 4-bromo-2-nitro-5-(trifluoromethyl)aniline (2.5 g, 8.7 mmol) in conc. HCl (15 mL) was treated with NaNO2 (0.7 g, 9.6 mmol) for 1 h at 0 °C under nitrogen atmosphere followed by the addition of CuCl (1.3 g, 13.1 mmol) at 0 °C. The resulting mixture was stirred for 30 min at 0 °C. The mixture was neutralized to pH 7 with saturated Na2CO3 (aq.). The aqueous layer was extracted with EtOAc (3 x 100 mL). The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 90% EtOAc in petroleum ether) to afford (1.3 g, 48% yield) of 1-bromo-4-chloro-5-nitro-2-(trifluoromethyl)benzene as a pink oil. Step 2: Preparation of 5-bromo-2-chloro-4-(trifluoromethyl)aniline. Compound 418.2
To a stirred solution of 1-bromo-4-chloro-5-nitro-2-(trifluoromethyl)benzene (2.1 g, 6.8 mmol) and tetrahydroxydiborane (2.5 g, 27.5 mmol) in DMF (10 mL) was added bipyridine (107.7 mg, 0.7 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 5 min at room temperature. The reaction was quenched with water (200 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (3 x 100 mL), dried
over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in CH2Cl2) to afford (800 mg, 42% yield) of 5-bromo-2-chloro-4- (trifluoromethyl)aniline as an off-white oil. LCMS observed m/z = 271.90 [M-H]-. 5 Step 3: Preparation of 2-chloro-5-cyclopropyl-4-(trifluoromethyl)aniline. Compound 418.3
To a stirred solution of 5-bromo-2-chloro-4-(trifluoromethyl)aniline (500 mg, 1.8 mmol) and cyclopropylboronic acid (625 mg, 7.2 mmol) in toluene (2 mL) were added Pd(AcO)2 (44 mg, 0.2 mmol), PCy3 (61 mg, 0.2 mmol), water (0.5 mL) and K2CO3 (881 mg, 6.4 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 80 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (260 mg, 60% yield) of 2-chloro-5-cyclopropyl-4-(trifluoromethyl)aniline as a colorless oil. LCMS observed m/z = 234.05 [M-H]-. Step 4: Preparation of tert-butyl 4-[4-({[2-chloro-5-cyclopropyl-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate. Compound 418.4
To a stirred solution of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (150 mg, 0.2 mmol) and 2-chloro-5-cyclopropyl-4-(trifluoromethyl)aniline (134 mg, 0.5 mmol)
in DCM (5 mL) were added bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (145 mg, 0.5 mmol) and DIEA (110 mg, 0.8 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified 5 by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (180 mg, 84% yield) of tert-butyl 4-[4-({[2-chloro-5-cyclopropyl-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a white solid. LCMS observed m/z = 740.25 [M-H]-. Step 5: Preparation of N-[2-chloro-5-cyclopropyl-4-(trifluoromethyl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 418.5
tert-butyl 4-[4-({[2-chloro-5-cyclopropyl-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (170 mg, 0.2 mmol) in TFA (5 mL) was stirred overnight at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 640.20 [M-H]-. Step 6: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-5-cyclopropyl-4- (trifluoromethyl)phenyl]acetamide. Compound 418.6
To a stirred solution of N-[2-chloro-5-cyclopropyl-4-(trifluoromethyl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (109 mg, 0.1 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4- carboxylic acid (62 mg, 0.2 mmol) in DCM (1 mL) were added DIEA (147 uL, 0.8 mmol) and bis(2-oxo-1,3-oxazolidin-3-yl)phosphinoyl chloride (86 mg, 0.3 mmol) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at room temperature under nitrogen atmosphere. After concentration the residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (100 mg, 67% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2- chloro-5-cyclopropyl-4-(trifluoromethyl)phenyl]acetamide as a yellow oil. LCMS observed m/z = 866.25 [M+H]+. Step 7: Preparation of N-[2-chloro-5-cyclopropyl-4-(trifluoromethyl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 418
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-
yl)-N-[2-chloro-5-cyclopropyl-4-(trifluoromethyl)phenyl]acetamide (110 mg, 0.1 mmol) in TFA (5 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Shield RP18, 5μm, 19*250mm; Mobile 5 Phase A: water(10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 45% B in 10 min; Wave Length: 254nm/220nm nm; RT1(min): 9.13) to afford (15.2 mg, 14% yield) of N-[2-chloro-5-cyclopropyl-4- (trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide as a white solid.1H NMR (400 MHz, Chloroform- d) δ 12.10 (s, 1H), 8.93 (s, 1H), 8.66 (s, 1H), 8.27 – 8.15 (m, 2H), 8.12 (s, 1H), 7.58 (s, 1H), 7.35 (d, J = 7.8 Hz, 1H), 5.68 (m, 1H), 5.18 (s, 6H), 4.84 (m, 1H), 3.87 (m, 2H), 3.55 (s, 1H), 3.24 (d, J = 8.0 Hz, 2H), 3.13 (s, 1H), 2.94 – 2.80 (m, 2H), 2.67 (s, 3H), 2.11 (s, 1H), 1.38 (t, J = 7.4 Hz, 3H), 1.11 – 1.02 (m, 2H), 0.80 (d, J = 5.6 Hz, 2H). LCMS observed m/z = 778.25 [M+H]+. EXAMPLE 419 Synthesis of N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 419
Step 1: Preparation of potassium trifluoro(3,3,3-trifluoroprop-1-en-2-yl)boranuide Compound 419.1
To a stirred mixture of Mg (1.7 g, 70.0 mmol) in THF (120 mL) was added trimethyl borate (19.5 mL) dropwise at 0 ℃ under N2 atmosphere. To the above mixture was added 2-
bromo-3,3,3-trifluoroprop-1-ene (10.2 g, 58.3 mmol) in THF (15 mL) dropwise over 30 min at 0 ℃. The resulting mixture was stirred for additional 6 hours at this temperature and allowed to warmed to room temperature and stirred 16 hours. The solution was cooled to 0 ℃ and was quenched with 6 M aq. HCl (40 mL) via slow addition through a syringe. The mixture was allowed to stir at 0 ℃ for 1 hour. The solution was transferred to a 1000 mL separatory funnel and diluted with deionized H2O (200 mL) and Et2O (120 mL). The layers were separated, and the aqueous layer was extracted with Et2O (120 mL x 2). The combined organic layers were transferred to a 1000 mL round bottom flask and were cooled to 0 ℃. To the flask was slowly added 4.5 M aq. KHF2 (80 mL). The solution was allowed to warm to room temperature and stirred overnight. The solvent was removed in vacuo to afford the crude organotrifluoroborate. The solids were washed with hot acetone (4 x 120 mL), and the filtrate was collected in a round bottom flask. The solvent was removed in vacuo to give (8.4 g, 71.1% yield) of potassium trifluoro(3,3,3-trifluoroprop-1-en-2-yl) boranuide as a light-yellow semi-solid. Step 2: Preparation of 2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)aniline Compound 419.2
To a stirred solution of 4-bromo-2-chloroaniline (1.8 g, 8.7 mmol) and potassium trifluoro(3,3,3-trifluoroprop-1-en-2-yl) boranuide (3.5 g, 17.3 mmol) in THF (30 mL) were added Pd(OAc)2 (97 mg, 0.4 mmol) and PPh3 (272 mg, 1.0 mmol) in one portion at room temperature under N2 atmosphere. To the above mixture was added a solution of Cs2CO3 (8.5 g, 26.0 mmol) in H2O (15 mL). The resulting mixture was stirred for additional 16 hours at 80 ℃. The reaction was quenched by the addition of H2O (50 mL) at room temperature. The resulting mixture was extracted with EA (50 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/ EA (5:1) to afford (1.2 g, 62.6% yield) of 2-chloro-4- (3,3,3-trifluoroprop-1-en-2-yl) aniline as a light-yellow oil. LCMS observed m/z =
. [M-H]-.
Step 3: Preparation of tert-butyl 4-[4-({[2-chloro-4-(3,3,3-trifluoroprop-1-en-2- yl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate Compound 419.3
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (135 mg, 0.3 mmol) and DIEA (100 mg, 0.8 mmol) in DCM (3 mL) was added bis(2-oxo-1,3- oxazolidin-3-yl) phosphinoyl chloride (131 mg, 0.5 mmol) and 2-chloro-4-(3,3,3- trifluoroprop-1-en-2-yl)aniline (114 mg, 0.5 mmol) in portions at room temperature under N2 atmosphere. The resulting mixture was stirred for additional 3 hours at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL), extracted with DCM (5 mL x 3). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 100% EtOAc) to afford (88 mg, 47.0% yield) tert-butyl 4-[4-({[2-chloro-4-(3,3,3- trifluoroprop-1-en-2-yl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate as a white solid. LCMS observed m/z = 728.25 [M+H]+. Step 4: Preparation of N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide Compound 419.4
A solution of tert-butyl 4-[4-({[2-chloro-4-(3,3,3-trifluoroprop-1-en-2- yl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (100 mg, 0.1 mmol) and TFA (0.5 mL) in DCM (2 mL) was stirred for 15 min at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 628.20 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2- yl)phenyl]acetamide Compound 419.5
To a stirred solution of N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (80 mg, 0.1 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4- carboxylic acid (31 mg, 0.1 mmol) in DMF (3 mL) was added HATU (58 mg, 0.2 mmol) and DIEA (82 mg 06 mmol) in portions at room temperatu for additional 1
hour at room temperature. Desired product could be detected by LCMS. The reaction was quenched with H2O (5 mL). The resulting mixture was extracted with EA (5 mL x 3). The combined organic layers were washed with brine (5 mL x 2), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to afford (62 mg, 57.0% yield) of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin- 1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]acetamide as a yellow solid. LCMS observed m/z = 854.27 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 419
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl)-N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]acetamide (60 mg, 0.1 mmol) in TFA (3 mL) was stirred for 18 hours at room temperature under N2 atmosphere. Desired product could be detected by LCMS. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 30*150mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 32% B to 47%B in 7 min; Wave Length: 254nm/ 220nm nm; RT1(min): 6.67) to afford (21.9 mg, 40.0% yield) N-[2-chloro-4-(3,3,3-trifluoroprop-1-en-2-yl)phenyl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo- solid. 1H NMR
(400 MHz, DMSO-d6) δ 10.29 (s, 1H), 8.54 (s, 1H), 8.05 (d, J = 8.0 Hz, 2H), 7.85 (m, 1H), 7.66 (d, J = 2.2 Hz, 1H), 7.46 (d, J = 7.9 Hz, 2H), 6.21 (s, 1H), 6.13 (s, 1H), 5.36 (s, 2H), 5.06 (d, J = 5.5 Hz, 4H), 4.53 (m, 1H), 3.52 (m, 3H), 3.24 (m, 1H), 3.01 (m, 3H), 2.84 (m, 1H), 2.69 – 2.62 (m, 2H), 2.43 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 764.25 [M+H]+. EXAMPLE 420 Synthesis of N-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 420
Step 1: Preparation of tert-butyl (4-(1- (trifluoromethyl)cyclopropyl)phenyl)carbamate. Compound 420.1
To a stirred mixture of 1-bromo-4-[1-(trifluoromethyl)cyclopropyl]benzene (300 mg, 1.1 mmol), XantPhos (130 mg, 0.2 mmol), Cs2CO3 (1.0 g, 3.3 mmol) and tert-butyl carbamate (159 mg, 1.3 mmol) in dioxane (3 mL) was added Pd2(dba)3 (103 mg, 0.1 mmol) in one portion at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 115 °C and stirred at 115 °C for 2 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to 25 °C and concentrated under vacuum. The residue was purified by silica gel column chromatography, eluted with PE/EA (10/1) to
afford tert-butyl (4-(1-(trifluoromethyl)cyclopropyl)phenyl)carbamate (300 mg, 87.7 % yield) as a white solid. LCMS observed m/z = 302.13 [M+H]+. Step 2: Preparation of 4-(1-(trifluoromethyl)cyclopropyl)aniline. Compound 420.2
To a stirred solution of tert-butyl N-{4-[1- (trifluoromethyl)cyclopropyl]phenyl}carbamate (150 mg, 0.5 mmol) in DCM (0.5 mL) was added TFA (0.5 mL) dropwise at 25 °C. The reaction mixture was stirred at 25 °C for 30 minutes. After completion of reaction, the mixture neutralized to PH = 8 with saturated NaHCO3 (aq.). The resulting mixture was extracted with EA (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 4-(1-(trifluoromethyl)cyclopropyl)aniline (100 mg, crude). The crude product was used in the next step directly without further purification. LCMS observed m/z = 202.08 [M+H]+. Step 3: Preparation of 2-chloro-4-(1-(trifluoromethyl)cyclopropyl)aniline. Compound 420.3
To a stirred solution of 4-[1-(trifluoromethyl)cyclopropyl]aniline (100 mg, 0.5 mmol) in AcOH (1 mL) was added NCS (63.3 mg, 0.4 mmol) dropwise at 0 °C. The reaction mixture was stirred at 25 °C for 4 hours. After completion of reaction, the mixture was neutralized to pH = 7 with saturated NaHCO3 (aq.). The resulting mixture was extracted with EA (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (6/1) to afford 2-chloro-4-[1- (trifluoromethyl)cyclopropyl]aniline (100 mg, 85.2% yield) as a brown yellow solid. LCMS observed m/z = 236.04 [M+H]+. Step 4: Preparation of tert-butyl 4-(4-(2-((2-chloro-4-(1- (trifluorom obenzofuran-5-
yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate. Compound 420.4
To a stirred solution of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (100 mg, 0.2 mmol) in ACN (1 mL) were added NMI (39.1 mg, 0.5 mmol) and TCFH (80.2 mg, 0.3 mmol) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 30 minutes. To the above mixture was added 2-chloro-4-[1-(trifluoromethyl)cyclopropyl]aniline (49 mg, 0.2 mmol) dropwise at 25 °C. The reaction mixture was heated to 70 °C and stirred at 70 °C for 2 hours. After completion of reaction, the resulting mixture was cooled to 25 °C. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 40% to 60% gradient in 10 minutes; detector, UV 254 nm) to give tert-butyl 4-(4-(2-((2-chloro-4-(1- (trifluoromethyl)cyclopropyl)phenyl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate (138 mg, 97% yield) as a white solid. LCMS observed m/z =
. [M+H]+. Step 5: Preparation of N-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenyl)-2-(2- (1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 420.5
of tert-butyl 4-(4-(2-((2-chloro-4-(1- (trifluoromethyl)cyclopropyl)phenyl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate (55 mg, 0.07 mmol) in DCM (0.5 mL) was added TFA (0.5 mL) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 30 minutes. After completion of reaction, the resulting mixture was concentrated under vacuum to afford N-(2-chloro-4-(1- (trifluoromethyl)cyclopropyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- 6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (50 mg, crude). The crude product was used in the next step directly without further purification. LCMS observed m/z = 642.21 [M+H]+. Step 6: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(1- (trifluoromethyl)cyclopropyl)phenyl)acetamide. Compound 420.6
To opyl)phenyl)-2- (2-(1,3-dih 4]triazolo[1,5-
a]pyrimidin-4(7H)-yl)acetamide (50 mg, 0.08 mmol) and 5-(benzyloxy)-6- methylpyrimidine-4-carboxylic acid (23 mg, 0.1 mmol) in DMF (1 mL) were added HATU (45 mg, 0.1 mmol) and DIEA (60 mg, 0.5 mmol) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 2 hours. After completion of reaction, the residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 50% to 80% gradient in 10 minutes; detector, UV 254 nm) to give 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(1- (trifluoromethyl)cyclopropyl)phenyl)acetamide (16 mg, 23.7 % yield) as a yellow solid. LCMS observed m/z = 868.29 [M+H]+. Step 7: Preparation of N-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenyl)-2-(2- (1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 420
To a mixture of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenyl)acetamide (16 mg, 0.02 mmol) in DCM (0.5 mL) was added TFA (0.5 mL) dropwise at 25 °C. The reaction mixture was heated to 80 °C and stirred at 80 °C for 30 minutes. After completion of reaction, the resulting mixture was concentrated under vacuum. The residue was purified by reversed- phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 40 % to 60 % gradient in 10 minutes; detector, UV 254 nm) to give N-(2-chloro-4-(1-(trifluoromethyl)cyclopropyl)phenyl)-2-(2-(1,3- dihydroiso 4- carbonyl)p cetamide (5.0
mg, 34.2 % yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.5 – 9.8 (m, 1H), 8.50 (s, 1H), 8.05 (d, J = 7.4 Hz, 2H), 7.75 (d, J = 8.4 Hz, 1H), 7.60 (d, J = 2.1 Hz, 1H), 7.50 – 7.40 (m, 2H), 5.34 (s, 2H), 5.07 (d, J = 5.3 Hz, 4H), 4.54 (d, J = 12.6 Hz, 1H), 3.62 – 3.45 (m, 3H), 3.28 – 3.25 (m, 2H), 3.12 – 2.89 (m, 3H), 2.84 (d, J = 11.4 Hz, 1H), 2.73 – 2.62 (m, 1H), 2.43 (s, 3H), 1.38 – 1.29 (m, 2H), 1.27 – 1.19 (m, 3H), 1.16 (s, 2H). LCMS observed m/z = 778.20 [M+H]+. EXAMPLE 421 Synthesis of N-[2-chloro-4-(2,2-difluoroethenyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl]-7-oxo-[1,2,4]triazolo[1,5-a] pyrimidin-4-yl]acetamide Compound 421
Step 1: Preparation of tert-butyl 4-(4-{[(4-bromo-2- chlorophenyl)carbamoyl]methyl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 421.1
To a stirred mixture of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (89 mg, 0.2 mmol mL) were added bis(2-oxo- d DIEA (54 mg,
0.4 mmol) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 2 h. Then the reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford tert-butyl 4- (4-{[(4-bromo-2-chlorophenyl)carbamoyl]methyl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyri midin-6-yl)piperazine-1-carboxylate (90 mg, 74.0% yield) as a white solid. LCMS observed m/z = 712.16 [M+H] +. Step 2: Preparation of tert-butyl 4-[4-({[2-chloro-4-(2,2- difluoroethenyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate. Compound 421.2
To a stirred solution of tert-butyl4-(4-{[(4-bromo-2- chlorophenyl)carbamoyl]methyl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (140 mg, 0.2 mmol) and 2- (2,2-difluoroethenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (44 mg, 0.2 mmol) in dioxane (10 mL) and H2O (0.9 mL) were added K2CO3 (67 mg, 0.5 mmol) and Pd(dppf)Cl2 (1 mg, 0.02 mmol) in one portion at 25 °C under nitrogen atmosphere. The resulting mixture was heated to 85 °C and stirred at 85 °C for 4 h under nitrogen atmosphere. Then the mixture was allowed to cool down to 25 °C and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford tert-butyl 4-[4-({[2-chloro-4-(2,2-difluoroethenyl)phenyl]carbamoyl}methyl) -2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl]piperazine-1-carboxylate (80 mg, 58.0% yield) as a white solid. LCMS observed m/z = 696.24 [M+H] +. Step 3: Preparation of N-[2-chloro-4-(2,2-difluoroethenyl)phenyl]-2-[2-(1,3- dihydro-2 benzofuran 5 yl) 5 ethyl 7 oxo 6 (piperazin 1 yl) [124]triazolo[1,5- a]pyrimid
Compound 421.3
of tert-butyl 4-[4-({[2-chloro-4-(2,2- difluoroethenyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (80 mg, 0.1 mmol) in DCM (2 mL) was added TFA (2 mL) dropwise at 25 °C. The resulting mixture was stirred at 25 °C for 3 h. After the reaction was completed, the resulting mixture was concentrated under reduced pressure to afford N-[2-chloro-4-(2,2-difluoroethenyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]tria zolo[1,5- a]pyrimidin-4-yl]ace tamide (60 mg, crude) as a colorless oil. LCMS observed m/z = 596.19 [M+H] +. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(2,2- difluoroethenyl)phenyl]acetamide. Compound 421.4
To a stirred mixture of N-[2-chloro-4-(2,2-difluoroethenyl)phenyl]-2-[2-(1,3- dihydro-2 o[1,5- a]pyrimid mol) in DMF (5
mL) were added HATU (102 mg, 0.3 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4- carboxylic acid (39 mg, 0.1 mmol) in portions at 25 °C. The resulting mixture was stirred at 25 °C for 2 h. The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 15 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with CH2Cl2/MeOH (10/1) to afford 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(2,2- difluoroethenyl)phenyl]acetamide (58 mg, 52.0% yield) as a white solid. LCMS observed m/z = 822.27 [M+H] +. Step 5: Preparation of N-[2-chloro-4-(2,2-difluoroethenyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin -4-yl]acetamide. Compound 421
Into a 8 mL vial were added 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(2,2-difluoroethenyl) phenyl]acetamide (140 mg, 0.1 mmol) and TFA (3 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 0.5 h. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X select CSH C185 m, 30 mm x 150 mm; mobile phase A: water (0.1% FA), mobile phase B: ACN; flow rate: 60 mL/min; gradient: 50% B to 58% B in 7 min; wave length: 254 nm / 220 nm; RT1 (min): 6.15) to afford N-[2-chloro-4-(2,2- difluoroethenyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)piperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a] pyrimidin-4- yl]acetamide (11.4 mg, 95.0% purity) as a white solid.1H NMR (400 MHz, Chloroform-d)
δ 12.28 (s, 1H), 8.88 (s, 1H), 8.68 (s, 1H), 8.30 (d, J = 8.7 Hz, 1H), 8.24 – 8.20 (m,2H), 7.38 – 7.33 (m, 2H), 7.23 (dd, J = 8.6, 2.0 Hz, 1H), 5.65 (d, J = 13.0 Hz, 1H), 5.18 (d, J = 2.2 Hz, 7H), 4.84 (d, J = 12.8 Hz, 1H), 3.93 – 3.83 (m, 2H), 3.58 (d, J = 12.7 Hz, 1H), 3.28 – 3.20 (m, 2H), 2.94 – 2.79 (m, 3H), 2.72 (s, 3H), 1.38 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 732.20 [M+H] +. EXAMPLE 422 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine-2- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 422
Step 1: Preparation of tert-butyl N-(benzyloxy)-N-(3-cyanopropyl) carbamate. Compound 422.1
To a stirred solution of 4-chlorobutanenitrile (2.0 g, 19.3 mmol) in DMF (10 mL) was added NaH (1.4 g, 57.9 mmol) and NaI (5.8 g, 38.6 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 10 min. To the above mixture was added tert-butyl N-(benzyloxy) carbamate (4.3 g, 19.3 mmol) in portions at room temperature over 3 min. The resulting mixture was heated to 80 °C and stirred at 80 °C for additional 12 h. Then the reaction was cooled to room temperature, quenched with water and extracted with EtOAc (3 x 30 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was conce lica gel column
chromatography and eluted with PE / EA (50:1) to afford tert-butyl N-(benzyloxy)-N-(3- cyanopropyl) carbamate (1.5 g, 26% yield) as a yellow solid. LCMS observed m/z = 290.36 [M+H]+. Step 2: Preparation of 1-(benzyloxy) pyrrolidin-2-imine. Compound 422.2
A solution of tert-butyl N-(benzyloxy)-N-(3-cyanopropyl) carbamate (1.0 g, 3.4 mmol) in dioxane (15 mL) and HCl (5 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. The residue was diluted with Et2O (10 mL). The precipitated solids were collected by filtration and washed with Et2O (3 x10 mL). This resulted in 1-(benzyloxy) pyrrolidin-2-imine (330 mg, 50% yield) as an off-white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 190.25 [M+H]+. Step 3: Preparation of 2-iminopyrrolidin-1-ol. Compound 422.3 To a stirred solution of 1-(benzyloxy)pyrrolidin-2-imine (320 mg, 1.7 mmol) in MeOH (32 mL) was added Pd/C (322 mg, 0.9 mmol) in one portion at room temperature under hydrogen atmosphere. The resulting mixture was stirred overnight at room temperature under hydrogen atmosphere. Then the resulting mixture was filtered and the filter cake was washed with MeOH (3 x 5 mL). The filtrate was concentrated under reduced pressure to afford 2-iminopyrrolidin-1-ol (120 mg, crude) as a yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 100.12 [M+H]+. Step 4: Preparation of methyl 3-hydroxy-4-oxo-6H,7H,8H-pyrrolo[1,2- a]pyrimidine-2-carboxylate. Compound 422.4
To a stirred solution of 2-iminopyrrolidin-1-ol (100 mg, 1.0 mmol) in CHCl3 (3 mL) was added TEA (151 mg, 1.5 mmol) in one portion at room temperature under air atmosphere. The mixture was cooled to -30 °C. To the above mixture was added dimethyl acetylenedicarboxylate (156 mg, 1.1mmol) in portions at -30 °C. After stirring for 1 h at - 30 °C, the resulting mixture was warmed to room temperature and concentrated under vacuum. Then the residue was diluted with o-xylene (2 mL) and heated to 150 °C. After stirring for 2 h at 150 °C, the mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% TFA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 3-hydroxy- 4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine-2-carboxylate (160 mg, 76% yield) as an off- white oil. LCMS observed m/z = 210.19 [M+H]+. Step 5: Preparation of methyl 3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2- a]pyrimidine-2-carboxylate. Compound 422.5
To a stirred solution of methyl 3-hydroxy-4-oxo-6H,7H,8H-pyrrolo[1,2- a]pyrimidine-2-carboxylate (60 mg, 0.3 mmol) and benzyl bromide (98 mg, 0.6 mmol) in ACN (5 mL) was added K2CO3 (118 mg, 0.9 mmol) in one portion at room temperature. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (1:3) to afford methyl 3-(benzyloxy)-4-oxo- 6H,7H,8H as an off-white solid. LCM
Step 6: Preparation of 3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine-2- carboxylic acid. Compound 422.6
To a stirred solution of methyl 3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2- a]pyrimidine-2-carboxylate (45 mg, 0.2 mmol) in THF (1 mL) and H2O (1 mL) was added LiOH (18 mg, 0.8 mmol) in one portion at room temperature under air atmosphere. The resulting mixture was stirred at room temperature for 4 h under air atmosphere. Then the resulting mixture was neutralized to pH 7 with sat. NH4Cl and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine-2-carboxylic acid (35 mg, crude) as an off-white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 286.29 [M+H]+. Step 7: Preparation of 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2- a]pyrimidine-2-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Compound 422.7
To a stirred solution of 3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine- 2-carboxylic acid (30 mg, 0.1 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-
dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (63 mg, 0.1 mmol) in ACN (3 mL) were added TCFH (29 mg, 0.1 mmol) and NMI (26 mg, 0.3 mmol) in portions at room temperature under air atmosphere. The resulting mixture was heated to 40 °C and stirred at 40 °C for 1 h under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (1:1) to afford 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2- a]pyrimidine-2-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (70 mg, 76% yield) as an off-white solid. LCMS observed m/z = 870.29 [M+H]+. Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine-2- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 422
A solution of 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,8H-pyrrolo[1,2-a]pyrimidine-2- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (15 mg, 0.1 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: Xselect CSH C18, 5 m, 30 mm x150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 42% B to 55% B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.2) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]- 2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,8H- pyrrolo[1,2-a]pyrimidine-2-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-
a]pyrimidin-4-yl]acetamide (5.6 mg, 40% yield ) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 9.71 (s, 1H), 8.06 (m, 1H), 8.06 (s, 2H), 7.98 (d, J = 2.1 Hz, 1H), 7.75 – 7.70 (m, 1H), 7.46 (d, J = 8.0 Hz, 1H), 5.47 – 5.33 (m, 2H), 5.06 (d, J = 5.1 Hz, 4H), 4.49 (m, 1H), 4.02 (t, J = 7.3 Hz, 2H), 3.58 (m, 1H), 3.46 (m, 2H), 3.25 (m, 1H), 3.05 – 2.96 (m, 5H), 2.82 (m, 1H), 2.68 (d, J = 9.2 Hz, 1H), 2.18 (p, J = 7.7 Hz, 2H), 1.22 (t, J = 7.5 Hz, 3H).LCMS observed m/z = 780.35 [M+H]+. EXAMPLE 423 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(3-hydroxy-5,6,7,8-tetrahydroquinoline-2- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 423
Step 1: Preparation of 5,6,7,8-tetrahydroquinoline 1-oxide. Compound 423.1
5,6,7,8-tetrahydroquinoline (5.0 g, 37.6 mmol) and m-CPBA (8.9 g, 41.3 mmol) were dissolved in DCM (20 mL) and stirred at 20 °C for 2 h. After completed, the mixture was basified to pH 7 with saturated NaHCO3 (aq.). The resulting mixture was extracted with DCM (2 x 20 mL). The combined organic layers were dried over anhydrous Na2SO4 and the solvent was evaporated under reduced pressure. This resulted in 5,6,7,8-tetrahydroquinoline 1-oxide (5.0 g, crude) as a yellow oil. LCMS observed m/z = 150.25 [M+H]+. Step 2: Preparation of 5,6,7,8-tetrahydroquinoline-2-carbonitrile. Compound 423.2
To a solution of 5,6,7,8-tetrahydroquinoline 1-oxide (1.5 g, 10.1 mmol) and TMSCN (3.0 g, 30.2 mmol) in DCE (10 mL) was added diethyl carbamoyl-chloride (2.7 g, 20.2 mmol) in one portion at 25°C. The resulting mixture was stirred at 25°C for 16 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (9/1) to afford 5,6,7,8-tetrahydroquinoline-2-carbonitrile (840 mg, 52% yield) as a white solid. LCMS observed m/z = 159.15 [M+H]+. Step 3: Preparation of 5,6,7,8-tetrahydroquinoline-2-carboxylic acid. Compound 423.3 A solution of 5,6,7,8-tetrahydroquinoline-2-carbonitrile (800 mg, 5.1 mmol) and NaOH (1.01 g, 25.3 mmol) in EtOH (10 mL) was stirred at 25°C for 2 hours under nitrogen atmosphere. The mixture was acidified to pH 2 with conc. HCl. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in EtOH (10 mL) and washed with 4x5 mL of MeOH. The resulting mixture was filtered, the filter cake was washed with MeOH (3x3 mL). The filtrate was concentrated under reduced pressure to afford 5,6,7,8-tetrahydroquinoline-2-carboxylic acid (800 mg, 89 % yield) as a yellow solid. LCMS observed m/z = 178.10 [M+H]+.. Step 4: Preparation of N,N-diethyl-5,6,7,8-tetrahydroquinoline-2-carboxamide. Compound 423.4
To a solution of 5,6,7,8-tetrahydroquinoline-2-carboxylic acid (800 mg, 4.7 mmol) in ACN (20 mL) were added DEA (364 mg, 5.0 mmol), NMI (1.11 g, 13.5 mmol) and TCFH (2.53 g, 9.1 mmol) at 10 °C under nitrogen atmosphere. The resulting mixture was stirred at 20 °C for 2 hours under nitrogen atmosphere. After completed, the resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/1) to afford N,N-diethyl-5,6,7,8-
tetrahydroquinoline-2-carboxamide (800 mg, 73% yield) as a white oil. LCMS observed m/z = 231.20 [M+H]+. Step 5: Preparation of N,N-diethyl-3-hydroxy-5,6,7,8-tetrahydroquinoline-2- carboxamide. Compound 423.5
To a solution of N,N-diethyl-5,6,7,8-tetrahydroquinoline-2-carboxamide (680 mg, 3.0 mmol) and iPrO3B (1.65 g, 8.8 mmol) in THF (5 mL) and cooled to 0° C under nitrogen atmosphere. LDA (4.1 mL, 17.6 mmol, 2M in THF) was added dropwise, keeping the temperature below 0° C under nitrogen atmosphere. The mixture was stirred at 0 °C for 20 min. To the above mixture was added H2O2 (88 mg, 2.6 mmol) in portions at 0 °C. The mixture was stirred at 20 °C for 2 h. The resulting mixture was quenched with water (5 mL). The mixture was acidified to pH 2 with conc. HCl. The resulting mixture was extracted with EA (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (9/1) to afford N,N-diethyl-3- hydroxy-5,6,7,8-tetrahydroquinoline-2-carboxamide (150 mg, 20% yield) as a white solid. LCMS observed m/z = 249.50 [M+H]+. Step 6: Preparation of 3-hydroxy-5,6,7,8-tetrahydroquinoline-2-carboxylic acid. Compound 423.6
A solution of N,N-diethyl-3-hydroxy-5,6,7,8-tetrahydroquinoline-2-carboxamide (150 mg, 0.6 mmol) in aq. HCl (5 ml, 6 M) was stirred at 110°C for overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 15% gradient in
10 min; detector, UV 254 nm to afford 3-hydroxy-5,6,7,8-tetrahydroquinoline-2-carboxylic acid (50 mg, 42% yield) as a white solid. LCMS observed m/z = 194.20 [M+H]+. Step 7: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(3-hydroxy-5,6,7,8-tetrahydroquinoline-2- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 423
To a solution of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (30 mg, 0.05 mmol, Intermediate) in DMF (2 mL) were added HOBT (12 mg, 0.1 mmol), 3-hydroxy-5,6,7,8-tetrahydroquinoline-2-carboxylic acid (30 mg, 0.1 mmol) and EDCI (15 mg, 0.08 mmol) at 20 °C. The resulting mixture was stirred for 5 h at 25 °C under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The resulting mixture was extracted with EA (3 x 4 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm to afford N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(3- hydroxy-5,6,7,8-tetrahydroquinoline-2-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (10.1 mg, 25% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.44 (s, 1H), 10.17 (s, 1H), 8.10 – 8.01 (m, 3H), 7.97 (d, J = 2.2 Hz, 1H), 7.71 (dd, J = 8.6, 2.2 Hz, 1H), 7.46 (d, J = 7.9 Hz, 1H), 6.97 (s, 1H), 5.40 (s, 2H), 5.06 (d, J = 5.1 Hz, 4H), 4.55 (m, 1H), 3.58 (m, 1H), 3.50 (m, 2H), 3.27 – 3.22 (m, 1H), 3.02 (q, J = 7.3 Hz, 3H), 2.82 (m, 1H), 2.71 (t, J = 6.2 Hz, 4H), 2.65 (s, 1H), 1.76 (m, 4H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 777.25 [M+H]+. EXAMPLE 424
Synthesis of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 424
Step 1: Preparation of tert-butyl 4-(4-(2-((5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate. Compound 424.1
To a stirred mixture of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid (100 mg, 0.2 mmol, Intermediate) and 5-chloro-7-(trifluoromethyl)-1,3-benzothiazol- 4-amine (63 mg, 0.3 mmol) in THF (3 mL) were added DIEA (0.1 mL, 0.6 mmol) and cyanuric chloride (46 mg, 0.3 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 12 h under nitrogen atmosphere. After completion of reaction, the reaction was quenched with water (10 mL) at room temperature, and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was
concentrated under reduced pressure. This resulted in tert-butyl 4-(4-(2-((5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate (150 mg, crude) as a yellow solid. LCMS observed m/z = 759.20 [M+H] +. Step 2: Preparation of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2- (1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 424.2
To a stirred solution of tert-butyl 4-(4-(2-((5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine -1- carboxylate (150 mg, 0.2 mmol) in DCM (5 mL) was added TFA (5 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 1 h and concentrated under reduced pressure subsequently. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 30% to 50% gradient in 10 min; detector, UV 254 nm) to afford N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (26 mg, 19% yield) as a colorless oil. LCMS observed m/z = 659.15 [M+H] +. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol- 4-yl)acetamide. Compound 424.3
To a stirred mixture of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (15 mg, 0.06 mmol, Intermediate) in DMF (2 mL) were added EDCI (11 mg, 0.06 mmol) and HOBT (8 mg, 0.06 mmol) in portions at room temperature. After stirring for 1 h, to the above mixture was added N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl) acetamide (26 mg, 0.04 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 12 h under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol- 4-yl)acetamide (56 mg, crude) as a colorless oil. LCMS observed m/z = 885.22 [M+H] +. Step 4: Preparation of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2- (1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 424
Into a 8 mL sealed tube was added 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol- 4-yl)acetamide (56 mg, 0.06 mmol) and TFA (2 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 3 h. Then the resulting mixture was cooled down to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column: X select CSH C185 m, 19 mm * 250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 48% B to 70% B in 8 min; Wave Length: 254 nm/220 nm; RT 1 (min): 6.8) to afford N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (7.3 mg, 14% yield) as a white solid.1H NMR (400 MHz, Methanol -d4) δ 9.47 (s, 1H), 8.59 (s, 1H), 8.25 – 8.21 (m, 1H), 8.18 (s, 1H), 8.04 (s, 1H), 7.43 (d, J = 7.9 Hz, 1H), 5.54 (s, 2H), 5.17 – 5.15 (m, 4H), 4.75 – 4.71 (m, 1H), 4.15 – 4.10 (m, 1H), 3.78 (s, 2H), 3.56 – 3.43 (m, 1H), 3.27 – 3.20 (m, 2H), 3.19 – 3.13 (m, 1H), 3.00 – 2.97 (m, 1H), 2.85 – 2.81 (m, 1H), 2.55 (s, 3H), 1.41 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 795.15 [M+H] +. EXAMPLE 425 Synthesis of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(1,3-dihydroisobenzofuran- 5-yl)-6-(4-(2,2-dioxido-3,4-dihydro-1H-benzo[c][1,2] thiazine-8-carbonyl)piperazin-1-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 425
To a solution of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4] triazolo[1,5-a] pyrimidin-4(7H)-yl)acetamide (40 mg, 66 µmol), 3,4-dihydro-1H-benzo[c][1,2]thiazine-8- carboxylic acid 2,2-dioxide (15 mg, 66 µmol), EDC ^HCl (23 mg, 100 µmol) and DMAP (1.6 mg, 13 µmol) in CH2Cl2 (0.7 mL) was added DIPEA (17 mg, 23 µL, 133 µmol). The mixture was stirred at 25 °C for 16 h. The mixture was concentrated in vacuo. The residue was purified by reversed phase flash chromatography (Column: Xbridge C185 µm, 19 mm X 100 mm; mobile phase: 5-95% MeCN in H2O with 0.1% NH4OH modifier) to afford (14 mg, 26% yield) of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(1,3-dihydroisobenzofuran-5- yl)-6-(4-(2,2-dioxido-3,4-dihydro-1H-benzo[c][1,2] thiazine-8-carbonyl)piperazin-1-yl)-5- ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 9.40 (s, 1H), 8.10 – 8.01 (m, 3H), 7.98 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 7.46 (d, J = 7.9 Hz, 1H), 7.31 (d, J = 7.3 Hz, 1H), 7.23 – 7.11 (m, 2H), 5.40 (s, 2H), 5.06 (d, J = 5.5 Hz, 4H), 4.54 (d, J = 12.6 Hz, 1H), 3.53 – 3.43 (m, 2H), 3.39 (s, 4H), 3.31 – 3.21 (m, 1H), 3.05 – 2.97 (m, 3H), 2.84 (d, J = 9.7 Hz, 1H), 2.65 (d, J = 13.4 Hz, 1H), 2.08 (s, 1H), 1.22 (t, J = 7.4 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 811.20 [M+H]+. EXAMPLE 426 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-6-(4-(2,2-dioxido-1H-benzo[c][1,2]thiazine-8- carbonyl)piperazin-1-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 426
The title compound was prepared using similar procedure as compound 425 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-6-(4-(2,2- dioxido-1H-benzo[c][1,2]thiazine-8-carbonyl)piperazin-1-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 11.16 (s, 1H), 10.42 (s, 1H), 8.10 – 8.01 (m, 3H), 7.98 (s, 1H), 7.72 (d, J = 8.7 Hz, 1H), 7.66 (d, J = 7.7 Hz, 1H), 7.53 (d, J = 10.4 Hz, 1H), 7.49 – 7.40 (m, 2H), 7.31 – 7.19 (m, 2H), 5.40 (s, 2H), 5.06 (d, J = 5.6 Hz, 4H), 4.55 (d, J = 11.1 Hz, 1H), 3.65 – 3.53 (m, 2H), 3.49 – 3.42 (m, 1H), 3.31 - 3.24 (m, 1H), 3.09 – 2.96 (m, 3H), 2.85 (d, J = 11.0 Hz, 1H), 2.65 (d, J = 13.5 Hz, 1H), 1.22 (t, J = 7.3 Hz, 3H). LCMS observed m/z = 809.2 [M+H]+. EXAMPLE 427 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,8H,9H-pyrido[1,2-a]pyrimidine- 2-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 427
Step 1: Preparation of tert-butyl N-(benzyloxy)-N-(4-cyanobutyl)carbamate. Compound 427.1
To a stirred solution of 5-bromopentanenitrile (2.2 g, 19.3 mmol) in DMF (10 mL) were added NaH (1.4 g, 57.9 mmol) and NaI (5.8 g, 38.6 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 10 min. To the above mixture was added tert-butyl N-(benzyloxy)carbamate (4.3 g, 19.3 mmol) in portions at room temperature over 3 min. The resulting mixture was heated to 80 °C and stirred at 80 °C for additional 12 h. Then the reaction was cooled to room temperature, quenched with water and extracted with EtOAc (3 x 30 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (50:1) to afford tert-butyl N-(benzyloxy)-N-(4- cyanobutyl)carbamate (1.5 g, 26% yield) as a yellow solid. LCMS observed m/z = 305.18 [M+H]+. Step 2: Preparation of 1-(benzyloxy)piperidin-2-imine. Compound 427.2
A solution of tert-butyl N-(benzyloxy)-N-(4-cyanobutyl)carbamate (1.0 g, 3.4 mmol) in dioxane (15 mL) and HCl (5 mL) was stirred overnight at room temperature. The resulting mixture was concentrated under vacuum. The residue was diluted with Et2O (10 mL). The precipitated solids were collected by filtration and washed with Et2O (3 x 10 mL). This resulted in 1-(benzyloxy)piperidin-2-imine (330 mg, 50% yield) as an off-white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 205.13 [M+H]+. Step 3: Preparation of 2-iminopiperidin-1-ol. Compound 427.3
To a stirred solution of 1-(benzyloxy)piperidin-2-imine (320 mg, 1.7 mmol) in MeOH (32 mL) was added Pd/C (10%, 322 mg, 0.9 mmol) in one portion at room temperature under nitrogen atmosphere. The mixture was hydrogenated at room temperature for 12 h under hydrogen atmosphere using a hydrogen balloon. Then the resulting mixture was filtered through a celite pad and the filter cake was washed with MeOH (3 x 100 mL). The filtrate was concentrated under reduced pressure to afford 2-iminopiperidin-1-ol (120 mg, crude) as a yellow solid. LCMS observed m/z = 115.08 [M+H]+. Step 4: Preparation of methyl 3-hydroxy-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carboxylate. Compound 427.4
To a stirred solution of 2-iminopiperidin-1-ol (100 mg, 1.0 mmol) in CHCl3 (3 mL) was added TEA (151 mg, 1.5 mmol) in one portion at room temperature under air atmosphere. The mixture was cooled to -30 °C. To the above mixture was added dimethyl acetylenedicarboxylate (156 mg, 1.1mmol) in portions at -30 °C. After stirring for 1 h at - 30 °C, the resulting mixture was warmed to room temperature and concentrated under vacuum. Then the residue was diluted with o-xylene (2 mL) and heated to 150 °C. After stirring for 2 h at 150 °C, the mixture was cooled to room temperature and concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% TFA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in methyl 3-hydroxy- 4-oxo-6H,7H,8H,9H-pyrido[1,2-a]pyrimidine-2-carboxylate (160 mg, 76% yield) as an off- white oil. LCMS observed m/z = 225.08 [M+H]+. Step 5: Preparation of methyl 3-(benzyloxy)-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carboxylate. Compound 427.5
To a stirred solution of methyl 3-hydroxy-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carboxylate (60 mg, 0.3 mmol) and benzyl bromide (98 mg, 0.6 mmol) in ACN (5 mL) was added K2CO3 (118 mg, 0.9 mmol) in one portion at room temperature. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. Desired product could be detected by LCMS. The reaction was quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (1:3) to afford methyl 3-(benzyloxy)-4-oxo- 6H,7H,8H,9H-pyrido[1,2-a]pyrimidine-2-carboxylate (50 mg, 58% yield) as an off-white solid. LCMS observed m/z = 315.31 [M+H]+. Step 6: Preparation of 3-(benzyloxy)-4-oxo-6H,7H,8H,9H-pyrido[1,2-a]pyrimidine- 2-carboxylic acid. Compound 427.6
To a stirred solution of methyl 3-(benzyloxy)-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carboxylate (45 mg, 0.2 mmol) in THF (1 mL) and H2O (1 mL) was added LiOH (18 mg, 0.8 mmol) in one portion at room temperature under air atmosphere. The resulting mixture was stirred at room temperature for 4 h under air atmosphere. Desired product could be detected by LCMS. Then the resulting mixture was neutralized to pH 7 with sat. NH4Cl and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate
was concentrated under reduced pressure. This resulted in 3-(benzyloxy)-4-oxo- 6H,7H,8H,9H-pyrido[1,2-a]pyrimidine-2-carboxylic acid (35 mg, crude) as an off-white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 301.11 [M+H]+. Step 7: Preparation of 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Compound 427.7
To a stirred solution of 3-(benzyloxy)-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carboxylic acid (30 mg, 0.1 mmol) and N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (63 mg, 0.1 mmol) in ACN (3 mL) were added TCFH (29 mg, 0.1 mmol) and NMI (26 mg, 0.3 mmol) in portions at room temperature under air atmosphere. The resulting mixture was heated to 40 °C and stirred at 40 °C for 1 h under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (1:1) to afford 2-(6-{4-[3-(benzyloxy)-4- oxo-6H,7H,8H,9H-pyrido[1,2-a]pyrimidine-2-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (70 mg, 76% yield) as an off-white solid. LCMS observed m/z = 884.28 [M+H]+.
Step 8: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-hydroxy-4-oxo-6H,7H,8H,9H-pyrido[1,2-a]pyrimidine- 2-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 427
A solution of 2-(6-{4-[3-(benzyloxy)-4-oxo-6H,7H,8H,9H-pyrido[1,2- a]pyrimidine-2-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (15 mg, 0.1 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 1 h under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep- HPLC with the following conditions (Column: Xselect CSH C18, 5 m, 30 mm X 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 42% B to 55%B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.2) to afford N-[2- chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-(4-{3- hydroxy-4-oxo-6H,7H,8H,9H-pyrido[1,2-a]pyrimidine-2-carbonyl}piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (1.7 mg, 12% yield ) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.41 (s, 1H), 9.61 (s, 1H), 8.10 – 8.01 (m, 3H), 7.98 (d, J = 2.1 Hz, 1H), 7.72 (dd, J = 8.8, 2.1 Hz, 1H), 7.46 (d, J = 8.0 Hz, 1H), 5.40 (s, 2H), 5.06 (d, J = 5.0 Hz, 4H), 4.49 (d, J = 12.4 Hz, 1H), 3.88 (dt, J = 6.2, 3.4 Hz, 2H), 3.57 (d, J = 12.6 Hz, 1H), 3.47 (t, J = 11.8 Hz, 2H), 3.25 (t, J = 12.0 Hz, 1H), 3.10 – 2.91 (m, 3H), 2.80 (q, J = 8.5, 6.6 Hz, 3H), 2.67 (t, J = 6.2 Hz, 1H), 1.93 – 1.74 (m, 4H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 794.15 [M+H]+. EXAMPLE 428 Synthesis of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide
Compound 428
Step 1: Preparation of tert-butyl 4-(4-(2-((5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)-2-(2,3-dihydrobenzofuran-5- yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate. Compound 428.1
To a stirred solution of 2-(6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetic acid (70 mg, 0.1 mmol) and 5-chloro-7-(trifluoromethyl)-1,3-benzothiazol-4-amine (34 mg, 0.1 mmol) in THF (2 mL) were added pyridine (105 uL, 1.3 mmol) and 2,4,6-trichloro- 1,3,5-triazine (74 mg, 0.4 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was heated to 60 °C and stirred at 60 °C for 3 hours under nitrogen atmosphere. Then the reaction mixture was cooled to 25 °C and quenched with H2O (2 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in
tert-butyl 4-(4-(2-((5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)amino)-2-oxoethyl)- 2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl)piperazine-1-carboxylate (30 mg, 29% yield) as a light yellow solid. LCMS observed m/z = 759.35 [M+H]+. Step 2: Preparation of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2- (2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide. Compound 428.2
A solution of tert-butyl 4-(4-(2-((5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4- yl)amino)-2-oxoethyl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (30 mg, 0.04 mmol) in TFA (0.5 mL) was stirred for 30 min at 25 °C. The resulting mixture was concentrated under reduced pressure to afford N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (30 mg, crude) as a brown yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 659.15 [M+H]+. Step 3: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol- 4-yl)acetamide. Compound 428.3
To a stirred solution of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (13 mg, 0.06 mmol) and pyridine (8 mg, 0.1 mmol) in DMF (1 mL) were added EDCI (8 mg, 0.05 mmol) and HOBT (6 mg, 0.05 mmol) in portions at 25 °C under nitrogen atmosphere. The reaction mixture was stirred for 30 min at 25 °C under nitrogen atmosphere. To the above mixture was added N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(2,3- dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide (30 mg, 0.05 mmol) in one portion at 25 °C. The resulting mixture was stirred for additional 2 hours at 25 °C under nitrogen atmosphere. Then the reaction was quenched with H2O (5 mL) at 25 °C and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (5 mL) and dried over anhydrous MgSO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-(6-(4-(5- (benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-(2,3-dihydrobenzofuran-5- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(5-chloro-7- (trifluoromethyl)benzo[d]thiazol-4-yl)acetamide (30 mg, crude) as a yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 885.40 [M+H]+. Step 4: Preparation of N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2- (2,3-dihydrobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 428
The solution of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin- 1-yl)-2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(5-chloro-7-(trifluoromethyl)benzo[d]thiazol-4-yl)acetamide (30 mg, 0.03 mmol) in TFA (1 mL) was stirred at 80 °C for 30 min under nitrogen atmosphere. The reaction mixture was cooled to 25 °C and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Mobile Phase A: 10 mmol/L NH4HCO3 in water; Mobile Phase B: ACN; Flow rate: 60 mL/min) to afford N-(5-chloro- 7-(trifluoromethyl)benzo[d]thiazol-4-yl)-2-(2-(2,3-dihydrobenzofuran-5-yl)-5-ethyl-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide (10.1 mg, 37% yield) as an off-white solid.1H NMR (400 MHz, Methanol-d4) δ 9.46 (d, J = 2.6 Hz, 1H), 8.29 (s, 1H), 8.09 (s, 1H), 8.02 (s, 2H), 6.83 (d, J = 8.4 Hz, 1H), 5.49 (s, 2H), 4.71 (dd, J = 15.0, 6.6 Hz, 1H), 4.63 (t, J = 8.7 Hz, 2H), 3.75 (q, J = 11.2 Hz, 2H), 3.54 (s, 2H), 3.29 (s, 2H), 3.22 (q, J = 12.2, 9.6 Hz, 3H), 2.97 (d, J = 11.0 Hz, 1H), 2.80 (d, J = 11.4 Hz, 1H), 2.46 (s, 3H), 1.36 (dt, J = 15.8, 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 795.15 [M+H]+. EXAMPLE 429 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(6-methoxy-1-methyl-1H- benzo[d][1,2,3]triazole-5-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)acetamide Compound 429
The title compound was prepared using similar procedure as compound 425 to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6- (4-(6-methoxy-1-methyl-1H-benzo[d][1,2,3]triazole-5-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.06 (d, J = 9.0 Hz, 3H), 7.98 (s, 1H), 7.88 (d, J = 25.4 Hz, 1H), 7.72 (d, J = 8.7 Hz, 1H), 7.46 (d, J = 7.0 Hz, 2H), 5.40 (s, 2H), 5.06 (s, 4H), 4.65 – 4.56 (m, 1H), 4.28 (s, 3H), 3.98 (d, J = 23.2 Hz, 3H), 3.59 - 3.36 (m, 2H), 3.30 – 3.16 (m, 2H), 3.10 – 2.93 (m, 3H), 2.83 (t, J = 12.8 Hz, 1H), 2.63 – 2.55 (m, 1H), 1.21 (t, J = 8.2 Hz, 3H). LCMS observed m/z = 791.2 [M+H]+. EXAMPLE 430 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(3,6-dihydro-2H- pyran-4-carbonyl)piperazin-1-yl)-5-ethyl-2-(2-hydroxypyrimidin-4-yl)-7-oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 430
Step 1: Preparation of 2-methoxy-4-(tributylstannyl)pyrimidine. Compound 430.1
To a stirred mixture of 4-bromo-2-methoxypyrimidine (500 mg, 2.6 mmol) and 1,1,1,2,2,2-hexabutyldistannane (4.6 g, 7.9 mmol) in dioxane (20 mL) was added Pd (PPh3)4 (305 mg, 0.3 mmol) in one portion at room temperature under nitrogen atmosphere. The mixture was heated to 80 °C and stirred at 80 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5/1) to afford 2-methoxy-4- (tributylstannyl)pyrimidine (200 mg, 18% yield) as a yellow oil. LCMS observed m/z = 401.15 [M+H] +. Step 2: Preparation of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 430.2
To a stirred mixture of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (600 mg, 1.4 mmol) and DIEA (544 mg, 4.2 mmol) in DMF (20 mL) was added N-(2-chloro-4- (trifluoromethyl)phenyl)-2-iodoacetamide (539 mg, 1.5 mmol) in portions at room temperature under nitrogen atmosphere. The mixture was heated to 50 °C and stirred at 50 °C for 4 h under nitrogen atmosphere. The reaction mixture was quenched by addition of water (30 mL). The aqueous layer was extracted with ethyl acetate (3 x 30 mL). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified silica gel column chromatography, eluted with PE / EA (5 / 1)to afford tert-butyl 4- (2-bromo-4-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (600 mg, 64% yield) as a yellow solid. LCMS observed m/z = 662.10 [M+H] +.
Step 3: Preparation of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-2-(2-methoxypyrimidin-4-yl)-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pipera zine-1-carboxylate. Compound 430.3
To a stirred mixture of tert-butyl 4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (300 mg, 0.4 mmol) and 2- methoxy-4-(tributylstannyl)pyrimidine (198 mg, 0.5 mmol) in DMF (15 mL) was added Pd(PPh3)4 (52 mg, 0.04 mmol) in portions at room temperature under nitrogen atmosphere. The mixture was heated to 130 °C and stirred at 130 °C for 2 h under nitrogen atmosphere. After completion of reaction, the reaction mixture was quenched by addition of water (30 mL). The aqueous layer was extracted with ethyl acetate (30 mL). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to give crude product which was further purified silica gel column chromatography, eluted with CH2Cl2 / MeOH (20/1) to afford tert-butyl 4-(4- (2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-2-(2- methoxypyrimidin-4-yl)-7-oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pipera zine-1-carboxylate (150 mg, 47.4% yield) as a yellow solid. LCMS observed m/z = 692.22 [M+H] +. Step 4: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(2- hydroxypyrimidin-4-yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 430.4
To a stirred mixture of tert-butyl 4-(4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-2-(2-methoxypyrimidin-4-yl)-7-oxo- 4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)pipera zine-1-carboxylate (140 mg, 0.2 mmol) in DCM (3 mL) was added BBr3 (4.0 mL, 4.0 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 4 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure to give N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(2-hydroxypyrimidin-4-yl)-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (80 mg, crude) as a yellow solid. The crude resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 578.16 [M+H]+. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(3,6-dihydro- 2H-pyran-4-carbonyl)piperazin-1-yl)-5-ethyl-2-(2-hydroxypyrimidin-4-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 430
To a stirred mixture of 3,6-dihydro-2H-pyran-4-carboxylic acid (10 mg, 0.07 mmol), NMI (17 mg, 0.2 mmol), TCFH (21 mg, 0.07 mmol) and in ACN (2 mL) was added N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(2-hydroxypyrimidin-4-yl)-7-oxo-6- (piperazin-1-yl)-[1,2,4]tria zolo[1,5-a]pyrimidin-4(7H)-yl) acetamide(40 mg, 0.069 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. The crude product
(40 mg) was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 ExRs 5 m, 19*250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 31% B to 50% B in 8min; Wave Length: 254 nm/220 nm; RT1(min): 7.37) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4- (3,6-dihydro-2H-pyran-4-carbonyl)piperazin-1-yl)-5-ethyl-2-(2-hydroxypyrimidin-4-yl)-7- oxo-[1,2,4] triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (5.9 mg, 12% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 8.10 (d, J = 8.5 Hz, 2H), 7.95 (s, 1H), 7.69 (d, J = 8.4 Hz, 1H), 7.06 (s, 1H), 5.90 (d, J = 2.4 Hz, 1H), 5.42 (s, 2H), 4.14 (q, J = 2.8 Hz, 4H), 3.75 (t, J = 5.4 Hz, 2H), 3.41 (t, J = 11.3 Hz, 2H), 3.03 (d, J = 8.1 Hz, 4H), 2.78 (d, J = 11.0 Hz, 2H), 2.24 (s, 2H), 1.22 (t, J = 7.4 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 688.25 [M+H]+. EXAMPLE 431 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(2- hydroxypyrimidin-4-yl)-7-oxo-6-(4-(tetrahydro-2H-pyran-4-carbonyl)piperazin-1-yl)- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 431
To a stirred mixture of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(2- hydroxy pyrimidin-4-yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl) acetamide(50 mg, 0.08 mmol) and TEA (26 mg, 0.2 mmol) in DCM (2 mL) was added oxane-4-carbonyl chloride (13 mg, 0.07 mmol) in DCM (1 mL) dropwise at 0°C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C for 2 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product (50 mg) was purified by Prep-HPLC with the following conditions (Column: YMC Triart C18 ExRs 5 m, 19*250 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 35% B to 58% B in 7min; Wave Length: 254 nm/220 nm; RT1(min): 5.42) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-
ethyl-2-(2-hydroxypyrimidin-4-yl)-7-oxo-6-(4-(tetrahydro-2H-pyran-4- carbonyl)piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (13.3 mg, 22.1% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ10.13(br, 1H), 8.10 (d, J = 8.6 Hz, 2H), 7.97 (d, J = 2.2 Hz, 1H), 7.70 (dd, J = 8.8, 2.1 Hz, 1H), 7.07 (s, 1H), 5.44 (s, 2H), 4.45 (d, J = 12.3 Hz, 1H), 4.06 (d, J = 12.8 Hz, 1H), 3.87 (s, 2H), 3.53 – 3.34 (m, 4H), 3.29 – 3.17 (m, 1H), 3.11 – 2.90 (m, 3H), 2.83 – 2.66 (m, 3H), 1.62 (d, J = 38.2 Hz, 4H), 1.22 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 690.30 [M+H] +. EXAMPLE 432 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{7-hydroxy-2H,3H,4H-pyrano[3,2-b]pyridine-6- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 432
Step 1: Preparation of 3,5-bis(benzyloxy)pyridine. Compound 432.1
A solution of 3,5-dichloropyridine (50.0 g, 337.8 mmol) in DMF (200 mL) was treated with NaH (33.2 g, 830.0 mmol, 60%) at 0 °C under nitrogen atmosphere followed by the addition of benzyl alcohol (91.3 g, 844.6 mmol) dropwise at room temperature. The resulting mixture was stirred for 1 h at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 16 h at 80 °C. The reaction was quenched with water (300 mL) at 0
°C. The resulting mixture was extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with brine (3 x 500 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The precipitated solids were collected by filtration and washed with Et2O (3 x 30 mL). This resulted in (11.0 g, 11% yield) of 3,5- bis(benzyloxy)pyridine as a white solid. LCMS observed m/z = 292.35 [M+H]+. Step 2: Preparation of 3,5-bis(benzyloxy)-2,6-dibromopyridine. Compound 432.2
A solution of KBr (16.5 g, 138.9 mmol) in H2O (1.0 L) was treated with potassium bromate (60.5 g, 0.7 mmol), H2O (190 mL) and H2SO4 (65.7 g, 670.4 mmol) at room temperature under nitrogen atmosphere followed by the addition of 3,5- bis(benzyloxy)pyridine (25.3 g, 86.8 mmol), H2O (193 mL) and HCl (16 mL) at room temperature. The resulting mixture was stirred for 1 h at room temperature under nitrogen atmosphere. The reaction was quenched with sat. sodium sulfite at room temperature. The precipitated solids were collected by filtration and washed with water (3 x 100 mL). The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (21.0 g, 54% yield) of 3,5-bis(benzyloxy)-2,6-dibromopyridine as a white solid. LCMS observed m/z = 449.14 [M+H]+. Step 3: Preparation of 3,5-bis(benzyloxy)-2-bromo-6-{3-[(trimethylsilyl)oxy]prop- 1-yn-1-yl}pyridine. Compound 432.3
A solution of 3,5-bis(benzyloxy)-2,6-dibromopyridine (19.0 g, 42.3 mmol) in DMF (60 mL) was treated with CuI (805 mg, 4.2 mmol) and Pd(PPh3)2Cl2 (2.9 g, 4.2 mmol) at room temperature under nitrogen atmosphere followed by the addition of trimethyl(prop-2- yn-1-yloxy)silane (6.5 g, 50.7 mmol) and TEA (7.7 g, 76.1 mmol) dropwise at room temperature. The resulting mixture was stirred for 8 h at 80 °C under nitrogen atmosphere.
The reaction was quenched with water (120 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 150 mL). The combined organic layers were washed with brine (3 x 150 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 497.10 [M+H]+. Step 4: Preparation of 3-[3,5-bis(benzyloxy)-6-bromopyridin-2-yl]prop-2-yn-1-ol. Compound 432.4
A solution of 3,5-bis(benzyloxy)-2-bromo-6-{3-[(trimethylsilyl)oxy]prop-1-yn-1- yl}pyridine (25.2 g, 50.8 mmol) in THF (200 mL) was treated with TBAF (14.6 g, 55.8 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 15 min at room temperature under nitrogen atmosphere. The reaction was quenched with water (300 mL) at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 200 mL). The organic layers were dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 50% EtOAc in petroleum ether) to afford (6.0 g, 28% yield) of 3-[3,5-bis(benzyloxy)-6-bromopyridin-2- yl]prop-2-yn-1-ol as a yellow solid. LCMS observed m/z = 425.10 [M+H]+. Step 5: Preparation of methyl 3,5-bis(benzyloxy)-6-(3-hydroxyprop-1-yn-1- yl)pyridine-2-carboxylate. Compound 432.5
A solution of 3-[3,5-bis(benzyloxy)-6-bromopyridin-2-yl]prop-2-yn-1-ol (6.0 g, 14.1 mmol) in MeOH (60 mL) and DMSO (30 mL) was treated with Pd(dppf)Cl2 (1.0 g, 1.4 mmol) at room temperature under nitrogen atmosphere followed by the addition of TEA (2.9 g, 28.3 mmol) dropwise at room temperature. The resulting mixture was stirred for 16 h at 100 °C under CO (50 atm) atmosphere. The resulting mixture was concentrated under reduced pressure. The reaction was quenched with water (50 mL) at room temperature. The
resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 38% EtOAc in petroleum ether) to afford (3.5 g, 61% yield) of methyl 3,5-bis(benzyloxy)-6-(3-hydroxyprop-1-yn-1-yl)pyridine-2-carboxylate as a black solid. LCMS observed m/z = 404.13 [M+H]+. Step 6: Preparation of methyl 3,5-dihydroxy-6-(3-hydroxypropyl)pyridine-2- carboxylate. Compound 432.6
A solution of methyl 3,5-bis(benzyloxy)-6-(3-hydroxyprop-1-yn-1-yl)pyridine-2- carboxylate (2.0 g, 1.2 mmol) in THF (20 mL) was treated with Pd/C (0.8 g, 0.2 mmol, 10%) and Pd(OH)2/C (1.6 g, 1.6 mmol, 50%) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 2 h at 45 °C under hydrogen atmosphere. The resulting mixture was filtered, the filter cake was washed with THF (3 x 20 mL). The filtrate was concentrated under reduced pressure. The crude product was purified by Prep- HPLC with the following conditions (Column: Xbridge BEH Shield RP18, 5μm, 19*250mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 2% B to 20%B in 7 min; Wave Length: 254nm/220nm nm; RT1(min): 6.53) to afford (400 mg, 37% yield) of methyl 3,5-dihydroxy-6-(3- hydroxypropyl)pyridine-2-carboxylate as a white solid. LCMS observed m/z = 228.22 [M+H]+. Step 7: Preparation of methyl 7-hydroxy-2H,3H,4H-pyrano[3,2-b]pyridine-6- carboxylate. Compound 432.7
A solution of methyl 3,5-dihydroxy-6-(3-hydroxypropyl)pyridine-2-carboxylate (400 mg, 1.7 mmol) in THF (10 mL) was treated with Ph3P (923 mg, 3.5 mmol) at room
temperature under nitrogen atmosphere followed by the addition of DIAD (0.7 mL, 3.5 mmol) dropwise at 0 °C. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The residue was purified by reversed phase C18 silica gel column chromatography to afford (134 mg, 36% yield) of methyl 7-hydroxy-2H,3H,4H-pyrano[3,2- b]pyridine-6-carboxylate as a yellow oil. LCMS observed m/z = 210.07 [M+H]+. Step 8: Preparation of methyl 7-[(4-methoxyphenyl)methoxy]-2H,3H,4H- pyrano[3,2-b]pyridine-6-carboxylate. Compound 432.8
A solution of methyl 7-hydroxy-2H,3H,4H-pyrano[3,2-b]pyridine-6-carboxylate (134 mg, 0.6 mmol) in DMF (3 mL) was treated with K2CO3 (221 mg, 1.6 mmol) at room temperature under nitrogen atmosphere followed by the addition of 4-methoxybenzyl chloride (150 mg, 0.9 mmol) dropwise at room temperature. The resulting mixture was stirred for 4 h at 60 °C under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 15 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 20% EtOAc in petroleum ether) to afford (130 mg, 62% yield) of methyl 7-[(4-methoxyphenyl)methoxy]-2H,3H,4H- pyrano[3,2-b]pyridine-6-carboxylate as a yellow solid. LCMS observed m/z = 330.13 [M+H]+. Step 9: Preparation of 7-[(4-methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2- b]pyridine-6-carboxylic acid. Compound 432.9
A solution of methyl 7-[(4-methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2- b]pyridine-6-carboxylate (117 mg, 0.4 mmol) in THF (5 mL) and H2O (2.5 mL) was treated with NaOH (43 mg, 1.1 mmol) at room temperature under nitrogen atmosphere. The
resulting mixture was stirred for 2 h at 40 °C under nitrogen atmosphere. The residue was neutralized to pH 7 with HCl (1M). The resulting mixture was filtered, the filter cake was washed with THF (3 x 9 mL). The filtrate was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: Water (10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 9% B to 36% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 6.53) to afford (26 mg, 22% yield) of 7-[(4-methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2-b]pyridine-6-carboxylic acid as a white solid. LCMS observed m/z = 316.33 [M+H]+. Step 10: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide. Compound 432.10
A solution of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (70 mg, 0.1 mmol) in DCM (1 mL) was treated with TFA (1 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 20 min at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was used in the next step directly without further purification. LCMS observed m/z = 602.02 [M+H]+. Step 11: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-6-(4-{7-[(4-methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2- b]pyridine-6-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide. Compound 432.11
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (60 mg, 0.1 mmol) in DMF (1 mL) was treated with 7-[(4- methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2-b]pyridine-6-carboxylic acid (21 mg, 0.1 mmol) and TCFH (28 mg, 0.1 mmol) at room temperature under nitrogen atmosphere followed by the addition of NMI (27 mg, 0.3 mmol) dropwise at room temperature. The resulting mixture was stirred for 2 h at room temperature under nitrogen atmosphere. The reaction was quenched with water (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (eluent: with 50% EtOAc in petroleum ether) to afford (54 mg, 90% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-(4-{7-[(4- methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2-b]pyridine-6-carbonyl}piperazin-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. LCMS observed m/z = 899.33 [M+H]+. Step 12: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-6-(4-{7-hydroxy-2H,3H,4H-pyrano[3,2-b]pyridine-6- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 432
A solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{7-[(4-methoxyphenyl)methoxy]-2H,3H,4H-pyrano[3,2- b]pyridine-6-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (50 mg, 0.1 mmol) in DCM (2 mL) was treated with TFA (2 mL) at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 30 min at room temperature under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 19 mm * 250 mm; Mobile Phase A: Water(10mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 58% B to 73% B in 7min; Wave Length: 254nm/220nm nm; RT1(min): 7.38) to afford (11.6 mg, 26% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-6-(4-{7-hydroxy-2H,3H,4H-pyrano[3,2-b]pyridine-6- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 8.11 – 8.01 (m, 3H), 7.98 (d, J = 2.2 Hz, 1H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.46 (d, J = 8.1 Hz, 1H), 6.67 (s, 1H), 5.40 (s, 2H), 5.07 (d, J = 4.9 Hz, 4H), 4.55 (s, 1H), 4.16 (t, J = 5.1 Hz, 2H), 3.90 (m 1H), 3.56 – 3.45 (m, 2H), 3.27 (s, 1H), 3.03 (d, J = 7.8 Hz, 3H), 2.77 (t, J = 6.5 Hz, 4H), 2.01 (m 2H), 1.22 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 779.20 [M+H]+. EXAMPLE 433 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-5-{4-[(6-hydroxy-1-methyl-1H-1,2,3-benzotriazol-5-yl) carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide. Compound 433
Step 1: Preparation of N-[2-chloro-4-(trifluoromethyl) phenyl] [5-(4-{[6- (benzyloxy)-1-methyl-1H-1,2,3-benzotriazol-5-yl] carbonyl}-1-piperazinyl)-2-(1,3- dihydro-5-isobenzofuranyl)-6-ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl] acetamide. Compound 433.1
To a solution of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-4-oxo-5-(1-piperazinyl)-1,3,3a,7-tetraaza-7-indenyl]acetamide (50 mg, 83 µmol), 6-(benzyloxy)-1-methyl-1H-1,2,3-benzotriazole-5-carboxylic acid (23.5 mg, 83 µmol) and PyBrop (58 mg, 125 µmol) in DMF (0.83 mL) was added DIPEA (54 mg, 72 µL, 415 µmol). The mixture was stirred at 25 °C for 16 h. The mixture was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-20% MeOH in CH2Cl2) to afford (70 mg, 97% yield) 2-(6-(4-(6- (benzyloxy)-1-methyl-1H-benzo[d][1,2,3]triazole-5-carbonyl)piperazin-1-yl)-2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide as a yellow solid. LCMS observed m/z = 867.3 [M+H]+. Step 2: N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-5-{4-[(6-hydroxy-1-methyl-1H-1,2,3-benzotriazol-5- yl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 433
The compound N-[2-chloro-4-(trifluoromethyl)phenyl][5-(4-{[6-(benzyloxy)-1- methyl-1H-1,2,3-benzotriazol-5-yl]carbonyl}-1-piperazinyl)-2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide (70 mg, 80 µmol) was added to TFA in one portion. The mixture was stirred at 60 °C for 2 hours. The mixture was concentrated in vacuo. The residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (20.3 mg, 28% yield) of N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(6-hydroxy-1- methyl-1H-benzo[d][1,2,3]triazole-5-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-
a]pyrimidin-4(7H)-yl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.61 (s, 1H), 10.36 (s, 1H), 8.03 – 7.94 (m, 3H), 7.91 (s, 1H), 7.75 (s, 1H), 7.64 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 7.9 Hz, 1H), 6.95 (s, 1H), 5.33 (s, 2H), 4.99 (d, J = 5.9 Hz, 4H), 4.53 (d, J = 12.5 Hz, 1H), 4.12 (s, 3H), 3.44 (s, 2H), 3.19 (s, 1H), 2.99 – 2.90 (m, 3H), 2.76 (d, J = 11.0 Hz, 1H), 2.60 – 2.54 (m, 1H), 1.14 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR spectra. LCMS observed m/z = 778.6 [M+H]+. EXAMPLE 434 Synthesis of 2-(6-(4-acetylpiperazin-1-yl)-5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide Compound 434
Step 1: Preparation of tert-butyl 4-(3-amino-6-ethyl-2-imino-4-oxo-1,2,3,4- tetrahydropyrimidin-5-yl)piperazine-1-carboxylate. Compound 434.1
To a stirred mixture of 1-aminoguanidine hydrochloride (2.1 g, 19.1 mmol, 2.0 equiv.) and TBAH (4.1 g, 6.4 mmol, 2.0 equiv.) in EtOH (25 mL) were added tert-butyl 4- (1-methoxy-1,3-dioxopentan-2-yl) piperazine-1-carboxylate (3.0 g, 9.5 mmol, 1.0 equiv.) in portions at room temperature. The resulting mixture was heated to 80 ℃ and stirred at 80 oC for 6 h. After completion of the reaction, the mixture was cooled to room temperature and filtered. The cake was collected. This resulted in tert-butyl 4-(1-amino-4-ethyl-2-imino- 6-oxo-3H-pyrimidin-5-yl) piperazine-1-carboxylate (1.2 g, crude) as a white solid. LCMS observed m/z = 338.21 [M+H]+.
Step 2: Preparation of tert-butyl 4-[5-ethyl-2-(5-methoxypyrimidin-2-yl)-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate. Compound 434.2
To a stirred solution of tert-butyl 4-(1-amino-4-ethyl-2-imino-6-oxo-3H-pyrimidin- 5-yl)piperazine-1-carboxylate (200 mg, 0.6 mmol, 1.0 equiv.) and methyl 5- hydroxypyrimidine-2-carboxylate (91 mg, 0.6 mmol, 1.0 equiv.) in MeOH (3 mL) were added NaOMe (80 mg, 1.5 mmol, 2.5 equiv.) in portions at room temperature. The resulting mixture was heated to 85 ℃ and stirred at 85 oC for 1 h under nitrogen atmosphere. After completion of the reaction, the mixture was cooled to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, FA in H2O, 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 4-[5-ethyl-2-(5-methoxypyrimidin-2-yl)-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-6-yl]piperazine-1-carboxylate (60 mg, 22% yield) as a brown solid. LCMS observed m/z = 442.21 [M+H]+. Step 3: Preparation of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate.
To a stirred solution of tert-butyl 4-[5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7-oxo- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (65 mg, 0.1 mmol, 1.0 equiv.) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (53 mg, 0.1 mmol, 1.0 equiv.) in DMF (2 ml) were added DIEA (50 uL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. After
completion of the reaction, the resulting mixture was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% NH3.H2O), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in tert-butyl 4-[4-({[2-chloro- 4-(trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (50 mg, 50% yield) as a yellow solid. LCMS observed m/z = 677.21 [M+H]+. Step 4: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(5-ethyl-2-(5- hydroxypyrimidin-2-yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 434.4
To a stirred solution of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (10 mg, 0.02 mmol, 1.0 equiv.) in DCM (0.8 ml) was added TFA (0.4 ml) dropwise at 0 oC. The resulting mixture was heated to 25 °C and stirred for 0.5 h at 25 °C. After completion of the reaction, the resulting mixture was concentrated under reduced pressure. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 577.16 [M+H]+. Step 5: Preparation of 2-(6-(4-acetylpiperazin-1-yl)-5-ethyl-2-(5-hydroxypyrimidin- 2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 434
To a stirred solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(5- hydroxypyrimidin-2-yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (10 mg, 0.02 mmol, 1.0 equiv.) and TEA (7.2 uL) in DCM (1 ml) were added acetyl chloride (5 ul, 0.04 mmol, 2.0 equiv.) in portions at 0 oC. The resulting mixture was heated to 25 °C and stirred for 1 h at 25 °C under nitrogen atmosphere. After completion of the reaction, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X Bridge BEH Shield RP185 m, 19 mm *250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 39% B to 62% B in 10 min; Wave Length: 254 nm/220 nm; RT 1(min): 7.88) to afford 2-(6-(4-acetylpiperazin-1-yl)-5-ethyl-2-(5- hydroxypyrimidin-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (2.8 mg, 97.7% purity) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.36 (s, 2H), 8.14 (d, J = 8.6 Hz, 1H), 7.97 (d, J = 2.1 Hz, 1H), 7.71 (dd, J = 8.8, 2.1 Hz, 1H), 5.42 (s, 2H), 4.42 (d, J = 12.1 Hz, 1H), 2.73 (d, J = 11.1 Hz, 3H), 2.17 (s, 3H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 620.30 [M+H]+. EXAMPLE 435 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{6-hydroxy-2H,3H-furo[3,2-b]pyridine-5- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 435
Step 1: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide.
of tert-butyl 4-[4-({[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl]piperazine-1-carboxylate (70 mg, 0.1 mmol) in DCM (1 mL) was added TFA (57 mg, 0.5 mmol) dropwise at room temperature. The resulting mixture was stirred at room temperature for additional 1 h. After completed, the resulting mixture was concentrated under vacuum to afford (50 mg, crude) of N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6- (piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 602.18 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{6-hydroxy-2H,3H-furo[3,2-b]pyridine-5- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 435.2
To a stirred solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (15 mg, 0.1 mmol) and 6-hydroxy-2H,3H-furo[3,2-b]pyridine-5-carboxylic acid (5 mg, 0.1 mmol) in ACN (2 mL) were added NMI (6 mg, 0.1 mmol) and TCFH (7 mg, 0.1 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for additional 1 h. After completed, poured the resulting mixture into water (3 mL) and extracted with EA (3 x 3 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography (Column: XBridge BEH C185μm, 19*250mm; mobile phase: 52%- 57% ACN in H2O) to afford (1.2 mg, 7% yield) of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl- 6-(4-{6-hydroxy-2H,3H-furo[3,2-b]pyridine-5-carbonyl}piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Acetonitrile-d3) δ 12.58 (s, 1H), 8.89 (s, 1H), 8.41 – 8.31 (m, 1H), 8.18 – 8.09 (m, 2H), 7.88 – 7.81 (m, 1H), 7.66 – 7.61 (m, 1H), 7.47 – 7.39 (m, 1H), 6.66 (s, 1H), 5.30 (s, 2H), 5.18 – 5.07 (m, 4H), 4.80 – 4.70 (m, 2H), 3.75 – 3.65 (m, 2H), 3.32 – 3.26 (m, 1H), 3.24 – 3.17 (m, 2H), 3.15 – 3.07 (m, 2H), 2.91 – 2.80 (m, 2H), 2.12 (s, 3H), 1.36 – 1.24 (m, 3H). LCMS observed m/z = 765.15 [M+H]+. EXAMPLE 436 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(6-(4-(3,6-dihydro-2H- pyran-4-carbonyl)piperazin-1-yl)-5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 436
To a stirred solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[5-ethyl-2-(5- hydroxypyrimidin-2-yl)-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (20 mg, 0.04 mmol) and 3,6-dihydro-2H-pyran-4-carboxylic acid (5 mg, 0.04 mmol) in ACN (1 ml) were added TCFH (12 mg, 0.04 mmol) and NMI (11 uL) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. After completion of the reaction, the mixture was quenched with water (10 mL) at room temperature and extracted with EA (3 x 15 mL). The combined organic layers were washed with brine (15 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep- HPLC with the following conditions (Column: Xselect CSH C185 m, 19 mm * 250 mm; Mobile Phase A: water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 42% B to 65% B in 7min; wave Length: 254/220 nm; RT1 (min): 5.85; 7.14) to afford N- [2-chloro-4-(trifluoromethyl)phenyl]-2-{6-[4-(3,6-dihydro-2H-pyran-4- carbonyl)piperazin-1-yl]-5-ethyl-2-(5-hydroxypyrimidin-2-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}acetamide (9.2 mg, 38% yield) as a white solid.1H NMR (400 MHz, Methanol-d4) δ 8.49 (s, 2H), 8.22 (d, J = 8.7 Hz, 1H), 7.83 (d, J = 2.1 Hz, 1H), 7.62 (dd, J = 8.7, 2.1 Hz, 1H), 5.99 (d, J = 2.4 Hz, 1H), 5.51 (s, 2H), 4.61 (s, 4H), 4.24 (t, J = 2.8 Hz, 2H), 3.88 (t, J = 5.4 Hz, 2H), 3.63 (m, 2H), 3.14 (d, J = 8.3 Hz, 2H), 2.90 (m, 2H), 2.37 (s, 2H), 1.35 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR spectra. LCMS observed m/z = 688.30 [M+H]+. EXAMPLE 437 Synthesis of 2-{6-[(3aS,7aS)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide Compound 437
EXAMPLE 438 Synthesis of 2-{6-[(3aR,7aS)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide
Step 1: Preparation of tert-butyl 1-(1-methoxy-1,3-dioxopentan-2-yl)-hexahydro- 2H-pyrrolo[3,2-b]pyridine-4-carboxylate. Compound 437.1/438.1
To a stirred solution of tert-butyl octahydropyrrolo[3,2-b]pyridine-4-carboxylate (2.2 g, 9.7 mmol) in ACN (12 mL) were added K2CO3 (4.0 g, 29.1 mmol) and methyl 2- chloro-3-oxopentanoate (1.7 g, 10.6 mmol) in portions at room temperature under air atmosphere. The resulting mixture was heated to 60 ℃ and stirred at 60 ℃ for 3 hours under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated
under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (5:1) to afford tert-butyl 1-(1-methoxy-1,3-dioxopentan-2-yl)- hexahydro-2H-pyrrolo[3,2-b]pyridine-4-carboxylate (2.0 g, 58% yield) as a colorless oil. LCMS observed m/z = 355.12 [M-H]-. Step 2: Preparation of 2-bromo-5-ethyl-6-{octahydropyrrolo[3,2-b]pyridin-1-yl}- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one. Compound 437.2/438.2
To a stirred mixture of tert-butyl 1-(1-methoxy-1,3-dioxopentan-2-yl)-hexahydro- 2H-pyrrolo[3,2-b]pyridine-4-carboxylate (1.0 g, 2.8 mmol) and 5-bromo-2H-1,2,4-triazol- 3-amine (505 mg, 3.1 mmol) in EtOH (5 mL) was added H3PO4 (1.4 g, 14.1 mmol) in portions at room temperature under air atmosphere. The resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 3 days under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was diluted with water (30 mL), neutralized to pH 6-7 with saturated NaHCO3 (aq.) and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.1% NH4HCO3), 20% to 40% gradient in 10 min; detector, UV 254 nm, to afford 2-bromo-5-ethyl-6- {octahydropyrrolo[3,2-b]pyridin-1-yl}-4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one (320 mg, 30% yield) as a yellow solid. LCMS observed m/z = 367.12 [M+H]+. Step 3: Preparation of 6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-4H-[1,2,4]triazolo[1,5- a]pyrimidin-7-one. Compound 437.3/438.3
To a stirred mixture of 2-bromo-5-ethyl-6-{octahydropyrrolo[3,2-b]pyridin-1-yl}- 4H-[1,2,4]triazolo[1,5-a]pyrimidin-7-one (320 mg, 0.8 mmol) and 5-(benzyloxy)-6-
methylpyrimidine-4-carboxylic acid (234 mg, 0.9 mmol) in DMF (4 mL) were added DIEA (337 mg, 2.6 mmol) and HATU (364 mg, 0.9 mmol) in portions at room temperature under air atmosphere. The resulting mixture was heated to 50 ℃ and stirred at 50 ℃ for 3 hours under nitrogen atmosphere. Then the mixture was cooled to room temperature, quenched with water (20 mL) and exacted with EA (3 x 20 mL). The organic layers were washed with brine (20 mL), dried over Na2SO4 and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.1% NH3.H2O), 50% to 70% gradient in 10 min; detector, UV 254 nm to afford 6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-4H-[1,2,4]triazolo[1,5- a]pyrimidin-7-one (280 mg, 54% yield) as a yellow solid. LCMS observed m/z = 592.82 [M+H]+. Step 4: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide. Compound 437.4/438.4
To a stirred mixture of 6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-4H-[1,2,4]triazolo[1,5- a]pyrimidin-7-one (280 mg, 0.4 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2- iodoacetamide (188 mg, 0.5 mmol) in DMF (4 mL) was added DIEA (183 mg, 1.4 mmol) in one portion at room temperature under air atmosphere. The resulting mixture was heated to 45 ℃ and stirred at 45 ℃ for 4 hours under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.1% NH3.H2O), 60% to 70% gradient in 10 min; detector, UV 254 nm to afford 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-
hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (290 mg, 74% yield) as a white solid. LCMS observed m/z = 828.12 [M+H]+. Step 5: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Compound 437.5/438.5
To a stirred mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (290 mg, 0.3 mmol) and 2-(1,3-dihydro-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (103 mg, 0.4 mmol) in 1,4-dioxane (2 mL) were added K2CO3 (145 mg, 1.0 mmol) and Pd(dppf)Cl2.CH2Cl2 (28 mg, 0.03 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 ℃ and stirred at 80 ℃ for 2 hours under nitrogen atmosphere. Then the mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, ACN in water (0.1% NH3.H2O), 60% to 80% gradient in 10 min; detector, UV 254 nm to afford 2-(6-{4-[5-(benzyloxy)-6- methylpyrimidine-4-carbonyl]-hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl}-2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide (200 mg, 65% yield) as a yellow solid. LCMS observed m/z = 868.32 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro- 2H-pyrrolo[3,2-b]pyridin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide.
Compound 437/438
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-hexahydro- 2H-pyrrolo[3,2-b]pyridin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (200 mg, 0.2 mmol) in TFA (3 mL) was stirred at room temperature for 1.5 h under nitrogen atmosphere. Then the mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: Xbridge BEH Phenyl 5 μm, 19 * 250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 30% B to 45%B in 10 min; Wave Length: 254 nm / 220 nm; RT1 (min): 8.73) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (85 mg, 47% yield) as a white solid LCMS observed m/z = 778.45 [M+H]+. Step 7: Preparation of 2-{6-[(3aS,7aS)-4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-hexahydro-2H-pyrrolo[3,2-b]pyridin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Compound 437
N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5- ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro-2H-pyrrolo[3,2- b]pyridin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (85 mg, 0.1 mmol) was resolved by chiral-HPLC with the following conditions (Column: CHIRALPAK IG, 3 * 25 cm, 10 μm; Mobile Phase A: MeOH: DCM = 1:1 HPLC, Mobile Phase B: MtBE (0.1% FA)-HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1(min): 7.2; RT2 (min): 9.3; Sample Solvent: MEOH; Injection Volume: 1 mL; Number of Runs: 6) to afford 2-{6-[(3aS,7aS)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro-2H- pyrrolo[3,2-b]pyridin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (30.8 mg, 35% yield) as off-white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 10.11 (s, 1H), 8.59 (m, 1H), 8.14 – 7.93 (m, 4H), 7.72 (d, J = 8.6 Hz, 1H), 7.46 (d, J = 7.7 Hz, 1H), 5.44 – 5.31 (m, 2H), 5.14 – 4.94 (m, 4H), 4.56 – 4.23 (m, 1H), 3.53 – 3.36 (m, 1H), 3.28 – 2.75 (m, 6H), 2.44 (s, 2H), 2.36 – 2.10 (m, 2H), 1.99 (s, 1H), 1.89 – 1.31 (m, 4H), 1.30 – 1.17 (m, 3H). LCMS observed m/z = 778.45 [M+H]+. Compound 438
N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5- ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro-2H-pyrrolo[3,2- b]pyridin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (85 mg, 0.1 mmol) was resolved by chiral-HPLC with the following conditions (Column: CHIRALPAK IG, 3 * 25 cm, 10 μm; Mobile Phase A: MeOH: DCM = 1:1 HPLC, Mobile Phase B: MtBE (0.1% FA)-HPLC; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1(min): 7.2; RT2 (min): 9.3; Sample Solvent: MEOH; Injection Volume: 1 mL; Number of Runs: 6) to afford 2-{6-[(3aR,7aS)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-hexahydro- 2H-pyrrolo[3,2-b]pyridin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (30.2 mg, 34% yield) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 10.09 (s, 1H), 8.59 (m, 1H), 8.12 – 7.95 (m, 4H), 7.72 (d, J = 8.6 Hz, 1H), 7.46 (d, J = 7.8 Hz, 1H), 5.44 – 5.32 (m, 2H), 5.12 – 4.98 (m, 4H), 4.52 – 4.26 (m, 1H), 3.41 (s, 1H), 3.31 – 2.84 (m, 6H), 2.44 (s, 2H), 2.35 – 2.12 (m, 2H), 1.99 – 1.68 (m, 2H), 1.64 - 142 (m, 2H), 1.36 (s, 1H), 1.29 – 1.19 (m, 3H). LCMS observed m/z = 778.45 [M+H]+. EXAMPLE 439 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(4-hydroxyisoxazole-3-carbonyl)piperazin-1-yl)- 7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 439
The title compound was prepared using similar procedure as compound 398 to afford of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl- 6-(4-(4-hydroxyisoxazole-3-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.43 (s, 1H), 10.42 (s, 1H), 8.11 – 8.02 (m, 3H), 8.01 – 7.94 (m, 2H), 7.72 (d, J = 8.6 Hz, 1H), 7.46 (d, J = 8.0 Hz, 1H), 5.40 (s, 2H), 5.06 (d, J = 4.9 Hz, 4H), 4.54 (d, J = 12.4 Hz, 1H), 3.65 (d, J = 12.6 Hz, 1H), 3.55 – 3.46 (m, 2H), 3.31 – 3.27 (m, 1H), 3.08 – 2.96 (m, 3H), 2.84
(d, J = 11.3 Hz, 1H), 2.72 (d, J = 11.1 Hz, 1H), 1.22 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 729.2 [M+H]+. EXAMPLE 440 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-5-{4-[(3-hydroxy-7,8-dihydro-5H-6-oxa-1-azanaphth-2- yl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 440
The title compound was prepared using a similar procedure as 425. 1H NMR (400MHz, MeOD) δ 8.20 – 8.08 (m, 3H), 7.83 (s, 1H), 7.66 – 7.53 (m, 2H), 7.39 (d, J = 8.0 Hz, 1H), 5.53 – 5.41 (m, 2H), 5.12 (s, 4H), 4.78 – 4.31 (m, 2H), 4.13 – 4.03 (m, 1H), 3.83 – 3.37 (m, 5H), 3.38 – 3.32 (m, 1H), 3.23 – 2.76 (m, 7H), 1.36 – 1.30 (m, 3H). Note: Two exchangeable protons were not observed in NMR Spectra. LCMS observed m/z = 779.3 [M+H]+. EXAMPLE 441 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-5-{4-[(3-hydroxy-6,8-dihydro-5H-7-oxa-1-azanaphth-2- yl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide. Compound 414
The title compound was prepared using a similar procedure as Example 425. 1H NMR (400MHz, MeOD) δ 8.20 – 8.08 (m, 3H), 7.83 (d, J = 2.0 Hz, 1H), 7.63 – 7.59 (m, 1H), 7.44 (s, 1H), 7.39 (d, J = 7.9 Hz, 1H), 5.46 (s, 2H), 5.12 (s, 4H), 4.77 – 4.32 (m, 2H), 4.03 – 3.95 (m, 2H), 3.83 – 3.61 (m, 3H), 3.61 – 3.37 (m, 2H), 3.20 – 3.04 (m, 3H), 3.03 – 2.76 (m, 4H), 1.36 – 1.31 (m, 3H). Note: Two exchangeable protons were not observed in NMR Spectra. LCMS observed m/z = 779.2 [M+H]+. EXAMPLE 442 Synthesis of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide Compound 442
Step 1: Preparation of 3-chloro-2-iodo-5-(trifluoromethyl) phenol. Compound 442.1
To a stirred mixture of 3-chloro-5-(trifluoromethyl) phenol (10.0 g, 50.8 mmol) in toluene (50 mL) was added sodium hydride (2.4 g, 101.7 mmol) in portions at 0 ℃ under N2 atmosphere. To the above mixture was added I2 (12.9 g, 50.8 mmol) in portions at 0 ℃. The resulting mixture was stirred at 0 ℃ for additional 3 hours under nitrogen atmosphere. The mixture was acidified to pH 4 with conc. HCl and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent:
with 10% EtOAc in petroleum ether) to afford (12.0 g, 73% yield) of 3-chloro-2-iodo-5- (trifluoromethyl)phenol as a yellow oil. LCMS observed m/z = 320.75 [M-H]-. Step 2: Preparation of 1-chloro-2-iodo-3-(prop-2-en-1-yloxy)-5-(trifluoromethyl) benzene. Compound 442.2
To a stirred solution of 3-chloro-2-iodo-5-(trifluoromethyl) phenol (12.0 g, 37.2 mmol) and allyl bromide (4.8 mL, 55.8 mmol) in DMF (70 mL) was added K2CO3 (15.4 g, 111.6 mmol) in one portion at 25 ℃ under nitrogen atmosphere. The resulting mixture was stirred at 25 ℃ for 4 hours under nitrogen atmosphere. Then the reaction was quenched with H2O (100 mL) at room temperature and extracted with EA (3 x 250 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (7.5 g, 55% yield) of 1-chloro-2-iodo-3-(prop-2-en-1- yloxy)-5-(trifluoromethyl) benzene as a colorless oil. Step 3: Preparation of 3-chloro-2-iodo-6-(prop-2-en-1-yl)-5-(trifluoromethyl) phenol. Compound 442.3
A solution of 1-chloro-2-iodo-3-(prop-2-en-1-yloxy)-5-(trifluoromethyl) benzene (7.5 g, 20.6 mmol) in 1,2-dichlorobenzene (20 mL) was stirred at 175 °C for 24 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (5.0 g, 67% yield) of 3-chloro-2-iodo-6-(prop-2-en-1- yl)-5-(trifluoromethyl) phenol as a brown yellow oil. LCMS observed m/z = 360.70 [M-H]- .
Step 4: Preparation of 2-[4-chloro-2-hydroxy-3-iodo-6-(trifluoromethyl)phenyl] acetaldehyde. Compound 442.4
A solution of 3-chloro-2-iodo-6-(prop-2-en-1-yl)-5-(trifluoromethyl) phenol (2.0 g, 5.5 mmol) in MTBE (15 mL) and H2O (15 mL) was treated with K2OsO4.2H2O (61 mg, 0.2 mmol) in one portion for 30 min at room temperature under nitrogen atmosphere followed by the addition of NaIO4 (1.4 g, 6.6 mmol). Then the resulting mixture was stirred at 25℃ for 6 hours under nitrogen atmosphere. The reaction was quenched with water at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the mixture was concentrated in vacuo to afford (2.0 g, crude) of 2-[4-chloro-2-hydroxy-3-iodo-6-(trifluoromethyl)phenyl] acetaldehyde as a brown yellow solid. The crude product was used in the next step directly without further purification. Step 5: Preparation of 3-chloro-6-(2-hydroxyethyl)-2-iodo-5-(trifluoromethyl) phenol. Compound 442.5
A solution of 2-[4-chloro-2-hydroxy-3-iodo-6-(trifluoromethyl) phenyl] acetaldehyde (2.0 g, 5.5 mmol) in MeOH (25 mL) was treated with NaBH4 (1.0 g, 27.4 mmol) in one portion at 25 ℃ for 2 hours. The reaction was quenched with 2N HCl. Then the resulting mixture was concentrated under reduced pressure. The resulting mixture was extracted with EA (3 x 10 ml). The combined organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (250 mg, 11% yield) of 3-chloro-6-(2-
hydroxyethyl)-2-iodo-5-(trifluoromethyl) phenol as a brown yellow oil. LCMS observed m/z = 364.80 [M-H]-. Step 6: Preparation of 6-chloro-7-iodo-4-(trifluoromethyl)-2,3-dihydro-1- benzofuran. Compound 442.6
To a stirred solution of 3-chloro-6-(2-hydroxyethyl)-2-iodo-5-(trifluoromethyl) phenol (250 mg, 0.7 mmol) and PPh3 (358 mg, 1.3 mmol) in THF (5 mL) were added DIAD (276 mg, 1.3 mmol) in one portion at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (95 mg, 40% yield) of 6-chloro-7-iodo-4-(trifluoromethyl)-2,3-dihydro-1-benzofuranas a white solid. Step 7: Preparation of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7- yl]-1,1-diphenylmethanimine. Compound 442.7
A solution of 6-chloro-7-iodo-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran (95 mg, 0.3 mmol) and Benzophenone imine (59 mg, 0.3 mmol) in dioxane (5 mL) was treated with XantPhos (32 mg, 0.05 mmol), sodium tert-butoxide (78 mg, 0.8 mmol) and Pd2(dba)3 (25 mg, 0.03 mmol) in one portion at 25 ℃. The resulting mixture was stirred at 100 °C for 16 hours under nitrogen atmosphere. The resulting mixture was diluted with H2O (10 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na2SO4. After filtration, the resulting mixture was concentrated in vacuo to afford (120 mg, crude) of N-[6-chloro-4-(trifluoromethyl)-1- benzofuran-7-yl]-1,1-diphenylmethanimine as a brown yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 401.95 [M+H]+.
Step 8: Preparation of 6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7- amine. Compound 442.8
A solution of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl]-1,1- diphenylmethanimine (120 mg, 0.3 mmol) in THF (2 mL) was treated with HCl (6 M) (2 mL) in one portion at 25 ℃. The resulting mixture was stirred at 25 ℃ for 1 hour. The mixture was acidified to pH 7~8 with NaHCO3 and extracted with EA (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (52 mg, 57% yield) of 6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-amine as a brown yellow oil. LCMS observed m/z = 238.01 [M+H]+. Step 9: Preparation of tert-butyl 4-[4-({[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1- benzofuran-7-yl] carbamoyl} methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo - [1,2,4] triazolo[1,5-a] pyrimidin-6-yl] piperazine-1-carboxylate. Compound 442.9
A solution of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (66 mg, 0.13 mmol) and 6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-amine (30 mg, 0.13 mmol) in DCM (3 mL) was treated with DIEA (49 mg, 0.4 mmol) and Bis(2-oxo-3- oxazolid (42 mg, 0.2 mmol) in one portion at 25 ℃. The resulting mixture was stirred at 25 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was concentrated under
reduced pressure and dissolved in DMF (3 mL). The mixture was purified by preparatory HPLC (Column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 60-75% MeCN in H2O) to afford (16 mg, 33% yield) of tert-butyl 4-[4-({[6-chloro- 4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl] carbamoyl} methyl)-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo - [1,2,4] triazolo[1,5-a] pyrimidin-6-yl] piperazine-1- carboxylate as a white solid. LCMS observed m/z = 742.05 [M-H]-. Step 10: Preparation of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran- 7-yl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl) - [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide. Compound 442.10
A solution of tert-butyl 4-[4-({[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl] carbamoyl} methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5- a] pyrimidin-6-yl] piperazine-1-carboxylate (35 mg, 0.05 mmol) in DCM (5 mL) was treated with Trifluoroacetic acid (2 mL) in one portion at 25 ℃. The resulting mixture was stirred at 25 ℃ for 30 min. The mixture was concentrated in vacuo to afford (38 mg, crude) of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl) - [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide as a brown yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 644.15 [M+H]+. Step 11: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl)-N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl] acetamide. Compound 442.11
solution of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (38 mg, 0.06 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4- carboxylic acid (17 mg, 0.07 mmol) in DMF (10 mL) was treated with TCFH (20 mg, 0.07 mmol) and NMI (24 mg, 0.3 mmol) in one portion at 25 ℃. The resulting mixture was stirred at 25 ℃ for 2 hours under nitrogen atmosphere. The mixture was purified by preparatory HPLC (Column: X Bridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; mobile phase: 10-60% MeCN in H2O) to afford (40 mg, 70% yield) of 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1-yl}-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl)-N-[6-chloro-4- (trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl] acetamide as a White solid. LCMS observed m/z = 870.00 [M+H]+. Step 12: Preparation of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran- 7-yl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine- 4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide. Compound 442
A solution of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-
yl)-N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7-yl] acetamide (36 mg, 0.04 mmol) in TFA (2 mL) was stirred at 25 ℃ for 12 hours. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparatory HPLC (Column: XBridge BEH C185 μm, 30*150mm; mobile phase: 10-50% ACN in H2O) to afford (4.7 mg, 14% yield) of N-[6-chloro-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran-7- yl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide as a White solid.1H NMR (400 MHz, DMSO-d6) δ 10.32 (s, 2H), 8.53 (s, 1H), 8.08 (d, J = 5.5 Hz, 2H), 7.49 (d, J = 8.2 Hz, 1H), 7.32 (s, 1H), 5.24 (s, 2H), 5.09 (d, 4H), 4.71 (t, 2H), 4.54 (d, 1H), 3.58 – 3.48 (m, 3H), 3.40 (d, 2H), 3.25 (d, 1H), 3.02 (d, 3H), 2.83 (d, 1H), 2.66 (d, 1H), 2.43 (s, 3H), 1.24 (t, 3H). LCMS observed m/z = 780.35 [M+H]+. EXAMPLE 443 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(4-hydroxy-5-(4-methylpiperazin-1- yl)isothiazole-3-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 443
Step 1: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl](5-{4-[(5-bromo-4- hydroxy-3-isothiazolyl)carbonyl]-1-piperazinyl}-2-(1,3-dihydro-5-isobenzofuranyl)-6- ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide
To a solution of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-4-oxo-5-(1-piperazinyl)-1,3,3a,7-tetraaza-7-indenyl]acetamide (500 mg, 831 µmol), 5-bromo-4-hydroxy-3-isothiazolecarboxylic acid (186 mg, 831 µmol) and HATU (475 mg, 1250 µmol) in DMF (8.3 mL) was added DIPEA (537 mg, 723 µL, 4150 µmol). The mixture was stirred at 35 °C for 18 h. The mixture was cooled to 25 °C and was concentrated in vacuo. Then, the mixture was diluted with H2O (50 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-100% Ethyl acetate in n-Hexanes) to afford (491 mg, 66% yield) N-[2-chloro-4-(trifluoromethyl)phenyl](5-{4-[(5-bromo-4- hydroxy-3-isothiazolyl)carbonyl]-1-piperazinyl}-2-(1,3-dihydro-5-isobenzofuranyl)-6- ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide as a yellow solid. LCMS observed m/z = 809.0 [M+H]+. Step 2: Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(4-hydroxy-5-(4-methylpiperazin-1- yl)isothiazole-3-carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 443
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl](5-{4-[(5-bromo-4- hydroxy-3-isothiazolyl)carbonyl]-1-piperazinyl}-2-(1,3-dihydro-5-isobenzofuranyl)-6- ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide (40 mg, 49 µmol, Intermediate 416.1), 1-methylpiperazine (7.5 mg, 49 µmol) and Cs2CO3 (32 mg, 99 µmol) in dioxane (0.5 mL) was added Pd(dppf)Cl2.CH2Cl2 (4 mg, 5 µmol). The mixture was stirred at 80 °C under N2 for 16 hours. The mixture was concentrated in vacuo. The residue was dissolved in CH2Cl2 and filtered through a pad of Celite and the filtrate was concentrated in vacuo. The residue was purified by preparatory HPLC (mobile phase: 0-100% MeCN in H2O) to afford (3.5 mg, 8% yield) of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran- 5-yl)-5-ethyl-6-(4-(4-hydroxy-5-(4-methylpiperazin-1-yl)isothiazole-3-
carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as a faint yellow solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.34 (s, 1H), 8.01 – 7.94 (m, 3H), 7.90 (d, J = 2.1 Hz, 1H), 7.64 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 7.9 Hz, 1H), 5.33 (s, 2H), 4.99 (d, J = 4.8 Hz, 4H), 4.46 (d, J = 12.5 Hz, 1H), 4.17 (d, J = 12.6 Hz, 1H), 3.55 – 3.39 (m, 3H), 3.16 (t, J = 5.0 Hz, 4H), 3.03 – 2.84 (m, 3H), 2.77 (d, J = 11.1 Hz, 1H), 2.67 (d, J = 11.1 Hz, 1H), 2.39 (t, J = 5.0 Hz, 4H), 2.15 (s, 3H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 827.3 [M+H]+. EXAMPLE 444 Synthesis of 2-(6-(4-(5H-pyrrolo[3,2-d]pyrimidine-4-carbonyl)piperazin-1-yl)-2- (1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)- N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide
The title compound was prepared using similar procedure as compound 425 to afford 2-(6-(4-(5H-pyrrolo[3,2-d]pyrimidine-4-carbonyl)piperazin-1-yl)-2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 12.06 (s, 1H), 10.37 (s, 1H), 8.81 (s, 1H), 8.03 – 7.94 (m, 3H), 7.90 - 7.88 (m, 2H), 7.65 - 7.63 (m, 1H), 7.39 (d, J = 8.0 Hz, 1H), 6.64 (d, J = 4.0 Hz, 1H), 5.33 (s, 2H), 4.99 (d, J = 4.0 Hz, 4H), 4.59 (d, J = 12.0 Hz, 1H), 3.96 (d, J = 12.0 Hz, 1H), 3.58 - 3.43 (m, 3H), 3.32 - 3.28 (m, 2H), 3.10 - 2.94 (m, 3H), 2.84 (d, J = 12.0 Hz, 1H), 2.63 (d, J = 12.0 Hz, 1H), 1.16 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 747.3 [M+H]+. EXAMPLE 445 Synthesis of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(4-hydroxy-5-morpholinoisothiazole-3- carbonyl)piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide Compound 445
The title compound was prepared using similar procedure as Compound 443 afford N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6- (4-(4-hydroxy-5-morpholinoisothiazole-3-carbonyl)piperazin-1-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide as an orange solid.1H NMR (400 MHz, DMSO) δ 10.41 (s, 1H), 8.42 (s, 1H), 8.02 – 7.94 (m, 3H), 7.87 (s, 1H), 7.62 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 7.9 Hz, 1H), 5.31 (s, 2H), 4.99 (d, J = 4.8 Hz, 4H), 4.46 (d, J = 12.5 Hz, 1H), 4.13 (d, J = 12.5 Hz, 1H), 3.71 – 3.64 (m, 4H), 3.45 (t, J = 11.6 Hz, 2H), 3.22 - 3.17 (m, 1H), 3.15 – 3.10 (m, 4H), 3.01 – 2.87 (m, 3H), 2.77 (d, J = 11.2 Hz, 1H), 2.67 (d, J = 11.2 Hz, 1H), 1.15 (t, J = 7.3 Hz, 3H). LCMS observed m/z = 814.3 [M+H]+. EXAMPLE 446 Synthesis of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(4-hydroxyisoxazole-3-carbonyl) piperazin-1-yl)- 7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4(7H)-yl)acetamide Compound 446
The title compound was prepared using similar procedure as compound 425 to afford of N-(2-chloro-4-(trifluoromethyl) phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-5-ethyl- 6-(4-(4-hydroxyisoxazole-3-carbonyl) piperazin-1-yl)-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4(7H)-yl)acetamide as a white solid. 1H NMR (400 MHz, DMSO) δ 10.34 (s, 1H), 9.69 (s, 1H), 8.53 (s, 1H), 8.03 – 7.94 (m, 3H), 7.93 – 7.88 (m, 1H), 7.65 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 7.9 Hz, 1H), 5.33 (s, 2H), 4.99 (d, J = 4.9 Hz, 4H), 4.46 (d, J = 12.4 Hz, 1H), 3.56 (d, J = 12.5 Hz, 1H), 3.43 - 3.40 (m, 2H), 3.29 - 3.26 (m, 1H), 3.01 – 2.90 (m,
3H), 2.78 (d, J = 11.2 Hz, 1H), 2.68 (d, J = 11.1 Hz, 1H), 1.15 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 713.2 [M+H]+. EXAMPLE 447 Synthesis of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1- yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide Compound 447
Step 1: Preparation of 3-chloro-2-iodo-5-(trifluoromethyl) phenol. Compound 447.1
To a stirred mixture of 3-chloro-5-(trifluoromethyl) phenol (10.0 g, 50.8 mmol) in toluene (50 mL) was added sodium hydride (2.4 g, 101.7 mmol) in portions at 0 ℃ under N2 atmosphere. To the above mixture was added I2 (12.9 g, 50.8 mmol) in portions at 0 ℃. The resulting mixture was stirred at 0 ℃ for additional 3 hours under nitrogen atmosphere. The mixture was acidified to pH 4 with conc. HCl and extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (12.0 g, 73% yield) of 3-chloro-2-iodo-5- (trifluoromethyl)phenol as a yellow oil. LCMS observed m/z = 320.75 [M-H]-. Step 2: Preparation of 1-chloro-2-iodo-3-(prop-2-en-1-yloxy)-5-(trifluoromethyl) benzene. Compound 447.2
To a stirred solution of 3-chloro-2-iodo-5-(trifluoromethyl) phenol (12.0 g, 37.2 mmol) and allyl bromide (4.8 mL, 55.8 mmol) in DMF (70 mL) was added K2CO3 (15.4 g, 111.6 mmol) in one portion at 25 ℃ under nitrogen atmosphere. The resulting mixture was stirred at 25 ℃ for 4 hours under nitrogen atmosphere. Then the reaction was quenched with H2O (100 mL) at room temperature and extracted with EA (3 x 250 mL). The combined organic layers were washed with brine (2 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (7.5 g, 55% yield) of 1-chloro-2-iodo-3-(prop-2-en-1- yloxy)-5-(trifluoromethyl) benzene as a colorless oil. Step 3: Preparation of 3-chloro-2-iodo-6-(prop-2-en-1-yl)-5-(trifluoromethyl) phenol. Compound 447.3
A solution of 1-chloro-2-iodo-3-(prop-2-en-1-yloxy)-5-(trifluoromethyl) benzene (7.5 g, 20.6 mmol) in 1,2-dichlorobenzene (20 mL) was stirred at 175 °C for 24 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (5.0 g, 67% yield) of 3-chloro-2-iodo-6-(prop-2-en-1- yl)-5-(trifluoromethyl) phenol as a brown yellow oil. LCMS observed m/z = 360.70 [M-H]- . Step 4: Preparation of 2-[4-chloro-2-hydroxy-3-iodo-6-(trifluoromethyl)phenyl] acetaldehyde. Compound 447.4
A solution of 3-chloro-2-iodo-6-(prop-2-en-1-yl)-5-(trifluoromethyl) phenol (2.0 g, 5.5 mmol) in MTBE (15 mL) and H2O (15 mL) was treated with K2OsO4.2H2O (61 mg, 0.2 mmol) in one portion for 30 min at room temperature under nitrogen atmosphere followed by the addition of NaIO4 (1.4 g, 6.6 mmol). Then the resulting mixture was stirred at 25℃ for 6 hours under nitrogen atmosphere. The reaction was quenched with water at room temperature. The aqueous layer was extracted with CH2Cl2 (3 x 50 mL), dried over anhydrous Na2SO4. After filtration, the mixture was concentrated in vacuo to afford (2.0 g, crude) of 2-[4-chloro-2-hydroxy-3-iodo-6-(trifluoromethyl)phenyl] acetaldehyde as a brown yellow solid. The crude product was used in the next step directly without further purification. Step 5: Preparation of 3-chloro-6-(2-hydroxyethyl)-2-iodo-5-(trifluoromethyl) phenol. Compound 447.5
A solution of 2-[4-chloro-2-hydroxy-3-iodo-6-(trifluoromethyl) phenyl] acetaldehyde (2.0 g, 5.5 mmol) in MeOH (25 mL) was treated with NaBH4 (1.0 g, 27.4 mmol) in one portion at 25 ℃ for 2 hours. The reaction was quenched with 2N HCl. Then the resulting mixture was concentrated under reduced pressure. The resulting mixture was extracted with EA (3 x 10 ml). The combined organic layers were concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (250 mg, 11% yield) of 3-chloro-6-(2- hydroxyethyl)-2-iodo-5-(trifluoromethyl) phenol as a brown yellow oil. LCMS observed m/z = 364.80 [M-H]-. Step 6: Preparation of 6-chloro-7-iodo-4-(trifluoromethyl)-2,3-dihydro-1- benzofuran.
Compound 447.6
To a stirred solution of 3-chloro-6-(2-hydroxyethyl)-2-iodo-5-(trifluoromethyl) phenol (250 mg, 0.7 mmol) and PPh3 (358 mg, 1.3 mmol) in THF (5 mL) were added DIAD (276 mg, 1.3 mmol) in one portion at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 0 °C overnight under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (95 mg, 40% yield) of 6-chloro-7-iodo-4-(trifluoromethyl)-2,3-dihydro-1-benzofuranas a white solid. Step 7: Preparation of 6-chloro-7-iodo-4-(trifluoromethyl)-1-benzofuran. Compound 447.7
A solution of 6-chloro-7-iodo-4-(trifluoromethyl)-2,3-dihydro-1-benzofuran (100 mg, 0.3 mmol) in chlorobenzene (5 mL) was treated with NBS (51 mg, 0.3 mmol) and AIBN (2 mg, 0.01 mmol) in portions at 25 ℃ under N2 atmosphere. The reaction mixture was heated to 70 ℃ and stirred at 70 ℃ for 1 hour under N2 atmosphere. The resulting mixture was cooled to room temperature and purified by silica gel column chromatography(eluent: with 5% EtOAc in petroleum ether) to afford(70 mg, 70% yield) of 6-chloro-7-iodo-4-(trifluoromethyl)-1-benzofuran as a brown yellow oil. Step 8: Preparation of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-1,1- diphenylmethanimine. Compound 447.8
A solution of 6-chloro-7-iodo-4-(trifluoromethyl)-1-benzofuran (60 mg, 0.2 mmol) and benzophenone imine (3 mg, 0.02 mmol) in dioxane (4 mL) was treated with
sodium tert-butoxide (3 mg, 0.017 mmol), XantPhos (20 mg, 0.03 mmol) and Pd2(dba)3 (16 mg, 0.02 mmol) in one portion at 25 ℃ under N2 atmosphere. The reaction mixture was heated to 100 ℃ and stirred at 100 ℃ for 16 hours under N2 atmosphere. The resulting mixture was cooled to room temperature and concentrated in vacuo to afford (50 mg, crude) of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-1,1-diphenylmethanimine as a brown yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 400.05 [M+H]+. Step 9: Preparation of 6-chloro-4-(trifluoromethyl)-1-benzofuran-7-amine. Compound 447.9
A solution of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-1,1- diphenylmethanimine (50 mg, 0.1 mmol) in tetrahydrofuran (3 mL) was treated with HCl (3 mL) in one portion at 25 ℃ for 1hour. The mixture was acidified to pH 7~8 with NaHCO3 and extracted with EA (3 x 5 mL). The combined organic layers were dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 15% EtOAc in petroleum ether) to afford (17 mg, 57% yield) of 6-chloro-4-(trifluoromethyl)-1- benzofuran-7-amine as a brown yellow oil. LCMS observed m/z = 234.03 [M-H]-. Step 10: Preparation of tert-butyl 4-[4-({[6-chloro-4-(trifluoromethyl)-1- benzofuran-7-yl] carbamoyl} methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-6-yl] piperazine-1-carboxylate. Compound 447.10
A solution of {6-[4-(tert-butoxycarbonyl)piperazin-1-yl]-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetic acid (33 mg, 0.06 mmol) and 6-chloro-4-(trifluoromethyl)-1-benzofuran-7-amine (15 mg, 0.06 mmol) in DCM (5 mL) was treated with Bis(2-oxo-3-oxazolid (19 mg, 0.08 mmol) and DIEA (24 mg, 0.2 mmol) in one portion at 25 ℃. The resulting mixture was stirred at 25 ℃ for 2 hours under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure and dissolved in DMF (3 mL). The mixture was purified by reversed phase C18 silica gel column chromatography to afford (16 mg, 33% yield) of tert-butyl 4-[4-({[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl] carbamoyl} methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-6-yl] piperazine-1-carboxylate as a white solid. LCMS observed m/z = 740.05 [M-H]-. Step 11: Preparation of tert-butyl N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7- yl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide. Compound 447.11
A solution of tert-butyl 4-[4-({[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl] carbamoyl} methyl)-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5- a] pyrimidin-6-yl] piperazine-1-carboxylate (15 mg, 0.02 mmol) in DCM (1 mL) was treated with trifluoroacetic acid (0.5 mL) in one portion at 25 ℃ for 30 min. The mixture was concentrated in vacuo to afford (12 mg, crude) of N-[6-chloro-4-(trifluoromethyl)-1- benzofuran-7-yl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide as a brown yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 639.95 [M-H]-.
Step 12: Preparation of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7- yl]acetamide. Compound 447.12
A solution of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-2-[2-(1,3- dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (12 mg, 0.02 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4- carboxylic acid (5 mg, 0.02 mmol) in DMF (1 mL) was treated with TCFH (6 mg, 0.02 mmol) and NMI (8 mg, 0.1 mmol) in one portion at 25 ℃. The resulting mixture was stirred at 25 ℃ for 2 hours under nitrogen atmosphere. The residue was purified by reversed phase C18 silica gel column chromatography to afford (8 mg, 49% yield) of 2-(6-{4-[5- (benzyloxy)-6-methylpyrimidine-4-carbonyl]piperazin-1-yl}-2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl)-N-[6-chloro-4- (trifluoromethyl)-1-benzofuran-7-yl]acetamide as a white solid. LCMS observed m/z = 868.20 [M+H]+. Step 13: Preparation of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide. Compound 447
A mixture of 2-(6-{4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl] piperazin-1- yl}-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4- yl)-N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl] acetamide (6 mg, 0.007 mmol) in trifluoroacetic acid (2 mL) was stirred at 25 ℃ for 16 hours. The resulting mixture was concentrated under reduced pressure. The residue was purified by preparatory HPLC (Column: XBridge BEH C185 μm, 30*150mm; mobile phase: 25-45% ACN in H2O) to afford (2.4 mg, 44% yield) of N-[6-chloro-4-(trifluoromethyl)-1-benzofuran-7-yl]-2-[2- (1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl) piperazin-1-yl]-7-oxo- [1,2,4] triazolo[1,5-a] pyrimidin-4-yl] acetamide as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 8.84 (s, 1H), 8.62 (s, 1H), 8.29 – 8.16 (m, 2H), 7.73 (d, J = 2.2 Hz, 1H), 7.60 (s, 1H), 7.35 (d, J = 7.8 Hz, 1H), 6.97 (s, 1H), 5.71 (d, J = 13.2 Hz, 1H), 5.27 (s, 2H), 5.18 (s, 4H), 4.83 (d, J = 12.7 Hz, 1H), 3.87 (t, J = 11.6 Hz, 2H), 3.53 (s, 1H), 3.28 (d, J = 7.7 Hz, 2H), 3.11 (s, 1H), 2.92 – 2.78 (m, 2H), 2.59 (s, 3H), 1.42 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 778.20 [M+H]+. EXAMPLE 448 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,3-dihydro-5- isobenzofuranyl)-6-ethyl-5-(4-{[4-hydroxy-5-(4-pyridyl)-3-isothiazolyl]carbonyl}-1- piperazinyl)-4-oxo-1,3,3a,7-tetraaza-7-indenyl]acetamide
To a mixture of N-[2-chloro-4-(trifluoromethyl)phenyl](5-{4-[(5-bromo-4- hydroxy-3-isothiazolyl)carbonyl]-1-piperazinyl}-2-(1,3-dihydro-5-isobenzofuranyl)-6- ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl)acetamide (80 mg, 99 µmol, Intermediate H), (4- pyridyl)boranediol (12 mg, 99 µmol) and K3PO4 (63 mg, 297 µmol) in a mixture of dioxane (0.7 mL) and H2O (0.1 mL) was added Pd(PPh3)4 (11 mg, 9.9 µmol). The mixture was stirred at 80 °C under N2 for 16 hours. The mixture was concentrated in vacuo. The residue was dissolved in CH2Cl2 and filtered through a pad of Celite and the filtrate was concentrated in vacuo. The residue was purified by silica gel column chromatography (mobile phase: 0-20% MeOH in CH2Cl2) to afford (5.2 mg, 6% yield) of N-[2-chloro-4- (trifluoromethyl)phenyl][2-(1,3-dihydro-5-isobenzofuranyl)-6-ethyl-5-(4-{[4-hydroxy-5- (4-pyridyl)-3-isothiazolyl]carbonyl}-1-piperazinyl)-4-oxo-1,3,3a,7-tetraaza-7- indenyl]acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.36 (s, 1H), 8.42 (d, J = 5.3 Hz, 2H), 8.03 – 7.94 (m, 3H), 7.90 (s, 1H), 7.72 – 7.61 (m, 3H), 7.38 (d, J = 7.8 Hz, 1H), 5.33 (s, 2H), 4.99 (d, J = 5.2 Hz, 4H), 4.48 (d, J = 12.5 Hz, 1H), 3.89 (d, J = 12.8 Hz, 1H), 3.53 – 3.42 (m, 3H), 3.03 – 2.86 (m, 3H), 2.77 (d, J = 11.7 Hz, 1H), 2.65 (d, J = 11.0 Hz, 1H), 1.16 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 806.4 [M+H]+. EXAMPLE 449 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{6-[4-(5-cyano-3-hydroxy- 2-methylpyridine-4-carbonyl)piperazin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide
Step 1: Preparation of 3-hydroxy-2-methylpyridine-4-carboxylic acid Compound 449.1
A mixture of 5-ethoxy-4-methyl-1,3-oxazole (200 mg, 1.6 mmol) and prop-2-enoic acid (197 mg, 2.7 mmol) was heated to 40 ℃ and stirred at 40 ℃ for 3 h under nitrogen atmosphere. After completion of the reaction, the mixture was cooled to room temperature. The precipitated solids were collected by filtration and washed with H2O (3 x 10 mL). The brown crystalline product was re-crystallized with water and dried under vacuum at 40 ℃. This resulted in 3-hydroxy-2-methylpyridine-4-carboxylic acid (160 mg, 66% yield) as a yellow solid. LCMS observed m/z = 154.04 [M+H]+. Step 2: Preparation of 5-bromo-3-hydroxy-2-methylpyridine-4-carboxylic acid. Compound 449.2
To a stirred mixture of 3-hydroxy-2-methylpyridine-4-carboxylic acid (200 mg, 1.3 mmol) in DMF (5 mL) was added NBS (256 mg, 1.4 mmol) in portions at room temperature. The resulting mixture was heated to 50 ℃ and stirred at 50 ℃ for 1 h under nitrogen atmosphere. After completion of the reaction, the mixture was cooled to room temperature, quenched with water (10 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 5-bromo-3-hydroxy-2-methylpyridine-4-carboxylic acid (201 mg, 66% yield) as a white solid. LCMS observed m/z = 231.95. [M+H]+. Step 3: Preparation of benzyl 3-(benzyloxy)-5-bromo-2-methylpyridine-4- carboxylate. Compound 449.3
To a stirred solution of 5-bromo-3-hydroxy-2-methylpyridine-4-carboxylic acid (50 mg, 0.2 mmol) and K2CO3 (60 mg, 0.43 mmol) in DMF (2 mL) was added benzyl bromide (92 mg, 0.5 mmol) in portions at room temperature. The resulting mixture was heated to 80 ℃ and stirred for 1 h at 80 °C under nitrogen atmosphere. After completion of the reaction, the mixture was cooled to room temperature, quenched with water (5 mL) and extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in benzyl 3-(benzyloxy)-5- bromo-2-methylpyridine-4-carboxylate (80 mg, 81% yield) as a white solid. LCMS observed m/z = 412.05 [M+H]+. Step 4: Preparation of benzyl 3-(benzyloxy)-5-cyano-2-methylpyridine-4- carboxylate. Compound 449.4
To a stirred solution of benzyl 3-(benzyloxy)-5-iodo-2-methylpyridine-4- carboxylate (300 mg, 0.7 mmol) in DMF (5 mL) was added CuCN (64 mg, 0.7 mmol) in one portion at room temperature. The resulting mixture was heated to 110 ℃ and stirred at 110 °C for 2 h under nitrogen atmosphere. After completion of the reaction, the mixture was cooled to room temperature, quenched with water (10 mL) and extracted with EA (3 x 20 mL). The combined organic layers were washed with brine (30 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE/EA (5/1) to afford benzyl 3-(benzyloxy)-5-cyano-2-methylpyridine-4-carboxylate (110 mg, 47% yield) as a white solid. LCMS observed m/z = 359.13 [M+H]+. Step 5: Preparation of 5-cyano-3-hydroxy-2-methylisonicotinic acid. Compound 449.5
To a stirred solution of benzyl 3-(benzyloxy)-5-cyano-2-methylpyridine-4- carboxylate (150 mg, 0.4 mmol) in EA (10 mL) was added Pd/C (10%, 34 mg, 0.04 mmol) in one portion at room temperature under nitrogen atmosphere. The mixture was hydrogenated at room temperature for 2 h under hydrogen atmosphere using a hydrogen balloon. Then the resulting mixture was filtered through a celite pad and the filter cake was washed with EA (3 x 100 mL). The filtrate was concentrated under reduced pressure to afford 5-cyano-3-hydroxy-2-methylisonicotinic acid (67 mg, crude) as a light-yellow solid. The crude product was used in the next step directly without further purification. LCMS observed m/z = 179.04 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-{6-[4-(5-cyano-3- hydroxy-2-methylpyridine-4-carbonyl)piperazin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)- 5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide.
To a stirred solution of 5-cyano-3-hydroxy-2-methylpyridine-4-carboxylic acid (30 mg, 0.2 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (100 mg, 0.2 mmol) in ACN (5 mL) were added TCFH (47 mg, 0.2 mmol) and NMI (41 mg, 0.5 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere. After completion of the reaction, the mixture was quenched with water (10 mL) and extracted with EA (3 x 15 mL). The combined organic layers were washed with brine (15 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C185 μm, 30 *150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29 % B to 45% B in 9 min; Wave Length: 254/220 nm; RT1 (min): 7.73) to afford N-[2-chloro-4-
(trifluoromethyl)phenyl]-2-{6-[4-(5-cyano-3-hydroxy-2-methylpyridine-4- carbonyl)piperazin-1-yl]-2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}acetamide (12.7 mg, 33% yield) as an off-white solid. 1H NMR (400 MHz, Methanol-d4) δ 8.16 (m, 3H), 7.84 (d, J = 2.1 Hz, 1H), 7.63 (t, J = 4.3 Hz, 2H), 7.40 (d, J = 7.9 Hz, 1H), 5.48 (s, 2H), 5.13 (s, 4H), 4.90-4.80 (m, 2H), 3.95-3.65 (m, 2H), 3.65-3.5 (m, 1H), 3.15 (q, J = 7.6 Hz, 3H), 2.89 (s, 2H), 2.50 (s, 3H), 1.35 (t, J = 7.5 Hz, 3H). Note: Two exchangeable protons were not observed in NMR Spectra. LCMS observed m/z = 760.05 [M+H]+. EXAMPLE 450 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-{4-[3-hydroxy-2-methyl-6-(4-methylpiperazin-1-yl)pyridine- 4-carbonyl]piperazin-1-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide Compound 450
Step 1: Preparation of 6-chloro-4-iodo-2-methylpyridin-3-ol. Compound 450.1
To a stirred mixture of 6-chloro-2-methylpyridin-3-ol (2.0 g, 13.9 mmol) and NaHCO3 (3.5 g, 41.7 mmol) in H2O (20 mL) was added I2 (3.9 g, 15.3 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 12 hours. After completion of reaction, the aqueous layer was extracted with EA (3 x 30 mL). The residue was purified by silica gel column chromatography, eluted with PE/EA (6/4) to afford 6- chloro-4-iodo-2-methylpyridin-3-ol (900 mg, 24.0% yield) as a yellow solid. LCMS observed m/z = 269.91 [M+H]+.
Step 2: Preparation of 6-chloro-4-iodo-3-[(4-methoxyphenyl)methoxy]-2- methylpyridine. Compound 450.2
To a stirred mixture of 6-chloro-4-iodo-2-methylpyridin-3-ol (900 mg, 3.3 mmol) and K2CO3 (1.4 g, 9.9 mmol) in acetone (10 mL) was added 4-methoxybenzyl chloride (785 mg, 5.0 mmol) in portions at room temperature. The reaction mixture was heated to 50 °C and stirred at 50 °C for 12 hours. After completion of reaction, the resulting mixture was cooled to room temperature and filtered, the filter cake was washed with MeOH (3 x 10 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (5/1) to afford 6-chloro-4-iodo-3-[(4- methoxyphenyl)methoxy]-2-methylpyridine (800 mg, 61.5% yield) as a white solid. LCMS observed m/z = 389.97 [M+H]+. Step 3: Preparation of methyl 6-chloro-3-[(4-methoxyphenyl)methoxy]-2- methylpyridine-4-carboxylate. Compound 450.3
To a solution of 6-chloro-4-iodo-3-[(4-methoxyphenyl)methoxy]-2-methylpyridine (300 mg, 0.8 mmol) in MeOH (10 mL) was added Pd(dppf)Cl2 (56 mg, 0.1 mmol) and TEA (156 mg, 1.6 mmol) dropwise at room temperature using a pressurized reactor. The mixture was degassed with nitrogen for 5 minutes and then was stirred at 80 °C for 2 hours under carbon monoxide atmosphere (10 atm). After completion of reaction, the reaction mixture was cooled to room temperature and filtered to remove insoluble solids. The filtrate was concentrated under reduced pressure and purified by silica gel column chromatography, eluted with PE/EA (7/1) to afford methyl 6-chloro-3-[(4-methoxyphenyl)methoxy]-2-
methylpyridine-4-carboxylate (140 mg, 56.5% yield) as a white solid. LCMS observed m/z = 322.08 [M+H]+. Step 4: Preparation of methyl 3-((4-methoxybenzyl)oxy)-2-methyl-6-(4- methylpiperazin-1-yl)isonicotinate. Compound 450.4
To a stirred mixture of methyl 6-chloro-3-[(4-methoxyphenyl)methoxy]-2- methylpyridine-4-carboxylate (140 mg, 0.4 mmol), 1-methylpiperazine (65 mg, 0.6 mmol) and Cs2CO3 (425 mg, 1.2 mmol) in dioxane (5 mL) were added Pd2(dba)3 (80 mg, 0.1 mmol) and BINAP (108 mg, 0.2 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100 °C for 2 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled down to room temperature and filtered through a celite pad and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (7/3) to afford methyl 3-((4-methoxybenzyl)oxy)-2-methyl-6-(4-methylpiperazin-1-yl)isonicotinate (120 mg, 71.5% yield) as a white solid. LCMS observed m/z = 386.20 [M+H]+. Step 5: Preparation of 3-[(4-methoxyphenyl)methoxy]-2-methyl-6-(4- methylpiperazin-1-yl)pyridine-4-carboxylic acid. Compound 450.5
To a stirred mixture of methyl 3-[(4-methoxyphenyl)methoxy]-2-methyl-6-(4- methylpiperazin-1-yl)pyridine-4-carboxylate (120 mg, 0.3 mmol) in THF (3 mL) was added LiOH aqueous solution (78 mg dissolved in H2O (1 mL), 1.8 mmol) dropwise at room temperature. The resulting mixture was stirred at room temperature for 5 hours. After completion of reaction, the mixture was concentrated under reduced pressure to remove the
solvent of THF. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 30% gradient in 20 minutes; detector, UV 254 nm) to afford 3-[(4- methoxyphenyl)methoxy]-2-methyl-6-(4-methylpiperazin-1-yl)pyridine-4-carboxylic acid (80 mg, 69.2% yield) as a brown oil. LCMS observed m/z = 372.18 [M+H]+. Step 6: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-(4-{3-[(4-methoxyphenyl)methoxy]-2-methyl-6-(4- methylpiperazin-1-yl)pyridine-4-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide. Compound 450.6
To a stirred mixture of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-7-oxo-6-(piperazin-1-yl)-[1,2,4]triazolo[1,5-a]pyrimidin-4- yl]acetamide (80 mg, 0.1 mmol) and 3-[(4-methoxyphenyl)methoxy]-2-methyl-6-(4- methylpiperazin-1-yl)pyridine-4-carboxylic acid (59 mg, 0.1 mmol) in DMF (2 mL) was added HATU (76 mg, 0.2 mmol) and DIEA (69 μL, 0.3 mmol) dropwise at room temperature. The resulting mixture was stirred at room temperature for 2 hours. After completion of reaction, the mixture was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% NH3.H2O), 60% to 80% gradient in 10 minutes; detector, UV 254 nm) to afford N-[2-chloro- 4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5-ethyl-6-(4-{3-[(4- methoxyphenyl)methoxy]-2-methyl-6-(4-methylpiperazin-1-yl)pyridine-4- carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (50 mg, 33.1% yield) as a white solid. LCMS observed m/z = 955.36 [M+H]+.
Step 7: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2- benzofuran-5-yl)-5-ethyl-6-{4-[3-hydroxy-2-methyl-6-(4-methylpiperazin-1-yl)pyridine- 4-carbonyl]piperazin-1-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 450
To a stirred solution of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro- 2-benzofuran-5-yl)-5-ethyl-6-(4-{3-[(4-methoxyphenyl)methoxy]-2-methyl-6-(4- methylpiperazin-1-yl)pyridine-4-carbonyl}piperazin-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl]acetamide (50 mg, 0.05 mmol) in DCM (1 mL) was added boron trichloride (1 mL, 1.0 mmol, 1 M in DCM) dropwise at 0 °C. The resulting mixture was stirred at room temperature for 1 hour. After completion of reaction, the resulting mixture was concentrated under vacuum. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% NH3.H2O), 60% to 70% gradient in 20 minutes; detector, UV 254 nm) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,3-dihydro-2-benzofuran-5-yl)-5- ethyl-6-{4-[3-hydroxy-2-methyl-6-(4-methylpiperazin-1-yl)pyridine-4-carbonyl]piperazin- 1-yl}-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (8.6 mg, 19.7% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 8.41 (s, 1H), 8.10 – 8.01 (m, 3H), 7.98 (d, J = 2.1 Hz, 1H), 7.76 – 7.69 (m, 1H), 7.46 (d, J = 8.3 Hz, 1H), 6.44 (s, 1H), 5.40 (s, 2H), 5.11 – 8.01 (m, 4H), 4.62 – 4.45 (m, 1H), 3.60 – 3.44 (m, 6H), 3.32 – 3.12 (m, 2H), 3.10 – 2.90 (m, 3H), 2.90 – 2.78 (m, 1H), 2.78 – 2.61 (m, 1H), 2.48 – 2.35 (m, 4H), 2.31 (s, 3H), 2.22 (s, 3H), 1.37 – 1.16 (m, 3H). LCMS observed m/z = 835.25 [M+H]+. EXAMPLE 451 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-3-methyl-5-oxoimidazo[1,2-a]pyrimidin-8(5H)-yl)acetamide Compound 451
Step 1: Preparation of 1-(1,3-dihydroisobenzofuran-5-yl)propan-1-one Compound 451.1
A solution of 5-bromo-1,3-dihydro-2-benzofuran (1 g, 5.0 mmol) in THF (25 mL) was treated with n-BuLi (2.5 M in n-hexane, 2.2 mL, 5.5 mmol) at -78°C for 40 min under nitrogen atmosphere followed by the addition of N-methoxy-N-methylpropionamide (588 mg, 5.0 mmol) dropwise at -78°C. The mixture was heated to 25°C and stirred at 25°C for 1h.The reaction was quenched by the addition of sat. NH4Cl (aq.) (15 mL) at 0°C.The resulting mixture was extracted with EtOAc (3 x 10mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (10/1) to afford 1-(1,3- dihydro-2-benzofuran-5-yl)propan-1-one (600 mg, 67.6% yield) as a white solid. LCMS observed m/z = 177.07 [M+H]+. Step 2: Preparation of 2-bromo-1-(1,3-dihydroisobenzofuran-5-yl)propan-1-one. Compound 451.2
To a stirred mixture of 1-(1,3-dihydro-2-benzofuran-5-yl)propan-1-one (600 mg, 3.4 mmol) in EA (30 mL) was added CuBr2 (1.5 g, 6.8 mmol) in portions at room temperature. The resulting mixture was heated to 70 °C and stirred at 70 °C for overnight under nitrogen atmosphere. The resulting mixture was washed with 3 x 15 mL of brine and dried over Na2SO4 After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5 / 1) to afford 2-
bromo-1-(1,3-dihydro-2-benzofuran-5-yl)ethanone (500 mg, 57.5%yield) as a white solid. LCMS observed m/z = 254.99 [M+H]+. Step 3: Preparation of tert-butyl 4-(2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- methyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 451.3
A solution of tert-butyl 4-(4-ethyl-2-imino-6-oxo-1,3-dihydropyrimidin-5- yl)piperazine-1-carboxylate (500 mg, 1.5 mmol) in DMF (30 mL) was treated with NaH (123 mg, 3.0 mmol, 60%Wt%) at 0°C for 1h under nitrogen atmosphere followed by the addition of 2-bromo-1-(1,3-dihydro-2-benzofuran-5-yl)ethanone (433 mg, 1.7 mmol) dropwise at 0°C. The resulting mixture was stirred at room temperature for 2h under nitrogen atmosphere. To the above mixture was added NaOH (2M/L, 1.39 mL, 2.7 mmol,) dropwise at room temperature. The resulting mixture was stirred at room temperature for additional 1h. The reaction was quenched by the addition of conc HCl(6M, 2 mL) at 0°C. The resulting mixture was extracted with EtOAc (3 x 10mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure, to afford tert-butyl 4-(2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin- 6-yl)piperazine-1-carboxylate (300 mg, crude) as a yellow solid. The crude resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 480.26 [M+H]+. Step 4: Preparation of tert-butyl 4-(8-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- methyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 451.4
To a stirred mixture of tert-butyl 4-(2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- methyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin-6-yl)piperazine-1-carboxylate (300 mg, 0.6 mmol) and DIEA (161 mg, 1.2 mmol) in DMF (10 mL) was added N-[2-chloro-4- (trifluoromethyl)phenyl]-2-iodoacetamide (272 mg, 0.7 mmol) in portions at room temperature under nitrogen atmosphere. The mixture was heated to 50°C and stirred at 50°C for overnight under nitrogen atmosphere. The reaction mixture was quenched by addition of water 20 mL. The aqueous layer was extracted with ethyl acetate (3 x 10 mL). The combined organic phase was washed with brine (3 x 10mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/1) to afford tert-butyl 4-(8-(2-((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin- 6-yl)pipera zine-1-carboxylate (40 mg, 9%yield) as a white solid. LCMS observed m/z = 715.26 [M+H] +. Step 5: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5-oxo-6-(piperazin-1-yl)imidazo[1,2- a]pyrimidin-8(5H)-yl)acetamide.
To a stirred mixture of tert-butyl 4-(8-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- methyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin-6-yl)piperazine-1-carboxylate (40 mg, 0.05 mmol) in DCM (2 mL) was added TFA (0.5 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure to give N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2- (1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5-oxo-6-(piperazin-1-yl)imidazo[1,2- a]pyrimidin-8(5H)-yl)acetamide (40 mg, crude) as a yellow solid. The crude resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 615.21 [M+H] +. Step 6: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5- oxoimidazo[1,2-a]pyrimidin-8(5H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 451.6
To a stirred mixture of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5-oxo-6-(piperazin-1-yl)imidazo[1,2- a]pyrimidin-8(5H)-yl)acetamide (40 mg, 0.06 mmol), DIEA (25 mg, 0.2 mmol) and 5- (benzyloxy)-6-methylpyrimidine-4-carboxylic acid (17 mg, 0.07 mmol) in DMF (3 mL) was added HATU (37 mg, 0.09 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 1h. The reaction was quenched by the addition of water (15mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 10mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The crude resulting mixture was used in the next step directly without further purification. This resulted in 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-2-
(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3-methyl-5-oxoimidazo[1,2-a]pyrimidin-8(5H)- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (30 mg,crude) as a white solid. LCMS observed m/z = 841.28 [M+H]+. Step 7: N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5- yl)-7-ethyl-6-(4-(5-hydro xy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-3-methyl-5- oxoimidazo[1,2-a]pyrimidin-8(5H)-yl)acetamide. Compound 451
Into a 4 mL sealed 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-2-(1,3-dihy droisobenzofuran-5-yl)-7-ethyl-3-methyl-5- oxoimidazo[1,2-a]pyrimidin-8(5H)-yl)-N-(2-chloro-4-(trifluoro methyl)phenyl)acetamide (30 mg, 0.03 mmol) was dissolved in TFA (1 mL) at room temperature. The resulting solution was heated to 80 °C stirred at 80 °C for 30 min. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X select CSH C185 μm, 30 mm X 150 mm; Mobile Phase A: Water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 55% B to 70% B in 7 min; Wave Length: 254 nm/220 nm; RT1(min): 5.73) to afford N-(2-chloro- 4-(trifluoromethyl)phenyl)-2-(2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-6-(4-(5-hydroxy- 6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-3-methyl-5-oxoimidazo[1,2-a]pyrimidin- 8(5H)-yl)acetamide (8.5 mg, 4%yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.87 (s, 1H), 9.23 (s, 1H), 8.61 (s, 1H), 8.41 (d, J = 8.6 Hz, 1H), 7.67 (d, J = 2.0 Hz, 1H), 7.56 (dd, J = 8.6, 2.1 Hz, 1H), 7.43 (d, J = 7.4 Hz, 1H), 7.35 – 7.29 (m, 2H), 5.62 (s, 1H), 5.20 (s, 4H), 4.79 (s, 3H), 3.78 (s, 2H), 3.53 (s, 1H), 3.13 (s, 1H), 2.93 (d, J = 7.6 Hz, 2H), 2.83 (d, J = 22.9 Hz, 2H), 2.70 (s, 3H), 2.59 (s, 3H), 1.28 (t, J = 7.2 Hz, 3H).LCMS observed m/z = 751.40 [M+H]+. EXAMPLE 452
Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-3-(hydroxymethyl)-5-oxoimidazo[1,2-a]pyrimidin-8(5H)- yl)acetamide
Step 1: Preparation of 1-(1,3-dihydroisobenzofuran-5-yl)ethan-1-one. Compound 452.1
To a stirred mixture of 5-bromo-1,3-dihydroisobenzofuran (1.0 g, 5.0 mmol), Et3N (1.5 g, 15.0 mmol,) and tributyl(1-ethoxyethenyl)stannane (2.0 g, 5.5 mmol) in dioxane (10 mL) were added Pd(PPh3)2Cl2 (0.2 g, 0.2 mmol) in one portions at room temperature under nitrogen atmosphere. The resulting mixture was heated to 100 °C and stirred at 100°C for 2 h under nitrogen atmosphere. After the reaction was completed, 4M HCl (3 mL) was added, the resulting mixture was heated to 50 °C and stirred at 50 °C for 30 min under nitrogen atmosphere. The cooled mixture was poured into water (50 mL) and extracted with EtOAc (3 x 50 mL). The combine organic phases were washed with brine (3 x 10 mL), dried over Na2SO4 and concentrated under reduce pressure. The crude product was purified by silica gel chromatography, eluted with PE / EA (4 / 1) to give1-(1,3-dihydroisobenzofuran-5- yl)ethan-1-one (450 mg, 55%yield) as a white solid. LCMS observed m/z = 163.07 [M+H] +. Step 2: Preparation of 2-bromo-1-(1,3-dihydroisobenzofuran-5-yl)ethan-1-one. Compound 452.2
To a stirred mixture of 1-(1,3-dihydroisobenzofuran-5-yl)ethan-1-one (450 mg, 2.7 mmol) in EA (15 mL) was added CuBr2 (743 mg, 3.3 mmol) in portions at room temperature. The resulting mixture was heated to 70 °C and stirred at 70 °C for overnight under nitrogen atmosphere. The resulting mixture was washed with 3 x 15 mL of brine and dried over Na2SO4 After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5 / 1) to afford 2-bromo-1-(1,3-dihydro-2-benzofuran-5-yl)ethanone (400 mg, 60%yield) as a yellow solid. LCMS observed m/z = 241.00 [M+H]+. Step 3: Preparation of tert-butyl 4-(2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-5- oxo-5,8-dihydroimidazo[1,2-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 452.3
To a solution of tert-butyl 4-(6-ethyl-2-imino-4-oxo-1,2,3,4-tetrahydropyrimidin-5- yl)piperazine-1-carboxylate (500 mg, 1.5 mmol) in DMF (15 mL) was added NaH (123 mg, 3.0 mmol, 60%Wt%) in portions at 0 °C under nitrogen atmosphere. The mixture was stirred at 0 °C for 0.5 hour. Then a solution of 2-bromo-1-(1,3-dihydro-2-benzofuran-5-yl)ethanone (447 mg, 1.8 mmol) in DMF (1 mL) was added dropwise at 0 °C. The reaction mixture was poured into ice sat. aq. NH4Cl (30 mL). The mixture was extracted with EA (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to afford tert- butyl 4-(2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-5-oxo-5,8-dihydroimidazo[1,2- a]pyrimidin-6-yl)piperazine-1-carbox ylate (400 mg, crude) as a yellow solid. The crude resulting mixture was used in the next step directly without further purification. LCMS observed m/z = 466.24 [M+H]+. Step 4: Preparation of tert-butyl 4-(2-(1,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- formyl-5-oxo-5,8-dihydroimidazo[1,2-a]pyrimidin-6-yl)piperazine-1-carboxylate. Compound 452.4
To a stirred solution of tert-butyl 4-(2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-5- oxo-5, 8-dihydroimidazo[l,2-a]pyrimidin-6-yl)piperazine-l-carboxylate (400 mg, 0.8 mmol) in DMF (10 mL) was added N-(chloromethylene)-N-methylmethanaminium chloride (274 mg, 2.1 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for Ih under nitrogen atmosphere. The residue was washed with water (20 mL). The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL) and dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1 / 1) to afford tert-butyl 4-(2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3-formyl-5- oxo-5, 8-dihydroimidazo[l,2-a]pyrimidin-6-yl)piperazine-l-carboxylate (320 mg, 75% yield) as a yellow solid. LCMS observed m/z = 494.24 [M+H]+.
Step 5: Preparation of tert-butyl 4-(2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- (hydroxymethyl)-5-oxo-5,8-dihydroimidazo[l,2-a]pyrimidin-6-yl)piperazine-l- carboxylate.
To a stirred mixture of tert-butyl 4-(2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- formyl-5-oxo-5,8-dihydroimidazo[l,2-a]pyrimidin-6-yl)piperazine-l -carboxylate (300 mg, 0.6 mmol) in THF (10 mL) was added LiAlEL (1.0 M in THF , 0.46 mL, 0.4 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at room temperature for Ih under nitrogen atmosphere. The reaction was quenched by the addition of water (0.02 mL) at 0 °C. The reaction was stirred at room temperature for 15 min, dried over anhydrous sodium sulfate. The resulting mixture was filtered, the filter cake was washed with THF (3 x 10 mL). The filtrate was concentrated under reduced pressure to give tert-butyl 4-(2-(l,3-
dihydroisobenzofuran-5-yl)-7-ethyl-3-(hydroxymethyl)-5-oxo-5,8-dihydroimidazo[l,2- a]pyrimidin-6-yl)pi perazine- 1 -carboxylate (300 mg, crude) as a yellow oil. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 496.25 [M+H]+.
Step 6: Preparation of tert-butyl 4-(8-(2-((2-chloro-4-
(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- (hydroxymethyl)-5-oxo-5,8-dihydroimidazo[l,2-a]pyrimidin-6-yl)pi perazine-1- carboxylate.
To a stirred mixture of tert-butyl 4-(2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- (hydroxymethyl)-5-oxo-5,8-dihydroimidazo[l,2-a]pyrimidin-6-yl)piperazine-l- carboxylate (290 mg, 0.6 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2- iodoacetamide (212 mg, 0.6 mmol) in DMF (5 mL) was added DIEA (226 mg, 1.7 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 50°C for overnight under nitrogen atmosphere. The residue was washed with water (20 mL) at 0°C. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous ISfeSCU After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/1) to afford tert-butyl 4-(8-(2- ((2-chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(l,3-dihydroisobenzofuran-5- yl)-7-ethyl-3-(hydroxymethyl)-5-oxo-5,8-dihydroimidazo[l,2-a]pyrimidin-6- yl)piperazine-l -carboxylate (40 mg, 9%yield) as a yellow solid. LCMS observed m/z = 731.25 [M+H]+.
Step 7 : Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(l,3- dihydroisobenzofuran-5-yl)-7-ethyl-3-(hydroxymethyl)-5-oxo-6-(piperazin-l- yl)imidazo[ 1 ,2-a]pyrimidin-8(5H)-yl)acetamide.
To a stirred mixture of tert-butyl 4-(8-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- (hydroxymethyl)-5-oxo-5,8-dihydroimidazo[l,2-a]pyrimidin-6-yl)piperazine-l- carboxylate (40 mg, 0.055 mmol) in DCM (2 mL) was added TFA (0.6 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. This resulted in N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3- (hydroxymethyl)-5-oxo-6-(piperazin-l-yl)imidazo[l,2-a]pyrimidin-8(5H)-yl)acetamide (40 mg, crude) as a yellow solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 631.20 [M+H]+.
Step 8: Preparation of 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-l-yl)-2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3-(hydroxymethyl)-5- oxoimidazo[l,2-a]pyrimidin-8(5H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide.
Compound 452.8
To a stirred mixture of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(l,3- dihydroisobenzofuran-5-yl)-7-ethyl-3-(hydroxymethyl)-5-oxo-6-(piperazin-l- yl)imidazo[l,2-a]pyrimidin-8(5H)-yl)acetamide (30 mg, 0.05 mmol), DIEA (18 mg, 0.1 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (12 mg, 0.05 mmol) in DMF (2 mL) was added HATU (21 mg, 0.05 mmol) in portions at room temperature .The resulting mixture was stirred at room temperature for Ih. The reaction was quenched by the addition of water (5 mL) at 0 °C. The resulting mixture was extracted with EtOAc (3 x 6 mL). The combined organic layers were washed with brine (3 x 3 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Q2 / MeOH 20/1) to afford 2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)piperazin-l-yl)-2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl- 3-(hydroxymethyl)-5-oxoimidazo[l,2-a]pyrimidin-8(5H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (20 mg, 49% yield) as a yellow solid LCMS observed m/z = 857.27 [M+H]+.
Step 9: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(l,3- dihydroisobenzofuran-5-yl)-7-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-l-yl)-3-(hydroxymethyl)-5-oxoimidazo[l,2-a]pyrimidin-8(5H)- yl)acetamide
Into a 4 mL sealed 2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-l-yl)-2-(l,3-dihydroisobenzofuran-5-yl)-7-ethyl-3-(hydroxymethyl)-5- oxoimidazo[l,2-a]pyrimidin-8(5H)-yl)-N-(2-chloro-4-(trifluorom ethyl) phenyl)acetamide (30 mg, 0.03 mmol) was dissolved in TFA (1 mL) at room temperature. The resulting solution was heated to 80 °C stirred at 80 °C for 30 min. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: X-Bridge BEH C18 5pm,
19*250mm; Mobile Phase A: Water(10 mmol/L NH4HCO3+0.1%NH3.H2O), Mobile Phase B: ACN; Flow rate: 25 mL/min; Gradient: 30% B to 45% B in 9 min; Wave Length: 254 nm/220 nm; RT1(min): 8.28) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,3- dihydroisobenzofuran-5-yl)-7-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-3-(hydroxymethyl)-5-oxoimidazo[1,2-a]pyrimidin-8(5H)- yl)acetamide (2.8 mg, 10% yield) as a white solid..1H NMR (400 MHz, DMSO-d6) δ 10.19 (s, 1H), 8.57 (s, 1H), 7.93 (d, J = 2.1 Hz, 1H), 7.86 (d, J = 8.6 Hz, 1H), 7.70 (dd, J = 9.1, 2.2 Hz, 1H), 7.54 – 7.39 (m, 3H), 5.07 (s, 4H), 4.94 (d, J = 5.7 Hz, 3H), 4.64 (s, 2H), 4.52 (d, J = 12.1 Hz, 1H), 3.55 (d, J = 12.2 Hz, 3H),3.33 – 3.01(m, 1H) 2.99 – 2.95 (m, 1H), 2.81 (q, J = 7.6 Hz, 3H), 2.61 (s, 1H), 2.44 (s, 3H), 1.18 (t, J = 7.5 Hz, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 767.20 [M+H]+. EXAMPLE 453 Synthesis of (S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,1-dimethyl-3H-2- benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2- methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 453
Step 1: 2-{2-bromo-5-ethyl-6-[(2S)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)- 2-methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4- (trifluoromethyl)phenyl]acetamide. Compound 453.1
A solution of 2-{6-[(2S)-4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-2- methylpiperazin-1-yl]-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in TFA (2 mL) was stirred at room temperature for 5 min. The reaction mixture was heated to 80 °C and stirred at 80 °C for 0.5 hours. After completed, the resulting mixture was concentrated under vacuum to afford (90 mg, crude) of 2-{2-bromo-5-ethyl-6-[(2S)-4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2-methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2- chloro-4-(trifluoromethyl)phenyl]acetamide as a light yellow oil. The crude product was used in the next step directly without further purification. LCMS observed m/z = 712.09 [M+H]+. Step 2: Preparation of (S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,1- dimethyl-3H-2-benzofuran-5-yl)-5-ethyl-6-[4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2-methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 453
To a stirred solution of (S)-2-{2-bromo-5-ethyl-6-[4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-2-methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) and 2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (42 mg, 0.2 mmol) in dioxane (2 mL) and H2O(0.4 mL)were added K2CO3 (58 mg, 0.4 mmol) and Pd(dppf)Cl2 (10 mg, 0.1 mmol) in portions at room temperature under nitrogen
atmosphere. The reaction mixture was heated to 80 °C and stirred at 80 °C for 2 hours. After completed, the resulting mixture was concentrated under vacuum. The residue was purified by prep-HPLC (Column: XBridge BEH C185μm, 19*250mm; mobile phase: 35-57% ACN in Water (10 mmol/L NH4HCO3+0.1%NH3.H2O)) to afford (63.6 mg, 58% yield) of (S)-N- [2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-5-ethyl-6- [4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2-methylpiperazin-1-yl]-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.93 (s, 1H), 8.99 (s, 1H), 8.68 – 8.60 (m, 1H), 8.54 – 8.49 (m, 1H), 8.24 – 8.19 (m, 1H), 8.13 (s, 1H), 7.65 – 7.62 (m, 1H), 7.58 – 7.53 (m, 1H), 7.25 – 7.21 (m, 1H), 5.75 – 5.56 (m, 1H), 5.35 – 5.23 (m, 1H), 5.18 (s, 1H), 5.13 (s, 2H), 4.89 – 4.74 (m, 1H), 4.01 – 3.91 (m, 1H), 3.86 – 3.74 (m, 1H), 3.57 – 3.44 (m, 1H), 3.18 – 3.01 (m, 2H), 2.96 – 2.67 (m, 2H), 2.62 (s, 3H), 1.55 (s, 6H), 1.45 – 1.34 (m, 3H), 1.06 – 0.92 (m, 3H). LCMS observed m/z = 780.45 [M+H]+. EXAMPLE 454 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] (5-{4-[(1H-1,6-diazainden-7- yl) carbonyl]-1-piperazinyl}-2-(1,1-dimethyl-1,3-dihydro-5-isobenzofuranyl)-6-ethyl-4- oxo-1,3,3a,7-tetraaza-7-indenyl) acetamide
The title compound was prepared using similar procedure as compound 398 using Intermediate AG to afford N-[2-chloro-4-(trifluoromethyl) phenyl] (5-{4-[(1H-1,6- diazainden-7-yl) carbonyl]-1-piperazinyl}-2-(1,1-dimethyl-1,3-dihydro-5- isobenzofuranyl)-6-ethyl-4-oxo-1,3,3a,7-tetraaza-7-indenyl) acetamide as a yellow solid. 1H NMR (400 MHz, DMSO) δ 10.43 (s, 1H), 8.27 (d, J = 5.9 Hz, 1H), 8.13 – 7.95 (m, 5H), 7.95 (s, 1H), 7.77 – 7.68 (m, 1H), 7.41 (d, J = 7.9 Hz, 1H), 6.83 (s, 1H), 5.41 (s, 2H), 5.02 (s, 2H), 4.67 (d, J = 12.5 Hz, 1H), 3.68 – 3.59 (m, 1H), 3.56 – 3.50 (m, 2H), 3.28 – 3.20 (m, 1H), 3.21 – 3.11 (m, 1H), 3.08 – 2.99 (m, 2H), 2.93 (d, J = 11.3 Hz, 1H), 2.73 – 2.65 (m, 1H), 1.45 (s, 6H), 1.23 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 774.3 [M+H]+.
EXAMPLE 455 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] {2-(1,1-dimethyl-1,3-dihydro- 5-isobenzofuranyl)-6-ethyl-5-[4-(3-hydroxy-2-methyl-isonicotinoyl)-1-piperazinyl]-4- oxo-1,3,3a,7-tetraaza-7-indenyl} acetamide Compound 455
The title compound was prepared using similar procedure as compound 455 using Intermediate AG to afford N-[2-chloro-4-(trifluoromethyl) phenyl] {2-(1,1-dimethyl-1,3- dihydro-5-isobenzofuranyl)-6-ethyl-5-[4-(3-hydroxy-2-methyl-isonicotinoyl)-1- piperazinyl]-4-oxo-1,3,3a,7-tetraaza-7-indenyl} acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 9.45 (s, 1H), 8.09 – 7.96 (m, 5H), 7.75 – 7.68 (m, 1H), 7.41 (d, J = 7.9 Hz, 1H), 7.04 (d, J = 4.8 Hz, 1H), 5.40 (s, 2H), 5.02 (s, 2H), 4.54 - 3.45 (m, 1H), 3.58 – 3.42 (m, 2H), 3.32 – 3.29 (m, 1H), 3.17 (d, J = 4.0 Hz, 1H), 3.03 – 2.99 (m, 3H), 2.87 – 2.82 (m, 1H), 2.71 – 2.62 (m, 1H), 2.43 (s, 3H), 1.45 (s, 6H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 765.3 [M+H]+. EXAMPLE 456 Synthesis of N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,1-dimethyl-1,3-dihydro- 5-isobenzofuranyl)-6-ethyl-4-oxo-5-{4-[(3H-1,3,5-triazainden-4-yl) carbonyl]-1- piperazinyl}-1,3,3a,7-tetraaza-7-indenyl] acetamide Compound 456
The title compound was prepared using similar procedure as compound 425 using Intermediate AG to afford N-[2-chloro-4-(trifluoromethyl) phenyl] [2-(1,1-dimethyl-1,3- dihydro-5-isobenzofuranyl)-6-ethyl-4-oxo-5-{4-[(3H-1,3,5-triazainden-4-yl) carbonyl]-1-
piperazinyl}-1,3,3a,7-tetraaza-7-indenyl] acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 8.64 (s, 1H), 8.45 (d, J = 5.7 Hz, 1H), 8.09 – 8.02 (m, 2H), 8.02 – 7.96 (m, 2H), 7.85 (d, J = 5.7 Hz, 1H), 7.75 – 7.68 (m, 1H), 7.41 (d, J = 7.9 Hz, 1H), 5.40 (s, 2H), 5.02 (s, 2H), 4.68 (d, J = 12.3 Hz, 1H), 3.61 – 3.54 (m, 3H), 3.38 – 3.27 (m, 1H), 3.18 – 3.08 (m, 1H), 3.07 – 2.99 (m, 2H), 2.91 (d, J = 11.3 Hz, 1H), 2.69 – 2.62 (m, 1H), 1.45 (s, 6H), 1.22 (t, J = 7.4 Hz, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 775.3 [M+H]+. EXAMPLE 457 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,1-dimethyl-3H-2- benzofuran-5-yl)-5-ethyl-6-[(3R)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3- methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide
Step 1: Preparation of 2-{6-[(3R)-4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-3-methylpiperazin-1-yl]-2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide.
To a stirred mixture of 2-{6-[(3R)-4-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-3-methylpiperazin-1-yl]-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-
4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) and 2-(1,1- dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (37 mg, 0.1 mmol) in 1,4-dioxane (1 mL) and H2O (0.2 mL) were added Cs2CO3 (121 mg, 0.3 mmol) and Pd(dppf)Cl2CH2Cl2 (10 mg, 0.01 mmol) in portions at room temperature under air atmosphere. The resulting mixture was heated to 80 °C and stirred at 80 °C for 2 hours under nitrogen atmosphere. The mixture was cooled to room temperature and filtered. The filter cake was washed with EtOH (3 x 5 mL) and the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18; mobile phase, MeOH in water (0.1% NH3.H2O), 70% to 85% gradient in 10 min; detector, UV 254 nm. The resulting mixture was concentrated under reduced pressure to afford 2-{6-[(3R)-4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-3- methylpiperazin-1-yl]-2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (95 mg, 86.4% yield) as a white solid. LCMS observed m/z = 870.40 [M+H]+. Step 2: Preparation of N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-5-ethyl-6-[(3R)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3- methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide. Compound 457
A solution of 2-{6-[(3R)-4-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-3- methylpiperazin-1-yl]-2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4-yl}-N-[2-chloro-4-(trifluoromethyl)phenyl]acetamide (100 mg, 0.1 mmol) in trifluoroacetic acid (2 mL) was heated to 80 °C and stirred at 80 °C for 20 min under nitrogen atmosphere. The mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 5 μm, 19*250 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1% NH3.H2O), Mobile Phase B: ACN; Flow rate: 25
mL/min; Gradient: 35% B to 57%B in 7 min; Wave Length: 254 nm/220 nm; RT1 (min): 6.25) to afford N-[2-chloro-4-(trifluoromethyl)phenyl]-2-[2-(1,1-dimethyl-3H-2- benzofuran-5-yl)-5-ethyl-6-[(3R)-4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-3- methylpiperazin-1-yl]-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4-yl]acetamide (38.0 mg, 41% yield) as a white solid.1H NMR (400 MHz, DMSO-d6) δ 10.42 (s, 1H), 10.09 (s, 1H), 8.58 (d, J = 4.1 Hz, 1H), 8.05 (t, J = 8.4 Hz, 2H), 8.01 – 7.96 (m, 2H), 7.75 – 7.69 (m, 1H), 7.41 (d, J = 7.9 Hz, 1H), 5.46 – 5.33 (m, 2H), 5.02 (s, 2H), 3.80 – 3.43 (m, 3H), 3.30 – 3.08 (m, 2H), 3.00 – 2.81 (m, 2H), 2.75 – 2.64 (m, 1H), 2.60 – 2.54 (m, 1H), 2.46 (d, J = 3.8 Hz, 3H), 1.53 – 1.35 (m, 9H), 1.24 (d, J = 7.4 Hz, 3H). LCMS observed m/z = 780.20. EXAMPLE 458 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-((1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide Compound 458
Step 1: Preparation of tert-butyl (1S,6S)-5-(1-methoxy-1,3-dioxopentan-2-yl)-2,5- diazabicyclo[4.2.0]octane-2-carboxylate. Compound 458.1
To a stirred solution of tert-butyl (1S,6S)-2,5-diazabicyclo[4.2.0]octane-2- carboxylate (100 mg, 0.5 mmol) and methyl 2-chloro-3-oxopentanoate (71 uL, 0.5 mmol) in ACN (1 mL) was added K2CO3 (195 mg, 1.5 mmol) in portions at room temperature
under nitrogen atmosphere. The reaction mixture was heated to 40 °C and stirred at 40 °C for 16 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to room temperature. After filtration, the filter cake was washed with EtOAc (3 x 20 mL). and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography and eluted with PE / EA (4:1) to afford tert-butyl (1S,6S)-5-(1-methoxy-1,3-dioxopentan-2-yl)-2,5-diazabicyclo[4.2.0]octane-2-carboxylate (100 mg, 62% yield) as a colorless oil. LCMS observed m/z = 341.05 [M+H]+. Step 2: Preparation of 6-((1S,6S)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5- ethyl-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one. Compound 458.2
To a stirred solution of tert-butyl (1S,6S)-5-(1-methoxy-1,3-dioxopentan-2-yl)-2,5- diazabicyclo[4.2.0]octane-2-carboxylate (800 mg, 2.4 mmol) and 3-bromo-1H-1,2,4- triazol-5-amine (421 mg, 2.6 mmol) in EtOH (2 mL) was added H3PO4 (1.2 g, 11.7 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 80 °C and stirred at 80 °C for 16 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to room temperature, and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 30% gradient in 10 min; detector, UV 254 nm. This resulted in 6-((1S,6S)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5-ethyl- [1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one (250 mg, 23% yield) as a light yellow solid. LCMS observed m/z = 352.90 [M+H]+. Step 3: Preparation of 6-((1S,6S)-5-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5-ethyl-[1,2,4]triazolo[1,5- a]pyrimidin-7(4H)-one. Compound 458.3
To a stirred solution of 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (274 mg, 1.1 mmol) and DIEA (488 uL, 2.8 mmol) in DMF (2 mL) were added HATU (710 mg, 1.8 mmol) and 6-((1S,6S)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5-ethyl- [1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one (330 mg, 0.9 mmol) in portions at room temperature under nitrogen atmosphere. The reaction mixture was heated to 45 °C and stirred at 45 °C for 2 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 6-((1S,6S)-5-(5- (benzyloxy)-6-methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo- 5-ethyl-[1,2,4]triazolo[1,5-a]pyrimidin-7(4H)-one (250 mg, 46% yield) as an off-white solid. LCMS observed m/z = 578.95 [M+H]+. Step 4: Preparation of 2-(6-((1S,6S)-5-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide.
To a stirred solution of 6-((1S,6S)-5-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5-ethyl-[1,2,4]triazolo[1,5- a]pyrimidin-7(4H)-one (320 mg, 0.5 mmol) and N-(2-chloro-4-(trifluoromethyl)phenyl)-2- iodoacetamide (301 mg, 0.8 mmol) in DMF (2 mL) was added DIEA (288 uL, 1.6 mmol) dropwise at room temperature under nitrogen atmosphere. The reaction mixture was heated to 45 °C and stirred at 45 °C for 2 hours under nitrogen atmosphere. After completion of reaction, the resulting mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, ACN in Water (10 mmol/L NH4HCO3), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in 2-(6- ((1S,6S)-5-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan- 2-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (300 mg, 66% yield) as an off-white solid. LCMS observed m/z = 814.10 [M+H]+. Step 5: Preparation of 2-(6-((1S,6S)-5-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 458.5
Step 6: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-((1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 458
A solution of 2-(6-((1S,6S)-5-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (100 mg, 0.1 mmol) in TFA (2 mL) was heated to 80 °C and stirred at 80 °C for 30 minutes under nitrogen atmosphere. Then the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 m, 30 mm * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 53% B in 8 min; Wave Length: 254/220 nm; RT1(min): 6.13) to afford N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6- ((1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan-2- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (63.7 mg, 71% yield) as an off-white solid.1H NMR (400 MHz, Methanol-d4) δ 8.45 (s, 1H), 8.18 (t, J = 7.2 Hz, 2H), 8.09 (s, 1H), 7.84 (d, J = 2.5 Hz, 1H), 7.63 (d, J = 6.8 Hz, 1H), 7.33 (d, J = 7.9 Hz, 1H), 5.47 (s, 2H), 5.11 (s, 2H), 4.70 (s, 1H), 3.91 (s, 1H), 3.68 (s, 2H), 3.55 (d, J = 6.4 Hz, 2H), 3.26 (m, 3H), 3.16 – 3.10 (m, 1H), 2.51 (s, 3H), 1.82 (s, 1H), 1.66 (s, 1H), 1.53 (s, 6H), 1.36 (t, J = 7.5 Hz, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 792.15 [M+H]+. EXAMPLE 459 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-((1S,6R)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 459
EXAMPLE 460 Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-((1R,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide.
Step 1: Preparation of 2-(6-(5-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide Compound 459.1/460.1
To a stirred solution of 2-(6-(5-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)- 2,5-diazabicyclo[4.2.0]octan-2-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-
4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide (100 mg, 0.1 mmol) and 2- (1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (37 mg, 0.1 mmol) in dioxane (0.5 mL) and H2O (0.1 mL) were added Cs2CO3 (120 mg, 0.4 mmol) and Pd(dppf)Cl2 (9 mg, 0.01 mmol) in one portion at room temperature under nitrogen atmosphere. The resulting mixture was heated to 80 °C stirred at 80 °C for 2 h under nitrogen atmosphere. After completion of the reaction, the mixture was allowed to cool down to room temperature and concentrated under reduced pressure. The residue was dissolved in water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with water (10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (eluent with 5% petroleum ether in EtOAc) to afford 2-(6-(5-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)acetamide (90 mg, 83.5% yield) as a white solid. LCMS observed m/z = 822.30 [M+H] +. Step 2: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)- 2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 459.2/460.2
Into a 8 mL vial were added 2-(6-(5-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5- yl)-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (90 mg, 0.1 mmol) and TFA (2 mL) dropwise at room temperature. The resulting mixture was heated to 80 °C and stirred at 80 °C for 1 h. After completion of the reaction, the mixture was allowed to cool down to room temperature and
concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column 130, 5 μm, 30 mm * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3+0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 30% B to 48% B in 8 min; Wave Length: 254 / 220 nm; RT1 (min): 6.95) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide (48.3 mg, 59.8% yield) as a white solid. LCMS observed m/z = 868.29 [M+H] +. Step 3: Preparation of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-((1S,6R)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 459
The solution of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (50 mg, 0.06 mmol, 1.0 equiv.) in MeOH was purified by Prep-Chiral-HPLC with the following conditions (Column: CHIRALPAK IF 2*25 cm, 5 μm; Mobile Phase A: Hex (0.3% FA)--HPLC, Mobile Phase B: MeOH: DCM=1:1; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1 (min): 5.7; RT2 (min): 8.5; Sample Solvent: MeOH; Injection Volume: 0.7 mL; Number of Runs: 3) to afford N-(2-chloro-4- (trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6- ((1S,6R)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan-2- yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (15.5 mg, 31.0% yield) as a light yellow solid.1H NMR (400 MHz, Methanol-d4) δ 8.62 – 8.55 (m, 1H), 8.18 (t, J = 9.0
Hz, 2H), 8.09 (s, 1H), 7.84 (s, 1H), 7.63 (d, J = 8.4 Hz, 1H), 7.32 (d, J = 7.8 Hz, 1H), 5.68 – 5.59 (m, 1H), 5.43 – 5.34 (m, 1H), 5.10 (s, 2H), 4.59 – 4.37 (m, 2H), 3.87 – 3.82 (m, 1H), 3.74 – 3.59 (m, 2H), 3.58 – 3.47 (m, 1H), 3.12 – 3.00 (m, 1H), 2.92 – 2.82 (m, 2H), 2.55 (d, J = 8.0 Hz, 3H), 2.44 – 2.20 (m, 1H), 1.91 – 1.77 (m, 1H), 1.52 (s, 7H), 1.43 – 1.33 (m, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 792.10 [M+H] +. Synthesis of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-((1R,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 460
The solution of N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (50 mg, 0.06 mmol) in MeOH was purified by Prep-Chiral-HPLC with the following conditions (Column: CHIRALPAK IF 2*25 cm, 5 μm; Mobile Phase A: Hex (0.3% FA)-- HPLC, Mobile Phase B: MeOH: DCM=1:1; Flow rate: 20 mL/min; Gradient: isocratic; Wave Length: 220 nm; RT1 (min): 5.7; RT2 (min): 8.5; Sample Solvent: MeOH; Injection Volume: 0.7 mL; Number of Runs: 3) to afford N-(2-chloro-4-(trifluoromethyl)phenyl)-2- (2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-((1R,6S)-5-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl)-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (17.6 mg, 35.2% yield) as a light yellow solid.1H NMR (400 MHz, Methanol-d4) 1H NMR (400 MHz, Methanol-d4) δ 8.62 – 8.54 (m, 1H), 8.18 (t, J = 8.8 Hz, 2H), 8.09 (s, 1H), 7.84 (s, 1H), 7.63 (d, J = 8.6 Hz, 1H), 7.32 (d, J = 7.8 Hz, 1H), 5.68 – 5.59 (m, 1H), 5.43 – 5.34 (m, 1H), 5.10 (s, 2H), 4.59 – 4.35 (m, 2H), 3.87 – 3.82 (m, 1H), 3.74 – 3.59 (m, 2H), 3.58 – 3.50 (m, 1H), 3.11 – 3.00 (m, 1H),
2.95 – 2.80 (m, 2H), 2.62 – 2.49 (m, 3H), 2.44 – 2.20 (m, 1H), 1.90 – 1.77 (m, 1H), 1.65 – 1.47 (m, 7H), 1.44 – 1.35 (m, 3H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 792.15 [M+H] +. EXAMPLE 461 Synthesis of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5- methyl-1,4-diazepan-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide. Compound 461
Step 1: Preparation of tert-butyl (7R)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7- methyl-1,4-diazepane-1-carboxylate. Compound 461.1
To a stirred solution of tert-butyl (7R)-7-methyl-1,4-diazepane-1-carboxylate (3.0 g, 14.0 mmol,) and methyl 2-chloro-3-oxopentanoate (2.1 mL, 15.4 mmol) in ACN (20 mL) was added K2CO3 (5.8 g, 42.0 mmol) in portions at room temperature. The resulting mixture was stirred at 40 °C for 16 h. The resulting mixture was filtered, the filter cake was washed with EA (3 x 20 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE/EA (3/1) to afford tert- butyl (7R)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7-methyl-1,4-diazepane-1-carboxylate (3.0 g, 62% yield) as a colorless oil. LCMS observed m/z = 343.22 [M+H]+. Step 2: Preparation of tert-butyl (R)-4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-7-methyl-1,4-diazepane-1-carboxylate.
Compound 461.2
To a stirred solution of tert-butyl (7R)-4-(1-methoxy-1,3-dioxopentan-2-yl)-7- methyl-1,4-diazepane-1-carboxylate (3.0 g, 8.7 mmol) and 3-bromo-1H-1,2,4-triazol-5- amine (1.6 g, 9.6 mmol) in EtOH (6 mL) was added H3PO4 (4.3 g, 43.8 mmol) in portions at room temperature. The resulting mixture was stirred at 80 °C for 16 h. Then the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 30% gradient in 10 min; detector, UV 254 nm) to afford tert-butyl (R)-4-(2-bromo-5-ethyl-7- oxo-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-7-methyl-1,4-diazepane-1- carboxylate (1.0 g, 25% yield) as an off-white solid. LCMS observed m/z = 455.36 [M+H]+. Step 3: Preparation of tert-butyl (R)-4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-7-methyl-1,4-diazepane-1-carboxylate. Compound 461.3
To a stirred solution of tert-butyl (R)-4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-7-methyl-1,4-diazepane-1-carboxylate (1.3 g, 2.8 mmol) and N-[2-chloro-4-(trifluoromethyl)phenyl]-2-iodoacetamide (1.1 g, 3.1 mmol) in DMF (10 mL) was added DIEA (1.5 mL, 8.5 mmol) dropwise at room temperature. The resulting mixture was stirred at 45 °C for 16 h. After completion of reaction, the resulting mixture was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (10 mmol/L NH4HCO3), 10% to 50%
gradient in 10 min; detector, UV 254 nm) to afford tert-butyl (R)-4-(2-bromo-4-(2-((2- chloro-4-(trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-7-methyl-1,4-diazepane-1-carboxylate (1.3 g, 65% yield) as an off-white solid. LCMS observed m/z = 690.13 [M+H]+. Step 4: Preparation of (R)-2-(2-bromo-5-ethyl-6-(5-methyl-1,4-diazepan-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide. Compound 461.4
To a stirred solution of tert-butyl (R)-4-(2-bromo-4-(2-((2-chloro-4- (trifluoromethyl)phenyl)amino)-2-oxoethyl)-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)-7-methyl-1,4-diazepane-1-carboxylate (1.5 g, 2.1 mmol) in DCM (5 mL) was added TFA (5 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 1 h. The resulting mixture was concentrated under reduced pressure. This resulted in (R)-2-(2-bromo-5-ethyl-6-(5-methyl-1,4-diazepan- 1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (1.0 g, crude) as an off-white solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 590.13 [M+H]+. Step 5: Preparatio of (R)-2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)- 5-methyl-1,4-diazepan-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin- 4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 461.5
To a stirred solution of (R)-2-(2-bromo-5-ethyl-6-(5-methyl-1,4-diazepan-1-yl)-7- oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (1.0 g, 1.7 mmol) and 5-(benzyloxy)-6- methylpyrimidine-4-carboxylic acid (496 mg, 2.0 mmol) in ACN (10 mL) were added TCFH (712 mg, 2.5 mmol) and NMI (675 uL, 8.5 mmol) dropwise at room temperature. The resulting mixture was stirred at 40 °C for 2 h under nitrogen atmosphere. Then the reaction mixture was cooled to room temperature and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with the following conditions (column, C18 silica gel; mobile phase, ACN in water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm) to afford (R)-2-(6-(4-(5-(benzyloxy)-6- methylpyrimidine-4-carbonyl)-5-methyl-1,4-diazepan-1-yl)-2-bromo-5-ethyl-7-oxo- [1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl) acetamide (650 mg, 47% yield) as an off-white solid. LCMS observed m/z = 816.16 [M+H]+. Step 6: Preparation of (R)-2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-5-methyl-1,4-diazepan-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)acetamide. Compound 461.6
Into a 20 mL vial were added (R)-2-(6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)-5-methyl-1,4-diazepan-1-yl)-2-bromo-5-ethyl-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl) acetamide (260 mg, 0.3 mmol) and TFA (5 mL) dropwise at room temperature. The resulting mixture was stirred at 80 °C for 20 min. Then the reaction mixture was cooled to room temperature and concentrated under reduced pressure. This resulted in (R)-2-(2-bromo-5-ethyl-6-(4-(5- hydroxy-6-methylpyrimidine-4-carbonyl)-5-methyl-1,4-diazepan-1-yl)-7-oxo- [1,2,4]triazolo [1,5-a]pyrimidin-4(7H)-yl)-N-(2-chloro-4- (trifluoromethyl)phenyl)acetamide (270 mg, crude) as a brown oil. LCMS observed m/z = 726.11 [M+H]+. Step 7: Preparation of (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-5-methyl-1,4-diazepan-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)- yl)acetamide. Compound 461
To a stirred mixture of (R)-2-(2-bromo-5-ethyl-6-(4-(5-hydroxy-6- methylpyrimidine-4-carbonyl)-5-methyl-1,4-diazepan-1-yl)-7-oxo-[1,2,4]triazolo[1,5- a]pyrimidin-4(7H)-yl)-N-(2-chloro-4-(trifluoromethyl) phenyl)acetamide (270 mg, 0.3 mmol) and 2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (204 mg, 0.7 mmol) in dioxane (5 mL) and H2O (1 mL) were added Pd(dppf)Cl2 (27 mg, 0.03 mmol) and K2CO3 (154 mg, 1.1 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 80 °C for 2 h under nitrogen atmosphere. The reaction was quenched with water (20 mL) at room temperature. The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by
reversed-phase flash chromatography with the following conditions (column: X Bridge BEH C18 OBD Prep Column 130 m , 5 um, 30 mm * 150 mm; Mobile Phase A: water (10 mmol/L NH4HCO3 + 0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 31% B to 44% B in 9 min; Wave Length: 254 nm /220 nm; RT 1 (min): 8.95) to afford (R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-5-ethyl-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-5- methyl-1,4-diazepan-1-yl)-7-oxo-[1,2,4]triazolo[1,5-a]pyrimidin-4(7H)-yl)acetamide (53.6 mg, 17% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.38 (s, 1H), 8.51 – 8.48 (m, 1H), 8.14 – 7.93 (m, 4H), 7.76 – 7.65 (m, 1H), 7.41 (d, J = 7.9 Hz, 1H), 5.40 – 5.37 (m, 2H), 5.02 (s, 2H), 4.64 – 4.14 (m, 1H), 3.67 – 3.46 (m, 3H), 3.24 – 3.10 (m, 1H), 2.98 – 2.96 (m, 2H), 2.81 – 2.62 (m, 2H), 2.46 – 2.41 (m, 3H), 2.37 – 2.02 (m, 1H), 1.80 – 1.76 (m, 1H), 1.45 (s, 6H), 1.29 – 1.16 (m, 5H), 1.09 (d, J = 6.4 Hz, 1H). Note: One exchangeable proton was not observed in NMR Spectra. LCMS observed m/z = 794.25 [M+H]+. EXAMPLE 462 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-dimethyl-1,3-dihydro- 5-isobenzofuranyl)-6-ethyl-5-[4-(3-hydroxy-4-oxo-7-oxa-1,4a-diaza-4,5,6,8-tetrahydro-2- naphthoyl)-1-piperazinyl]-4-oxo-1,3,3a,7-tetraaza-4,7-dihydro-7-indenyl]acetamide.
The title compound was prepared using similar procedure as compound 32 using the intermediate AG to afford N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-dimethyl-1,3- dihydro-5-isobenzofuranyl)-6-ethyl-5-[4-(3-hydroxy-4-oxo-7-oxa-1,4a-diaza-4,5,6,8- tetrahydro-2-naphthoyl)-1-piperazinyl]-4-oxo-1,3,3a,7-tetraaza-4,7-dihydro-7- indenyl]acetamide as a yellow solid.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 9.91 (s, 1H), 8.16 – 7.89 (m, 4H), 7.76 – 7.69 (m, 1H), 7.41 (d, J = 7.9 Hz, 1H), 5.40 (s, 2H), 5.03 (s, 2H), 4.60 (s, 2H), 4.49 (d, J = 12.4 Hz, 1H), 4.05 (t, J = 5.3 Hz, 2H), 3.87 (t, J = 5.4 Hz, 2H), 3.57 – 3.41 (m, 3H), 3.29 – 3.20 (m, 1H), 3.08 – 2.89 (m, 3H), 2.82 (d, J = 10.9 Hz, 1H), 2.68 (d, J = 10.0 Hz, 1H), 1.45 (s, 6H), 1.22 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 825.1 [M+H]+.
EXAMPLE 463 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-dimethyl-1,3-dihydro- 5-isobenzofuranyl)-6-ethyl-5-{4-[(3-hydroxy-7-oxa-1-aza-5,8-dihydro-6H-naphth-2- yl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-4,7-dihydro-7-indenyl]acetamide. Compound 463
The title compound was prepared using similar procedure as compound 32 using the intermediate AG to afford N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-dimethyl-1,3- dihydro-5-isobenzofuranyl)-6-ethyl-5-{4-[(3-hydroxy-7-oxa-1-aza-5,8-dihydro-6H- naphth-2-yl)carbonyl]-1-piperazinyl}-4-oxo-1,3,3a,7-tetraaza-4,7-dihydro-7- indenyl]acetamide.1H NMR (400 MHz, DMSO) δ 10.42 (s, 1H), 10.25 (s, 1H), 8.05 (t, J = 7.9 Hz, 2H), 8.01 – 7.97 (m, 2H), 7.72 (dd, J = 8.7, 2.1 Hz, 1H), 7.41 (d, J = 7.9 Hz, 1H), 7.07 (s, 1H), 5.40 (s, 2H), 5.02 (s, 2H), 4.70 – 4.40 (m, 3H), 3.87 (t, J = 5.7 Hz, 2H), 3.57 – 3.43 (m, 3H), 3.30 – 3.19 (m, 1H), 3.09 – 2.90 (m, 3H), 2.82 (d, J = 5.9 Hz, 3H), 2.65 (d, J = 11.2 Hz, 1H), 1.45 (s, 6H), 1.21 (t, J = 7.5 Hz, 3H). LCMS observed m/z = 807.2 [M+H]+. EXAMPLE 464 Synthesis of N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-dimethyl-1,3-dihydro- 5-isobenzofuranyl)-6-ethyl-5-[4-({3-[(p-methoxyphenyl)methoxy]-6-oxa-1-aza-7,8- dihydro-5H-naphth-2-yl}carbonyl)-1-piperazinyl]-4-oxo-1,3,3a,7-tetraaza-4,7-dihydro-7- indenyl]acetamide. Compound 464
The title compound was prepared using similar procedure as compound 32 using the intermediate AG to afford N-[2-chloro-4-(trifluoromethyl)phenyl][2-(1,1-dimethyl-1,3- dihydro-5-isobenzofuranyl)-6-ethyl-5-[4-({3-[(p-methoxyphenyl)methoxy]-6-oxa-1-aza- 7,8-dihydro-5H-naphth-2-yl}carbonyl)-1-piperazinyl]-4-oxo-1,3,3a,7-tetraaza-4,7- dihydro-7-indenyl]acetamide as a white solid.1H NMR (400 MHz, DMSO) δ 10.49 – 10.27 (m, 1H), 10.27 – 10.11 (m, 1H), 8.06 – 7.79 (m, 4H), 7.68 – 7.61 (m, 1H), 7.34 (d, J = 7.9 Hz, 1H), 6.90 (s, 1H), 5.32 (s, 2H), 4.95 (s, 2H), 4.60 (s, 2H), 4.48 (d, J = 12.4 Hz, 1H), 3.88 (t, J = 5.8 Hz, 2H), 3.53 – 3.37 (m, 3H), 3.22 – 3.10 (m, 1H), 3.01 – 2.84 (m, 3H), 2.79 – 2.67 (m, 3H), 2.59 (d, J = 11.4 Hz, 1H), 1.38 (s, 6H), 1.14 (t, J = 7.4 Hz, 3H). LCMS observed m/z = 807.2 [M+H]+. 1.465 Example 465 Synthesis of (10R,12R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide.
Step 1: Preparation of tert-butyl (4R)-5-[(4S)-4-benzyl-2-oxo-1,3-oxazolidin-3-yl]- 4-methyl-5-oxopentanoate. Compound 465.1
A solution of tetrakis(propan-2-yloxy)titanium (4.4 mL, 14.9 mmol) in DCM (250 mL) was treated with Titanium(IV) chloride (41.0 mL, 41.0 mmol, 1.0 M in DCM) at 0 °C for 10 min under nitrogen atmosphere followed by the addition of DIEA (11.5 mL, 66.0 mmol) in portions at 0 °C. The resulting mixture was stirred at 0 °C for 10 min and ((4S)- 4-benzyl-3-propanoyl-1,3-oxazolidin-2-one (13.5 g, 57.9 mmol) was added as a solid. The resulting mixture was stirred at 0 °C for 1.5 h, then tert-butyl prop-2-enoate (12.5 mL, 85.0 mmol) was added dropwise. The mixture was stirred at room temperature for 16 hours. The reaction was quenched with 100 mL of sat. NH4Cl (aq.) at 0 °C. The aqueous layer was extracted with CH2Cl2 (2 x 100 mL). The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 17% EtOAc in petroleum ether) to afford (17.1 g, 82% yield) of tert-butyl (4R)-5-[(4S)-4-benzyl- 2-oxo-1,3-oxazolidin-3-yl]-4-methyl-5-oxopentanoate as a yellow oil. LCMS observed m/z = 362.05 [M+H]+. Step 2: Preparation of (2R)-5-(tert-butoxy)-2-methyl-5-oxopentanoic acid. Compound 465.2
A solution of tert-butyl (4R)-5-[(4S)-4-benzyl-2-oxo-1,3-oxazolidin-3-yl]-4- methyl-5-oxopentanoate (17.0 g, 47.0 mmol) in H2O (75 mL) and THF (150 mL) was treated with H2O2 (37.3 mL, 329.2 mmol, 30%) at 0 °C for 10 min followed by the addition of LiOH.H2O (4.0 g, 94.0 mmol) in portions at 0 °C. The resulting mixture was stirred at 0 °C for 1 h. The reaction was quenched by the addition of 10% Na2SO3 solution (200 mL) at 0 °C. The resulting mixture was concentrated under reduced pressure. To the resulting aqueous residue was added 2N NaOH (6 mL) and the aqueous mixture was washed twice with DCM. The aqueous layer was acidified to pH 1 with HCl (aq.). The aqueous layer was extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (3 x 400 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated
under reduced pressure. This resulted in (7.8 g, 82% yield) of (2R)-5-(tert-butoxy)-2- methyl-5-oxopentanoic acid as a colorless oil. LCMS observed m/z = 203.10 [M+H]+. Step 3: Preparation of 7-tert-butyl 1-ethyl (4R)-4-methyl-3-oxoheptanedioate. Compound 465.3
A solution of (2R)-5-(tert-butoxy)-2-methyl-5-oxopentanoic acid (3.0 g, 14.8 mmol) in THF (60 mL) was treated with N,N-Carbonyldiimidazole (3.6 g, 22.3 mmol) at room temperature overnight. To a stirred solution of potassium 3-ethoxy-3-oxopropanoate (2.5 g, 14.8 mmol) and MgCl2 (2.8 g, 29.7 mmol) in THF (60 mL) were added TEA (8.3 mL, 59.3 mmol) and DMAP (181 mg, 1.5 mmol) in portions at room temperature. The resulting mixture was stirred at room temperature for 2 h under nitrogen atmosphere, then the first solution was added and the mixture was stirred at 80 °C for 22 hours. The reaction was quenched by the addition of water (100 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% EtOAc in petroleum ether) to afford (2.6 g, 65% yield) of 7-tert-butyl 1-ethyl (4R)-4-methyl-3-oxoheptanedioate as a yellow oil. LCMS observed m/z = 270.90 [M-H]-. Step 4: Preparation of (4R)-4-{2-bromo-7-oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin- 5-yl}pentanoate. Compound 465.4
To a stirred solution of 7-tert-butyl 1-ethyl (4R)-4-methyl-3-oxoheptanedioate (2.1 g, 7.8 mmol) and 5-bromo-2H-1,2,4-triazol-3-amine (4.1 g, 25.0 mmol) in t-BuOH (5.4 mL) were added tetraethyl silicate (3.5 mL, 15.6 mmol) and TFA (599 uL, 7.8 mmol) dropwise at room temperature. The resulting mixture was stirred at 85 °C for 18 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (eluent: with 10% MeOH in DCM) to
afford (1.2 g, 42% yield) of tert-butyl (4R)-4-{2-bromo-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-5-yl}pentanoate as a yellow solid. LCMS observed m/z = 370.95 [M+H]+. Step 5: Preparation of tert-butyl (4R)-4-[2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-7- oxo-4H-[1,2,4]triazolo[1,5-a]pyrimidin-5-yl]pentanoate. Compound 465.5
To a stirred solution of 2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane (2.1 g, 7.0 mmol) and tert-butyl (4R)-4-{2-bromo-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-5-yl}pentanoate (2.2 g, 6.0 mmol) in dioxane (20 mL) and water (4 mL) were added Cs2CO3 (2.0 g, 6.0 mmol) and Pd(dppf)Cl2CH2Cl2 (491 mg, 0.6 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 100 °C overnight under nitrogen atmosphere. The reaction mixture was purified by reversed phase C18 silica gel column chromatography to afford (1.4 g, 53% yield) of tert-butyl (4R)-4-[2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo-4H- [1,2,4]triazolo[1,5-a]pyrimidin-5-yl]pentanoate as an off-white oil. LCMS observed m/z = 439.05 [M+H]+. Step 6: Preparation of tert-butyl (10R)-8-bromo-4-(1,1-dimethyl-3H-2-benzofuran- 5-yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12- carboxylate. Compound 465.6
A solution of LiHMDS (9.6 mL, 9.6 mmol, 1M in THF) was treated with a solution of tert-butyl (4R)-4-[2-(1,1-dimethyl-3H-2-benzofuran-5-yl)-7-oxo-4H-[1,2,4]triazolo[1,5- a]pyrimidin-5-yl]pentanoate (14 g 32 mmol) in THF (10 mL) at -78 °C for 2 h under
nitrogen atmosphere followed by the addition of NBS (852 mg, 4.8 mmol) dropwise at -78 °C. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. The reaction was quenched by the addition of MeOH (10 mL) at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (800 mg, 48% yield) of tert-butyl (10R)-8-bromo-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10- methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylate as a yellow oil. LCMS observed m/z =514.95 [M+H]+. Step 7: Preparation of tert-butyl (10R)-8-[(1S,6S)-5-(tert-butoxycarbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo- 1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylate. Compound 465.7
To a stirred solution of tert-butyl (10R)-8-bromo-4-(1,1-dimethyl-3H-2-benzofuran- 5-yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12- carboxylate (799 mg, 1.6 mmol) and tert-butyl (1S,6S)-2,5-diazabicyclo[4.2.0]octane-2- carboxylate (987 mg, 4.7 mmol) in ACN (10 mL) was added AcOK (456 mg, 4.7 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at 90 °C for 4 days. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (400 mg, 38% yield) of tert-butyl (10R)-8-[(1S,6S)-5-(tert-butoxycarbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo- 1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylate as a yellow solid. LCMS observed m/z = 647.15 [M+H]+. Step 8: Preparation of (10R)-8-[(1S,6S)-5-(tert-butoxycarbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo- 1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylic acid.
Compound 465.8
A solution of tert-butyl (10R)-8-[(1S,6S)-5-(tert-butoxycarbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo- 1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylate (400 mg, 0.6 mmol) in DCM (5 mL) was treated with trifluoroacetic acid (5 mL) at room temperature for 1.5 h under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. To the above mixture was added DCM (5 mL), di-tert-butyl dicarbonate (405 mg, 1.9 mmol) and Et3N (430 uL, 3.1 mmol). The resulting mixture was stirred at room temperature for additional 1 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (240 mg, 66% yield) of (10R)-8-[(1S,6S)-5-(tert-butoxycarbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo- 1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylic acid as a yellow white solid. LCMS observed m/z = 591.15 [M+H]+. Step 10: Preparation of tert-butyl (1S,6S)-5-[(10R)-12-{[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7- oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-trien-8-yl]-2,5- diazabicyclo[4.2.0]octane-2-carboxylate.
To a stirred solution of (10R)-8-[(1S,6S)-5-(tert-butoxycarbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo- 1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxylic acid (280 mg, 0.5 mmol) and DIEA (248 uL, 1.4 mmol) in THF (5 mL) were added trichloro-1,3,5-triazine (131 mg, 0.7 mmol) and 2-chloro-4-(trifluoromethyl)aniline (131 uL, 0.9 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 3 h under nitrogen atmosphere. The reaction was quenched with water (20 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 20 mL). The combined organic layers dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (100 mg, 27% yield) of tert-butyl (1S,6S)-5-[(10R)- 12-{[2-chloro-4-(trifluoromethyl)phenyl]carbamoyl}-4-(1,1-dimethyl-3H-2-benzofuran-5- yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-trien-8-yl]-2,5- diazabicyclo[4.2.0]octane-2-carboxylate as a yellow oil. LCMS observed m/z = 768.15 [M+H]+. Step 11: Preparation of (10R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-8-[(1S,6S)- 2,5-diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7- oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide.
A solution of tert-butyl (1S,6S)-5-[(10R)-12-{[2-chloro-4- (trifluoromethyl)phenyl]carbamoyl}-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7- oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-trien-8-yl]-2,5- diazabicyclo[4.2.0]octane-2-carboxylate (100 mg, 0.01 mmol) and TFA (1 mL) in DCM (1 mL) was stirred at room temperature for 30 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. This resulted in (160 mg, crude) of (10R)-
N-[2-chloro-4-(trifluoromethyl)phenyl]-8-[(1S,6S)-2,5-diazabicyclo[4.2.0]octan-2-yl]-4- (1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide as a brown oil. LCMS observed m/z = 668.00 [M+H]+. Step 11: Preparation of (10R)-8-[(1S,6S)-5-[5-(benzyloxy)-6-methylpyrimidine-4- carbonyl]-2,5-diazabicyclo[4.2.0]octan-2-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]-4- (1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. Compound 465.11
To a stirred solution of (10R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-8-[(1S,6S)- 2,5-diazabicyclo[4.2.0]octan-2-yl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7- oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide (106 mg, 0.2 mmol) and NMI (63 uL, 0.8 mmol) in ACN (1 mL) were added TCFH (53 mg, 0.2 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (46 mg, 0.2 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred at room temperature for 3 h under nitrogen atmosphere. The reaction was quenched by the addition of water (20 mL) at room temperature. The aqueous layer was extracted with EtOAc (3 x 20 mL). The resulting mixture was concentrated under reduced pressure. The residue was purified by reversed phase C18 silica gel column chromatography to afford (120 mg, 84% yield) (10R)-8-[(1S,6S)-5-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-2,5- diazabicyclo[4.2.0]octan-2-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca- 2,4,8-triene-12-carboxamide as a yellow oil. LCMS observed m/z = 894.05 [M+H]+. Step 11: Preparation of (10R,12R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1- dimethyl-3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-
carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide.
A solution of (10R)-8-[(1S,6S)-5-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]- 2,5-diazabicyclo[4.2.0]octan-2-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1- dimethyl-3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide (120 mg, 0.134 mmol) and trifluoroacetic acid (5 mL) in DCM (5 mL) was stirred at 80 °C for 30 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse phase flash with the following conditions (Column: Xselect CSH C185 μm, 30 mm X 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 30%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.72) to afford (6.5 mg, 6% yield) of (10R,12R)-N-[2-chloro- 4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl-3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5- hydroxy-6-methylpyrimidine-4-carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7- oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide 1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 10.27 (s, 1H), 8.56 (s, 1H), 8.02 – 7.91 (m, 4H), 7.77 – 7.70 (m, 1H), 7.39 (d, J = 7.9 Hz, 1H), 5.67 (dd, J = 8.6, 5.1 Hz, 1H), 5.00 (s, 2H), 4.39 (m, 1H), 3.72 (m, 2H), 3.38 (s, 2H), 3.25 (s, 2H), 2.63 (d, J = 6.7 Hz, 2H), 2.44 (s, 3H), 1.67 (d, J = 7.5 Hz, 1H), 1.57 (d, J = 6.9 Hz, 3H), 1.44 (s, 6H), 1.37 (d, J = 7.9 Hz, 1H), 1.26 – 1.14 (m, 2H). LCMS observed m/z = 804.40 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.466 Example 465
Synthesis of (10S,12R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. Compound 466
Step 1: Preparation of (10S,12R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1- dimethyl-3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. A solution of 8-[(1S,6S)-5-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-2,5- diazabicyclo[4.2.0]octan-2-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca- 2,4,8-triene-12-carboxamide (120 mg, 0.134 mmol) and trifluoroacetic acid (5 mL) in DCM (5 mL) was stirred at 80 °C for 30 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse phase flash with the following conditions (Column: Xselect CSH C185 μm, 30 mm X 150 mm; Mobile Phase A: water(0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 30%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.72) to afford (3.6 mg, 3% yield) of (10S,12R)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl-3H-2- benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. 1H NMR (400 MHz, DMSO-d6) δ 10.57 (s, 1H), 10.25 (s, 1H), 8.57 (s, 1H), 8.07 – 7.90 (m, 4H), 7.75 (d, J = 8.6 Hz, 1H), 7.40 (d, J = 7.9 Hz, 1H), 5.65 – 5.57 (m, 1H), 5.02 (s, 2H), 4.41 (m, 1H), 3.73 (s, 2H), 339 (s 2H) 325 – 298 (m 3H) 245 (s 3H) 217 (m 1H) 177 – 169 (m, 1H), 1.49
(d, J = 7.3 Hz, 3H), 1.45 (s, 5H), 1.37 (s, 2H), 1.22 (m, 2H). LCMS observed m/z = 804.40 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.467 Example 467 Synthesis of (10R,12S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide.
Step 1: Preparation of (10R,12S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1- dimethyl-3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. A solution of 8-[(1S,6S)-5-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-2,5- diazabicyclo[4.2.0]octan-2-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca- 2,4,8-triene-12-carboxamide (120 mg, 0.134 mmol) and trifluoroacetic acid (5 mL) in DCM (5 mL) was stirred at 80 °C for 30 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse phase flash with the following conditions (Column: Xselect CSH C185 μm, 30 mm X 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 30%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.72). The resulting crude product was purified by Prep-CHIRAL-SFC with the following conditions (Column: CHIRALPAK IG, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MEOH: DCM=1: 1(0.1% 7M NH3-MeOH); Flow rate: 100 mL/min; Gradient: isocratic 50% B; Column Temperature(℃): 35; Back Pressure(bar): 100; Wave Length: 254 nm; RT1(min): 2.9;
RT2(min): 6.7; Sample Solvent: MEOH; Injection Volume: 1 mL) to afford (2.4 mg, 2% yield) of (10R,12S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl-3H-2- benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. 1H NMR (400 MHz, DMSO-d6) δ 10.55 (s, 2H), 8.57 (s, 1H), 8.16 – 7.93 (m, 4H), 7.73 (d, J = 8.6 Hz, 1H), 7.40 (d, J = 7.8 Hz, 1H), 5.69 (dd, J = 8.8, 4.6 Hz, 1H), 5.01 (s, 2H), 4.44 (s, 1H), 3.73 (q, J = 7.3 Hz, 2H), 3.52 (m, 3H), 2.65 – 2.59 (m, 1H), 2.44 (s, 4H), 1.66 (d, J = 7.5 Hz, 1H), 1.55 (d, J = 7.0 Hz, 3H), 1.45 (s, 6H), 1.35 (s, 2H), 1.22 (m, 2H). LCMS observed m/z = 804.45 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.468 Example 468 Synthesis of (10S,12S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide.
Step 1: Preparation of (10S,12S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1- dimethyl-3H-2-benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4- carbonyl)-2,5-diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. A solution of 8-[(1S,6S)-5-[5-(benzyloxy)-6-methylpyrimidine-4-carbonyl]-2,5- diazabicyclo[4.2.0]octan-2-yl]-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl- 3H-2-benzofuran-5-yl)-10-methyl-7-oxo-1,3,5,6-tetraazatricyclo[7.3.0.0^{2,6}]dodeca- 2,4,8-triene-12-carboxamide (120 mg, 0.134 mmol) and trifluoroacetic acid (5 mL) in DCM
(5 mL) was stirred at 80 °C for 30 min under nitrogen atmosphere. The resulting mixture was concentrated under reduced pressure. The crude product was purified by reverse phase flash with the following conditions (Column: Xselect CSH C185 μm, 30 mm X 150 mm; Mobile Phase A: water (0.1%FA), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 15% B to 30%B in 8 min; Wave Length: 254nm/220nm nm; RT1(min): 7.72). The resulting crude product was purified by Prep-CHIRAL-SFC with the following conditions (Column: CHIRALPAK IG, 3*25 cm, 5 μm; Mobile Phase A: CO2, Mobile Phase B: MEOH: DCM=1: 1(0.1% 7M NH3-MeOH); Flow rate: 100 mL/min; Gradient: isocratic 50% B; Column Temperature(℃): 35; Back Pressure(bar): 100; Wave Length: 254 nm; RT1(min): 2.9; RT2(min): 6.7; Sample Solvent: MEOH; Injection Volume: 1 mL) to afford (5.0 mg, 5% yield) of ((10S,12S)-N-[2-chloro-4-(trifluoromethyl)phenyl]-4-(1,1-dimethyl-3H-2- benzofuran-5-yl)-8-[(1S,6S)-5-(5-hydroxy-6-methylpyrimidine-4-carbonyl)-2,5- diazabicyclo[4.2.0]octan-2-yl]-10-methyl-7-oxo-1,3,5,6- tetraazatricyclo[7.3.0.0^{2,6}]dodeca-2,4,8-triene-12-carboxamide. 1H NMR (400 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.52 (s, 1H), 8.30 (s, 1H), 7.98 (m, 4H), 7.74 (d, J = 8.5 Hz, 1H), 7.40 (d, J = 7.9 Hz, 1H), 5.62 (d, J = 9.9 Hz, 1H), 5.02 (s, 2H), 4.46 (s, 2H), 3.77 (s, 2H), 3.05 (m, 2H), 2.36 (m, 3H), 2.15 (m, 2H), 1.71 (m, 2H), 1.46 (m, 10H), 1.27 (m, 2H). LCMS observed m/z = 804.45 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.469 Example 469 Synthesis of (7R,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropy rrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide. Compound 469
Step 1: Preparation of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate. Compound 469.1
To a stirred mixture of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4,7-dihydro- [1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (6.0 g, 14.0 mmol) and Et3N (4.3 g, 42.1 mmol) in DMF (50 mL) was added SEMCl (2.8 g, 16.8 mmol) dropwise at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at 60 °C for 16 h under nitrogen atmosphere. The reaction was quenched by the addition of ice-water (100 mL) at room temperature and extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (5 / 1) to afford tert-butyl 4-(2-bromo-5-ethyl- 7-oxo-4-((2-(trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (4.2 g, 53% yield) as an off-white solid. LCMS observed m/z = 557.18/559.18 [M+H]+. Step 2: Preparation of tert-butyl 4-(2-bromo-7-oxo-5-(pent-4-en-2-yl)-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate. Compound 469.2
To a solution of tert-butyl 4-(2-bromo-5-ethyl-7-oxo-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (4.2 g, 7.5 mmol) in THF (60 mL) was added dropwise
NaHMDS (4.2 mL, 8.2 mmol, 1 M in THF) over 10 min at -78 °C and the mixture was stirred at -78 °C for 2 h under nitrogen atmosphere. Then added dropwise a solution of 3- iodoprop-1-ene (1.5 g, 9.0 mmol) in THF (10 mL) at -78 °C, and allowed the resulting mixture stirred at -78 °C for another 2 h under nitrogen atmosphere. The reaction was quenched by the addition of sat. aq. NH4Cl (100 mL) at 0 °C, then extracted with EtOAc (3 x 100 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (3/1) to afford tert-butyl 4-(2-bromo-7-oxo-5-(pent-4-en-2-yl)-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (2.8 g, 62% yield) as a white solid. LCMS observed m/z = 597.21/599.21 [M+H]+. Step 3: Preparation of tert-butyl 4-(2-bromo-5-(1-(oxiran-2-yl)propan-2-yl)-7-oxo- 4-((2-(trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate. Compound 469.3
To a stirred solution of tert-butyl 4-(2-bromo-7-oxo-5-(pent-4-en-2-yl)-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (2.8 g, 4.6 mmol) in DCM (30 mL) was added ethaneperoxoic acid (28 mL, 46 mmol, concentration 18%) at 0 °C. The resulting mixture was stirred at room temperature for 16 h. The reaction was quenched by the addition of sat. aq. NaHCO3 (30 mL) and sat. aq. Na2S2O3 (30 mL) in sequence at 0 °C. The resulting mixture was extracted with CH2Cl2 (3 x 40 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure to give tert-butyl 4-(2-bromo-7-oxo-5-(pent-4-en-2-yl)-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxy late (2.3 g, crude) as a white solid. The crude resulting mixture was
used in the next step directly without further purification. LCMS observed m/z = 613.21/615.21 [M+H] +. Step 4: Preparation of tert-butyl 4-(2-bromo-9-(hydroxymethyl)-7-methyl-5-oxo- 5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate. Compound 469.4
To a stirred mixture of tert-butyl 4-(2-bromo-7-oxo-5-(pent-4-en-2-yl)-4-((2- (trimethylsilyl)ethoxy)methyl)-4,7-dihydro-[1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (2.3 g, 3.7 mmol) in DMF (15 mL) was added KF (1.1 g, 18.7 mmol) in portions at room temperature. The resulting mixture was stirred at 60 °C for 16 h. The reaction was quenched by the addition of water (30 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1 / 1) to afford tert-butyl 4-(2-bromo-9- (hydroxymethyl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a] pyrimidin-6-yl)piperazine-1-carboxylate (1.2 g, 66% yield) as an off-white solid. LCMS observed m/z = 483.13/485.13 [M+H] +. Step 5: Preparation of 2-bromo-6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-7-methyl- 5-oxo-5,7,8,9-tetrahy dropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxylic acid. Compound 469.5
To a stirred solution of tert-butyl 4-(2-bromo-9-(hydroxymethyl)-7-methyl-5-oxo- 5,7,8,9-tetrahydro pyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1- carboxylate (1.2 g, 2.4 mmol) in DCM (20 mL) were added 5% aq. NaHCO3 (2.09 g, 24.8 mmol), methyltrioctylazanium chloride (50 mg, 0.1 mmol), TEMPO (4 mg, 0.02 mmol), KBr (30 mg, 0.2 mmol). The biphasic resulting mixture was cooled in an ice bath to 0 °C. Under vigorous stirring, 8% aq. sodium hypochlorite solution (0.4 mL, 6.2 mmol) was added dropwise within 2 hours, so that the pH did not rise above 8.6. The resulting mixture was stirred for 30 min. 8% aq. sodium hypochlorite solution (0.4 mL, 6.2 mmol) was added dropwise within 1 hour maintaining the pH below 8.6. The resulting mixture was stirred for another 2 hours. Then the resulting mixture was diluted with DCM (20 mL) and stirred vigorously for 2 minutes. Then the phases were separated. The aqueous phase was extracted with DCM (20 mL). The combined organic phases were extracted with aq. NaHCO3 (3 x 30 mL). Then the combined aqueous phases were diluted with DCM (30 mL). The biphasic mixture was adjusted under vigorous stirring to pH 3 by addition of 5 N aq. HCl. The aqueous phase was extracted with DCM (3 x 50 mL). The combined organic layers dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 2-bromo-6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-7-methyl-5- oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxylic acid (400 mg crude) as a yellow solid.. LCMS observed m/z = 497.11/499.11 [M+H]+, 397.11/399.11[M+H-Boc]+. Step 6: Preparation of 6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxylic acid. Compound 469.6
To a stirred mixture of 2-bromo-6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxylic acid (395 mg, 0.8 mmol) and Pd(dppf)Cl2 (29 mg, 0.04 mmol), K3PO4 (421 mg,
1.9 mmol) in dioxane (8 mL) and H2O (4 mL) was added 2-(1,1-dimethyl-3H-2-benzofuran- 5-yl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (261 mg, 0.9 mmol) at 80 °C, and the resulting mixture was stirred at 80 °C for 5 minutes under nitrogen atmosphere. The resulting mixture was diluted water (10 mL), and extracted with EtOAc (2 x 5 mL). The aqueous layer was acidified to pH 2 with 2N HCl, and then extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3x10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. This resulted in 6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5- yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxylic acid. (300 mg, crude) as a yellow solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 565.27 [M+H]+. Step 7: Preparation of tert-butyl 4-((7R,9S)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate, tert-butyl 4-((7R,9R)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate, tert-butyl 4-((7S,9S)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate, tert-butyl 4-((7S,9R)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate. Compound 469.7
To a stirred mixture of 6-(4-(tert-butoxycarbonyl)piperazin-1-yl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxylic acid (300 mg, 0.5 mmol), TEA (537 mg,
5.3 mmol) and 2-chloro-4-(trifluoromethyl)aniline (114 mg, 0.5 mmol) in EtOAc (10 mL) was added dropwise T3P (845 mg, 2.655 mmol) at room temperature. The resulting mixture was stirred at 50 °C for 6 h. Then the reaction was quenched by the addition of aq. sat. NaHCO3 (10 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography, eluted with PE / EA (1/1) to afford tert-butyl 4-(9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (110 mg, 28% yield, mixture) as a yellow solid. LCMS observed m/z = 742.27 [M+H]+. The mixture (110 mg) was separated twice by preparative chiral SFC (Column: CHIRAL ART Amylose-SA, 2*25 cm, 5 μm; Mobile Phase A: Hex (10 mM NH3-MeOH), Mobile Phase B: MEOH: DCM=1: 1 (0.1% 7M NH3-MeOH); Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 220 nm; RT1 (min): 5.1; RT2 (min): 7.5; Sample Solvent: MEOH; Injection Volume: 1 mL; Number Of Runs: 2) and (Column: CHIRALPAK ID, 3*25 cm, 5 μm; Mobile Phase A: Hex (10 mM NH3-MeOH), Mobile Phase B: MEOH: DCM=1: 1; Flow rate: 20 mL/min; Gradient: isocratic ; Wave Length: 254 nm; RT1(min): 17; RT2 (min): 20; Sample Solvent: MEOH; Injection Volume: 0.2 mL; Number Of Runs: 5)
Peak 1: tert-butyl 4-((7R,9S)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2- (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate or tert-butyl 4-((7S,9R)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (36 mg, 95% purity, 32.7% yield).
Peak 2: tert-butyl 4-((7R,9R)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2- (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate or tert-butyl 4-((7S,9S)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (12 mg, 95% purity, 10.9% yield).
Peak 3: tert-butyl 4-((7S,9S)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2- (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate or tert-butyl 4-((7R,9R)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (16 mg, 95% purity, 14.5%
Peak 4: tert-butyl 4-((7S,9R)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2- (1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate or tert-butyl 4-((7R,9S)-9-((2-chloro-4-(trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-
tetrahydropyrrolo[1,2c][1,2,4]triazolo[1,5-a]pyrimidin-6-yl)piperazine-1-carboxylate (34 mg, 95% purity, 30.9% yield). Step 8: Preparation of (7R,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 469.8
To a stirred mixture of tert-butyl 4-((7R,9S)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate (36 mg, 0.05 mmol) in DCM (2 mL) was added TFA (0.6 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. This resulted in (7R,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (35 mg, crude) as a yellow solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 642.22 [M+H]+. Step 9: Preparation of (7R,9S)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 469.9
To a stirred mixture of (7R,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (35 mg, 0.05 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (15 mg, 0.06 mmol) in DCM (2 mL) were added DIEA (21 mg, 0.2 mmol) and HATU (31 mg, 0.08 mmol) at room temperature under air atmosphere. The resulting mixture was stirred at room temperature for 1 h. The reaction was quenched with water (2 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 2 mL). The combined organic layers were washed with brine (3 x 2 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Cl2 / MeOH 20/1) to afford (7R,9S)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (20 mg, 42% yield) as a yellow solid. LCMS observed m/z = 868.29 [M+H]+. Step 9: Preparation of (7R,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 469
(7R,9S)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-N- (2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide (20 mg, 0.02 mmol) was dissolved in TFA (1 mL) at room temperature. The resulting solution was heated to 80 °C stirred at 80 °C for 30 min. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column, 5 μm, 30 mm * 150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29% B to 52% B in 8 min; Wave Length: 254/220 nm; RT1 (min): 6.07) to afford (7R,9S)-N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (8.2 mg, 45.7% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.82 (s, 1H), 9.20 (s, 1H), 8.62 (s, 1H), 8.46 (d, J = 8.7 Hz, 1H), 8.18 (dd, J = 7.9, 1.5 Hz, 1H), 8.10 (s, 1H), 7.65 (d, J = 2.1 Hz, 1H), 7.54 (dd, J = 8.8, 2.1 Hz, 1H), 7.21 (d, J = 7.9 Hz, 1H), 5.44 (dd, J = 8.4, 2.1 Hz, 1H), 5.12 (s, 2H), 4.00 – 3.07 (m, 7H), 2.59 (s, 3H), 2.29 (dt, J = 13.0, 8.8 Hz, 1H), 1.75 (d, J = 6.9 Hz, 4H), 1.71 – 1.68 (m, 1H),1.61 – 1.58 (m, 1H), 1.55 (s, 6H). LCMS observed m/z = 778.15 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.470 Example 470 Synthesis of (7R,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-
yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide. Compound 470
Step 1: Preparation of (7R,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 470.1
To a stirred mixture of tert-butyl 4-((7R,9R)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate, Compound 469.7, Peak-2 (12 mg, 0.01 mmol) in DCM (1 mL) was added TFA (0.3 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. This resulted in (7R,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (12 mg, crude) as
a yellow solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 642.22 [M+H]+. Step 2: Preparation of (7R,9R)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 470.2
To a stirred mixture of (7R,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (10 mg, 0.01 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (5 mg, 0.01 mmol) in DCM (2 mL) were added DIEA (6 mg, 0.04 mmol) and HATU (8 mg, 0.02 mmol) at room temperature under air atmosphere. The resulting mixture was stirred at room temperature for 1 h. The reaction was quenched with water (1 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 2 mL). The combined organic layers were washed with brine (3 x 2 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM / MeOH =20/1) to afford (7R,9R)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5- yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide (10 mg, 74% yield) as a yellow solid. Step 3: Preparation of (7R,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl 13 dihydroisobenzofuran 5 yl) 6 (4 (5 hydroxy 6 methylpyrimidine-4-
carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 470
(7R,9R)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-N- (2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide (10 mg, 0.01 mmol) was dissolved in TFA (1 mL) at room temperature. The resulting solution was heated to 80 °C stirred at 80 °C for 30 min. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column, 5 μm, 30 mm * 150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29% B to 52% B in 8 min; Wave Length: 254/220 nm; RT1(min): 6.38) to afford (7R,9R)-N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (3.8 mg, 42.4% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) 1H NMR (400 MHz, Chloroform-d) δ 11.79 (s, 1H), 9.62 (s, 1H), 8.62 (s, 1H), 8.46 (d, J = 8.7 Hz, 1H), 8.21 (dd, J = 7.9, 1.5 Hz, 1H), 8.13 (s, 1H), 7.67 – 7.62 (m, 1H), 7.55 (dd, J = 8.9, 2.0 Hz, 1H), 7.23 (d, J = 7.9 Hz, 1H), 5.55 – 5.46 (m, 1H), 5.13 (s, 2H), 3.92 – 3.78 (m, 2H), 3.64 – 2.79 (m, 6H), 2.59 (s, 3H), 1.60 (d, J = 7.3 Hz, 6H) 1.57 (s, 6H). LCMS observed m/z = 778.20 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.471 Example 471 Synthesis of (7S,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-
yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide. Compound 471
Step 1: Preparation of (7S,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 471.1
To a stirred mixture of tert-butyl 4-((7S,9S)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate, Compound 469.7, Peak-3 (12 mg, 0.02 mmol) in DCM (1 mL) was added TFA (0.3 mL) dropwise at room temperature .The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. This resulted in (7S,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (15 mg, crude) as
a yellow solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 642.22 [M+H]+. Step 2: Preparation of (7S,9S)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 471.2
To a stirred mixture of (7S,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (15 mg, 0.02 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (6 mg, 0.02 mmol) in DCM (2 mL) were added DIEA (9 mg, 0.06 mmol) and HATU (13 mg, 0.03 mmol) at room temperature under air atmosphere. The resulting mixture was stirred at room temperature for 1h. The reaction was quenched with water (1 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 2 mL). The combined organic layers were washed with brine (3 x 2 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM / MeOH = 20/1) to afford (7S,9S)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1- yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5- yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide (10 mg, 49% yield) as a yellow solid. Step 3: Preparation of (7S,9S)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl 13 dihydroisobenzofuran 5 yl) 6 (4 (5 hydroxy 6 methylpyrimidine-4-
carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 471
(7S,9S)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide (10 mg, 0.01 mmol) was dissolved in TFA (1 mL) at room temperature. The resulting solution was heated to 80 °C stirred at 80 °C for 30 min. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column, 5 μm, 30 mm * 150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29% B to 52% B in 8 min; Wave Length: 254/220 nm; RT1 (min): 6.38) to afford (7S,9S)-N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (5.7 mg, 63.6% yield) as a white solid. 1H NMR (400 MHz, Chloroform-d) δ 11.82 (s, 1H), 9.62 (s, 1H), 8.62 (s, 1H), 8.46 (d, J = 8.7 Hz, 1H), 8.21 (dd, J = 7.9, 1.5 Hz, 1H), 8.13 (s, 1H), 7.65 (d, J = 2.1 Hz, 1H), 7.55 (dd, J = 8.8, 2.2 Hz, 1H), 7.23 (d, J = 7.9 Hz, 1H), 5.54 – 5.45 (m, 1H), 5.13 (s, 2H), 3.90 – 2.74 (m, 9H), 2.59 (s, 3H), 1.73 – 1.70 (m, 1H),1.65 – 1.60 (m, 4H), 1.57 (s, 6H). LCMS observed m/z = 778.10 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. 1.472 Example 472 Synthesis of (7S,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-
yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide. Compound 472
Step 1: Preparation of (7S,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 472.1
To a stirred mixture of tert-butyl 4-((7S,9R)-9-((2-chloro-4- (trifluoromethyl)phenyl)carbamoyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidin-6- yl)piperazine-1-carboxylate, Compound 469.7, Peak-4 (34 mg, 0.05 mmol) in DCM (1 mL) was added TFA (0.3 mL) dropwise at room temperature .The resulting mixture was stirred at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. This resulted in (7S,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl- 1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (35 mg, crude) as
a yellow solid. The crude product mixture was used in the next step directly without further purification. LCMS observed m/z = 642.22 [M+H]+. Step 2: Preparation of (7S,9R)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 472.2
To a stirred mixture of (7S,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-6-(piperazin-1-yl)-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (35 mg, 0.05 mmol) and 5-(benzyloxy)-6-methylpyrimidine-4-carboxylic acid (15 mg, 0.06 mmol) in DCM (2 mL) were added DIEA (21 mg, 0.2 mmol) and HATU (31 mg, 0.08 mmol) at room temperature under air atmosphere. The resulting mixture was stirred at room temperature for 1h. The reaction was quenched with water (1 mL) at room temperature. The resulting mixture was extracted with EtOAc (3 x 2 mL). The combined organic layers were washed with brine (3 x 2 mL), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (CH2Cl2 / MeOH = 20/1) to afford (7S,9R)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4- carbonyl)piperazin-1-yl)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3- dihydroisobenzofuran-5-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (20 mg, 42% yield) as a yellow solid. Step 3: Preparation of (7S,9R)-N-(2-chloro-4-(trifluoromethyl)phenyl)-2-(1,1- dimethyl-1,3-dihydroisobenzofuran-5-yl)-6-(4-(5-hydroxy-6-methylpyrimidine-4-
carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2- c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide. Compound 472
(7S,9R)-6-(4-(5-(benzyloxy)-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-7- methyl-5-oxo-5,7,8,9-tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9- carboxamide (10 mg, 0.01 mmol) was dissolved in TFA (1 mL) at room temperature. The resulting solution was heated to 80 °C stirred at 80 °C for 30 min. After the reaction was completed, the resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions (Column: XBridge BEH C18 OBD Prep Column, 5 μm, 30 mm * 150 mm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1% NH4OH), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 29% B to 52% B in 8 min; Wave Length: 254/220 nm; RT1 (min): 6.38) to afford (7S,9R)-N-(2- chloro-4-(trifluoromethyl)phenyl)-2-(1,1-dimethyl-1,3-dihydroisobenzofuran-5-yl)-6-(4- (5-hydroxy-6-methylpyrimidine-4-carbonyl)piperazin-1-yl)-7-methyl-5-oxo-5,7,8,9- tetrahydropyrrolo[1,2-c][1,2,4]triazolo[1,5-a]pyrimidine-9-carboxamide (17.1 mg, 95.4% yield) as a white solid.1H NMR (400 MHz, Chloroform-d) δ 11.83 (s, 1H), 9.20 (s, 1H), 8.62 (s, 1H), 8.47 (d, J = 8.7 Hz, 1H), 8.19 (dd, J = 8.0, 1.5 Hz, 1H), 8.10 (s, 1H), 7.65 (d, J = 2.1 Hz, 1H), 7.59 – 7.52 (m, 1H), 7.22 (d, J = 7.9 Hz, 1H), 5.43 (dd, J = 8.5, 2.1 Hz, 1H), 5.12 (s, 2H), 3.95 – 3.09 (m, 8H), 2.60 (s, 3H), 2.29 (dt, J = 13.0, 8.8 Hz, 1H), 1.75 (d, J = 6.9 Hz, 3H),1.73 – 1.71(m, 1H) 1.60 – 1.58 (s, 1H).1.57 (s, 6H). LCMS observed m/z = 778.15 [M+H]+. Note: The absolute stereochemistry was assigned tentatively. EXAMPLES 2.1-2.4 2.1 WRN Protein Purification and Biochemical Activity Assays
WRN helicase core (8xHis-tev-WRN (480-1251)) was expressed in Sf9 insect cells and purified using HiTrap TALON affinity purification followed by heparin column. The pure fractions were pooled and desalted/buffer exchanged into storage buffer using a HiTrap desalting column. WRN activity assays were performed in a multiplex fashion by monitoring both DNA unwinding using a quenched dsDNA substrate (dsDNArev-Cy3: 5’-Cy3- GAACGAACACATCGGGTACGTTTTTTTTTTTTTTTTTTTTTTTTTTTTTT; dsDNAfwd-FQ: 5’- TTTTTTTTTTTTTTTTTTTTTTTTTTTTTTCGTACCCGATGTGTTCGTTC- IowaBlackFQ-3’, Integrated DNA Technologies) and ATPase activity using an ADP-Glo Kinase Assay (Promega). First, WRN alone was pre-incubated with DMSO or compound of interest for 15 minutes. To initiate the reaction, ATP, dsDNA substrate, and ssDNA competitor (ssDNA_comp: GAACGAACACATCGGCTAC) were added and incubated at room temperature for 20 minutes at which point DNA unwinding activity was monitored by measuring Cy3 fluorescence. After obtaining the reading, the ATPase activity was measured using the ADP-Glo Kinase Assay following the manufacturers recommended protocol. The final reaction conditions were: 12.5nM WRN, 1mM ATP, 15nM dsDNA substrate, 1.5uM ssDNA competitor in reaction buffer composed of 50mM Tris-HCl pH 8.0, 50mM NaCl, 2mM MgCl2, 0.01% v/v Tween-20, 2.5µg/mL poly(dI-dC), 1mM DTT. Compounds were assayed in a 10-point dose response in duplicate at a 1% DMSO final assay concentration. 2.2 Differential Viability All cell lines used were obtained from ATCC. To assess microsatellite unstable sensitivity, the human colorectal cancer cell lines HCT-116 (CVCL_1724), SW48 (CVCL_1724), and RKO (CVCL_0504) were used. The MSS cell lines HT-29, SW480, and U2OS were used to assess general off-target cytotoxic effects. All cell lines were cultured in McCoy 5A media (ThermoFisher Cat# 16600082) supplemented with 10% fetal bovine serum (Cytiva Cat# SH30071.03IH25), 1x penicillin/streptomycin (ThermoFisher Cat# 15140122), 1x non-essential amino acids (ThermoFisher Cat# 11140050), 2 mM GlutaMAX (ThermoFisher Cat# 35050079). Cells were cultured at 37 ºC in a humidified 5% CO2 incubator. To seed cells, cells were trypsinized and resuspended in complete culture media to the desired concentration (HCT-116: 10,000 cells/mL; RKO: 15,000 cells/mL; SW-48, HT-
29, and SW-480: 25,000 cells/mL; U2OS: 7,000 cells/mL). Cell suspensions were seeded in 50 μL of complete culture media and onto 384-well white clear-bottom plates (ThermoFisher Cat# 164610) using a MultidropTM Combi liquid dispenser in the slowest setting in triplicate. After 24 h, cells were treated with compounds of interest in a 10-pt, 3.16 step serial dilution using an Echo acoustic liquid dispenser (Beckman). DMSO was backfilled to a final concentration of 0.1%. 10 μM etoposide (Cpos) and DMSO (Cneg) were used as reference compounds. 96 h post compound addition, all media was decanted from plates. To measure viability, cellular ATP concentrations were measured by adding 20 μL of 1x CellTiterGlo 2.0 solution (1 part PBS, 1 part CellTiterGlo2.0 stock solution; Promega Cat# G9241) using a MultidropTM Combi liquid dispenser and measuring the luminescence on a SpectraMax iD3 (Molecular Devices). Percent viability was calculated by the following equation: %V = (T - Cpos)/(Cpos – Cneg) * 100, where %V is percent viability and T is the measured luminescence of the wells treated with test compound. The effective compound concentration leading to a 50% reduction in viability (EC50), and the resulting cell viability was measured at the highest compound concentration tested (cmax), which was carried out by fitting a 4-parameter non- linear regression using GraphPad Prism. At least three biological replicates were done per compound tested. 2.3 DNA Damage Induction Assay Cell lines and growth conditions are identical to the conditions in Example 2.2. To seed cells for the assay, cells were trypsinized and resuspended in complete culture media to the desired concentration (HCT-116: 50,000 cells/mL; RKO: 55,000 cells/mL; SW-48, HT-29, and SW-480: 60,000 cells/mL; U2OS: 30,000 cells/mL). Cell suspensions were seeded in 50 μL of complete culture media and onto 384-well black clear- bottom optimal plastic plates (Greiner Bio-One Cat# 781097) using a MultidropTM Combi liquid dispenser in the slowest setting in triplicate. After 24 h, cells were treated with compounds in a 10-pt, 3.16 step serial dilution using an Echo acoustic liquid dispenser and incubated at 37 ºC in a humidified 5% CO2 incubator. 10 μM etoposide and DMSO were used as reference compounds. 24 h after compound treatments, cells were fixed in 4% paraformaldehyde for 10 min, washed three times with PBS, then blocked and permeabilized in PBS containing 10% goat serum and 0.1% triton X-100 for at least 30 min. After blocking and permeabilization,
all media was decanted from the plates. Primary antibody against γH2A.X (Ser 139) raised in mice (Millipore Sigma, Cat# 05-636, clone JBW3201) in blocking/permeabilization solution was added to plates and incubated for 3 h at 25 ºC. After 1º antibody incubation, plates were evacuated and decanted to remove all media. Blocking/permeabilization solution containing the DNA counterstain Hoechst, and a secondary antibody against mice (raised in goats) conjugated to a AlexaFluor 647 fluorophore (ThermoFisher Cat# A32728), were added to empty plates, and incubated for 30 min at 25 ºC, in the dark. After 2º antibody incubation, plates were washed 3 times in PBS, and sealed with thermal foil seals. Sealed plates were imaged using an ImageXpress Micro slit confocal microscope (Molecular Devices) using a 40x water immersion objective, and 6 fields of views per well. Exposure parameters were optimized to prevent pixel saturation for each channel. Images were analyzed using MetaXpress Custom Module Editor, by using a Hoechst mask to identify nuclei, then measuring the integrated AlexaFluor 648 intensity across all nuclei in the FOV and then averaged. Percent γH2A.X signal was calculated by using the following equation: %S = (T - Cpos)/(Cpos – Cneg) * 100, where %S is percent γH2A.X signal and T is the measured γH2A.X fluorescence of the wells treated with test compound. The effective compound concentration leading to a 50% induction of γH2A.X signal (EC50), and the resulting cell γH2A.X signal was measured at the highest compound concentration tested (cmax), which was carried out by fitting a 4-parameter non-linear regression using GraphPad Prism. At least three biological replicates were done per compound tested. 2.4 Results Results associated with compounds corresponding to Examples presented above, in connection with the biochemical activity assays described in this Example are provided in Table 1. Table 1.
EXAMPLES 3.1-3.2 3.1 In Vivo Efficacy and Pharmacodynamics Methods Efficacy Studies To assess the in vivo efficacy and pharmacodynamics of compounds, MSI-h HCT116 or MSS HT-29 tumors were established in 4–5-week-old female Crl:NU(NCr)- Foxn1nu athymic nude mice (Charles River Laboratories). Mice were housed in 5 animals per cage and food and water was provided ad libitum. All procedures were performed at the Mispro Vivarium facility in New York, NY and were conducted according to the guidelines of the Mispro Institutional Animal Care and Use Committee and Eikon Therapeutics protocol. HCT116 and HT-29 cells were sourced from ATCC and cultured in McCoy’s 5a Medium (ATCC) with 10% FBS. To establish the tumors, mice were injected with 2 x 106 HCT116 or HT-29 cells in 50% Geltrex/50% HBSS in the right flank. Animal body weights and tumor volumes were measured twice weekly throughout the study and recorded in StudyLog. Tumor volume was calculated using the formula TV = 0.5*length*width2. Tumor Growth Inhibition (TGI) was calculated using the formula TGI = (1-((TD25- Tinitial)/(CD25-Cinitial)))*100, where T is the Test group TV and C is the Control
group TV at Day 25 vs the initial TV at study commencement. Percent tumor regression was calculated using the formula % regression = 100-((Tfinal/Tinitial)x100), where Tfinal is the latest TV measurement and Tinitial is the initial TV at study commencement. After HCT116 tumors grew to an average volume of 150-200 mm3 (7 days post-implantation), mice were randomized into treatment groups (n=8 mice/group) and received either test compounds or vehicle control daily via oral gavage at 10mL/kg body weight. Compounds were dissolved to required concentrations in an aqueous solution of 20% 2-hydroxypropyl- beta-cyclodextrin (HP-β-CD) w/v in water and pH adjusted to 7.4. Compound 13 was administered at 15, 30 or 90 mg/kg while compounds 164 and 169 were administered at 5, 15 or 30 mg/kg. Pharmacodynamic and Pharmacokinetic Study To assess possible degradation of WRN mediated by WRN inhibitors, the level of WRN protein in tumor lysate samples was determined via an MSD assay. Animals were treated with Compound 164 at 5, 15, or 30 mg/kg QDx3 and then tumor and blood samples were taken 2, 8 and 48 hours post final dose. Tumor lysates were prepared by lysing tumors in lysis buffer (Buffer 9803, CST) and protein amounts were quantified (Pierce BCA, Thermo) and equalized across samples by dilution. For the MSD assay, a WRN capture antibody ( PA5-27319, Thermo) was incubated overnight on MSD assay plates and plates were then blocked for non-specific binding for 1 h using MSD Blocker A solution. Tumor lysate samples were added at 0.5 mg/mL and incubated for 1.5 h at room temperature. Bound WRN was then detected using 10 mg/ml detection antibody (195C, Thermo). Goat anti-mouse sulfo-tag antibody was added at 1:1000 and incubated for 1 h at room temperature. Plates were washed with MSD wash buffer between every assay step. MSD Read buffer was added after the final washing step and chemiluminescence measured on an MSD plate reader. Relative chemiluminescence levels in treated samples were compared to vehicle controls. For assessment of plasma levels of compounds, mice were bled via cardiac puncture under terminal anesthesia and blood collected in an EDTA tube for plasma collection. Plasma concentration of compounds was assessed at Meadowhawk Biolabs by mass spectroscopy. 3.2 Results Efficacy Studies
As evident in Table 3 and Fig.1, all WRN inhibitor compounds assessed in vivo mediated significant anti-tumor efficacy in the MSI-h HCT116 xenograft model. Compound 13 caused TGI of 60.4, 96.8 and greater than 100 % at 15, 30 and 90 mg/kg respectively. The 90 mg/kg dose of Compound 13 caused 45.2 % tumor regression at Day 25 post dosing start and none at Day 60 of the study. Compound 164 mediated TGI of 28.9 % at 5 mg/kg and greater than 100 % at 15 and 30 mg/kg. Both 15 and 30 mg/kg of Compound 164 caused tumor regression at Day 25 post dosing start: 48.7 and 47.6 %, and regression was still observed in tumors treated with 30 mg/kg at Day 60 (26.7%). Compound 169 caused TGI of 8.9, 69.6 and greater than 100 % at 5, 15 and 30 mg/kg respectively. The 30 mg/kg dose of Compound 169 caused 43.1 % tumor regression at Day 25 post dosing start and none at Day 60 of the study. To validate that efficacy of WRN inhibitors is limited to MSI-h tumors, we also assessed the activity of Compound 164 in the MSS HT-29 tumor model. Very limited activity of Compound 164 was observed in the HT-29 tumor model as shown in Figure 2. Tumor volume was not significantly different than vehicle control by ANOVA testing and 30 mg/kg Compound 164 only mediated TGI of 15% and no tumor regression (Table 4). Pharmacodynamic and Pharmacokinetic Study Pharmacodynamic assessment of WRN levels in tumor samples demonstrated specific degradation of WRN protein upon treatment with Compound 164 (Figure 3). At the initial 2 h timepoint, moderate reduction in WRN relative vehicle control is observed, with a dose responsive decrease of approximately 20-35 %. At 8 h post final dose, a further reduction in WRN protein in observed, with a 40% reduction seen following 5 mg/kg Compound 164 and 50% reduction seen for both 15 and 30 mg/kg of Compound 164. A rebound in overall WRN levels is seen at the final 48 h timepoint, with WRN levels in tumors treated with 5 mg/kg Compound 164 being equivalent to vehicle control. In the higher doses, an around 20 % reduction in WRN level relative to vehicle was observed at the 48 h timepoint. Plasma concentrations of Compound 164 demonstrated clear dose dependent increase from 5 to 30 mg/kg and showed expected decrease over the time-course of the study. Table 3.
Table 4.
EXAMPLE 4.1 4.1 In vivo mouse PK assay Study was conducted to characterize the single dose pharmacokinetics of Compound 164 in the male CD1 mice after intravenous (1 mg/kg, injection) or oral administration (5, 15, and 30 mg/kg). A group of male CD1 mice (n=3, Approximately 20-30 g) were administered a single intravenous or oral dose of Compound 164, as a solution in 5% N- methyl pyrrolidone (NMP)/20% PEG400/5% Solutol/70% water, or in 20% HP-β-CD w/v in water, pH 7.4. Formulations were freshly prepared on the day of dosing or no earlier than 24 hours prior to dosing and were maintained at room temperature during the study. Animals had free access to food and water prior and during the study. Approximately 0.03 mL blood was collected at each time point (0.083, 0.25, 0.5, 1, 2, 4, 8, 24 hr post dose IV and 0.25, 0.5, 1, 2, 4, 8, 24 hr post dose for PO groups) through Dorsal metatarsal vein. Blood of each sample was transferred into plastic micro centrifuge tubes containing anticoagulant of K2-EDTA and mixed well with anticoagulant. Blood samples were centrifuged at 4,000g for 5 minutes at 4°C to obtain plasma samples. All plasma samples were stored in a freezer at -75±15°C prior to analysis. Plasma samples were prepared for analysis by protein precipitation method. 20 μL unknown samples (10 µL plasma with 10 µL blank solution) were added to 200 μL of acetonitrile containing IS mixture for protein precipitation. Samples then were vortexed for
30 seconds prior to centrifugation at 4 degree Celsius, 4000 rpm for 15 min. The supernatant was diluted with H2O at a ratio of 1:2 (V/V, 1:2), Then 10 µL of diluted supernatant was injected into the LC/MS/MS system for quantitative analysis. Concentrations of test article in the plasma samples were analyzed using a LC-MS/MS method. WinNonlin (PhoenixTM, version 8.3) was used for pharmacokinetic calculations. Table 5.
EXAMPLE 5.1 5.1 10-Day Colony Formation Assay Experiment Cell seeding Cells were transferred into a clean 50ml tube and centrifuged at 1200 rpm (225Xg) for 5 min. The cell pellet was resuspended in an appropriate volume of pre-warmed complete growth medium. A sample was aliquoted for counting using the Countess® Automated Cell Counter. The cells were then seeded into a 24-well plate at the desired density and incubated overnight. Compound preparation and treatment For the preparation of test compounds, a 10 mM DMSO stock solution was prepared. From this stock solution, 36 μL was transferred to a 384-well polypropylene plate. An 8-point, 3-fold serial dilution was performed using a TECAN (EVO200) liquid handler by transferring 12 μL of the compound into 24 μL of DMSO. The source plates were spun at room temperature at 1,000 RPM for 1 minute. Subsequently, the serially diluted compounds were transferred from the source plate to the cell plate, resulting in a final concentration series with a top dose of 10 μM. Each concentration was tested in duplicate wells, and the medium was refreshed every 3 days. Detection
After 10 days in culture, the media was aspirated off, and the cells were fixed with 0.5 mL/well of 70% ethanol. The fixation process was carried out for 15 minutes at room temperature. Following ethanol aspiration, 0.5 mL of 0.1% crystal violet was added to each well, and the plates were incubated for another 15 minutes at room temperature. The crystal violet was then removed, and the plates were washed four times with distilled water (dH2O). The plates were allowed to dry overnight at room temperature. The dried plates were scanned using the Li-Cor instrument with the 680(700) channel to obtain images of each well. To quantify the staining, 0.5 mL/well of freshly prepared 10% acetic acid was added, and the plates were incubated for 20 minutes at room temperature. Following incubation, 200 μL of each sample was aliquoted into two wells of a 96-well plate, and the absorbance was read at 590 nm using a TECAN plate reader. Data analysis The inhibition activity was calculated using the formula: %Inhibition = 100 x (LumDMSO – LumSample) / (LumDMSO –LumInhibition), where DMSO is obtained from cells treated with 0.1% DMSO only; and Inhibition is obtained from cells treated with 7.5uM Etoposide. The IC50 values were calculated by fitting the data to a dose-response curve using Xlfit (v5.3.1.3) with equation 201: Y = Bottom + (Top - Bottom)/(1 + 10^((LogIC50 - X)*HillSlope)) Table 6.10-Day Colony Formation Assay Data:
* * *
Although the presently disclosed subject matter and its advantages have been described in detail, it should be understood that various changes, substitutions, and alterations can be made herein without departing from the spirit and scope of the present disclosure. Moreover, the scope of the present application is not intended to be limited to the particular embodiments of the process, machine, manufacture, and composition of matter, means, methods and steps described in the specification. As one of ordinary skill in the art will readily appreciate from the presently disclosed subject matter, processes, machines, manufacture, compositions of matter, means, methods, or steps, presently existing or later to be developed that perform substantially the same function or achieve substantially the same result as the corresponding embodiments described herein can be utilized according to the presently disclosed subject matter. Accordingly, the appended claims are intended to include within their scope such processes, machines, manufacture, compositions of matter, means, methods, or steps. Various patents, patent applications, publications, product descriptions, protocols, and sequence accession numbers are cited throughout this application, the contents of which are incorporated by reference in their entirety for all purposes.
Claims
CLAIMS: What is claimed is: 1. A compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl
ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, , wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4--membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6-
membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring,
wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; (B) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra and Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, - NR8R9, -C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the
substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C2-C6 alkenyl, a C3-C7 cycloalkyl ring, C1-C6 hydroxyalkyl, , wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
wherein W, U, V, and Z are independently selected from H, C, N, S, or O and refers to a single or a double bond; wherein when
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20,U V is a double-bond and R16, R17, R18, are independently
selected from H, -CH3, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring, and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring;
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-
C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, or -C(O), R26 and R27 are independently selected from H, -OCF2Cl, or -S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen; wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring.
2. A compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R1 is NR30R31, aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl, the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is optionally substituted with one or more R36, or the said aryl, C3-C7 cycloalkyl, 5-10 membered heteroaryl, or 3-8 membered heterocyclyl is linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or said the aryl C3-C7 cycloalkyl 5-10 membered heteroaryl or 3-8 membered heterocyclyl
is fused with a second 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring or a C3-C8 cycloalkenyl ring, wherein R36 is independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, 3-8 membered heterocyclyl, -O-(C1-C6)alkyl-COOH, -C(O)-R33, -C(O)- NH-R33, -S(O)R33, -S(O2)R33, -NH-S(O2)R33, -NH-C(O)-NH-R33, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; and either (B) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the
substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
, wherein W, U, V, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; ; wherein when
wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20 U V is a double-bond and R16, R17, and R18 are independently
selected from H, -CH3, -C(O)R21, -S(O2)R21, and - S(O)R21R22, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring; or (C) R4 is selected from
L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2; wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl;
wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring. 3. A compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or - S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 3-8 membered heterocyclyl ring wherein when the 3-8 membered heterocyclyl ring is substituted,
the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein R8 and R9 are independently selected from H, C1-C6 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein R10 is selected from C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3- C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C3-C8 cycloalkyl, halogen, a 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or R10 is fused with a second C3-C8 cycloalkyl, C3-C8 cycloalkenyl, C6-C10 aryl, a 3-8 membered heterocyclyl, or a 5-10 membered heteroaryl; and either (B) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl;
C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered
heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring; and wherein R4 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring or
L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 1 or 2;
wherein when
alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R23 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, bridged C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
C1-C4 alkyl, wherein the C1-C4 alkyl is optionally substituted with halogens selected from F or Cl, or R24 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1- C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring, wherein L is optionally substituted with -OH, halogen, C1-C6 alkyl; wherein when
6 alkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, 3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl or the said R24 is a spiro ring or a fused ring; wherein when
single or double bond; wherein when L G is a double bond, G and L are independently selected from C or N, R25 and R29 are independently selected from H, halogen, =O, or -C(O), R26 and R27 are independently selected from H, -C(F2)-C(F2Cl), -OC(F2Cl), -OC(HF2), or - S(O)mR28, wherein m is 0-2, R28 is a methyl or trihalomethyl group, or either R25 or R27 join with R26 to form a 5- or 6- membered heterocyclyl ring, optionally substituted with -OH, alkyl, haloalkyl, or halogen; and
wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring. 4. A compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof:
wherein K and J are independently selected from C, N, O, or S; R, R0 and R5 are independently selected from H, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1- C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, or C3-C6 cycloalkyl; R6 and R7 are independently selected from H or halogen; wherein R6 or R7 can optionally join with R5 to form a C3-C8 cycloalkyl, 4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl ring; and wherein (A) R4 is selected from
, wherein L and G are independently selected from C, N, or O, n is 0, 1, or 2, and x is 0, 1 or 2;
wherein R23 and R24 are independently selected from -OH, -COOH, -NH2, - CN, oxo, halogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 alkoxy, C1-C6 haloalkoxy, C1-C6 hydroxyalkyl, C6-C10 aryl, 5-10 membered heteroaryl, a bridged or unbridged C3-C8 cycloalkyl, a 3-8 membered heterocyclyl,
3-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl; wherein when
single or double bond; R25, R29, and R35 are independently selected from H, halogen, -OH, =O, -C(O); C1-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; R26 and R27 are independently selected from H, C1-C6 alkyl, C1-C6 haloalkyl, C2-C6 haloalkenyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl, -OR28, or -S(O)mR28, wherein m is 0-2, R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, alkoxy, halogenated alkoxy, or C3-C8 cycloalkyl; wherein R25 or R27 join with R26 to form a C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; and either (B) R1 is NR30R31, aryl, C3-C7 cycloalkyl ring, 5-10 membered heteroaryl, or 4-, 5-, or 6- membered heterocyclyl ring; wherein when R1 is NR30R31, R30 and R31 are independently selected from optionally substituted C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, or H; wherein when R1 is aryl, the aryl is substituted by one or more -O-(C1-C6)alkyl- COOH, three C1-C6 alkoxy, -C(O)-R33, -C(O)-NH-R33, -S(O)R33, -S(O2)R33, -NH- S(O2)R33, -NH-C(O)-NH-R33, or the aryl is fused with a 5-10 membered heteroaryl, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen and/or at least one nitrogen atom, wherein R33 is selected from C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1- C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl,
4-8 membered heterocyclyl ring, C6-C10 aryl, or 5-10 membered heteroaryl, and wherein the said aryl, 5-10 membered heteroaryl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl ring, and 5- or 6-membered heterocyclyl ring are optionally
substituted with one or more, C1-C3 alkyl, halogen, -OH, =O, -C(O)-C1-C6 alkyl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl; C3-C8 cycloalkenyl, a second C6-C10 aryl, a second 5-10 membered heteroaryl, or 3-8 membered heterocyclyl; wherein when R1 is C3-C7 cycloalkyl ring, the said C3-C7 cycloalkyl is a fully saturated ring, wherein the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a spiro ring, or the said C3-C7 cycloalkyl ring is optionally linked with a second C3-C8 cycloalkyl ring, a 3-8 membered heterocyclyl, C6-C10 aryl, or 5-10 membered heteroaryl to form a fused ring, unless R4 is a substituted phenyl group; wherein when R1 is a 5-10 membered heteroaryl ring, the said 5-10 membered heteroaryl ring is substituted with -OH, -C(O)-C1-C6 alkyl, or -O-(C1-C6)alkyl- COOH, or the said 5-10 heteroaryl ring is fused with a second 5-10 membered heteroaryl ring, a C6-C10 aryl ring, a C3-C8 cycloalkyl ring, a C3-C8 cycloalkenyl ring, or 4-8 membered heterocyclyl ring; wherein when R1 is a 4-membered heterocyclyl ring, the said 4-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, or the said 4-membered heterocyclyl ring is optionally linked with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen or oxygen atom, wherein the said 4- or 7-membered heterocyclyl ring is linked with the said second 4-, 5-, or 6- membered heterocyclyl ring by one carbon atom to form a spiro ring or two carbon atoms to form a fused ring; wherein when R1 is a 5- or 6-membered heterocyclyl ring, the said 5- or 6-membered heterocyclyl ring is a fully saturated ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein the said 5- or 6-membered heterocyclyl ring is substituted with one or more C2-C6 alkenyl, or C2-C6 halogenated alkenyl or is linked with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring
to form a spiro ring, or the said 5- or 6-membered heterocyclyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6-membered heterocyclyl ring to form a fused ring, wherein the said second 4-, 5-, or 6-membered heterocyclyl ring comprises carbon atoms and at least one of a nitrogen atom or oxygen atom, wherein when said 5- or 6-membered heterocyclyl ring is linked with a second 5 membered heterocyclyl ring by one carbon atom to form a spiro ring and the second 5 membered heterocyclyl ring is fully saturated, the one or more heteroatoms in the second 5 membered heterocyclyl ring is selected from N or S; wherein when said 5- or 6-membered -membered heterocyclyl ring is linked with a second 4-, 5-, or 6-membered heterocyclyl ring by two carbon atoms to form a fused ring; the second 4-, 5-, or 6-membered heterocyclyl ring is partially unsaturated or fully unsaturated; or the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one silicon atom; or the 5-membered heterocyclyl ring is a pyrazole ring, wherein the pyrazole ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring, wherein the said second 4- , 5-, or 6- membered heterocyclyl ring is a saturated ring comprising carbon atoms and at least one of a nitrogen atom or oxygen atom, or the 6-membered heterocyclyl ring is a pyridinyl ring, wherein the pyridinyl ring is joined with a second substituted or unsubstituted 4-, 5-, or 6- membered heterocyclyl ring to form a fused ring; and wherein n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, - OH, =O, a substituted or unsubstituted alkyl or join together to form a fused cycloalkyl ring, a cycloalkenyl ring, a aryl ring, a heterocyclyl ring, or a heteroaryl ring; R2 and R3 are independently selected from H, -OH, =O, an substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl, a substituted or unsubstituted heterocyclyl ring, or join together to form an unsubstituted or substituted fused heterocyclyl ring or
(C) n is 0, 1, 2, or 3; refers to a single or a double bond; B, D, E, X and Y are independently selected from C or N; Ra, Rb are independently selected from H, -OH, =O, C1-C6 alkyl; R2 and R3 are independently selected from H, -OH, =O, C1-C6 alkyl, -NR8R9, - C(O)R10, -S(O2)R10, substituted or unsubstituted C2-C3 alkenyl, or substituted or unsubstituted 5- or 6-membered heterocyclyl ring, or Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; or R2 and R3 join together to form an unsubstituted or substituted fused 4-, 5-, or 6-membered heterocyclyl ring, wherein when the 4-, 5-, or 6-membered heterocyclyl ring is substituted, the substituents are optionally selected from -OH, =O, C1-C6 alkyl, a 5- or 6- membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring; wherein when Ra and R2 join together to form a fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring, the fused C3-C8 cycloalkyl ring, C3-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 3-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring is substituted by -C(O)R34 or -S(O2)R34, wherein R34 is a C1-C6 alkyl, C3-C8 cycloalkyl ring, C4-C8 cycloalkenyl ring, a C6-C10 aryl ring, a 4-8 membered heterocyclyl ring, or a 5-10 membered heteroaryl ring; wherein when n is 0, X is N, Y is C, R2 is NR8R9, and R3 is H; wherein when n is 1, X and Y are C, R2 is NR8R9 and R3 is H or R2 and R3 join together to form a substituted fused 4-membered heterocyclyl ring, wherein the substituted 4-membered heterocyclyl ring is substituted with -C(O)R11, and wherein R11 is a substituted 5- or 6-membered heterocyclyl ring; wherein when R2 is NR8R9, R8 and R9 are independently selected from H, C1-C4 alkyl, and -C(O)R11, wherein R11 is a substituted or unsubstituted 5- or 6-membered heterocyclyl ring; wherein when n is 2, X is C, Y is N, and R2 and R3 join together to form an unsubstituted or substituted fused 4- or 5-membered heterocyclyl ring, wherein when the 5- membered heterocyclyl ring is substituted, the substituents are selected from a 5- or 6-membered heterocyclyl ring or -C(O)R11, wherein R11 is a C1-C3 alkyl or substituted 5- or 6-membered heterocyclyl ring, or
X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is C1-C6 hydroxyalkyl, C2-C6 alkenyl, a C3-C7 cycloalkyl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl, C1-C6 hydroxyalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, C1-C3 alkoxy, an optionally substituted C3-C5 cycloalkyl ring, halogen, an optionally substituted 3-8 membered heterocyclyl, -S(O2)CH3, or -C(O)N(CH3)2, or X is C, Y is N, R2 is H, R3 is -C(O)R10, or -S(O2)R10, and R10 is
, wherein W, U, V, and Z are independently selected from H, C, N, S, or O and wherein refers to a single or a double bond; wherein when U is N+, R15 is O-, R16 and R17 are H, and U V is a double bond; wherein when U is C, R15 is -NR19R20 U V is a double-bond and R16, R17, and R18 are independently selected from H, -CH3, -C(O)R21, -S(O2)R21, and -S(O)R21R22, R19, and R20 are independently selected from -C(O)R21, -S(O2)R21, and -S(O)R21R22, wherein R21 and R22 are independently selected from C1-C3 alkyl, C3-C5 cycloalkyl ring, or 5- or 6- membered heterocyclyl ring or R21 and R22 join together to form a substituted or unsubstituted C3-C5 cycloalkyl ring; and wherein R1 is H, -OH, =O, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkenyl; a substituted or unsubstituted aryl ring, a substituted or unsubstituted heterocyclyl ring, a substituted or unsubstituted heteroaryl ring.
5. The compound of claims 1-4, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, and wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl.
6. The compound of claims 1-4, wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is -C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, and wherein R10 is optionally substituted with -OH, C1-C6 alkyl.
8. The compound of claims 1-4, wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
9. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, and wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; and wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is - C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, and wherein R10 is optionally substituted with -OH, C1-C6 alkyl.
10. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, and wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; and
double bond; wherein when L G is a double bond; R25 and R33 are H and R29 is halogen; and R26 is H and R27 is C1-C6 haloalkyl.
11. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, and wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; and wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
12. The compound of claim 6, wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is -C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl; and
double bond; wherein when L G is a double bond; R25 and R33 are H and R29 is halogen; R26 is H and R27 is C1-C6 haloalkyl.
13. The compound of claim 6, wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is -C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl; and wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
15. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is - C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl; and
wherein
, wherein when
double bond; wherein when L G is a double bond; R25 and R33 are H and R29 is halogen; R26 is H and R27 is C1-C6 haloalkyl.
16. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is - C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl; and wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
17. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; wherein
double bond; wherein when L G is a double bond; R25 and R33 are H and R29 is halogen; and R26 is H and R27 is C1-C6 haloalkyl. wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
18. The compound of claim 6, wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is -C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, wherein R10 is optionally substituted with -OH, C1-C6 alkyl;
wherein
, wherein when
double bond; wherein when L G is a double bond; R25 and R33 are H and R29 is halogen; and R26 is H and R27 is C1-C6 haloalkyl; and wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
19. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, and wherein the said 5- or 6-membered heterocyclyl ring is optionally substituted with C1-C3 alkyl; wherein n is 2; B, D, E, and X are C; Y is N; Ra and Rb are H; R2 is H; and R3 is - C(O)R10, wherein R10 is a 5-10 membered heteroaryl ring, and wherein R10 is optionally substituted with -OH, C1-C6 alkyl; wherein
, wherein when
double bond; wherein when L G is a double bond; R25 and R33 are H and R29 is halogen; and R26 is H and R27 is C1-C6 haloalkyl; and wherein K and J are N; R0 is H, R is =O, and R5 is C1-C6 alkyl; and R6 and R7 are H.
20. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen.
21. The compound of claim 5, wherein R1 is aryl fused with a 5- or 6-membered heterocyclyl ring, wherein the said 5- or 6-membered heterocyclyl ring comprises carbon atoms and at least one oxygen, and wherein the said 5- or 6-membered heterocyclyl ring is substituted with C1-C3 alkyl.
22. The compound of any of the preceding claims, wherein when R1 is an aryl fused with a 5- or 6-membered heterocyclyl ring, the 5- or 6-membered heterocyclyl ring is a 5- membered heterocyclyl ring.
23. The compound of any of the preceding claims, wherein when R10 is a 5-10 membered heteroaryl ring, the 5-10 membered heteroaryl ring is a 6-membered heteroaryl ring.
24. The compound of any of the preceding claims, wherein when R10 is optionally substituted with C1-C6 alkyl, the C1-C6 alkyl is C1 alkyl.
25. The compound of any of the preceding claims, wherein when R29 is halogen, the halogen is Cl.
26. The compound of any of the preceding claims, wherein when R27 is C1-C6 haloalkyl, the C1-C6 haloalkyl is trifluoromethyl.
27. The compound of any of the preceding claims, wherein when R5 is C1-C6 alkyl, the C1- C6 alkyl is C2 alkyl.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363535307P | 2023-08-29 | 2023-08-29 | |
| US63/535,307 | 2023-08-29 | ||
| US202463665217P | 2024-06-27 | 2024-06-27 | |
| US63/665,217 | 2024-06-27 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025049746A1 true WO2025049746A1 (en) | 2025-03-06 |
Family
ID=92801311
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/044435 Pending WO2025049746A1 (en) | 2023-08-29 | 2024-08-29 | Compositions comprising werner syndrome helicase inhibitors and methods of using the same |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025049746A1 (en) |
Cited By (10)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US12344609B2 (en) | 2023-06-08 | 2025-07-01 | Nimbus Wadjet, Inc. | WRN inhibitors |
| WO2025140518A1 (en) * | 2023-12-29 | 2025-07-03 | 勤浩医药(苏州)有限公司 | Fused ring compound, pharmaceutical composition containing same and use thereof |
| WO2025152932A1 (en) * | 2024-01-15 | 2025-07-24 | 上海齐鲁制药研究中心有限公司 | Triazolopyrimidine derivative, preparation method therefor and use thereof |
| WO2025162253A1 (en) * | 2024-02-02 | 2025-08-07 | 中国科学院上海药物研究所 | Spiro compound, pharmaceutical composition containing same and use thereof |
| WO2025162478A1 (en) * | 2024-02-04 | 2025-08-07 | InventisBio Co., Ltd. | Compounds, preparation methods and uses thereof |
| WO2025223396A1 (en) * | 2024-04-22 | 2025-10-30 | 3H Pharmaceuticals Co., Ltd. | Fused tricyclic compounds and methods of use thereof |
| WO2025232693A1 (en) * | 2024-05-08 | 2025-11-13 | Nuphase Therapeutics (Shanghai) Limited., Co. | Novel compounds, compositions comprising the same and uses thereof |
| WO2025237389A1 (en) * | 2024-05-17 | 2025-11-20 | 成都微芯药业有限公司 | Tricyclic wrn inhibitor, and preparation method therefor and use thereof |
| WO2025242186A1 (en) * | 2024-05-24 | 2025-11-27 | 浙江海正药业股份有限公司 | Aza-tricyclic derivative, and preparation method therefor and use thereof |
| WO2025247335A1 (en) * | 2024-05-30 | 2025-12-04 | Allorion Therapeutics Inc | Compounds and use thereof as wrn inhibitors |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022249060A1 (en) * | 2021-05-26 | 2022-12-01 | Novartis Ag | Triazolo-pyrimidine analogues for treating diseases connected to the inhibiton of werner syndrome recq helicase (wrn) |
-
2024
- 2024-08-29 WO PCT/US2024/044435 patent/WO2025049746A1/en active Pending
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2022249060A1 (en) * | 2021-05-26 | 2022-12-01 | Novartis Ag | Triazolo-pyrimidine analogues for treating diseases connected to the inhibiton of werner syndrome recq helicase (wrn) |
Cited By (11)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US12344609B2 (en) | 2023-06-08 | 2025-07-01 | Nimbus Wadjet, Inc. | WRN inhibitors |
| US12421233B2 (en) | 2023-06-08 | 2025-09-23 | Nimbus Wadjet, Inc. | WRN inhibitors |
| WO2025140518A1 (en) * | 2023-12-29 | 2025-07-03 | 勤浩医药(苏州)有限公司 | Fused ring compound, pharmaceutical composition containing same and use thereof |
| WO2025152932A1 (en) * | 2024-01-15 | 2025-07-24 | 上海齐鲁制药研究中心有限公司 | Triazolopyrimidine derivative, preparation method therefor and use thereof |
| WO2025162253A1 (en) * | 2024-02-02 | 2025-08-07 | 中国科学院上海药物研究所 | Spiro compound, pharmaceutical composition containing same and use thereof |
| WO2025162478A1 (en) * | 2024-02-04 | 2025-08-07 | InventisBio Co., Ltd. | Compounds, preparation methods and uses thereof |
| WO2025223396A1 (en) * | 2024-04-22 | 2025-10-30 | 3H Pharmaceuticals Co., Ltd. | Fused tricyclic compounds and methods of use thereof |
| WO2025232693A1 (en) * | 2024-05-08 | 2025-11-13 | Nuphase Therapeutics (Shanghai) Limited., Co. | Novel compounds, compositions comprising the same and uses thereof |
| WO2025237389A1 (en) * | 2024-05-17 | 2025-11-20 | 成都微芯药业有限公司 | Tricyclic wrn inhibitor, and preparation method therefor and use thereof |
| WO2025242186A1 (en) * | 2024-05-24 | 2025-11-27 | 浙江海正药业股份有限公司 | Aza-tricyclic derivative, and preparation method therefor and use thereof |
| WO2025247335A1 (en) * | 2024-05-30 | 2025-12-04 | Allorion Therapeutics Inc | Compounds and use thereof as wrn inhibitors |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2025049746A1 (en) | Compositions comprising werner syndrome helicase inhibitors and methods of using the same | |
| TWI887559B (en) | Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators | |
| AU2019240299B2 (en) | SHP2 phosphatase inhibitors and methods of use thereof | |
| ES2442501T3 (en) | Thienopyrimidine derivatives as PI3K inhibitors | |
| KR101787680B1 (en) | Kinase Inhibitor and Use Thereof | |
| KR101750667B1 (en) | Syk inhibitors | |
| KR101548439B1 (en) | Tricyclic pi3k inhibitor compounds and methods of use | |
| AU2018388406B2 (en) | Aryl-bipyridine amine derivatives as phosphatidylinositol phosphate kinase inhibitors | |
| JP2021503004A5 (en) | ||
| JP2024096702A (en) | Monoacylglycerol lipase regulators | |
| WO2016171755A1 (en) | Fused-bicyclic aryl quinolinone derivatives as mutant-isocitrate dehydrogenase inhibitors | |
| BR112021002408A2 (en) | carboxamides as ubiquitin-specific protease inhibitors | |
| WO2022167682A1 (en) | Azabicyclic shp2 inhibitors | |
| AU2023364551A1 (en) | Methods for treating cancer | |
| KR20230107291A (en) | PPARγ modulators and methods of use | |
| TWI864633B (en) | Tricyclic heterocyclic compounds as IAP antagonists | |
| JPWO2020065613A5 (en) | ||
| CN118488947A (en) | Condensed ring substituted six-membered heterocyclic compound and its preparation method and use | |
| CA3250387A1 (en) | Tetrahydropyrido[3,4-d]pyrimidines compounds as hpk1 inhibitors | |
| WO2023247590A1 (en) | Triazolopyridine and benzoisoxazole carboxamides and their uses as pdgfr inhibitors | |
| HK40031814A (en) | Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators | |
| HK40007020A (en) | Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators | |
| HK40003108B (en) | Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators | |
| HK40003108A (en) | Pyrrolopyrrole compositions as pyruvate kinase (pkr) activators |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24772841 Country of ref document: EP Kind code of ref document: A1 |